text
stringlengths
0
1.67M
1. A method for transmitting data between a first and second device by traversing firewalls, said method comprising the steps of: a. said first and second device establishing a communication link with a proxy server over a network, each of said first and second devices accessing said network over a firewall; b. said proxy server inspecting said firewalls and identifying an external mapped addresses BP associated with said first device and identifying an external mapped address CP associated with said second device; c. said proxy server notifying said first device regarding said identified external mapped address CP and said proxy server notifying said second device regarding said identified external mapped address BP, and d. said first or second device spoofing TCP packets via transmitting data with said notified external mapped address as the destination address. 2. A method for transmitting data between a first and second device by traversing firewalls, as per claim 1, wherein said step of spoofing TCP packets is done via Raw-IP datagrams. 3. A method for transmitting data between a first and second device by traversing firewalls, as per claim 1, wherein said communication link is established via TCP. 4. A method for transmitting data between a first and second device by traversing firewalls, as per claim 1, wherein said communication link is established via TCP/HTTP. 5. A method for transmitting data between a first and second device by traversing firewalls, as per claim 1, wherein said firewall is equipped with a network address translation device (NAT). 6. A method for transmitting data between a first and second device by traversing firewalls, as per claim 1, wherein said network is any of: local area network (LAN), wide area network (WAN), wireless network, or the Internet. 7. A method for transmitting data between a first and second device by traversing firewalls, as per claim 1, wherein said data is streaming multimedia data. 8. A method for forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, said method comprising the steps of: a. said first and second device establishing a communication link with a proxy server over a network, each of said first and second devices accessing said network over a firewall; b. said proxy server inspecting said firewalls and identifying an external mapped addresses BP associated with said first device and identifying an external mapped address CP associated with said second device; c. said proxy server notifying said packet forwarder regarding said identified external mapped addresses CP and BP, and d. said first device forwarding TCP packets via transmitting data with said packet forwarder as said destination address and said packet forwarder forwarding said data with CP as the destination address, or said second device forwarding TCP packets via transmitting data with said packet forwarder as said destination address and said packet forwarder forwarding said data with BP as the destination address. 9. A method for forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, as per claim 8, wherein said step of forwarding TCP packets is done via Raw-IP datagrams. 10. A method for forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, as per. Claim 8, wherein said communication link is established via TCP. 11. A method for forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, as per claim 8, wherein said communication link is established via TCP/HTTP. 12. A method for forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, as per claim 8, wherein said firewall is equipped with a network address translation device (NAT). 13. A method for forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, as per claim 8, wherein said network is any of: local area network (LAN), wide area network (WAN), wireless network, or the Internet. 14. A method for forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, as per claim 8, wherein said data is streaming multimedia data. 15. A system for transmitting data between a first and second device by traversing firewalls, said system comprising: a. a first host and an associated first firewall; b. a second host and an associated second firewall; c. a proxy server that establishes a communication link with said first and second host, identifies from said first and second firewalls external mapped addresses BP and CP respectively, and forwards said CP to said first device and forwards said BP to said second device, whereupon said first and second host utilize said forwarded external mapped addresses to spoof TCP packets and forward data directly between each other. 16. A system for transmitting data between a first and second device by traversing firewalls, as per claim 15, wherein said proxy server forwards TCP packets via Raw-IP datagrams. 17. A system for transmitting data between a first and second device by traversing firewalls, as per claim 15, wherein said communication link is established via TCP. 18. A system for transmitting data between a first and second device by traversing firewalls, as per claim 15, wherein said communication link is established via TCP/HTTP. 19. A system for transmitting data between a first and second device by traversing firewalls, as per claim 15, wherein said firewall is equipped with a network address translation device (NAT). 20. A system for transmitting data between a first and second device by traversing firewalls, as per claim 15, wherein said network is any of: local area network (LAN), wide area network (WAN), wireless network, or the Internet. 21. A system for transmitting data between a first and second device by traversing firewalls, as per claim 15, wherein said data is streaming multimedia data. 22. A system for transmitting data between a first and second device by traversing firewalls, said system comprising: a. a first host and an associated first firewall; b. a second host and an associated second firewall; c. a proxy server that establishes a communication link with said first and second host, identifies from said first and second firewalls external mapped addresses BP and CP respectively; d. a packet forwarder receiving BP and CP from said proxy server and using BP and CP to forward incoming communications from said first device to second device with CP as destination address, or forwarding incoming communications from said second device to first device with BP as the destination address. 23. A system for transmitting data between a first and second device by traversing firewalls, as per claim 22, wherein said packet forwarder forwards TCP packets via Raw-IP datagrams. 24. A system for transmitting data between a first and second device by traversing firewalls, as per claim 22, wherein said communication link is established via TCP. 25. A system for transmitting data between a first and second device by traversing firewalls, as per claim 22, wherein said communication link is established via TCP/HTTP. 26. A system for transmitting data between a first and second device by traversing firewalls, as per claim 22, wherein said firewall is equipped with a network address translation device (NAT). 27. A system for transmitting data between a first and second device by traversing firewalls, as per claim 22, wherein said network is any of: local area network (LAN), wide area network (WAN), wireless network, or the Internet. 28. A system for transmitting data between a first and second device by traversing firewalls, as per claim 22, wherein said data is streaming multimedia data. 29. An article of manufacture comprising a computer usable medium having computer readable program code embodied therein for assisting in the transmission of data between a first and second device by traversing firewalls, said article further comprising: a. computer readable program code aiding in establishing a communication link with a proxy server over a network, each of said first and second devices accessing said network over a firewall; b. computer readable program code inspecting said firewalls and identifying an external mapped addresses BP associated with said first device and identifying an external mapped address CP associated with said second device; c. computer readable program code notifying said first device regarding said identified external mapped address CP and computer readable program code notifying said second device regarding said identified external mapped address BP, and d. computer readable program code aiding said first or second device in spoofing TCP packets via transmitting data with said notified external mapped address as the destination address. 30. An article of manufacture comprising a computer usable medium having computer readable program code embodied therein for aiding in forwarding data between a first and second device by traversing firewalls, said data forwarded via a packet forwarder, said medium further comprising: a. computer readable program code aiding in establishing a communication link with a proxy server over a network, each of said first and second devices accessing said network over a firewall; b. computer readable program code inspecting said firewalls and identifying an external mapped addresses BP associated with said first device and identifying an external mapped address CP associated with said second device; c. computer readable program code notifying said packet forwarder regarding said identified external mapped addresses CP and BP, and d. computer readable program code forwarding TCP packets via transmitting data with said packet forwarder as said destination address and computer readable program code aiding said packet forwarder in forwarding said data with CP as the destination address, or computer readable program code forwarding TCP packets via transmitting data with said packet forwarder as said destination address and computer readable program code aiding said packet forwarder in forwarding said data with BP as the destination address.
<SOH> BACKGROUND OF THE INVENTION <EOH>1. Field of Invention The present invention relates generally to the field of network communications. More specifically, the present invention is related to a system and method for traversing firewalls and network address translators (NATs). 2. Discussion of Prior Art NATs and firewalls present a challenge to a network software programming, while their functions and operations are different: firewalls filter information into and out of the private network, while NATs hide or encapsulate a private network behind a single (or few) “real” Internet Protocol addresses. Their effect on many network applications is the same: The inability to send and receive information when receiving information using UDP (e.g., UDP data-grams coming into the private network). The inability to send and receive information when opening TCP communications into the private network. Each of the below described references teach the method of firewalls in general. However, none of the references provide or suggest the present invention method of ATM over IP traversing firewalls and network address translators (NATs). U.S. Pat. No. 5,898,830, assigned to Network Engineering Software describes a system, which allows connectionless traffic across a firewall. Rule checking is performed on the first packet entering, and if it is determined that the packet needs to be sent, a virtual host sends it to the destination computer. A time limit is set and so long as the set time limit does not run out, the communication is allowed. Addressing is accomplished utilizing name based addressing for end-to-end communication, with virtual hosts/DNS servers providing the intermediate address routing information. A connection type session does not appear to be initiated for the UDP transport. U.S. Pat. No. 5,915,087 discloses a firewall system, which allows communication, using a connectionless protocol. The firewall holds a list of servers located on the private side, and intercepts any communications addressed to the servers. The firewall then binds a port and notes it in a link table. The packet is passed to a stack, on the private side, which forwards the packet to the server. Any communications from the server to the originating client is sent to the firewall, where the originating clients address is determined using the link table. U.S. Pat. No. 5,778,174 describes a system, which utilizes an external machine, located on a public network to bypass a router firewall. A client on the public network connects to the external machine. A private channel is opened between the external machine and a machine internal to the private network. The internal machine connects to the destination server, and communication between the client and server is conducted through the external and internal machines. U.S. Pat. No. 5,941,988 provides for a proxy system that “glues” together two separate TCP connections terminating at a common host (proxy). When communications from one connection are received at the proxy, the headers are altered to address the socket at the end of the second connection, and the sequence numbers of the first connection are mapped to the sequence space of the second connection. The non-patent literature entitled, “A Weakness in the 4.2 BSD Unix TCP/IP Software” describes the spoofing of a trusted host to communicate with a system, having a list of the trusted hosts, from a host that is not on the trusted list. It should however be noted that the prior art described above fails to provide many features, for example an explicit recitation of opening a connection-oriented session in order to allow connectionless data-grams to pass through a NAT/firewall is not provided. Additionally, none of the prior art described above uses a proxy server to exchange respective address information between two hosts and the hosts communicating directly via the address information and spoofing the proxy, in order to traverse at least one firewall. Whatever the precise merits, features and advantages of the above cited references, none of them achieve or fulfills the purposes of the present invention.
<SOH> SUMMARY OF THE INVENTION <EOH>The present invention provides for a method and a system for allowing an incoming UDP packet to traverse a NAT/firewall comprising, opening a TCP connection and utilizing a Raw-IP interface which builds the UDP packet utilizing the parameters of the TCP connection (e.g., session number, port, etc.). Furthermore, the present system provides for a method and system for allowing communication between two machines, at least one of which is behind a firewall. Connections are established between each machine and a proxy server sitting on a public network. The proxy then communicates the port and address information of each machine to the other machine, after which, each machine sends directly to each other using the supplied port and address information, while using the proxy servers port and address information as the source port and address.
Novel nucleic acids and polypeptides
The present invention provides novel nucleic acids, novel polypeptide sequences encoded by these nucleic acids and uses thereof.
1. An isolated polynucleotide comprising a nucleotide sequence selected from the group consisting of SEQ ID NO: 1-30368, a mature protein coding portion of SEQ ID NO: 1-30368, an active domain of SEQ ID NO: 1-30368, and complementary sequences thereof. 2. An isolated polynucleotide encoding a polypeptide with biological activity, wherein said polynucleotide hybridizes to the polynucleotide of claim 1 under stringent hybridization conditions. 3. An isolated polynucleotide encoding a polypeptide with biological activity, wherein said polynucleotide has greater than about 90% sequence identity with the polynucleotide of claim 1. 4. The polynucleotide of claim 1 wherein said polynucleotide is DNA. 5. An isolated polynucleotide of claim 1 wherein said polynucleotide comprises the complementary sequences. 6. A vector comprising the polynucleotide of claim 1. 7. An expression vector comprising the polynucleotide of claim 1. 8. A host cell genetically engineered to comprise the polynucleotide of claim 1. 9. A host cell genetically engineered to comprise the polynucleotide of claim 1 operatively associated with a regulatory sequence that modulates expression of the polynucleotide in the host cell. 10. An isolated polypeptide, wherein the polypeptide is selected from the group consisting of: (a) a polypeptide encoded by any one of the polynucleotides of claim 1; and (b) a polypeptide encoded by a polynucleotide hybridizing under stringent conditions with any one of SEQ ID NO: 1-30368. 11. A composition comprising the polypeptide of claim 10 and a carrier. 12. An antibody directed against the polypeptide of claim 10. 13. A method for detecting the polynucleotide of claim 1 in a sample, comprising: a) contacting the sample with a compound that binds to and forms a complex with the polynucleotide of claim 1 for a period sufficient to form the complex; and b) detecting the complex, so that if a complex is detected, the polynucleotide of claim 1 is detected. 14. A method for detecting the polynucleotide of claim 1 in a sample, comprising: a) contacting the sample under stringent hybridization conditions with nucleic acid primers that anneal to the polynucleotide of claim 1 under such conditions; b) amplifying a product comprising at least a portion of the polynucleotide of claim 1; and c) detecting said product and thereby the polynucleotide of claim 1 in the sample. 15. The method of claim 14, wherein the polynucleotide is an RNA molecule and the method further comprises reverse transcribing an annealed RNA molecule into a cDNA polynucleotide. 16. A method for detecting the polypeptide of claim 10 in a sample, comprising: a) contacting the sample with a compound that binds to and forms a complex with the polypeptide under conditions and for a period sufficient to form the complex; and b) detecting formation of the complex, so that if a complex formation is detected, the polypeptide of claim 10 is detected. 17. A method for identifying a compound that binds to the polypeptide of claim 10, comprising: a) contacting the compound with the polypeptide of claim 10 under conditions sufficient to form a polypeptide/compound complex; and b) detecting the complex, so that if the polypeptide/compound complex is detected, a compound that binds to the polypeptide of claim 10 is identified. 18. A method for identifying a compound that binds to the polypeptide of claim 10, comprising: a) contacting the compound with the polypeptide of claim 10, in a cell, under conditions sufficient to form a polypeptide/compound complex, wherein the complex drives expression of a reporter gene sequence in the cell; and b) detecting the complex by detecting reporter gene sequence expression, so that if the polypeptide/compound complex is detected, a compound that binds to the polypeptide of claim 10 is identified. 19. A method of producing the polypeptide of claim 10, comprising, a) culturing a host cell comprising a polynucleotide sequence selected from the group consisting of a polynucleotide sequence of SEQ ID NO: 1-30368, a mature protein coding portion of SEQ ID NO: 1-30368, an active domain of SEQ ID NO: 1-30368, complementary sequences thereof and a polynucleotide sequence hybridizing under stringent conditions to SEQ ID NO: 1-30368, under conditions sufficient to express the polypeptide in said cell; and b) isolating the polypeptide from the cell culture or cells of step (a). 20. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 30369-60736, the mature protein portion thereof, or the active domain thereof. 21. The polypeptide of claim 20 wherein the polypeptide is provided on a polypeptide array. 22. A collection of polynucleotides, wherein the collection comprises the sequence information of at least one of SEQ ID NO: 1-30368. 23. The collection of claim 22, wherein the collection is provided on a nucleic acid array. 24. The collection of claim 23, wherein the array detects full-matches to any one of the polynucleotides in the collection. 25. The collection of claim 23, wherein the array detects mismatches to any one of the polynucleotides in the collection. 26. The collection of claim 22, wherein the collection is provided in a computer-readable format. 27. A method of treatment comprising administering to a mammalian subject in need thereof a therapeutic amount of a composition comprising a polypeptide of claim 10 or 20 and a pharmaceutically acceptable carrier. 28. A method of treatment comprising administering to a mammalian subject in need thereof a therapeutic amount of a composition comprising an antibody that specifically binds to a polypeptide of claim 10 or 20 and a pharmaceutically acceptable carrier.
<SOH> 2. BACKGROUND <EOH>Technology aimed at the discovery of protein factors (including e.g. cytokines, such as lymphokines, interferons, CSFs, chemokines, and interleukins) has matured rapidly over the past decade. The now routine hybridization cloning and expression cloning techniques clone novel polynucleotides “directly” in the sense that they rely on information directly related to the discovered protein (i.e., partial DNA/amino acid sequence of the protein in the case of hybridization cloning; activity of the protein in the case of expression cloning). More recent “indirect” cloning techniques such as signal sequence cloning, which isolates DNA sequences based on the presence of a now well-recognized secretory leader sequence motif, as well as various PCR-based or low stringency hybridization-based cloning techniques, have advanced the state of the art by making available large numbers of DNA/amino acid sequences for proteins that are known to have biological activity, for example, by virtue of their secreted nature in the case of leader sequence cloning, by virtue of their cell or tissue source in the case of PCR-based techniques, or by virtue of structural similarity to other genes of known biological activity. Identified polynucleotide and polypeptide sequences have numerous applications in, for example, diagnostics, forensics, gene mapping; identification of mutations responsible for genetic disorders or other traits, to assess biodiversity, and to produce many other types of data and products dependent on DNA and amino acid sequences.
<SOH> 3. SUMMARY OF THE INVENTION <EOH>The compositions of the present invention include novel isolated polypeptides, novel isolated polynucleotides encoding such polypeptides, including recombinant DNA molecules, cloned genes or degenerate variants thereof, especially naturally occurring variants such as allelic variants, antisense polynucleotide molecules, and antibodies that specifically recognize one or more epitopes present on such polypeptides, as well as hybridomas producing such antibodies. The compositions of the present invention additionally include vectors, including expression vectors, containing the polynucleotides of the invention, cells genetically engineered to contain such polynucleotides and cells genetically engineered to express such polynucleotides. The present invention relates to a collection or library of at least one novel nucleic acid sequence assembled from expressed sequence tags (ESTs) isolated mainly by sequencing by hybridization (SBH), and in some cases, sequences obtained from one or more public databases. The invention relates also to the proteins encoded by such polynucleotides, along with therapeutic, diagnostic and research utilities for these polynucleotides and proteins. These nucleic acid sequences are designated as SEQ ID NO: 1-30368. The polypeptides sequences are designated SEQ ID NO: 30369-60736. The nucleic acids and polypeptides are provided in the Sequence Listing. In the nucleic acids provided in the Sequence Listing, A is adenosine; C is cytosine; G is guanine; T is thymine; and N is any of the four bases. In the amino acids provided in the Sequence Listing, * corresponds to the stop codon. The nucleic acid sequences of the present invention also include, nucleic acid sequences that hybridize to the complement of SEQ ID NO: 1-30368 under stringent hybridization conditions; nucleic acid sequences which are allelic variants or species homologues of any of the nucleic acid sequences recited above, or nucleic acid sequences that encode a peptide comprising a specific domain or truncation of the peptides encoded by SEQ ID NO: 1-30368. A polynucleotide comprising a nucleotide sequence having at least 90% identity to an identifying sequence of SEQ ID NO: 1-30368 or a degenerate variant or fragment thereof. The identifying sequence can be 100 base pairs in length. The nucleic acid sequences of the present invention also include the sequence information from the nucleic acid sequences of SEQ ID NO: 1-30368. The sequence information can be a segment of any one of SEQ ID NO: 1-30368 that uniquely identifies or represents the sequence information of SEQ ID NO: 1-30368. A collection as used in this application can be a collection of only one polynucleotide. The collection of sequence information or identifying information of each sequence can be provided on a nucleic acid array. In one embodiment, segments of sequence information is provided on a nucleic acid array to detect the polynucleotide that contains the segment. The array can be designed to detect full-match or mismatch to the polynucleotide that contains the segment. The collection can also be provided in a computer-readable format. This invention also includes the reverse or direct complement of any of the nucleic acid sequences recited above; cloning or expression vectors containing the nucleic acid sequences; and host cells or organisms transformed with these expression vectors. Nucleic acid sequences (or their reverse or direct complements) according to the invention have numerous applications in a variety of techniques known to those skilled in the art of molecular biology, such as use as hybridization probes, use as primers for PCR, use in an array, use in computer-readable media, use in sequencing full-length genes, use for chromosome and gene mapping, use in the recombinant production of protein, and use in the generation of anti-sense DNA or RNA, their chemical analogs and the like. In a preferred embodiment, the nucleic acid sequences of SEQ ID NO: 1-30368 or novel segments or parts of the nucleic acids of the invention are used as primers in expression assays that are well known in the arL In a particularly preferred embodiment, the nucleic acid sequences of SEQ ID NO: 1-30368 or novel segments or parts of the nucleic acids provided herein are used in diagnostics for identifying expressed genes or, as well known in the art and exemplified by Vollrath et al., Science 258:52-59 (1992), as expressed sequence tags for physical mapping of the human genome. The isolated polynucleotides of the invention include, but are not limited to, a polynucleotide comprising any one of the nucleotide sequences set forth in SEQ ID NO: 1-30368; a polynucleotide comprising any of the full length protein coding sequences of SEQ ID NO: 1-30368; and a polynucleotide comprising any of the nucleotide sequences of the mature protein coding sequences of SEQ ID NO: 1-30368. The polynucleotides of the present invention also include, but are not limited to, a polynucleotide that hybridizes under stringent hybridization conditions to (a) the complement of any one of the nucleotide sequences set forth in SEQ ID NO: 1-30368; (b) a nucleotide sequence encoding any one of the amino acid sequences set forth in the Sequence Listing (e.g., SEQ ID NO: 30369-60736); (c) a polynucleotide which is an allelic variant of any polynucleotides recited above; (d) a polynucleotide which encodes a species homolog (e.g. orthologs) of any of the proteins recited above; or (e) a polynucleotide that encodes a polypeptide comprising a specific domain or truncation of any of the polypeptides comprising an amino acid sequence set forth in the Sequence Listing. The isolated polypeptides of the invention include, but are not limited to, a polypeptide comprising any of the amino acid sequences set forth in the Sequence Listing; or the corresponding full length or mature protein. Polypeptides of the invention also include polypeptides with biological activity that are encoded by (a) any of the polynucleotides having a nucleotide sequence set forth in SEQ ID NO: 1-30368; or (b) polynucleotides that hybridize to the complement of the polynucleotides of (a) under stringent hybridization conditions. Biologically or immunologically active variants of any of the polypeptide sequences in the Sequence Listing, and “substantial equivalents” thereof (e.g., with at least about 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% amino acid sequence identity) that preferably retain biological activity are also contemplated. The polypeptides of the invention may be wholly or partially chemically synthesized but are preferably produced by recombinant means using the genetically engineered cells (e.g. host cells) of the invention. The invention also provides compositions comprising a polypeptide of the invention. Polypeptide compositions of the invention may further comprise an acceptable carrier, such as a hydrophilic, e.g., pharmaceutically acceptable, carrier. The invention also provides host cells transformed or transfected with a polynucleotide of the invention. The invention also relates to methods for producing a polypeptide of the invention comprising growing a culture of the host cells of the invention in a suitable culture medium under conditions permitting expression of the desired polypeptide, and purifying the polypeptide from the culture or from the host cells. Preferred embodiments include those in which the protein produced by such process is a mature form of the protein. Polynucleotides according to the invention have numerous applications in a variety of techniques known to those skilled in the art of molecular biology. These techniques include use as hybridization probes, use as oligomers, or primers, for PCR, use for chromosome and gene mapping, use in the recombinant production of protein, and use in generation of anti-sense DNA or RNA, their chemical analogs and the like. For example, when the expression of an mRNA is largely restricted to a particular cell or tissue type, polynucleotides of the invention can be used as hybridization probes to detect the presence of the particular cell or tissue mRNA in a sample using, e.g., in situ hybridization. In other exemplary embodiments, the polynucleotides are used in diagnostics as expressed sequence tags for identifying expressed genes or, as well known in the art and exemplified by Vollrath et al., Science 258:52-59 (1992), as expressed sequence tags for physical mapping of the human genome. The polypeptides according to the invention can be used in a variety of conventional procedures and methods that are currently applied to other proteins. For example, a polypeptide of the invention can be used to generate an antibody that specifically binds the polypeptide. Such antibodies, particularly monoclonal antibodies, are useful for detecting or quantitating the polypeptide in tissue. The polypeptides of the invention can also be used as molecular weight markers, and as a food supplement. Methods are also provided for preventing, treating, or ameliorating a medical condition which comprises the step of administering to a mammalian subject a therapeutically effective amount of a composition comprising a polypeptide of the present invention and a pharmaceutically acceptable carrier. In particular, the polypeptides and polynucleotides of the invention can be utilized, for example, in methods for the prevention and/or treatment of disorders involving aberrant protein expression or biological activity. The present invention further relates to methods for detecting the presence of the polynucleotides or polypeptides of the invention in a sample. Such methods can, for example, be utilized as part of prognostic and diagnostic evaluation of disorders as recited herein and for the identification of subjects exhibiting a predisposition to such conditions. The invention provides a method for detecting the polynucleotides of the invention in a sample, comprising contacting the sample with a compound that binds to and forms a complex with the polynucleotide of interest for a period sufficient to form the complex and under conditions sufficient to form a complex and detecting the complex such that if a complex is detected, the polynucleotide of interest is detected. The invention also provides a method for detecting the polypeptides of the invention in a sample comprising contacting the sample with a compound that binds to and forms a complex with the polypeptide under conditions and for a period sufficient to form the complex and detecting the formation of the complex such that if a complex is formed, the polypeptide is detected. The invention also provides kits comprising polynucleotide probes and/or monoclonal antibodies, and optionally quantitative standards, for carrying out methods of the invention. Furthermore, the invention provides methods for evaluating the efficacy of drugs, and monitoring the progress of patients, involved in clinical trials for the treatment of disorders as recited above. The invention also provides methods for the identification of compounds that modulate (i.e., increase or decrease) the expression or activity of the polynucleotides and/or polypeptides of the invention. Such methods can be utilized, for example, for the identification of compounds that can ameliorate symptoms of disorders as recited herein. Such methods can include, but are not limited to, assays for identifying compounds and other substances that interact with (e.g., bind to) the polypeptides of the invention. The invention provides a method for identifying a compound that binds to the polypeptides of the invention comprising contacting the compound with a polypeptide of the invention in a cell for a time sufficient to form a polypeptide/compound complex, wherein the complex drives expression of a reporter gene sequence in the cell; and detecting the complex by detecting the reporter gene sequence expression such that if expression of the reporter gene is detected the compound that binds to a polypeptide of the invention is identified. The methods of the invention also provides methods for treatment which involve the administration of the polynucleotides or polypeptides of the invention to individuals exhibiting symptoms or tendencies. In addition, the invention encompasses methods for treating diseases or disorders as recited herein comprising administering compounds and other substances that modulate the overall activity of the target gene products. Compounds and other substances can effect such modulation either on the level of target gene/protein expression or target protein activity. The polypeptides of the present invention and the polynucleotides encoding them are also useful for the same functions known to one of skill in the art as the polypeptides and polynucleotides to which they have homology (set forth in the sequence listing). If no homology is set forth for a sequence, then the polypeptides and polynucleotides of the present invention are useful for a variety of applications, as described herein, including use in arrays for detection. detailed-description description="Detailed Description" end="lead"?
Novel means for the diagnosis and therapy of ctcl
A means for the diagnosis and therapy of T lymphomas, in particular Cutaneous T Cell Lymphomas (“CTCL”) are provided. Tumor markers which are universal for CTCL are described. More particularly, a molecule, termed SC5 by the inventors, an allelic form of p140, and biological applications of SC5 and p140 molecules, notably in the diagnosis and therapy of CTCL are described.
1 A monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M. 2 A hybridoma deposited as deposit number I-2575 at the C.N.C.M. 3 An isolated protein obtainable by: (i) collecting cells selected from the group consisting of total blood cells and peripheral blood lymphocytes (PBL), and stimulating them with PHA at 1 microgram/ml, (ii) lysing the cells by incubation in a lysis buffer containing Triton X-100 at 1%, (iii) recovering from the lysate the compound onto which the mAb of claim 1 binds under conditions enabling this mAb to perform reactions of the antigen-antibody type. 4 An isolated protein obtainable by recovering the isolated protein compound of claim 3 under non reducing conditions. 5 An isolated cDNA obtainable by: collecting cells selected from the group consisting of total blood cells and PBL5 incubating the collected cell population with a CD3 activator at 1 microgram per ml, extracting and purifying the whole mRNA population from said cells, synthesizing every complementary cDNA, operably cloning each cDNA so that expression of this cDNA in this clone is under appropriate clone culture conditions, and cultivating every clone accordingly, selecting those clones which express a compound onto which the monoclonal antibody of claim 1 binds when placed under conditions suitable for this monoclonal antibody to perform reactions of the antigen-antibody type, and/or of which lysate would give an immuno-precipitation reaction with this mAb, optionally, amplifying those clones that have been thus selected, and recovering the inserted cDNA from the selected clones. 6 An isolated mRNA obtainable by selection among the mRNA population of cells selected from the group consisting of total blood cells and PBL, of a mRNA which is complementary to a cDNA of claim 5. 7 An isolated genomic DNA encoding an isolated mRNA of claim 6. 8 An engineered cell in which a cDNA according to claim 5, has been transfected. 9 An isolated protein encoded by an isolated cDNA according to claim 5. 10 A bank of polypeptide compounds, obtainable by cleavage of a protein according to claim 3, with an enzyme selected from the group consisting of V8 protease and alpha chymotrypsine. 11 An isolated polypeptide compound obtainable by: collecting PBL cells or total blood cells, incubating the collected cells with a CD3 activator at 1 microgram par ml, labeling the cells with a polypeptide-specific label, lysing the cells in a lysis buffer comprising Trition X-100 at 1%, and submitting the lysate to an immuno-precipitation reaction with the mAb of claim 1, and recovering from the digested immuno-precipitate any compound onto which bear a label. 12 A monoclonal antibody obtainable by: (i) immunizing an animal against a protein according to claim 3, (ii) producing hybridomas from the spleen cells of this animal, and cultivating them to produce monoclonal antibodies in their culture supernatants, (iii) evaluating the supernatants for the presence of an antibody which is capable of binding to the protein or to the polypeptide compound that has been used as an immunogen in step (i), and which has at least one property selected from the group consisting of: binding resting non-tumoral T cells in their cytoplasmic compartment, and binding malignant CD4+ CTCL cells maizy at their cell surface, modulating the CD3 activation pathway of T cells, modulating IL-2 production from T cells, modulating the CD3-induced proliferation of T cells, modulating the CD3-induced in vitro proliferation of CTCL cells, modulating the proliferation of CTCL cells in a non-human animal, and competing with We a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M. for binding to a protein according to claim 3, (iv) selecting and cloning hybridomas producing the desired antibody, and (v) recovering the antibody from the supernatant above said clones. 13 A fragment of a monoclonal antibody according to claim 1, selected from the group consisting of heavy chains, light chains, VH, VL, Fab, F(ab′)2, CD1, CDR2, and CDR3. 14 An isolated compound, comprising a fragment according to claim 13. 15 The isolated compound according to claim 14, wherein it is a humanized antibody. 16 The isolated compound according to claim 14, wherein it further comprises at least one anti-CD4 fragment selected from the group consisting of heavy chains, light chains, VH, VL, Fab, F(ab′)2, CD1, CDR2, and CDR3 fragments of anti-CD4 antibodies. 17 An isolated protein of SEQ ID No. 2. 18 The isolated DNA encoding an isolated protein according to claim 17. 19 An isolated DNA of SEQ No. 1. 20 A polypeptidic vector comprising a monoclonal antibody according to claim 1, or a fragment thereof selected from the group consisting of heavy chains, light chains, VH, VL, Fab, F(ab′)2, CD1, CDR2 and CDR3. 21 A polypeptidic vector for use in the treatment of CTCL, wherein said vector comprises a p140 binding compound. 22 The polypeptidic vector according to claim 20, further comprising an element selected from the group consisting of tumoral toxins and radioelements. 23 The polypeptidic vector according to claim 20, further comprising an enzyme capable of transforming an anti-mitotic pro-drug into an active drug form. 24 A medical kit comprising a polypeptidic vector according to claim 23 and an antimitotic pro-drug. 25 A medicament comprising, as an active principle, an element selected from the group consisting of a monoclonal antibody according to claim 1, a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody according to claim 1. 26 A method for manufacturing an anti-CTCL medicament comprising including a p140 binding compound in said medicament. 27 A method for evaluating the percentage of malignant CD4+ CTCL cells preset within a body compartment of a patient, comprising: measuring the percentage of CD4+ cells expressing a protein according to claim 3 in a biological sample collected from said patient, wherein the percentage of malignant CD4+ CTCL cells present in said body compartment falls within a ±10% range around said measured percentage. 28 A method for CTCL diagnosis, comprising measuring the percentage of T cells expressing a protein according to claim 3, in a biological sample collected from said patient, and comparing said percentage measured to the average percentage observed in non-CTCL humans, a CTCL-positive diagnosis being made when the measured percentage is significantly higher than the average percentage. 29 A method for CTCL diagnosis, comprising measuring the percentage of CD4+ cells expressing an element selected from the group consisting of the proteins according to claim 3, in a biological sample collected from a patient, wherein a CD4+ CTCL positive diagnosis is made when the measured percentage is higher than the average percentage usually observed in non-CTCL humans. 30 A method for CTCL diagnosis, comprising determining the presence of CD4+ cells expressing p140 in a biological sample which comprises potential CTCL cells, a CTCL-positive diagnosis being made when such a presence is significantly detected. 31 A method according to claim 27, wherein said percentage measurement makes use of an element selected from the group consisting of a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M., a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody according to claim 1 and the DNA encoding SEQ ID No. 3 or SEQ ID No. 4. 32 A kit for assessing the development level of a CTCL, or for CTCL diagnosis, which comprises an element selected from the group consisting of the monoclonal antibodies according to claim 1, a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody according to claim 1. 33 A method for the identification of a compound which is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells, said method comprising detecting a compound that is capable of binding to a protein according to claim 3, or to a p140 molecule, wherein said compound which binds is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells. 34 The isolated protein of claim 4, wherein said non reducing conditions are of chromatography by affinity with a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M. 35 The isolated cDNA of claim 5, wherein said CD3 activator is PHA at 1 microgram per ml. 36 An engineered cell in which a mRNA according to claim 6 has been transfected. 37 An engineered cell in which a DNA according to claim 7 has been transfected. 38 An isolated protein encoded by an isolated mRNA according to claim 6. 39 An isolated protein encoded by an isolated cDNA according to claim 7. 40 A bank of polypeptide compounds, obtainable by cleavage of a protein according to claim 4, with an enzyme selected from the group consisting of V8 protease and alpha chymotrypsine. 41 A bank of polypeptide compounds, obtainable by cleavage of a protein according to claim 9, with an enzyme selected from the group consisting of V8 protease and alpha chymotrypsine. 42 A monoclonal antibody obtainable by: (i) immunizing an animal against a protein according to claim 4, (ii) producing hybridomas from the spleen cells of this animal, and cultivating them to produce monoclonal antibodies in their culture supernatants, (iii) evaluating the supernatants for the presence of an antibody which is capable of binding to the protein compound that has been used as an immunogen in step (i), and which has at least one property selected from the group consisting of: binding resting non-tumoral T cells in their cytoplasmic compartment, and binding malignant CD4+ CTCL cells at their cell surface, modulating the CD3 activation pathway of T cells, modulating IL-2 production from T cells, modulating the CD3-induced proliferation of T cells, modulating the CD3-induced in vitro proliferation of CTCL cells, modulating the proliferation of CTCL cells in a non-human animal, and competing with a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M. for binding to a protein according to claim 4, (iv) selecting and cloning hybridomas producing the desired antibody, and (v) recovering the antibody from the supernatant above said clones. 43 A monoclonal antibody obtainable by: (i) immunizing an animal against a protein according to claim 9, (ii) producing hybridomas from the spleen cells of this animal, and cultivating them to produce monoclonal antibodies in their culture supernatants, (iii) evaluating the supernatants for the presence of an antibody which is capable of binding to the protein compound that has been used as an immunogen in step (i), and which has at least one property selected from the group consisting of: binding resting non-tumoral T cells in their cytoplasmic compartment, and binding malignant CD4+ CTCL cells at their cell surface, modulating the CD3 activation pathway of T cells, modulating IL-2 production from T cells, modulating the CD3-induced proliferation of T cells, modulating the CD3-induced in vitro proliferation of CTCL cells, modulating the proliferation of CTCL cells in a non-human animal, and competing with a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M. for binding to a protein according to claim 9, (iv) selecting and cloning hybridomas producing the desired antibody, and (v) recovering the antibody from the supernatant above said clones. 44 A monoclonal antibody obtainable by: (i) immunizing an animal against a polypeptide compound according to claim 11, (ii) producing hybridomas from the spleen cells of this animal, and cultivating them to produce monoclonal antibodies in their culture supernatants, (iii) evaluating the supernatants for the presence of an antibody which is capable of binding to the polypeptide compound that has been used as an immunogen in step (i), and which has at least one property selected from the group consisting of: binding resting non-tumoral T cells in their cytoplasmic compartment, and binding malignant CD4+ CTCL cells at their cell surface, modulating the CD3 activation pathway of T cells, modulating IL-2 production from T cells, modulating the CD3-induced proliferation of T cells, modulating the CD3-induced in vitro proliferation of CTCL cells, modulating the proliferation of CTCL cells in a non-human animal, and competing with a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M. for binding to a polypeptide according to claim 11, (iv) selecting and cloning hybridomas producing the desired antibody, and (v) recovering the antibody from the supernatant above said clones. 45 A fragment of a monoclonal antibody according to claim 12, selected from the group consisting of heavy chains, light chains, VH, VL, Fab, F(ab′)2, CD1, CDR2, and CDR3. 46 The isolated compound according to claim 15, wherein it further comprises at least one anti-CD4 fragment selected from the group consisting of heavy chains, light chains, VH, VL, Fab, F(ab′)2, CD1, CDR2, and CDR3 fragments of anti-CD4 antibodies. 47 The polypeptidic vector according to claim 20, wherein said fragment is the Fab or F(ab′)2 fragment. 48 A polypeptidic vector comprising an isolated compound according to claim 15, optionally including at least one anti-CD4 fragment selected from the group consisting of heavy chains, light chains, VH, VL, Fab, F(ab′)2, CD1, CDR2, and CDR3 fragments of anti-CD4 antibodies. 49 The polypeptidic vector according to claim 21, further comprising an element selected from the group consisting of tumoral toxins and radioelements. 50 The polypeptidic vector according to claim 21, further comprising an enzyme capable of transforming an anti-mitotic pro-drug into an active drug form 51 The polypeptidic vector according to claim 23, wherein said enzyme capable of transforming an anti-mitotic pro-drug into an active drug form is a carboxypeptidase. 52 The polypeptidic vector according to claim 50, wherein said enzyme capable of transforming an anti-mitotic pro-drug into an active drug form is a carboxypeptidase. 53 The medical kit according to claim 24, wherein said antimitotic pro-drug is phenol mustard pro-drug. 54 A medicament comprising, as an active principle, an element selected from the group consisting of a monoclonal antibody according to claim 12, a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody. 55 A method for evaluating the percentage of malignant CD4+ CTCL cells preset within a body compartment of a patient, comprising: measuring the percentage of CD4+ cells expressing a protein according to claim 4 in a biological sample collected from said patient, wherein the percentage of malignant CD4+ CTCL cells present in said body compartment falls within a ±10% range around said measured percentage. 56 A method for evaluating the percentage of malignant CD4+ CTCL cells preset within a body compartment of a patient, comprising: measuring the percentage of CD4+ cells expressing a protein according to claim 9 in a biological sample collected from said patient, wherein the percentage of malignant CD4+ CTCL cells present in said body compartment falls within a ±10% range around said measured percentage. 57 A method for evaluating the percentage of malignant CD4+ CTCL cells preset within a body compartment of a patient, comprising: measuring the percentage of CD4+ cells expressing a member selected from the group consisting of the polypeptide compounds of claim 11 and the p140 molecules in a biological sample collected from said patient, wherein the percentage of malignant CD4+ CTCL cells present in said body compartment falls within a ±10% range around said measured percentage. 58 A method for CTCL diagnosis, comprising measuring the percentage of T cells expressing a protein according to claim 4, in a biological sample collected from said patient, and comparing said percentage measured to the average percentage observed in non-CTCL humans, a CTCL-positive diagnosis being made when the measured percentage is significantly higher than the average percentage. 59 A method for CTCL diagnosis, comprising measuring the percentage of T cells expressing a protein according to claim 9, in a biological sample collected from said patient, and comparing said percentage measured to the average percentage observed in non-CTCL humans, a CTCL-positive diagnosis being made when the measured percentage is significantly higher than the average percentage. 60 A method for CTCL diagnosis, comprising measuring the percentage of T cells expressing a member selected from the group consisting of the polypeptide compounds of claim 11 and the p140 molecules in a biological sample collected from said patient, and comparing said percentage measured to the average percentage observed in non-CTCL humans, a CTCL-positive diagnosis being made when the measured percentage is significantly higher than the average percentage. 61 A method for CTCL diagnosis comprising measuring the percentage of CD4+ cells expressing a protein according to claim 4 in a biological sample collected from a patient, wherein a CD4+ CTCL positive diagnosis is made when the measured percentage is higher than the average percentage usually observed in non-CTCL humans. 62 A method for CTCL diagnosis comprising measuring the percentage of CD4+ cells expressing a protein according to claim 9 in a biological sample collected from a patient, wherein a CD4+ CTCL positive diagnosis is made when the measured percentage is higher than the average percentage usually observed in non-CTCL humans. 63 A method for CTCL diagnosis comprising measuring the percentage of CD4+ cells expressing the polypeptide compounds of claim 11 in a biological sample collected from a patient, wherein a CD4+ CTCL positive diagnosis is made when the measured percentage is higher than the average percentage usually observed in non-CTCL humans. 64 A method according to claim 28, wherein said percentage measurement makes use of an element selected from the group consisting of a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M., a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody. 65 A method according to claim 29, wherein said percentage measurement makes use of an element selected from the group consisting of a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M., a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody. 66 A method according to claim 30, wherein said percentage measurement makes use of an element selected from the group consisting of a monoclonal antibody (mAb) produced by a hybridoma deposited as deposit number I-2575 at the C.N.C.M., a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody, and the DNA encoding SEQ ID No. 3 or SEQ ID No. 4. 67 A kit for assessing the development level of a CTCL, or for CTCL diagnosis, which comprises an element selected from the group consisting of the monoclonal antibodies according to claim 12, a fragment thereof selected from the group consisting of Fab and F(ab′)2 fragments of said antibody, and a humanized antibody comprising a Fab or F(ab′)2 fragment of said monoclonal antibody. 68 A kit for assessing the development level of a CTCL, or for CTCL diagnosis, which comprises at least one polypeptide compound selected from the bank of polypeptide compounds according to claim 10. 69 A kit for assessing the development level of a CTCL, or for CTCL diagnosis, which comprises an element selected from the group consisting the cDNA according to claim 5, an mRNA complementary to said cDNA, a p140 binding compound, a p140 DNA, an isolated DNA encoding an isolated protein of SEQ ID NO: 2, an isolated DNA of SEQ ID NO: 1, the DNA encoding SEQ ID No. 3 and the DNA encoding SEQ ID No. 4. 70 A method for the identification of a compound which is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells, said method comprising detecting a compound that is capable of binding to a protein according to claim 4, wherein said compound which binds is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells. 71 A method for the identification of a compound which is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells, said method comprising detecting a compound that is capable of binding to a protein according to claim 9, wherein said compound which binds is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells. 72 A method for the identification of a compound which is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells, said method comprising detecting a compound that is capable of binding to a polypeptide compound according to claim 10, wherein said compound which binds is useful in the palliation, prevention, relief and/or therapy of a proliferation of malignant CTCL cells. 73 The method according to claim 33, wherein said p140 molecule is of SEQ ID No. 2 or SEQ ID No. 4.
<SOH> BACKGROUND OF THE INVENTION <EOH>CTCL is a group of T lymphomas which primarily involve the skin. The CTCL group namely comprises transformed Mycosis Fungoides (abbreviated into transformed MF), Sézary Syndrome (abbreviated into SS), Lymphomatoide Papulosis (abbreviated into LP), and CD30+ lymphomas. Transformed MF is characterized by skin invasion of clonally-derived malignant T lymphocytes that phenotypically resemble mature T helper cells. LP and CD30+ lymphomas also develop in the skin. More aggressive forms of CTCL develop when the malignant cells become non-epidermotropic, and are associated with extra-cutaneous involvement. SS is a more aggressive form of CTCL that is characterized by a clonal expansion of CD4+/CD45RO+ T cells and the appearance of these malignant T cells in the blood. Most CTCL are CD4+ CTCL, but some rare CD8+ CTCL cases exist. The biology of CTCL disease remains poorly understood, as it is difficult to identify the malignant cells, due to the lack of specific cell surface markers. In cutaneous lesions, it is therefore difficult to distinguish CTCL cells from reactive infiltrating (non-tumoral) T lymphocytes. Diagnosis of T lymphomas such as CTCL is at present time mainly based on cytological and histological observations of the presence or absence of tumoral cells in a sample collected from a suspected body area (observation of histopathological aspect on skin biopsis and/or presence of SS cells in the blood, via detection of cells showing a cerebriform nucleus). Such a diagnosis method is not fully reliable, notably at the early stages of the transformation of skin lymphocytes into malignant lymphocytes. Such a diagnosis method does also not enable to stage the disease. Technically speaking, it is also time-consuming. Today therapy of CTCL is tentatively achieved by induction of tumoral cell apoptosis via non specific chemotherapy. There is therefore still a need for more accurate and appropriate solutions to the problem of the diagnosis and therapy of diseases involving the proliferation of malignant T cells, such as CTCL.
<SOH> SUMMARY OF THE INVENTION <EOH>In view of this prior art situation, the inventors now provide with two molecules which give solutions to the problem of CTCL diagnostic and therapy: one has been termed SC5 by the inventors, and the other one is the p140 molecule (p140 is also referred to as KIR3DL2). The SC5 molecule of the invention appears as a biochemically and functionally new protein, of which apparent molecular weight is of 96 kD under reducing conditions. It has been isolated as the antigen of a monoclonal antibody that has been produced by the inventors. The hybridoma producing this monoclonal antibody has been deposited on Oct. 30, 2000 at the C.N.C.M. (C.N.C.M. Institut Pasteur; 25, rue du Docteur Roux, F-75724 Paris Cedex 15, France) in accordance with the Budapest Treaty (C.N.C.M. deposit number: I-2575). When expressed at the cell surface, and made to aggregate, SC5 molecules act as inhibitory receptors for cell activity and proliferation. The second marker of the invention, i.e. p140, was already known to be an inhibitory receptor, but has been previously described only on sub-groups of NK cells and of CD3+ CD8+ cells from healthy humans. The inventors now demonstrate that p140 is expressed at the surface of tumoral T cells such as CTCL cells. They further demonstrate that p140 is expressed at the surface of malignant CD4+ T cells, such as CD4+ CTCL cells, whereas those other receptors that are usually observed at the surface of NK cells (such as p58.1, p58.2, p70 KIRs, CD94/NKG2A) are not found at the surface of malignant CD4+ T cells. Of further note is that p140 has not been observed by the inventors at the surface of CD4+ T cells collected from patients suffering from non-tumoral dermatological diseases such as inflammatory skin diseases (e.g. lupus, lichen), or toxic epidermal necrolysis. At the surface of tumoral CD4+ T cells, two allelic forms of p140 have been identified by the inventors: allelic form KIR3D clone 24 (SEQ ID No.1), and allelic form p140 clone 1.1 (SEQ ID No.3). Allelic form KIR3D clone 24 (SEQ ID No. 1) is a new polynucleotide encoding a new protein (SEQ ID No.2): KIR3D clone 24 (SEQ ID No. 1) displays five differences when compared to the previously described p140 clone 1.1 DNA sequence (SEQ ID No. 3), resulting in four amino acid substitutions in the mature protein (SEQ ID No. 2) when compared to p140 clone 1.1. mature protein (SEQ ID No. 4). The inventors demonstrate that the SC5 and p140 both share the same technical following features: SC5 and p140 both are membranar differentiation antigens which are characteristic of malignant T cells, and notably of malignant CD4+ T cells, whichever form of CD4+ CTCL is concerned, there are malignant CD4+ T cells which express SC5 or p140 at their surface (usually both SC5 and p140 are expressed): SC5 as well as p140 indeed cover the whole range of CD4+ CTCL, and notably the Sézary Syndrome (abbreviated into “SS”), transformed Mycosis Fungoides (abbreviated into “transformed MF”), Lymphomatoide Papulosis (abbreviated onto “LP”), and CD30+ lymphomas, there exists such a link between the presence of SC5 or p140 at the surface of CD4+ T cells and the existence of a CD4+ CTCL, that a CD4+ CTCL diagnosis based on the analysis of the presence of SC5 or p140 at the surface of CD4+ cells collected from the suspected body area (e.g. sample of skin erythroderma when transformed MF is suspected, or sample of peripheral blood when a more aggressive CTCL form, such as SS, is suspected) has a reliability of more than 90%, preferably of more than of 95%, most preferably of 100% (among those patients who were tested up to now, the reliability is of 100%): indeed, according to the invention, it can be concluded that a CD4+ T cell is tumoral as soon as there are p140 molecules detected at the surface of these CD4+ T cells, or as soon as a percentage of SC5+ CD4+ T cells higher than the average standard level is measured (the average standard level is in the 1-15% range, generally in the 5-10% range), and there exists such a link between the presence of SC5 or p140 at the surface of CD4+ T cells and the existence of a CD4+ CTCL, that the percentage of CD4+ SC5+ T cells, as well as of CD4+ p140+ T cells, that is measured in a sample of peripheral blood collected from a patient for whom a Sézary Syndrome (SS) is suspected, substantially corresponds to the percentage of malignant SS cells that are actually present in the peripheral blood of this patient (within a ±10% range for SC5+ CD4+ cells, within a ±5% range for p140+ CD4+ cells): the SC5 and p140 markers of the invention therefore share the common particular technical feature of enabling to assess the staging of a SS. To the best of the inventors' knowledge, SC5 and p140 are the first molecules which are described as having these common technical features. The invention thus provides with the first CTCL universal markers. No prior art product was known to be such a CTCL universal marker. The closest prior art product in this respect would be CD30 of which presence at the surface of malignant CD4+ T cells directs to the conclusion that the patient has a particular form of CD4+ CTCL which is referred to in the art as CD30+ lymphoma. CD30 is therefore a CTCL marker, but its reliability is limited to a particular form of CTCL (CD30+ lymphomas), and CD30 does not cover every form of CD4+ CTCL: for CD4+ CTCL such as SS, transformed MF, or LP, there does not necessarily exist a malignant CD4+ T cell which would express CD30 at its surface. CD30 uses in CTCL diagnosis and therapy is thus restricted to a particular form of CTCL, whereas the markers of the invention advantageously cover the whole range of CD4+ CTCL. In addition to the technical features shared with p140, SC5 has the further characteristic of being a positive indicator of proliferation and/or functional activity (e.g. cytokine profile) of non-tumoral T cells. The presence of SC5 at the surface of T cells is indeed positively linked with the activation status of non-tumoral T cells, such as normal (CD4+ and CD8+) T cells or virus-infected CD4+ T cells. SC5 is thus also a useful novel means for the diagnostic and therapy of T cell viral infections such as HIV-infection, and of inflammatory diseases such as those of the auto-immune type (e.g. rhumatoid arthritis such as spondyloarthropathis, or skin immune mediated diseases such as psoriasis, eczema, atopic dermatitis). SC5 is also a useful target for modulating graft-host reactions: the activation of SC5 transduces an inhibitory signal that can be used to inhibit the reactions that effector recipient cells may develop against a graft. The invention also provides with products capable of binding to the SC5 new molecule, and in particular with monoclonal antibodies directed against SC5 (anti-SC5 mAbs). It is also provided with anti-SC5 monoclonal antibodies which are capable of modulating the proliferation and/or the functional activity of T cells, and notably of tumoral T cells such as malignant CTCL cells. In particular, the monoclonal antibody produced by hybridoma I-2575 is capable of inhibiting the proliferation and/or functional activity of T cells, and according to a particularly useful feature, is capable of inhibiting the proliferation and/or activity of malignant T cells such as malignant CTCL cells (see examples below). Compounds binding to SC5, but not aggregating them will prevent SC5 inhibitory signal transduction, or will prevent compounds to reach SC5+ cells (target masking). More generally, compounds binding to SC5 or p140 may, according to the invention, be used as complement recruiting agents, as ADCC stimulators, or as vectors for therapeutic agents so as to prevent, palliate, treat T lymphomas, such as CTCL. The invention therefore also relates to the diagnostic and therapeutic uses of SC5- or p140-binding compounds such as anti-SC5 and/or anti-p140 monoclonal antibodies, and of products directly-derived therefrom (e.g. humanized monoclonal antibodies, or monoclonal antibodies with a double CD4-p140/SC5 specificity).
Methods for the production of multimeric proteins and related compositions
Improved methods for the production of multimeric-protein-complexes, such as redox proteins and immunoglobins, in association with oil bodies are described. The redox protein is enzymatically active when prepared in association with the oil bodies. Also provided are related nucleic acids, proteins, cells, plants, and compositions.
1-266. (canceled) 267. A method of producing an oil body associated with a recombinant multimeric-protein-complex, said method comprising: (a) producing in a cell comprising oil bodies, a first recombinant polypeptide and a second recombinant polypeptide wherein said first recombinant polypeptide is capable of associating with said second recombinant polypeptide to form said multimeric-protein-complex; and (b) associating said multimeric-protein-complex with an oil body through an oil-body-targeting-protein capable of associating with said oil body and said first recombinant polypeptide. 268. A method of expressing a recombinant multimeric-protein-complex comprising a first and second recombinant polypeptide in a cell, said method comprising: (a) introducing into a cell a first chimeric nucleic acid sequence comprising: (i) a first nucleic acid sequence capable of regulating transcription in said cell operatively linked to; (ii) a second nucleic acid sequence encoding a first recombinant polypeptide; (b) introducing into said cell a second chimeric nucleic acid sequence comprising: (i) a third nucleic acid sequence capable of regulating transcription in said cell operatively linked to; (ii) a fourth nucleic acid sequence encoding a second recombinant polypeptide; (c) growing said cell under conditions to permit expression of said first and second recombinant polypeptide in a progeny cell comprising oil bodies wherein said first recombinant polypeptide and said second recombinant polypeptide are capable of forming a multimeric-protein-complex; and (d) associating said first recombinant polypeptide with an oil body through an oil-body-targeting-protein capable of associating with said oil body and said first recombinant polypeptide. 269. A method of producing in a plant a recombinant multimeric-protein-complex, said method comprising: (a) preparing a first plant comprising cells, said cells comprising oil bodies and a first recombinant polypeptide wherein said first recombinant polypeptide is capable of associating with said oil bodies through an oil-body-targeting-protein; (b) preparing a second plant comprising cells, said cells comprising oil bodies and a second recombinant polypeptide; and (c) sexually crossing said first plant with said second plant to produce a progeny plant comprising cells, said cells comprising oil bodies, wherein said oil bodies are capable of associating with said first recombinant polypeptide, and said first recombinant polypeptide is capable of associating with said second recombinant polypeptide to form said recombinant multimeric-protein-complex. 270. A chimeric nucleic acid sequence encoding a multimeric-fusion-protein, said nucleic acid comprising: (a) a first nucleic acid sequence encoding an oil-body-targeting-protein operatively linked in reading frame to; (b) a second nucleic acid sequence encoding a first recombinant polypeptide; linked in reading frame to; (c) a third nucleic acid sequence encoding a second recombinant polypeptide, wherein said first and second recombinant polypeptide are capable of forming a multimeric-protein-complex. 271. A recombinant multimeric-fusion-protein comprising (i) an oil-body-targeting-protein, or fragment thereof, (ii) a first recombinant polypeptide and a (iii) second recombinant polypeptide, wherein said first and second recombinant polypeptides are capable of forming a multimeric-protein-complex. 272. Isolated oil bodies comprising a multimeric-protein-complex comprising (i) an oil-body-targeting-protein and (ii) a first recombinant polypeptide, said oil bodies further comprising a second recombinant polypeptide, wherein said first and second recombinant polypeptide are capable of forming a multimeric-protein-complex. 273. Isolated oil bodies comprising (a) a first fusion protein comprising a first oil-body-targeting-protein fused to a first recombinant polypeptide; and (b) a second fusion protein comprising a second oil-body-targeting-protein fused to a second recombinant polypeptide, wherein said first and second recombinant polypeptide are capable of forming a multimeric-protein-complex. 274. A cell comprising oil bodies and (i) an oil-body-targeting-protein, (ii) a first recombinant polypeptide and (iii) a second recombinant polypeptide wherein (1) said first recombinant polypeptide is capable of associating with said oil-body-targeting-protein; and (2) said first recombinant polypeptide is capable of associating with said second recombinant polypeptide to form a multimeric-protein-complex. 275. A composition comprising isolated oil bodies, thioredoxin and thioredoxin-reductase. 276. A food product, personal care product or pharmaceutical composition comprising the composition of claim 275. 277. A method of reducing allergenicity of a food comprising the steps of: providing the isolated oil bodies of claim 273; and adding the isolated oil bodies to the food, whereby allergenicity of the food is reduced. 278. A method of treating or protecting a target against oxidative stress, comprising the steps of: providing the recombinant fusion polypeptide of claim 269; and contacting the recombinant fusion polypeptide with a target, wherein the target is susceptible to oxidative stress, thereby treating or protecting against the stress. 279. A method for preparing an enzymatically active redox protein associated with oil bodies comprising: a) producing in a cell a redox fusion polypeptide comprising a first redox protein linked to a second redox protein; b) associating said redox fusion polypeptide with oil bodies through an oil-body-targeting-protein capable of associating with said redox fusion polypeptide and said oil bodies; and c) isolating said oil bodies associated with said redox fusion polypeptide. 280. A method for preparing a redox protein associated with oil bodies comprising: a) introducing into a cell a chimeric nucleic acid sequence comprising: 1) a first nucleic acid sequence capable of regulating transcription in said cell operatively linked to; 2) a second nucleic acid sequence encoding a recombinant fusion polypeptide comprising (i) a nucleic acid sequence encoding a sufficient portion of an oil-body-protein to provide targeting of said recombinant fusion polypeptide to an oil body linked to (ii) a nucleic acid sequence encoding a redox fusion polypeptide comprising a first redox protein linked to a second redox protein operatively linked to; 3) a third nucleic acid sequence capable of terminating transcription in said cell; b) growing said cell under conditions to permit expression of said redox fusion polypeptide in a progeny cell comprising oil bodies; and c) isolating from said progeny cell said oil bodies comprising said redox fusion polypeptide. 281. A chimeric nucleic acid comprising: 1) a first nucleic acid sequence capable of regulating transcription in a host cell operatively linked to; 2) a second nucleic acid sequence encoding a recombinant fusion polypeptide comprising (i) a nucleic acid sequence encoding a sufficient portion of an oil-body-protein to provide targeting of said recombinant fusion polypeptide to an oil body linked to (ii) a nucleic acid sequence encoding a redox fusion polypeptide comprising a first redox protein linked to a second redox protein operatively linked to; 3) a third nucleic acid sequence capable of terminating transcription in said cell. 282. A transgenic plant comprising the chimeric nucleic acid sequence of claim 281. 283. A safflower plant comprising the chimeric nucleic acid of anyone of claim 281. 284. A plant seed comprising the chimeric nucleic acid of claim 281. 285. A safflower seed comprising the chimeric nucleic acid of claim 281. 286. An oil body preparation obtained by the method of claim 279. 287. A food product comprising an oil body preparation of claim 286. 288. A composition comprising an oil body preparation of claim 286 (New). 289. A personal care product comprising an oil body preparation of claim 286. 290. A product capable of treating oxidative stress in a target comprising an oil body preparation of claim 286. 291. A product capable of chemically reducing a target comprising an oil body preparation of claim 286. 292. A detergent composition comprising the product of claim 286. 293. A method of cleansing an item, comprising administering the product of claim 290 to said item under conditions that promote cleansing. 294. An emulsion formulation prepared by the method of claim 279. 295. A nucleic acid construct comprising a gene fusion, wherein the gene fusion comprises a first region encoding an oil-body-protein or an active fragment thereof, operably linked to a second region encoding at least one thioredoxin-related protein or an active fragment thereof. 296. The seed of the plant of claim 281. 297. An extract of the seed of claim 296, wherein the extract comprises an activity of a thioredoxin-related protein. 298. An oil body from the seed of claim 296. 299. Oil produced from the seed of claim 296. 300. A method of making a fusion protein comprising a thioredoxin-related activity, the method comprising the steps of: providing a transgenic plant comprising a nucleic acid construct comprising a seed-specific promoter operably linked to a gene fusion, wherein the gene fusion comprises a region encoding an oil-body-protein or an active fragment thereof, operably linked to a region encoding a first thioredoxin-related protein or an active fragment thereof, the gene fusion encoding a fusion protein comprising a thioredoxin-related activity; obtaining seeds from the plant; and recovering the fusion protein by isolating oil bodies from the seeds. 301. Oil bodies in association with a fusion protein, obtained by the method of claim 300. 302. A method of reducing allergenicity of a food comprising the steps of: providing a preparation comprising oil bodies associated with a fusion protein, the fusion protein comprising an oil-body protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof; and adding the preparation to the food, whereby allergenicity of the food is reduced due to activity of the thioredoxin-related protein or fragment. 303. A composition comprising a fusion protein, the fusion protein comprising an oil-body-protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof, in a pharmaceutically acceptable carrier. 304. A cosmetic formulation comprising oil bodies associated with a fusion protein, the fusion protein comprising an oil-body-protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof, in an acceptable carrier. 305. A method of treating or protecting a target against oxidative stress, comprising the steps of: providing a preparation comprising a fusion protein, the fusion protein comprising an oil-body-protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof; and contacting the preparation with a target, wherein the target is susceptible to oxidative stress, thereby treating or protecting against the stress. 306. A nucleic acid construct comprising a gene fusion, wherein the gene fusion comprises a first region encoding an oil-body-protein or an active fragment thereof, operably linked to a second region encoding at least one recombinant polypeptide and an oil-body-surface-avoiding linker in frame between the first and second region polypeptides.
<SOH> BACKGROUND <EOH>Multimeric proteins (i.e. proteins comprising multiple polypeptide chains) are a biologically and commercially important class of proteins. Antibodies for example are multimeric proteins which are used to treat a wide range of disease conditions. However in view of their complexity, multimeric proteins frequently represent significant manufacturing challenges. Redox proteins are also a commercially important class of proteins with applications in a variety of different industries including the pharmaceutical, personal care and food industry. For example, the redox protein thioredoxin may be used in the manufacture of personal care products (Japanese Patent Applications JP9012471A2, JP103743A2, JP1129785A2), pharmaceutical compositions/products (Aota et al. (1996) J. Cardiov. Pharmacol. (1996) 27: 727-732) as well as to reduce protein allergens present in food products such as milk (del Val et al. (1999) J. Allerg. Vlin. Immunol. 103: 690-697) and wheat (Buchanan et al. (1997) Proc. Natl. Acad. Sci. USA 94: 5372-5377). However, there is a need in the art to further improve the methods for the recombinant expression of multimeric proteins, including redox proteins. The present invention satisfies this need and provides related advantages as well.
<SOH> SUMMARY OF THE INVENTION <EOH>The present invention relates to novel and improved methods of producing a first and/or second recombinant polypeptides, multimeric-protein-complexes, heteromultimeric-protein-complexes, multimeric-fusion-proteins, heteromultimeric-fusion-proteins, immunoglobulin-polypeptide-chains, immunoglobulins, redox-fusion-polypeptides, and/or thioredoxin-related proteins; in association with oil bodies. Accordingly, provided herein are methods of producing a recombinant multimeric-protein-complex, said method comprising: (a) producing in a cell comprising oil bodies, a first recombinant polypeptide and a second recombinant polypeptide wherein said first recombinant polypeptide is capable of associating with said second recombinant polypeptide to form said multimeric-protein-complex; and (b) associating said multimeric-protein-complex with an oil body through an oil-body-targeting-protein capable of associating with said oil bodies and said first recombinant polypeptide. The method further contemplates isolating the oil bodies associated with said recombinant multimeric-protein-complex. The second recombinant polypeptide can be associated with a second oil-body-targeting-protein capable of associating with an oil body and said second recombinant polypeptide. Each of said oil-body-targeting-proteins can be an oil-body-protein or an immunoglobulin. The oil-body-targeting-protein can be an oleosin or caleosin. When the oil-body-targeting-protein can be an oleosin or caleosin, the first recombinant polypeptide can be fused to said oleosin or caleosin. Likewise, the second recombinant polypeptide can be fused to a second oleosin or second caleosin capable of associating with an oil body. The first and second recombinant polypeptides can be produced as a multimereic-fusion-protein comprising said first and second polypeptide, and can form a multimeric-protein-complex. The multimeric-protein-complex can be a heteromultimeric-protein-complex, and the heteromultimeric-protein-complex can be an enzymatically active redox complex or an immunoglobulin. In one embodiment, the first recombinant polypeptide is capable of associating with said second recombinant polypeptide in the cell. In another embodiment, the first recombinant polypeptide can be a thioredoxin and the second recombinant polypeptide can be a thioredoxin-reductase. In particular embodiments, the thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194; and the thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 50 and SEQ ID NOs:195-313. In another embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. The cell can be a plant cell, such as a safflower cell, and the like. Also provided herein is a method of expressing a recombinant multimeric-protein-complex comprising a first and second recombinant polypeptide in a cell, said method comprising: (a) introducing into a cell a first chimeric nucleic acid sequence comprising: (i) a first nucleic acid sequence capable of regulating transcription in said cell operatively linked to; (ii) a second nucleic acid sequence encoding a first recombinant polypeptide; (b) introducing into said cell a second chimeric nucleic acid sequence comprising: (i) a third nucleic acid sequence capable of regulating transcription in said cell operatively linked to; (ii) a fourth nucleic acid sequence encoding a second recombinant polypeptide; (c) growing said cell under conditions to permit expression of said first and second recombinant polypeptide in a progeny cell comprising oil bodies wherein said first recombinant polypeptide and said second recombinant polypeptide are capable of forming a multimeric-protein-complex; and (d) associating said first recombinant polypeptide with an oil body through an oil-body-targeting-protein capable of associating with said oil bodies and said first recombinant polypeptide. This method further contemplates isolating from the progeny cell, oil bodies comprising the multimeric-protein-complex. The second recombinant polypeptide can be associated with a second oil-body-targeting-protein capable of associating with an oil body and second recombinant polypeptide. Each of said oil-body-targeting-proteins can be an oil-body-protein or an immunoglobulin. The oil-body-targeting-protein can be an oleosin or caleosin. When the oil-body-targeting-protein is an oleosin or caleosin, the first recombinant polypeptide can be fused to said oleosin or caleosin. Likewise, the second recombinant polypeptide can be fused to a second oleosin or second caleosin capable of associating with an oil body. The first and second recombinant polypeptides can be produced as a multimereic-fusion-protein comprising said first and second polypeptide, and can form a multimeric-protein-complex. The multimeric-protein-complex can be a heteromultimeric-protein-complex, and the heteromultimeric-protein-complex can be an enzymatically active redox complex or an immunoglobulin. In one embodiment, the first recombinant polypeptide and said second recombinant polypeptide are capable of forming a multimeric-protein-complex in said progeny cell. In another embodiment, the first recombinant polypeptide can be a thioredoxin and the second recombinant polypeptide can be a thioredoxin-reductase. In particular embodiments, the thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194; and the thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 60 and SEQ ID NOs:195-313. In another embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. The cell can be a plant cell, such as a safflower cell, and the like. Also provided herein are methods of producing in a plant a recombinant multimeric-protein-complex, said method comprising: (a) preparing a first plant comprising cells, said cells comprising oil bodies and a first recombinant polypeptide wherein said first recombinant polypeptide is capable of associating with said oil bodies through an oil-body-targeting-protein; (b) preparing a second plant comprising cells, said cells comprising oil bodies and a second recombinant polypeptide; and (c) sexually crossing said first plant with said second plant to produce a progeny plant comprising cells, said cells comprising oil bodies, wherein said oil bodies are capable of associating with said first recombinant polypeptide, and said first recombinant recombinant polypeptide is capable of associating with said second recombinant polypeptide to form said recombinant multimeric-protein-complex. The second recombinant polypeptide can be associated with oil bodies through a second oil-body-targeting-protein in the second plant. The oil bodies can be isolated from the progeny plant comprising said multimeric-protein-complex. The oil-body-targeting-protein can be selected from an oil-body-protein or an immunoglobulin, wherein the oil-body-protein can be an oleosin or caleosin. The first recombinant polypeptide can be fused to the oleosin or caleosin; and the second recombinant polypeptide can be fused to a second oleosin or second caleosin capable of associating with an oil body. The first and second recombinant polypeptide can form a multimeric-protein-complex, such as a heteromultimeric-protein-complex, wherein the heteromultimeric-protein-complex can be an enzymatically active redox complex or an immunoglobulin. In a particular embodiment, the first recombinant polypeptide is a thioredoxin and the second recombinant polypeptide is a thioredoxin-reductase. The thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194; and the thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 50 and SEQ ID NOs:195-313. In another embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. The plant can be a safflower plant. Also provided herein are chimeric nucleic acids encoding a multimeric-fusion-protein as described herein, said nucleic acid comprising: (a) a first nucleic acid sequence encoding an oil-body-targeting-protein operatively linked in reading frame to; (b) a second nucleic acid sequence encoding a first recombinant polypeptide; linked in reading frame to; (c) a third nucleic acid sequence encoding a second recombinant polypeptide, wherein said first and second recombinant polypeptide are capable of forming a multimeric-protein-complex. The oil-body-targeting-protein can be selected from an oil-body-protein or an immunoglobulin. The oil-body-protein can be an oleosin or caleosin. The multimeric-protein-complex can be a heteromultimeric-protein-complex, and the first and second recombinant polypeptide can form an enzymatically active heteromultimeric redox complex or an immunoglobulin. In a particular embodiment, the first recombinant polypeptide is a thioredoxin and the second recombinant polypeptide is a thioredoxin-reductase. The thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194; and the thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 50 and SEQ ID NOs:195-313. In another embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. In yet another embodiment, positioned between the nucleic acid sequence encoding an oil-body-targeting-protein and the nucleic acid sequence encoding a first recombinant polypeptide can be a linker nucleic acid sequence encoding an oil-body-surface-avoiding linker amino acid sequence. The oil-body-surface-avoiding linker amino acid sequence can be substantially negatively charged, or have a molecular weight of at least 35 kd. Optionally, the gene fusion further comprises a linker nucleic acid sequence encoding an amino acid sequence that is specifically cleavable by an enzyme or a chemical, wherein the linker sequence is positioned between the oil-body-surface-avoiding linker amino acid sequence that is also a non-proteolytic linker and said sequence encoding the first recombinant polypeptide. Also provided herein are recombinant multimeric-fusion-proteins comprising (i) an oil-body-targeting-protein, or fragment thereof, (ii) a first recombinant polypeptide and a (iii) second recombinant polypeptide, wherein said first and second recombinant polypeptides are capable of forming a multimeric-protein-complex. The oil-body-targeting-protein can be selected from an oil-body-protein or an immunoglobulin, and the oil-body-protein can be an oleosin or a caleosin. The multimeric-fusion-protein can be a heteromultimeric-fusion-protein, wherein said first and second recombinant polypeptide form an enzymatically active heteromultimeric redox complex or an immunoglobulin. In a particular embodiment, the first recombinant polypeptide is a thioredoxin and the second recombinant polypeptide is a thioredoxin-reductase. The thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194; and the thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 50 and SEQ ID NOs:195-313. In another embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. In yet another embodiment, positioned between the nucleic acid sequence encoding an oil-body-targeting-protein and the nucleic acid sequence encoding a first recombinant polypeptide can be a linker nucleic acid sequence encoding an oil-body-surface-avoiding linker amino acid sequence. The oil-body-surface-avoiding linker amino acid sequence can be substantially negatively charged, or have a molecular weight of at least 35 kd. Optionally, the gene fusion further comprises a linker nucleic acid sequence encoding an amino acid sequence that is specifically cleavable by an enzyme or a chemical, wherein the linker sequence is positioned between the oil-body-surface-avoiding linker amino acid sequence and said sequence encoding the first recombinant polypeptide. Also provided herein are isolated oil bodies comprising a multimeric-protein-complex comprising (i) an oil-body-targeting-protein and (ii) a first recombinant polypeptide, said oil bodies further comprising a second recombinant polypeptide, wherein said first and second recombinant polypeptide are capable of forming a multimeric-protein-complex. The oil-body-targeting-protein can be selected from an oil-body-protein or an immunoglobulin, and the oil-body-protein can be an oleosin or a caleosin. The multimeric-fusion-protein can be a heteromultimeric-fusion-protein, wherein said first and second recombinant polypeptide form an enzymatically active heteromultimeric redox complex or an immunoglobulin. In a particular embodiment, the first recombinant polypeptide is a thioredoxin and the second recombinant polypeptide is a thioredoxin-reductase. In another embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. Also provided herein are isolated oil bodies comprising (a) a first fusion protein comprising a first oil-body-targeting-protein fused to a first recombinant polypeptide; and (b) a second fusion protein comprising a second oil-body-targeting-protein fused to a second recombinant polypeptide, wherein said first and second recombinant polypeptide are capable of forming a multimeric-protein-complex. The oil-body-targeting-protein can be selected from an oil-body-protein or an immunoglobulin, and the oil-body-protein can be an oleosin or a caleosin. The multimeric-fusion-protein can be a heteromultimeric-fusion-protein, wherein said first and second recombinant polypeptide form an enzymatically active heteromultimeric redox complex or an immunoglobulin. In a particular embodiment, the first recombinant polypeptide is a thioredoxin and the second recombinant polypeptide is a thioredoxin-reductase. The thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194; and the thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 50 and SEQ ID NOs:195-313. In another embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. Also provided are cells and transgenic plants comprising oil bodies, multimeric-protein-complexes, and multimeric-fusion-proteins, set forth herein. In one embodiment, the first recombinant polypeptide can be an immunoglobulin-polypeptide-chain. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. In embodiments, wherein said first recombinant polypeptide is a thioredoxin and said second recombinant polypeptide is a thioredoxin-reductase, the methods described herein can be used to formulate the oil bodies for use in the preparation of a food product, personal care product or pharmaceutical composition. These formulations can further comprise the addition of NADP or NADPH. The food product can be a milk or wheat based food product. The personal care product can reduce the oxidative stress to the surface area of the human body or can be used to lighten the skin. The pharmaceutical composition can be used to treat chronic obstructive pulmonary disease (COPD), cataracts, diabetes, envenomation, bronchiopulmonary disease, malignancies, psoriasis, reperfusion injury, wound healing, sepsis, GI bleeding, intestinal bowel disease (IBD), ulcers, GERD (gastro esophageal reflux disease). Also provided herein are compositions comprising isolated oil bodies, thioredoxin and thioredoxin-reductase, wherein said thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194, and said thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 50 and SEQ ID NOs:195-313. The composition can further comprise NADP or NADPH. In another embodiment, the composition comprises a first recombinant polypeptide that can be an immunoglobulin-polypeptide-chain and a second recombinant polypeptide. For example, the first recombinant polypeptide can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second recombinant polypeptide can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. Also provided are multimeric-fusion-proteins, wherein the fusion-protein contains two or more polypeptide chains selected from the group of proteins set forth in FIG. 5 . Methods are also provided of reducing allergenicity of a food comprising the steps of providing the isolated oil bodies set forth herein; and adding the isolated oil bodies to the food, whereby allergenicity of the food is reduced. The food can be selected from the group consisting of wheat flour, wheat dough, milk, cheese, yogurt and ice cream. The various methods of treating food can further comprise providing NADH as a co-factor in the substantial absence of NADPH. Also provided herein are methods of treating or protecting a target against oxidative stress, comprising the steps of providing the recombinant redox fusion polypeptide comprising thioredoxin and thioredoxin-reductase; and contacting the recombinant fusion polypeptide with a target, wherein the target is susceptible to oxidative stress, thereby treating or protecting against the stress. The target can be selected from the group consisting of a molecule, a molecular complex, a cell, a tissue, and an organ. Also provided herein are methods for preparing an enzymatically active redox protein associated with oil bodies comprising: a) producing in a cell a redox fusion polypeptide comprising a first redox protein linked to a second redox protein; b) associating said redox fusion polypeptide with oil bodies through an oil-body-targeting-protein capable of associating with said redox fusion polypeptide and said oil bodies; and c) isolating said oil bodies associated with said redox fusion polypeptide. The first redox protein can be a thioredoxin and the second redox protein can be a thioredoxin-reductase. Also, provided herein are methods of producing an immunoglobulin, said method comprising: (a) producing in a cell comprising oil bodies, a first immunoglobulin-polypeptide-chain and a second immunoglobulin-polypeptide-chain wherein said first immunoglobulin-polypeptide-chain is capable of associating with said second immunoglobulin-polypeptide-chain to form said immunoglobulin; and (b) associating said immunoglobulin with an oil body through an oil-body-targeting-protein capable of associating with said oil bodies and said first immunoglobulin-polypeptide-chain. For example, the first immunoglobulin-polypeptide-chain can be an immunoglobulin light chain, or an immunologically active portion thereof, and the second immunoglobulin-polypeptide-chain can be an immunoglobulin heavy chain, or an immunologically active portion thereof. In this embodiment, the oil-body-targeting-protein can comprise protein A, protein L or protein G. Also provided herein are methods for preparing a redox protein or an immunoglobulin associated with oil bodies comprising: a) introducing into a cell a chimeric nucleic acid sequence comprising: 1) a first nucleic acid sequence capable of regulating transcription in said cell operatively linked to; 2) a second nucleic acid sequence encoding a recombinant fusion polypeptide comprising (i) a nucleic acid sequence encoding a sufficient portion of an oil-body-protein to provide targeting of said recombinant fusion polypeptide to an oil body linked to (ii) a nucleic acid sequence encoding a redox fusion polypeptide comprising a first redox protein linked to a second redox protein, or a nucleic acid sequence encoding a immunoglobulin comprising a first immunoglobulin-polypeptide-chain linked to a second immunoglobulin-polypeptide-chain, operatively linked to; 3) a third nucleic acid sequence capable of terminating transcription in said cell; b) growing said cell under conditions to permit expression of said redox fusion polypeptide or immunoglobulin in a progeny cell comprising oil bodies; and c) isolating from said progeny cell said oil bodies comprising said redox fusion polypeptide or immunoglobulin. In certain embodiments, positioned between said nucleic acid sequence encoding a sufficient portion of an oil-body-protein and said nucleic acid sequence encoding a redox fusion polypeptide or immunoglobulin can be a linker nucleic acid sequence encoding an oil-body-surface-avoiding linker amino acid sequence. The oil-body-surface-avoiding linker amino acid sequence can be substantially negatively charged or have a molecular weight of at least 35 kd. Optionally, the gene fusion further comprises a linker nucleic acid sequence encoding an amino acid sequence that is specifically cleavable by an enzyme or a chemical, wherein the linker sequence is positioned between the oil-body-surface-avoiding linker amino acid sequence and said nucleic acid sequence encoding a redox fusion polypeptide. In this optional embodiment, also contemplated is the introduction of an enzyme or chemical that cleaves said redox fusion polypeptide from said oil body, thereby obtaining isolated redox fusion polypeptide. The first redox protein can be a thioredoxin and said second redox protein can be a thioredoxin-reductase. In one embodiment, the thioredoxin and thioredoxin-reductase can be obtained from Arabidopsis . In another embodiment, the first redox protein is at least 5 times more active when produced as a redox fusion polypeptide as compared to the production of the first redox protein without the second redox protein. Also provided herein, for use with the various methods set forth herein is the formulation of an emulsion of the oil bodies associated with the redox fusion polypeptide for use in the preparation of a product capable of treating oxidative stress in a target, a product capable of chemically reducing a target, pharmaceutical composition, a personal care product or a food product. Accordingly, an emulsion formulation composition is provided. Also provided herein is a chimeric nucleic acid comprising: 1) a first nucleic acid sequence capable of regulating transcription in a host cell operatively linked to; 2) a second nucleic acid sequence encoding a recombinant fusion polypeptide comprising (i) a nucleic acid sequence encoding a sufficient portion of an oil-body-protein to provide targeting of said recombinant fusion polypeptide to an oil body linked to (ii) a nucleic acid sequence encoding a redox fusion polypeptide comprising a first redox protein linked to a second redox protein operatively linked to; 3) a third nucleic acid sequence capable of terminating transcription in said cell. The oil-body-protein can be an oleosin or a caleosin, the first redox protein can be a thioredoxin and said second redox protein can be a thioredoxin-reductase. In certain embodiments, positioned between said nucleic acid sequence encoding a sufficient portion of an oil-body-protein and said nucleic acid sequence encoding a redox fusion polypeptide is a linker nucleic acid sequence encoding an oil-body-surface-avoiding linker amino acid sequence. The oil-body-surface-avoiding linker amino acid sequence can be substantially negatively charged, or have a molecular weight of at least 35 kd. In one embodiment, the gene fusion optionally further comprises a linker nucleic acid sequence encoding an amino acid sequence that is specifically cleavable by an enzyme or a chemical, wherein the linker sequence is positioned between the oil-body-surface-avoiding linker amino acid sequence and said nucleic acid sequence encoding a redox fusion polypeptide. Also provided herein are transgenic plants, e.g., safflower plants, comprising any of the chimeric nucleic acid sequences and constructs described herein. The chimeric nucleic acids can be contained within a plastid. Accordingly, isolated plastids are provided having chimeric nucleic acids therein. Also provided are plant seeds comprising the chimeric nucleic acids provided herein. Also provided are oil body preparations obtained using any of the methods provided herein, and food products, pharmaceutical compositions, and personal care products containing the oil body preparations. The products and/or compositions provided herein are capable of treating oxidative stress in a target, capable of chemically reducing a target. Also provided is a detergent composition comprising an oil body preparation capable of chemically reducing a target, and related methods of cleansing an item, comprising administering such product to the item under conditions that promote cleansing. Also provided herein are nucleic acid constructs comprising a gene fusion, wherein the gene fusion comprises a first region encoding an oil-body-protein or an active fragment thereof, operably linked to a second region encoding at least one thioredoxin-related protein or an active fragment thereof. In one embodiment, the at least one thioredoxin-related protein can be thioredoxin. The thioredoxin can be selected from the group consisting of SEQ ID NOs:38, 42, 46, 50 and SEQ ID NOs:52-194. The thioredoxin can be obtained from Arabidopsis or wheat. In another embodiment, the at least one thioredoxin-related protein can be thioredoxin-reductase. The thioredoxin-reductase can be selected from the group consisting of those set forth in SEQ ID NOs:8, 9, 10, 40, 44, 48, 50 and SEQ ID NOs:195-313 and/or derived from Arabidopsis or wheat. The thioredoxin-reductase can be an NADPH-dependent thioredoxin-reductase. The second region can encode a thioredoxin and thioredoxin-reductase. In one embodiment, the thioredoxin and thioredoxin-reductase is obtained from Mycobacterium leprae . In another embodiment, the at least one thioredoxin-related protein can be an engineered fusion protein. The first region can precede, in a 5′ to 3′ direction, the second region. Alternatively, the first region follows, in a 5′ to 3′ direction, the second region. The gene fusion can optionally further comprise a third region encoding a second thioredoxin-related protein or an active fragment thereof, operably linked to the first region, or to the second region, or to both. A seed-specific promoter, such as a phaseolin promoter, can be operably linked to the gene fusion. In one embodiment, at least one thioredoxin-related protein is derived from a plant species selected from the group consisting of Arabidopsis and wheat. In another embodiment, at least one thioredoxin-related protein can be derived from E. coli. In one embodiment, the gene fusion further comprises a nucleic acid sequence encoding an oil-body-surface-avoiding linker amino acid sequence, wherein the linker amino acid sequence is positioned between the first region and the second region. The oil-body-surface-avoiding linker amino acid sequence can be substantially negatively charged, or have a molecular weight of at least 35 kd. In addition, the gene fusion can further comprise a linker nucleic acid sequence encoding an amino acid sequence that is specifically cleavable by an enzyme or a chemical, wherein the linker sequence is positioned between the oil-body-surface-avoiding linker amino acid sequence and the second region. Also provided herein are transgenic plants containing a nucleic acid construct comprising a gene fusion, wherein the gene fusion comprises a region encoding an oil-body-protein or an active fragment thereof, operably linked to a region encoding a first thioredoxin-related protein or an active fragment thereof. The thioredoxin-related protein can be thioredoxin. The nucleic acid construct can be contained within a plastid. In one embodiment, when the first thioredoxin-related protein is thioredoxin and the construct can further comprise a region encoding a thioredoxin-reductase. The gene fusion can optionally further comprise a third region encoding a second thioredoxin-related protein or an active fragment thereof, operably linked to the first region, or to the second region, or to both. The gene fusion can optionally further comprise a nucleic acid sequence encoding an oil-body-surface-avoiding linker amino acid sequence, wherein the nucleic acid encoding the linker amino acid sequence is positioned between the region encoding an oil-body-protein and the region encoding a first thioredoxin-related protein. The oil-body-surface-avoiding linker amino acid sequence can be substantially negatively charged, or have a molecular weight of at least 35 kd. The gene fusion can optionally further comprise a linker nucleic acid sequence encoding an amino acid sequence that is specifically cleavable by an enzyme or a chemical, wherein the linker sequence is positioned between the oil-body-surface-avoiding linker amino acid sequence and the region encoding a first thioredoxin-related protein. Also provided is a transgenic plant comprising a nucleic acid construct, a seed-specific promoter operably linked to a gene fusion, wherein the gene fusion comprises a region encoding an oil-body-protein or an active fragment thereof, operably linked to a region encoding a first thioredoxin-related protein or an active fragment thereof, wherein a fusion protein comprising activities of oleosin and the thioredoxin-related protein is produced in a seed of the plant. In another embodiment, a thioredoxin-related protein having concentration of at least about 0.5% of total cellular seed protein is provided. Also provided herein is an extract comprising an activity of a thioredoxin-related protein. Also provided are oil bodies and/or oil obtained from various seeds. Also provided herein are methods of making a fusion protein comprising a thioredoxin-related activity, the method comprising the steps of: a) providing a transgenic plant comprising a nucleic acid construct comprising a seed-specific promoter operably linked to a gene fusion, wherein the gene fusion comprises a region encoding an oil-body-protein or an active fragment thereof, operably linked to a region encoding a first thioredoxin-related protein or an active fragment thereof, the gene fusion encoding a fusion protein comprising a thioredoxin-related activity; b) obtaining seeds from the plant; and c) recovering the fusion protein by isolating oil bodies from the seeds. In one embodiment, the oil bodies are fractionated to achieve partial purification of the fusion protein. The oil bodies can be in association with a fusion protein. The oil-body-protein can be cleaved from the thioredoxin-related protein after fractionation of the oil bodies. The cleaving step can make use of a protease or chemical proteolysis. Also provided herein are methods of reducing allergenicity of a food comprising the steps of: a) providing a preparation comprising oil bodies associated with a fusion protein, the fusion protein comprising an oil-body-protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof; and b) adding the preparation to the food, whereby allergenicity of the food is reduced due to activity of the thioredoxin-related protein or fragment. The food can be wheat flour, wheat dough, milk, cheese, yogurt and ice cream. In one embodiment, NADH is used as a co-factor in the substantial absence of NADPH. Also provided herein are pharmaceutical compositions comprising a fusion protein, the fusion protein comprising an oil-body-protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof, in a pharmaceutically acceptable carrier. The oil bodies can be associated with the fusion protein. Also provided is a cosmetic formulation comprising oil bodies associated with a fusion protein, the fusion protein comprising an oil-body-protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof, in a pharmaceutically acceptable carrier. Also provided are methods of treating or protecting a target against oxidative stress, comprising the steps of: a) providing a preparation comprising a fusion protein, the fusion protein comprising an oil-body-protein or an active fragment thereof and a thioredoxin-related protein or an active fragment thereof; and b) contacting the preparation with a target, wherein the target is susceptible to oxidative stress, thereby treating or protecting against the stress. The target can be selected from the group consisting of a molecule, a molecular complex, a cell, a tissue, and an organ. Also provided is a nucleic acid construct comprising a gene fusion, wherein the gene fusion comprises a first region encoding an oil-body-protein or an active fragment thereof, operably linked to a second region encoding at least one polypeptide or an active fragment thereof, and an oil-body-surface-avoiding linker in frame between the first and second region polypeptides. Also provided are methods of expressing this construct into the encoded amino acid sequence; and oil bodies, formulations, emulsions, cells, and plants comprising the construct and encoded amino acid sequence. These particular constructs, oil bodies, formulations, emulsions, cells, and plants can be produced according to the methods described herein. The second region can encode any polypeptide, for example, a therapeutically, nutritionally, industrially or cosmetically useful peptide as set forth herein. For example, the second region can encode a redox protein, an immunoglobulin, a thioredoxin-related protein or any one or more recombinant polypeptides of a multimeric-protein-complex. Other features and advantages of the present invention will become readily apparent from the following detailed description. It should be understood however that the detailed description and the specific examples while indicating particular embodiments of the invention are given by way of illustration only.
Adaptive comparator circuit and acoustic distance sensor comprising said circuit
The invention relates to an adaptive comparator circuit comprising a comparator with a threshold value input which receives a threshold voltage and a signal input which receives a voltage signal. The threshold value input is connected to one pole by means of a switch and connected to the other pole of a first voltage source by means of a capacitor. The threshold value input is also connected to the signal input of the comparator by means of a diode or a second voltage source. The switch is controlled by the control signal of a signal transmitter. When the switch is in a closed position, the capacitor is charged with the voltage of the first voltage source. When the switch is in an open position, the capacitor is discharged in such a way that the threshold voltage of the signal voltage is corrected at a given time interval.
1. An adaptive comparator circuit, particularly for an acoustic distance sensor, comprising a first comparator (1), having a first threshold voltage input (1a) to which a first threshold voltage (Uref1) is applied and a first signal input (1b) to which a voltage signal (Usignal) is applied and, when a voltage signal (Usignal) is present that is greater than the first threshold voltage (Uref1), said comparator emits a first switching signal (S1), characterized by a signal transmitter (2), especially a microcontroller or microprocessor, that at times emits a control signal (SSt), a first switch (3, 103) that has a control means (3a, 103a), a switch input (3b, 103b) and a switch output (3c, 103c), and that can be regulated or activated through the effect of the control signal (SSt) on the control means (3a, 103a) in such a way that it is in the closed state when the signal transmitter (2) emits a control signal (SSt), and otherwise it is in the open state, or vice versa, whereby the switch input (3b, 103b) is connected to the positive or negative first pole (4a) of a first direct voltage source and the switch output (3c, 103c) is connected to the first threshold voltage input (1a), so that the first threshold voltage (Uref1) is equal to the voltage present at the switch output (3c, 103c), a capacitor (6) via which the switch output is connected to the negative or positive pole (4b) of the first direct voltage source, and a diode (5) via which the switch output (3c, 103c) is connected to the first signal input (1b) in such a way that the anode (5a) or the cathode (5b) of the diode (5) is connected to the switch output (3c, 103c). 2. The adaptive comparator according to claim 1, characterized in that, instead of the diode (5), a second direct voltage source (7) is used whose positive pole (7a) is in the place of the anode (5a) of the diode (5) and whose negative pole (7b) is in the place of the cathode (5b) of the diode (5). 3. The adaptive comparator according to claim 2, characterized in that the voltage supplied by the second voltage source (7) can be regulated or adjusted. 4. The adaptive comparator according to claim 1, characterized in that the signal transmitter (2) emits an electric control signal (SSt) at regular intervals. 5. The adaptive comparator according to claim 1, characterized in that the first switch (3, 103) is a switch that can be electrically or electronically controlled by the control means (3a, 103a) and the control signal (SSt) is an electric signal that is transmitted by the signal transmitter (2) via a control output (2a) and fed to the control input (3a, 103a). 6. The adaptive comparator according to claim 5, characterized in that the first switch (3, 103) is a transistor (103) and the control-current terminal (3a, 103a) is the base (103) or the gate terminal of the transistor (103). 7. The adaptive comparator according to claim 1, characterized in that a first resistor (11) is connected between the first pole (4a) and the first threshold voltage input (1a), and a second resistor (12) is connected between the first threshold voltage input (1a) and the second pole (4b), so that the first resistor (11) and the second resistor (12) form a first voltage divider (11, 12). 8. The adaptive comparator according to claim 7, characterized in that first voltage divider (11, 12) is an adjustable potentiometer whose pick-up is connected to the switch output (3c, 103c). 9. The adaptive comparator according to claim 1, characterized by a second comparator (20) that has a second threshold voltage input (20a) and a second signal input (20b), and that emits a second switching signal S2 when a greater voltage is present at the second signal input (20b) than at the second threshold voltage input (20a), a third resistor (13) via which the first pole (4a) is connected to the second threshold voltage input (20a), a fourth resistor (14) that, on the one hand, is connected to the second threshold voltage input (20a) and, on the other hand, to the second pole (4b) of the first voltage source, so that the third resistor (13) and the fourth resistor (14) form a second voltage divider (13, 14), a second switch (23) that is connected in series to the diode (5) and that can be regulated or activated by the second switching signal S2 in such a way that it is in the closed state when the second comparator (20) emits the second switching signal S2, and otherwise it is in the open state. 10. The adaptive comparator according to claim 9, characterized in that a fifth resistor (15) is connected between the switch output (3c, 103c) and the second threshold voltage input (20a). 11. The adaptive comparator according to claim 9, characterized in that the second voltage divider (13, 14) is a potentiometer. 12. The adaptive comparator according to claim 1, characterized in that a sixth resistor (16) is connected between the control output (2a) and the control-current terminal (3a, 103a). 13. The adaptive comparator according to claim 6, characterized in that a seventh resistor (17) is connected between the control-current terminal (3a, 103a) and the first pole (4a). 14. The adaptive comparator according to claim 1, characterized in that the diode (5) or the second voltage source (7) is connected in series to an eighth resistor (18). 15. The adaptive comparator according to claim 1, characterized in that, before reaching the first signal input (1b), the voltage signal Usignal passes through a voltage follower or impedance transformer (21). 16. The adaptive comparator according to claim 1, characterized in that the voltage supplied by the first voltage source (4a, 4b) is greater in magnitude than the voltage of the maximum of the voltage signal Usignal. 17. The adaptive comparator according to claim 1, characterized in that the diode (5) is connected to at least another diode codirectionally in series. 18. An acoustic distance sensor, comprising a control unit (31) that at times transmits signal pulses (41) to an oscillator (32) which, during the presence of a signal pulse (41), transmits an alternating voltage to a sound transducer (34) that is excited by the alternating voltage to emit sound waves and that is capable of receiving sound waves coming back to the sound transducer (34) as an echo as a result of reflection, and of converting them into an electric received signal, and also comprising an envelope curve shaper (37) to which the received signal is fed and which forms the envelope curve of the received signal and emits it as a voltage signal Usignal, characterized in that the voltage signal is fed to an adaptive comparator circuit comprising a first comparator (1) having a first threshold voltage input (1a) to which a first threshold voltage (Uref1) is applied and a first signal input (1b) to which a voltage signal (Usignal) is applied and, when a voltage signal (Usignal) is present that is greater than the first threshold voltage Uref1, said comparator (1) emits a first switching signal (S1), a signal transmitter (2), especially a microcontroller or microprocessor, that at times emits a control signal (SSt), a first switch (3, 103) that has a control means (3a, 103a), a switch input (3b, 103b) and a switch output (3c, 103c), and that can be regulated or activated through the effect of the control signal (SSt) on the control means (3a, 103a) in such a way that it is in the closed state when the signal transmitter (2) emits a control signal (SSt), and otherwise it is in the open state, or vice versa, whereby the switch input (3b, 103b) is connected to the positive or negative first pole (4a) of a first direct voltage source and the switch output (3c, 103c) is connected to the first threshold voltage input (1a), so that the first threshold voltage (Uref1) is equal to the voltage present at the switch output (3c, 103c), a capacitor (6) via which the switch output (3c, 103c) is connected to the negative or positive pole (4b) of the first direct voltage source, and a diode (5) via which the switch output (3c, 103c) is connected to the first signal input (1b) in such a way that the anode (5a) or the cathode (5b) of the diode (5) is connected to the switch output (3c, 103c). 19. The acoustic distance sensor according to claim 18, characterized in that, instead of the diode (5), a second direct voltage source (7) is used whose positive pole (7a) is in the place of the anode (5a) of the diode (5) and whose negative pole (7b) is in the place of the cathode (5b) of the diode (5). 20. The acoustic distance sensor according to claim 18 characterized in that the alternating voltage emitted by the oscillator (32) reaches the sound transducer (34) after passing through a driver stage (33). 21. The acoustic distance sensor according to claim 18, characterized in that, before entering the envelope curve shaper (37), the received signal passes through an amplifier (36c). 22. The acoustic distance sensor according to claim 21, characterized in that the amplifier (36c) is a logarithmic amplifier. 23. The acoustic distance sensor according to claim 18, characterized in that, before entering the envelope curve shaper (37), the received signal passes through a voltage limiter (35) that limits the amplitude of the received signal to a maximum value. 24. The acoustic distance sensor according to claim 18, characterized in that the signal transmitter (2) is integrated into the control unit (31) or is a component of the control unit (31). 25. The acoustic distance sensor according to claim 18, characterized in that the first switching signal (S1) is transmitted to the control unit (31) for evaluation purposes. 26. The acoustic distance sensor according to claim 18, characterized in that the signal pulses (41) are synchronized with the control signals (SSt) in such a way that the control signals (SSt) each begin before the signal pulses (41). 27. The acoustic distance sensor according to claim 18, characterized in that the signal pulses 41 are synchronized with the control signals SSt in such a way that the control signals SSt each begin before or simultaneously with the end of the signal pulses (41). 28. An adaptive comparator circuit for an acoustic distance sensor, comprising a first comparator (1) having a first signal input and a first threshold voltage input; a first direct voltage source (4) for delivering a threshold voltage to the first threshold voltage input, wherein the first direct voltage source comprises a first and a second pole; a first switch (3), wherein the first switch is connected to the first direct voltage source and to the first threshold voltage input; a signal transmitter (2), wherein the signal transmitter controls the first switch through an input of the switch; a capacitor (6), wherein a first plate of the capacitor is connected to the a first threshold voltage input and wherein a second plate of the capacitor is connected to second pole the first direct voltage source; a diode (5) connected the first signal input and to the first threshold voltage input of the first comparator. 29. The adaptive comparator circuit according to claim 28, wherein the diode is connected in series to a first resistor. 30. The adaptive comparator circuit according to claim 1, wherein the signal transmitter emits an electric control signal at regular intervals. 31. The adaptive comparator circuit according to claim 28 further comprising a second resistor (11) connected between the first pole of the first direct voltage source and the first threshold voltage input, and a third resistor (12) connected between the first threshold voltage input and the second pole of the first direct voltage source, so that the second resistor (11) and the third resistor (12) form a first voltage divider (11, 12). 32. The adaptive comparator circuit according to claim 31, wherein the first voltage divider (11, 12) is an adjustable potentiometer whose pick-up is connected to the first threshold voltage input. 33. The adaptive comparator circuit according to claim 28 further comprising a fourth resistor (17) connected between the first pole of the first direct voltage source and the input of the first switch; a fifth resistor (16) connected between the input of the first switch and the output of the signal transmitter (2).
<SOH> BRIEF DESCRIPTION OF THE DRAWING IN WHICH THE FOLLOWING IS SHOWN <EOH>FIG. 1 —a schematic block diagram of a variant of an acoustic distance sensor according to the state of the art, FIG. 2 —a schematic representation of a typical course over time of a) a signal pulse, b) an amplification factor, c) an envelope curve and d) a comparator output signal, all of which can occur during the operation of the distance sensor shown in FIG. 1 , FIG. 3 —a schematic block diagram of another variant of an acoustic distance sensor according to the state of the art, FIG. 4 —a schematic representation of a typical course over time of a) a timing pulse, b) an envelope curve and c) a comparator output signal, all of which can occur during the operation of the distance sensor shown in FIG. 3 , FIG. 5 —a circuit diagram of an embodiment of a comparator circuit according to the invention, FIG. 6 —a circuit diagram of an alternative embodiment of a comparator circuit according to the invention, FIGS. 7 and 8 —a schematic block diagram of embodiments of an acoustic distance sensor according to the invention, FIG. 9 —a schematic representation of a typical course over time of a) a signal pulse, b) a control signal, c) an envelope curve and threshold voltage as well as d) a comparator switching signal, all of which can occur during the operation of the distance sensor according to the invention shown in FIG. 8 , whereby an echo occurs during the quiescent phase, FIG. 10 —a schematic representation of a typical course over time of a) an envelope curve and threshold voltage as well as b) a comparator switching signal, all of which can occur during the operation of the distance sensor according to the invention shown in FIG. 8 , whereby an echo additionally occurs during the relaxation phase, and FIGS. 11 to 13 —circuit diagrams of further embodiments of comparator circuits according to the invention. detailed-description description="Detailed Description" end="lead"? FIGS. 1 to 4 serve to further illustrate the state of the art. FIG. 1 shows a schematic block diagram of a variant of an acoustic distance sensor 30 a with which a measure according to the state of the art has been taken in order to reduce the short range. The distance sensor 30 a shown in FIG. 1 comprises a control unit 31 , an oscillator 32 , a driver 33 , a sound transducer 34 , a voltage limiter 35 , an amplifier 36 a with regulatable amplification factor, an envelope curve shaper 37 , a comparator 1 as well as an amplification factor actuation stage 38 a. The control unit 31 transmits short signal pulses 41 to the oscillator 32 . The latter oscillates when a signal pulse is applied, thus emitting an alternating voltage of, for example, 400 kHz, which is then amplified by the driver 33 and subsequently used to excite the sound transducer 34 so that the latter is excited so as to oscillate, i.e. to emit sound waves. Corresponding to the short signal pulses, the sound transducer 34 emits short sound wave packets that constitute the transmittal pulse of the distance sensor 30 a. During the transmitting phase, the sound transducer 34 oscillates at a high oscillation amplitude. This period of time is followed by a relaxation time in which the oscillation amplitude dies out—usually exponentially. In the subsequent quiescent phase, the oscillation amplitude generally remains at a constant mean disturbance level. The signal emitted by the sound transducer and thus also echoes returning after the distance-proportional propagation time are fed via the voltage limiter 35 to the amplifier 36 a with a regulatable amplification factor. The voltage limiter keeps the high voltage used for exciting the sound transducer away from the amplifier 36 a Using, for example, an internal rectifier circuit, the envelope curve shaper 37 forms the envelope curve of the voltage limiter 35 on the basis of the alternating voltage signal emitted by said voltage limiter 35 and transmits to the comparator 1 an envelope curve voltage signal whose course over time corresponds to that of the envelope curve. The comparator 1 transmits a switching signal to the control unit 31 for evaluation purposes when the envelope curve voltage signal is greater than a first steadily supplied threshold voltage. In particular, the comparator then emits a switching signal when an echo signal is received whose magnitude exceeds that of the threshold voltage. The amplification factor of the amplifier 36 a is regulated by the amplification factor actuation stage 38 a in a time-dependent manner such that the amplification factor is minimal during and immediately after the transmittal pulse and grows as soon as the oscillation amplitude of the sound transducer 34 has died out to such an extent that the magnitude of the received signal is no longer limited by the voltage limiter 35 . Therefore, the growth of the amplification factor does not begin completely simultaneously with the end of the signal pulse, but rather delayed by a certain lag time dt with respect thereto ( FIG. 2 ). The amplification factor actuation stage 38 a receives the time information necessary for synchronizing the time behavior of the amplification factor with the rhythm of the transmittal pulse from the control unit 31 . In this manner a reduction of the short range can be achieved. The course over time can advantageously be selected in such a way that the distance-dependent attenuation of the echo is countered and the system sensitivity remains constant. FIG. 2 shows a schematic representation of a course over time of a) a signal pulse, b) an amplification factor of the amplifier 36 a , c) an envelope curve and threshold voltage as well as d) a comparator output signal, that can typically occur during the operation of the distance sensor shown in FIG. 1 . The time axes of the curves a) to d) are selected identically in order to illustrate the courses over time. As was already explained with reference to FIG. 1 , during the signal pulse 41 , the amplification factor 42 reaches a minimum 42 a that lasts longer than the signal pulse 41 by a certain lag time dt, and then it grows monotonously over time. During the signal pulse 41 , the envelope curve voltage 43 reaches a maximum (envelope curve segment 43 a ) at which it still remains during the lag time dt after the end of the signal pulse 41 . Then the envelope curve voltage 43 drops during the relaxation phase (envelope curve segment 43 b ), whereby the course of the envelope curve 43 b results from the multiplication of, for example, the exponential decay of the sound transducer amplitude by the growing amplification factor. The temporary lowering of the amplification factor during and after the signal pulse causes the envelope curve voltage 43 to fall below the threshold voltage 44 earlier than would be the case if the amplification factor were not lowered. The advantageous result of this is a reduction of the short range. In the quiescent phase, the envelope curve voltage 43 finally reaches a low constant level (envelope curve segment 43 b ). The threshold voltage 44 is constant over time. Since the comparator 1 emits a switching signal, as soon as the envelope curve voltage 43 exceeds the threshold voltage 44 , in a disadvantageous manner, at the beginning of the signal pulse 41 , a dummy switching signal 45 is always triggered that lasts until the envelope curve voltage 43 has dropped below the threshold value 44 . An echo signal (envelope curve segment 43 d ) causes a useful switching signal 46 so that the control unit has to be capable of distinguishing a useful switching signal 46 from a dummy switching signal 45 which, without the presence of an echo signal, was caused exclusively by the signal pulse 41 . The capability to make such a distinction requires complex hardware or software resources and thus entails disadvantages. Further drawbacks associated with a distance sensor of the type illustrated in FIG. 1 were already explained above. As already explained above, another method of reducing the short range lies in providing a threshold voltage that can be changed over time. For purposes of further illustrating the state of the art, FIG. 3 shows a schematic block diagram of a corresponding variant of an acoustic distance sensor 30 b . The distance sensor 30 b shown in FIG. 3 comprises a control unit 31 , an oscillator 32 , a driver 33 , a sound transducer 34 , a voltage limiter 35 , a preferably logarithmic amplifier 36 b , an envelope curve shaper 37 , a comparator 1 as well as a threshold value actuation stage 38 b which emits a threshold voltage that can be changed over time and transmits it to the comparator 1 . The essential difference from the distance sensor 30 a illustrated in FIG. 1 lies in the fact that the short range reduction is not achieved by an amplification factor that can be changed over time, but rather by a threshold voltage that can be changed over time. FIG. 4 shows a schematic representation of a course over time of a) a signal pulse, b) an envelope curve and threshold value as well as c) a comparator output signal, that can typically occur during the operation of the distance sensor 30 b shown in FIG. 3 . The time axes of the curves a) to c) are selected identically in order to illustrate the courses over time. During a signal pulse 41 , the envelope curve voltage 53 reaches a maximum (envelope curve segment 53 a ) at which it still remains during the lag time dt after the end of the signal pulse 41 . Then the envelope curve voltage 53 drops linearly during the relaxation phase (envelope curve segment 53 b ), since the amplifier 36 b is a logarithmic amplifier. Finally, in the quiescent phase, the envelope curve voltage 53 reaches a low constant level (envelope curve segment 53 c ). The threshold voltage 54 is not constant over time but rather is regulated by the threshold value actuation stage 38 b in such a way that it rises before every signal pulse 41 , remains at a maximum value for a certain period of time (envelope curve segment 54 a ), then drops (curve segment 54 b ) and finally reaches a constant level (envelope curve segment 54 c ) during the quiescent phase, whereby the threshold voltage 54 remains greater than the envelope curve voltage 53 , as long as no echo is received. The threshold value actuation stage 38 b receives the necessary time information for synchronizing the time behavior of the threshold voltage 54 with the rhythm of the signal pulse 41 from the control unit 31 . The advantageous result is a reduction of the short range. Moreover, advantageously, no dummy switching signal is triggered by the signal pulse 41 . Rather, a switching signal 56 is triggered only by the rise in the envelope curve 53 (envelope curve segment 53 d ) associated with an echo, so that the switching signal 56 is a useful switching signal 56 . Drawbacks associated with a distance sensor of the type illustrated in FIG. 3 were already explained above. Now reference is made to FIG. 5 , which illustrates a schematic circuit diagram of an embodiment 10 a of an adaptive comparator circuit according to the invention It comprises a signal transmitter 2 , a first regulatable switch 3 , a capacitor 6 , a diode 5 and a first comparator 1 , that has a first threshold value input 1 a and a first signal input 1 b. At the first threshold value input 1 a , a threshold voltage U ref1 , is present that is emitted internally in the adaptive comparator circuit according to the invention. At the signal input 1 b , a voltage signal U signal is present that can be, for example, an envelope curve of the received signal of a sound transducer and that is fed via a first electric terminal 8 to the adaptive comparator circuit according to the invention. The first comparator 1 emits a first switching signal S 1 when the voltage signal U signal is greater than the first threshold voltage U ref1 . The first switching signal S 1 can be tapped, for example; via a second electric terminal 9 and transmitted for evaluation purposes. Via a control output 2 a , the signal transmitter 2 emits an electric control signal S St at times, for example, at regular intervals. In the simplest case, the signal transmitter 2 can be, for instance, a manually operated signaling key. Moreover, the signal transmitter can be, for example, an electronic square-wave timing generator circuit with a suitable duty cycle. Preferably, the signal transmitter 2 is a microcontroller or microprocessor that is programmed in such a way that, at regular intervals, it emits an electric control signal S St having a time duration of typically, for example, 20 microseconds. The first switch 3 has a control means 3 a , a switch input 3 b and a switch output 3 c . The control means 3 a is connected to the control output 2 a and can be regulated by the control signal S St in such a way that it is in the closed state when the signal transmitter 2 emits a control signal S St , and otherwise it is in the open state. The first switch 3 can be, for example, a transistor. The switch input 3 b is connected to the positive first pole 4 a of a first direct voltage source 4 a , 4 b (not shown here). The switch output 3 c is connected to the first threshold value input 1 a of the comparator 1 , so that the first threshold voltage U ref1 is equal to the voltage present at the switch output 3 c . Thus, when the switch 3 is closed, the potential of the positive first pole 4 a is present at the first threshold value input 1 a of the comparator 1 . The switch output 3 c is also applied via a capacitor 6 to the negative second pole 4 b of the first direct voltage source so that the capacitor 6 is charged with the voltage supplied by the first voltage source when the switch 3 is closed. The switch output 3 c is also connected to the anode 5 a of a diode 5 . The cathode 5 b of the diode 5 is connected to the first signal input 1 b of the comparator. As soon as the signal transmitter 2 emits a control signal S St , the first switch 3 is in the closed state. Therefore, the capacitor 6 is charged with the voltage supplied by the first voltage source. When the switch 3 is closed, this voltage is also present at the threshold value input 1 a. After the end of the control signal S St , the switch 3 is in the open state. The capacitor 6 can now discharge according to the invention via the diode 5 to such an extent until the first U ref1 reaches a value that corresponds to the sum of the voltage signal U signal and the diode flow voltage dU. This means that the voltage that is established at the capacitor 6 depends on the magnitude of the voltage signal U signal : the smaller the voltage signal U signal , the more the voltage and thus the threshold voltage U ref1 drop according to the invention at the capacitor. Hence, a threshold voltage U ref1 is established that is greater than the voltage signal U signal by the fundamental voltage distance dU. According to the invention, the task of the diode 5 is to allow the discharging of the capacitor 6 and thus a drop in the threshold voltage U ref1 for as long as and only until the difference of U ref1 −U signal has dropped to the fundamental voltage distance dU, but to prevent a recharging of the capacitor and thus an increase in the threshold voltage if the voltage signal U signal increases. If the voltage signal U signal drops, then the threshold voltage U ref1 follows the voltage signal U signal at the distance dU. In contrast, if the voltage signal U signal increases, then the threshold voltage U ref1 remains constant because of the blocking effect of the diode 5 . Of course, the polarity of the first direct voltage source can also be the opposite from the one in FIG. 1 , so that the first pole 4 a is negative and the second pole 4 b is positive. In this case, the diode 5 in the circuit shown in FIG. 1 has to be arranged in the reverse direction, so that its cathode is connected to the switch output 3 c and its anode to the second pole 4 b. In another embodiment of the invention, the fundamental voltage distance dU is augmented by a codirectional series connection of a plurality of diodes 5 or second direct voltage sources 7 . According to the invention, as an alternative, the function of the diode 5 can be assumed by a second direct voltage source whose positive pole is in the place of the anode 5 a of the diode 5 and whose negative pole is in the place of the cathode 5 b of the diode 5 . FIG. 6 shows an embodiment 10 b of an adaptive comparator circuit according to the invention, which can be used as an alternative to that shown in FIG. 1 . The diode shown in FIG. 5 has been replaced by a second voltage source 7 , whereby its positive pole 7 a has been put in the place of the anode 5 a and the negative pole 7 b has been put in the place of the cathode 5 b . The fundamental voltage distance dU shown in FIG. 1 has now been replaced by the voltage of the voltage source 7 , that is to say, the fundamental voltage distance dU between U ref1 and U signal is now defined by the voltage supplied by the second direct voltage source. If the voltage supplied by the second voltage source 7 can be regulated or adjusted, then the fundamental voltage distance dU can be varied as needed. FIG. 7 shows a schematic block diagram of an embodiment of an acoustic distance sensor according to the invention in which the adaptive comparator circuit 10 a shown in FIG. 5 is used. A control unit 31 transmits short signal pulses at times, preferably at regular intervals, each lasting, for example, 25 microseconds, to an oscillator 32 . When a signal pulse is present, the oscillator 32 transmits an alternating voltage to a sound transducer 34 that is excited by the latter to emit sound waves. The sound transducer 34 is capable of receiving sound waves reflected, for example, from an object and coming back to the sound transducer 34 as an echo, and of converting them into an electric received signal, whereby the propagation time of the sound waves is a measure of the distance from the object. The received signal is fed to an envelope curve shaper 37 which forms the envelope curve of the received signal and transmits it via an electric terminal 8 to the adaptive comparator circuit 10 a shown in FIG. 5 . In a preferred embodiment of the invention, the signal pulses 41 that the control unit 31 transmits to the oscillator 32 are synchronized with the control signals S St that the control unit 2 transmits to the switch 3 and this is done in such a way that the control signals S St each begin shortly before the signal pulses 41 as will still be explained below with reference to FIGS. 9 a and 9 b . In this manner, it is ensured that, already at the beginning of the transmittal phase, the threshold voltage U ref1 is greater than the voltage signal U signal , so that no dummy switching signal is triggered. In another preferred embodiment of the invention, the signal pulses 41 are advantageously synchronized with the control signals S St in such a way that the control signals S St each begin before or simultaneously with the end of the signal pulses 41 . This ensures that, without any time lag, the threshold voltage U ref1 is supplied to the dropping voltage signal U signal at the fundamental voltage distance dU already at the beginning of the relaxation phase. The sensitivity of the distance sensor is defined by the fundamental voltage distance dU. FIG. 8 shows a schematic block diagram of another embodiment of an acoustic distance sensor according to the invention, in which the adaptive comparator circuit 10 a shown in FIG. 5 is likewise used. The distance sensor shown in FIG. 8 —in comparison to that shown in FIG. 7 —additionally has a driver 33 that amplifies the alternating voltage supplied by the oscillator 32 and transmits it to the sound transducer 34 . Moreover, before the received signal is fed into the envelope curve shaper 37 , it is passed through a voltage limiter 35 and through a logarithmic amplifier 36 c. Among other things, the voltage limiter 35 counters an overmodulation of the amplifier. The amplifier 36 c serves to adapt the low input level of just a few microvolts to the higher input level of the subsequent evaluation circuit. In FIG. 8 , in order to facilitate the synchronization, the signal transmitter 2 is accommodated in the control unit 31 . For example, the signal transmitter 2 and the control unit 31 can be combined in one single unit, for example, in a microprocessor, that concurrently fulfills the function of the signal transmitter 2 as well as that of the control unit 31 . In FIG. 8 , the first switching signal S 1 of the control unit 31 is also supplied for evaluation purposes, especially for determining the propagation time and calculating the distance. FIGS. 9 and 10 serve to further explain the advantageous effects of the invention, whereby the time axes of all of the curves have been selected identically in order to illustrate the course over time. FIG. 9 shows a schematic representation of a typical course over time of a) a signal pulse, b) a control signal, c) a voltage signal and threshold voltage as well as d) a comparator switching signal, all of which can occur during the operation of the distance sensor according to the invention shown in FIG. 8 , whereby an echo occurs during the quiescent phase. The control unit 31 transmits short signal pulses 41 to the oscillator 32 , as a result of which the sound transducer 34 is excited so as to emit a threshold voltage packet ( FIG. 9 , Curve a). Moreover, the signal transmitter 2 transmits a control signal S St to the switch 3 , which is synchronized with the signal pulse 41 in such a way that the control signal S St begins shortly before the signal pulse 41 and ends at the same time as or during the presence of the former ( FIG. 9 , Curve b). FIG. 9 , Curve c, shows the course of the voltage signal U signal (solid curve) that is identical to the envelope curve, and the course of the threshold voltage U ref1 (dotted curve). The course of the U signal during the transmittal and relaxation phase and at the beginning of the quiescent phase corresponds essentially to that of the envelope curve 53 , which was explained with reference to FIG. 4 . While the control signal S St is present at the switch 3 , the latter is opened, so that a threshold voltage U ref1 is established which is identical to the voltage supplied by the first direct voltage source 4 a , 4 b ( FIG. 9 , Curve c). Therefore, this has been advantageously selected in such a way that it is greater than the maximum of the voltage signal, so that the adaptation of the threshold voltage to the voltage signal begins immediately after the opening of the switch 3 . In the present example, the maximum possible magnitude of the voltage signal is supplied by the voltage limiter 35 or by the overmodulation limit of the amplifier 36 c ( FIG. 8 ). According to the invention, after the end of the control signal S St , a threshold voltage U ref1 is established that is greater than the voltage signal U signal by the voltage distance dU. Therefore, the threshold voltage U ref1 drops immediately after the end of the control signal by a certain quantity. From this point in time on, the threshold voltage proceeds parallel-offset by a fundamental voltage distance dU with respect to that of the voltage signal U signal as long as the latter does not increase. A dummy switching signal is not triggered. If, however, the voltage signal U signal increases, the threshold voltage remains constant so that a first echo peak 60 of the voltage signal U signal leads to a triggering of the comparator 1 as soon as the voltage signal U signal exceeds the threshold voltage U ref1 . In this case, the comparator 1 emits a first switching signal S 1 ( FIG. 9 , Curve d). FIG. 10 shows a schematic representation of a typical course over time of a) a voltage signal and threshold voltage as well as b) a comparator switching signal, all of which can occur during the operation of the distance sensor according to the invention shown in FIG. 8 , whereby an echo occurs during the relaxation phase and another echo occurs during the quiescent phase. The courses over time of the signal pulse 41 and of the control signal S St are identical to those shown in FIG. 9 . During the relaxation time, an echo signal occurs, which manifests itself in a second echo peak 61 of the voltage signal U signal ( FIG. 10 , Curve a). Up until the beginning of the second echo peak 61 , the curves correspond to those shown in FIG. 9 , Curve c. According to the invention, however, the decrease of the threshold voltage U ref1 comes to a halt as soon as the voltage signal U signal is no longer dropping, and a first switching signal S 1a is triggered by the second echo peak 61 . After the second echo peak 61 that occurs during the relaxation time, the fundamental voltage distance dU is once again established between the threshold voltage U ref1 and the voltage signal U signal . During the quiescent phase, another, third echo peak 62 of the voltage signal U signal occurs, as a result of which another switching signal S 1b is triggered. Now reference will be made to FIGS. 11 to 13 , which show additional advantageous embodiments of an adaptive comparator circuit according to the invention. FIG. 11 shows a preferred embodiment 10 c of the invention which differs from the adaptive comparator circuit 10 a shown in FIG. 6 as follows: a) The first switch is a transistor 103 . The base 103 a of the transistor 103 serves as the control-current terminal, the emitter 103 b serves as the switch input and the collector 103 c as the switch output. b) A first resistor 11 is connected between the first pole 4 a and the first threshold value input 1 a , and a second resistor 12 is connected between the first threshold value input 1 a and the second pole 4 b , so that the first resistor 11 and the second resistor 12 form a first voltage divider 11 , 12 . Therefore, the magnitude of the first threshold voltage U ref1 counters the voltage that is supplied by the first voltage divider. Thus, through the use of the first voltage divider 11 , 12 , the voltage distance by which the threshold voltage is parallel-offset with respect to the voltage signal after the voltage signal U signal has died out can advantageously be changed by the fundamental voltage distance dU, to a value dU a . When this embodiment 10 c of an adaptive comparator circuit according to the invention is used in an acoustic distance sensor, the sensitivity of the distance sensor that is achieved during the quiescent phase can thus be changed and, in particular increased. This is advantageous for many applications such as, for example, for the detection of small objects. The first voltage divider can be an adjustable potentiometer whose pick-up is connected to the switch output. c) A sixth resistor 16 is connected between the control output 2 a of the signal transmitter 2 and the base 103 a , and a seventh resistor 17 is connected between the base 103 a and the first pole 4 a , which is a plus pole in the example shown. The purpose of these resistors was already explained above. d) The diode 5 is connected in series to an eighth resistor 18 which serves to slow down the discharging of the capacitor 6 and thus to reduce the speed with which the threshold voltage U ref1 is adapted to the voltage signal U signal . The eighth resistor also serves to smooth the course of the threshold voltage U ref1 with respect to very rapid fluctuations of the voltage signal U signal . FIG. 12 shows another preferred embodiment 10 d of the invention, which differs from the adaptive comparator circuit 10 c shown in FIG. 11 in that, before the voltage signal U signal reaches the first signal input 1 b , it passes through an impedance transformer. This embodiment is especially advantageous when the voltage signal arriving at the electric terminal 8 is high-ohmic, whereby said voltage signal can be the envelope curve voltage generated, for example, by an envelope curve shaper. FIG. 13 shows another preferred embodiment 10 e of the invention which differs from the adaptive comparator circuit 10 a shown in FIG. 6 as follows: a) The first switch is a transistor 103 . The base 103 a of the transistor 103 serves as the control-current terminal, the emitter 103 b serves as the switch input and the collector 103 c as the switch output. b) A second comparator 20 is used that has a second threshold value input 20 a and a second signal input 20 b , and that emits a second switching signal S 2 when a greater voltage is present at the second signal input 20 b than at the second threshold value input 20 a. c) The first pole 4 a of the first voltage source 4 a , 4 b is connected to the second threshold value input 20 a via a third resistor 13 . The second threshold value input 20 a is connected to the second pole 4 b via a fourth resistor 14 , so that the third and the fourth resistors 13 , 14 form a second voltage divider which can be configured as a potentiometer. d) The collector 103 c that serves as the switch output and the second threshold value input 20 a are connected via a fifth resistor 15 . e) A second switch 23 is connected in series to the diode 5 and the former can be regulated or activated by the second switching signal S 2 in such a way that it is in the closed state when the second comparator 20 emits the second switching signal S 2 , and otherwise it is in the open state. The second switch 23 can be a transistor. f) A sixth resistor 16 is connected between the control output 2 a of the signal transmitter 2 and the base 103 a , and a seventh resistor 17 is connected between the base 103 a and the first pole 4 a , which is a plus pole in the example shown. The purpose of these resistors was already explained above. g) The diode 5 is connected in series to an eighth resistor 18 . The purpose of this resistor was already explained with reference to FIG. 11 . h) Before reaching the first signal input 1 b , the voltage signal passes through an impedance transformer 21 whose purpose was already explained with reference to FIG. 12 . Due to the second voltage divider 13 , 14 , a voltage U ref2 is present at the second threshold value input 20 a . As long as the second switch 23 is closed, the first threshold voltage U ref1 follows the dropping voltage signal U signal , parallel-offset by the fundamental voltage distance dU, as explained above. According to the invention, however, the second switch opens as soon as the voltage signal U signal exceeds the second threshold voltage U ref2 . As a result, the discharge of the capacitor 6 and thus the dropping of the first threshold voltage U ref1 are ended. Hence, in this embodiment of the invention, the capacitor 6 can only discharge until the first threshold voltage U ref1 has dropped to the magnitude of the second threshold voltage U ref2 supplied by the second potentiometer 13 , 14 . Thus, through the use of the second voltage divider 13 , 14 , the voltage distance by which the first threshold voltage is advantageously changed parallel-offset after the decay of the voltage signal U signal with respect to voltage signal U signal , from the fundamental voltage distance dU to a value dU b . The second voltage divider 13 , 14 can be an adjustable potentiometer whose pick-up is connected to the second threshold value input 20 a. The fifth resistor 15 is connected between the switch output 103 c and the second threshold value input 20 a . Therefore, the magnitude of the first threshold voltage U ref1 counters the voltage that is supplied by the second voltage divider 13 , 14 . In particular, through the use of the fifth resistor 15 , it can additionally be achieved that the voltage distance by which the first threshold voltage is parallel-offset with respect to the voltage signal U signal strives towards a value dU b that—depending on the voltage supplied by the second voltage divider 13 , 14 —can advantageously be not only larger but also smaller than the fundamental voltage distance dU. When this embodiment 10 e of an adaptive comparator circuit according to the invention is used in an acoustic distance sensor, the sensitivity of the distance sensor that is achieved during the quiescent phase can thus be either increased or decreased by means of the second voltage divider 13 , 14 . All of the embodiments of an adaptive comparator circuit according to the invention and thus also the embodiments 10 b , 10 c , 10 d and 10 e illustrated in FIG. 6 and FIGS. 11 to 13 can be used according to the invention in the acoustic distance sensor shown in FIG. 8 instead of the adaptive comparator circuit 10 a shown there by way of an example.
Fuel injector and method for installing a fuel injector in a valve seat
A fuel injector, which, at its inflow-side end, has an intake connector at whose outer circumference at least two sealing rings are provided, which seal the fuel injector from a valve seat. Due to an intermediate ring, the two sealing rings are axially set apart from one another in a respective ring chamber. The intermediate ring is embodied independently of the intake connector and is made up of two parts. A flexible outer ring is locked in place on an inner extruded support ring, the outer ring only being mounted once the first sealing ring has already been moved across the support ring and has been stripped off. The fuel injector is particularly suited for the use in mixture-compressing internal combustion engines having external ignition.
1-13. (canceled) 14. A fuel injector for injecting fuel into a combustion chamber of a mixture-compressing internal combustion engine having external ignition, the fuel injector having a longitudinal valve axis, the fuel injector comprising: an intake connector situated at an inflow-side end, and having at least two sealing rings provided at an outer periphery of the intake connector to seal the fuel injector from a valve seat; and an intermediate ring, wherein, via the intermediate ring, the two sealing rings are axially set apart from one another, being situated in each case in a ring chamber, the intermediate ring being embodied independently of the intake connector and including two parts. 15. The fuel injector of claim 14, wherein the intermediate ring includes an inner support ring and an outer ring. 16. The fuel injector of claim 15, wherein the inner support ring, via an inwardly projecting lip, engages in a groove of the intake connector. 17. The fuel injector of claim 15, wherein the inner support ring is made of plastic. 18. The fuel injector of claim 17, wherein the inner support ring is made of the same plastic as a plastic extrusion coat partially surrounding the fuel injector. 19. The fuel injector of claim 14, further comprising: an outwardly projecting locking bead, at an outer diameter of the support ring, at which the outer ring is able to lock into place. 20. The fuel injector of claim 19, wherein an upper locking surface of the locking bead extends largely perpendicularly to the longitudinal valve axis and a lower locking surface has a slanted arrangement. 21. The fuel injector of claim 15, wherein the outer ring is made of flexible plastic. 22. The fuel injector of claim 14, wherein the intermediate ring forms a support shoulder for at least one of the at least two sealing rings. 23. A method for installing a fuel injector in a valve seat, the fuel injector, at the inflow-side end, having an intake connector at whose outer circumference at least two sealing rings are provided, which are axially set apart from one another by an intermediate ring and which seal the fuel injector from the valve seat, the method comprising: affixing an inner support ring of the intermediate ring on a circumference of the intake connector; spreading open a first one of the at least two sealing rings, mounting it on the intake connector, moving it across the inner support ring and stripping it into a ring chamber, and mounting an outer ring on the inner support ring to complete the intermediate ring; spreading open a second sealing ring, mounting it on the intake connector and stripping it into an additional ring chamber; and subsequently inserting the fuel injector in the valve seat together with a sealing system thus mounted on the intake connector. 24. The method of claim 23, wherein the inner support ring of the intermediate ring is extruded on a circumference of the intake connector in a same extrusion procedure as a plastic extrusion coat at least partially surrounding the fuel injector. 25. The method of claim 23, wherein the outer ring of the intermediate ring is locked into place on a locking bead of the inner support ring. 26. The method of claim 23, wherein the fuel injector is secured after having been inserted in the valve seat with corresponding affixation elements provided on the valve seat and on the fuel injector.
<SOH> BACKGROUND INFORMATION <EOH>At the outer periphery of various fuel injectors, a plurality of sealing elements in the form of sealing rings may be arranged in succession so as to ensure an effective sealing of the fuel injector from a valve seat. For instance, German Patent Application No. 28 27 789 refers to providing two sealing rings at a stepped inflow connector of the fuel injector. These may be embodied as whole ring stacks, which then include a stuffing-box ring, an o-ring, an asbestos ring cord, and a plastic fusion ring arranged in immediate succession. German Patent Application No. 28 27 878 refers to fuel injectors whose sealing is accomplished via sealing stacks, additional rings, having so-called emergency-sealing properties, being provided in addition to the actual sealing rings in o-form. Furthermore, German Patent Application No. 28 27 850 refers to spacing two sealing rings at the periphery of the fuel injector by using a shoulder that radially projects beyond the outer circumference of the inflow-connecting piece. Furthermore, a sealing system on a fuel injector is referred to in German Patent Application No. 37 03 615, in which two sealing rings in each case surround one of two connecting pieces independently from one another and appropriately seal it from a fuel-supply line. Moreover, German Patent Application No. 197 35 665 refers to a fuel-injection system in which a fuel injector is connected via an intermediate piece to a valve-seat connector of a fuel-supply line and sealed therefrom. In this arrangement, both the inflow connector of the fuel injector and also the intermediate pieces each have a sealing ring at the outer circumference, which spatially are thus clearly located at a distance from one another.
<SOH> SUMMARY OF THE INVENTION <EOH>The exemplary fuel injector according to the present invention provides a sealing system having two sealing rings at its outer periphery, which may be especially easy to install, avoid excessive expansion and excessive spread of the sealing rings and may therefore basically avoid damage of the sealing rings. Nevertheless, it may guarantee sufficiently large support shoulders for the sealing rings and completely prevent the release of hydrocarbons into the environment, in compliance with ever stricter environmental demands. Of particular advantage may be the simple handling of the two partial components of the intermediate ring. While the inner support ring may be installed in a very simple manner in a groove on the intake connector of the fuel injector by way of an inwardly projecting lip, the outer ring may be snapped into place on the support ring in an uncomplicated and reliable manner. The locking bead on the support ring used for locking is arranged such that the high compression forces of the one sealing ring during the installation of the fuel injector in the receiving socket are absorbed by an upper horizontal locking surface. The lower axial demounting forces of the other sealing ring are sufficiently absorbed by a lower locking surface of the locking bead, which has a slanted configuration. The exemplary method according to the present invention for installing a fuel injector in a valve seat may have the advantage that the overall installation may be performed in a fully automatic manner. In an especially advantageous manner, the sealing rings are handled very gently during the mounting, so that damage to the sealing rings is excluded. It may be especially advantageous that the sealing rings, despite being inserted in separate ring chambers, are barely spread open, so that there is no need to stretch them to a dangerous extent. Because the intermediate ring is arranged or configured in two parts, it is ensured that the first sealing ring is able to be installed without being overstretched, yet sufficiently large support shoulders are subsequently available for both sealing rings.
PROCESS FOR THE PRODUCTION OF GRAIN ORIENTED ELECTRICAL STEEL STRIPS
A process for the production of grain oriented electrical Fe—Si strips in which a Si-containing alloy is directly cast as a strip between 2.5-5.0 mm thick and cold rolled in one stage, or in more stages with intermediate annealing, to a final thickness of between 0.15-1.0 mm. The strip is then continuously annealed to carry out the primary recrystallization and then annealed to carry out the oriented secondary recrystallization. The process further includes that after solidification of the strip, and before its coiling, a phase transformation from Ferrite to Austenite is induced into the metal matrix for a volume fraction between 25-60%, obtained by controlling the alloy composition so that the Austenite fraction is allowed within the stability equilibrium between the two phases. The strip is then deformed by rolling in-line with the casting step to obtain a deformation higher than 20% in the temperature interval 1000-1300° C.
1-8. (canceled) 9. A process for the production of electrical grain oriented Fe—Si strips in which a Si-containing molten alloy composition is directly cast as continuous strips 2.5 to 5 mm thick, cold rolled in one step or more steps with intermediate annealing to a final thickness of between 1 and 0.15 mm, the strip being then continuously annealed to carry out oriented secondary recrystallisation, characterised in that after the strip solidification and before coiling of said strip in a coiling phase, a ferrite to austenite transformation is induced in the metal matrix via deformation, by rolling said strip between two cooled rolls to obtain a deformation over 20% in a temperature range of 1000-1300° C., thereby producing a volume fraction of austenite to be between 25 and 60%, said molten alloy composition being chosen such that said volume fraction of austenite is stable in a temperature interval of between 1100 and 1200° C. 10. The process according to claim 9, in which between the rolling phase and the coiling one, the strip is held between 1100 and 1200° C. for at least 5 s. 11. The process according to claim 9, in which the as-solidified strip thickness is comprised between 1.5 and 4.0 mm and after the rolling phase the strip is quenched to obtain a volume fraction of martensite comprised between 5 and 15%. 12. The process according to claim 9, in which before cold rolling the strip is annealed at a maximum temperature of 1200° C. 13. The process according to claim 12, in which after said annealing the strip is continuously quenched from a temperature comprised between 750 and 950° C. down to 400° C. in less than 12 s. 14. The process according to claim 9, in which the cast alloy comprises 2.5-5.0 wt % Si, 200-1000 ppm C, 0.05-0.5 wt % Mn, 0.07-0.5 wt % Cu, less than 2 wt % Cr+Ni+Mo. Less than 30 ppm O, less than 500 ppm S+Se, 50-400 ppm Al, less than 100 ppm N. 15. The process according to claim 9, in which in the alloy at least an element is added chosen in the group consisting of Zr, Ti, Ce, B, Ta, Nb, V, Co. 16. The process according to claim 9, in which in the alloy at least an element is added chosen between Sn, Sb, P, Bi. 17. A process for the production of grain oriented electrical Fe—Si strip, in which a Si-containing alloy is directly cast as continuous strip 2.5 to 5 mm thick, in-line hot-rolled and then cold-rolled in one step or more steps, with intermediate annealing, to a final thickness of between 1 and 0.15 mm, the cold-rolled strip being, then continuously annealed to carry out primary recrystallisation and subsequently again annealed to carry out secondary recrystallisation, characterized in that the Si-containing alloy composition is selected to induce in the alloy, during the hot-rolling step in which a deformation rate of at least 20% is utilized in a temperature interval of between 1000 and 1300° C., a ferrite to austenite phase transformation with an austenite volume fraction of between 25 to 60% stable in a temperature interval of between 1100 to 1200° C. 18. The process according to claim 17, in which between the hot-rolling and the coiling steps the strip is held in the temperature range of 1100 to 1200° C. for at least 5 s. 19. The process according to claim 17, in which an as-cast strip 2.5 to 4 mm thick is in-line hot-rolled and the thus obtained hot-rolled strip is quenched to obtain a volume fraction of martensite comprised between 5 and 15%. 20. The process according to claim 17, in which before cold-rolling the strip is annealed at a maximum temperature of 1200° C. 21. The process according to claim 20, in which, after said annealing, the strip is continuously quenched from a temperature of between 750 and 950° C. down to 400° C. in less than 12 s. 22. The process according to claim 17, in which the cast alloy comprises 2.5-5.0 wt % Si, 200-1000 ppm C, 0.05-0.5 wt % Mn, 0.07-0.5% Cu, less than 2.0% Cr+Ni+Mo, less than 30 ppm O, less than 500 ppm S+Se, 50-400 ppm Al, less than 100 ppm N. 23. The process according to claim 22, in which in the alloy at least an element is added chosen in the group consisting of Zr, Ti, Ce, B, Ta, Nb, V, Co. 24. The process according to claim 22, in which in the allot at least an element is added chosen between Sn, Sb, P, Bi.
<SOH> FIELD OF THE INVENTION <EOH>Present invention refers to the production of grain oriented electrical steel strips havig excellent magnetic characteristics, dedicated to the production of transformer cores. More precisely, rhe invention refers to a process in which a Fe—Si alloy is continuously cast directly as strip and, before coiling, the strip itself is continuously deformed by rolling to induce the formation in the metal matrix of a given fraction of Austenite, controlled as amount and distribution, thus obtaining a strip micro-structure stably and uniformly recrystallised before cold rolling.
<SOH> SUMMARY OF THE INVENTION <EOH>The aim of present invention is to solve the inconveniences due to the quality of electrical steel strips deriving from strip casting. Thus, it is an object of present invention a process for the production of electrical steel strips in which, through an in-line thickness reduction of the strip between casting and coiling stations, a significant level of recrystallisation by means of phase transformation is induced, thus normalising the crystalline structure before cold rolling, so that possible fluctuations in the process conditions are substantially non-influent with respect to the quality of the final product. Another object of present invention is to make it possible to industrially produce grain oriented electrical steel strips having excellent magnetic characteristics and constant quality, the process being stable and simplyfied with respect to the conventional processes currently utilised. Further objects of present invention will be evident from the following description of the invention. detailed-description description="Detailed Description" end="lead"?
Generic architecture for adaptable software
This invention concerns adaptable software. In particular it concerns a method for constructing an adaptable soft-ware application. A first step involves instantiating a first set of knowledge elements (kitems) related to the type of business objects or operations which will be handled by the adaptable software application, from a knowledge application development environment identifying the business needs of the application and used to produce dynamic and flexible templates. A second step involves instantiating a second set of kitems related to business objects, from the first set by specifying parameters to determine the features of the business objects. In a further aspect it concerns an adaptable software application comprising at least two levels of instantiation, where the templates for the second level instantiations are the first level instantiations.
1. A method for constructing an adaptable software application, comprising the steps of: instantiating a first set of kitems related to the type of business objects or operations which will be handled by the adaptable software application, from a knowledge application development environment identifying the business needs of the application; and, instantiating a second set of kitems related to business objects, from the first set by specifying parameters to determine the features of the business objects. 2. A method according to claim 1, where the step of instantiating the second set of kitems occurs dynamically, during the use of the application. 3. A method according to claim 1, where further instantiating steps produce further sets of kitems from the immediately preceding set. 4. A method according to claim 1, comprising the further step of directing enquiries against kitems at any level of instantiation using a consultation process. 5. A method according to claim 1, comprising the further step of accessing the business objects using a consultation process during use of the application. 6. A method according to claim 1, comprising the further step of accessing kitems related to the type of business objects for the purposes of inspecting or managing the design of the application. 7. An adaptable software application, comprising: at least two levels of instantiation, where the templates for the second level instantiations are the first level instantiations. 8. An application comprising adaptable, intelligent modules or agents that communicate with each other, where each module has its business logic defined and implemented using the adaptable system of claim 7. 9. An application according to claim 8, further comprising adaptable meta-module to define and implement the logic between the other modules. 10. An application according to claim 9, where the meta-module monitors a part of, or the whole chain of modules.
<SOH> BACKGROUND ART <EOH>An application is a software package designed for the manipulation, management and processing of business elements or objects. These are often, but not exclusively, documents. Manipulation, management and processing covers operations such as definition, access, editing, display, distribution, etc. Such applications are normally hard-coded using standard programming techniques. The knowledge of the business requirements to be met by the application is designed into the architecture of the application and the code itself. The consequence of this approach is that systems are difficult to design and develop. They are also difficult to maintain and modify. When change is required to software, due to changing business conditions for example, designers must carry out extensive redesign and coding. In many cases, the effort is so difficult and time consuming that it is not attempted. Another approach is to develop an expert system in which the business rules are separate from the hard-coded part of the application. This has the advantage that the business rules can, in principle, be changed without having to change the application. In practice, the process of entering and testing the rules is difficult and time consuming, and their reliability can only be ascertained through extensive testing. Expert systems are used to build only these applications for which knowledge is explicit and available, and where the effort required to enter and maintain the rules is clearly offset by big savings during operations. The difficulties associated with developing, implementing and maintaining expert systems are well documented in the expert system literature of the past twenty years. Few applications use expert systems technology; it is a niche technology. A further approach is to build knowledge applications as described in co-pending Australian patent application PR2152 entitled Generic Knowledge Agents, International patent application PCT/AU99/00501 entitled Generic Knowledge Management System, and International patent application PCT/AU01/01155 entitled Intelligent Courseware Development and Delivery. The contents of these three applications are incorporated into this specification by reference. A computerised generic knowledge management system, comprises: a multi-dimensional global space within computer memory defined by attributes, where each attribute defines a feature of the external world or the internal state of the system, or actions that can be taken to modify them, and each attribute is a dimension of the global space; a source space, within the global space, made up of selected ones of the attributes to define a context in which to state problems; a destination space, within the global space, made of selected ones of the attributes to define a context in which to provide answers to problems stated in the source space; mappings between defined parts of the source space which each represent one or more stated problems, to defined parts of the destination space which each represent one or more answers expressing and embodying knowledge supplied by experts appropriate to the respective problems stated in the part of the source space. The process for building knowledge applications starts in a knowledge application development environment where the business needs of the application determine the knowledge elements, or ‘kitems’, to be instantiated, or created, from contexts and templates. The outcome is an application or knowledge base in which the kitems are regions, knowledge items etc in the application—one can think of these kitems as documents. A knowledge application is typically characterised by a single instantiation process for all the kitems. The kitems can also be used as contexts or templates from which further kitems can be instantiated. Once the knowledge application is built the kitems can be accessed and managed, that is edited, displayed, distributed, etc. This entails defining an enquiry in a consultation process that retrieves the knowledge elements of interest for perusal and further manipulation. The enquiry is itself a kitem instantiated from a kitem template. Although designed and implemented in a radically different way from traditional expert systems, the business role of these knowledge applications and the way knowledge is accessed are similar to that of expert systems.
<SOH> SUMMARY OF THE INVENTION <EOH>The invention, in a first aspect, is a method for constructing an adaptable software application, comprising the steps of: Instantiating a first set of kitems related to the type of business objects or operations which will be handled by the adaptable software application, from a knowledge application development environment identifying the business needs of the application. Instantiating a second set of kitems related to business objects, from the first set by specifying parameters to determine the features of the business objects. The first level of knowledge and the corresponding instantiations in effect produce dynamic templates that work as dynamic adaptations to the needs of users. Alternatively, these templates can be activated manually. The business objects in an application are treated as knowledge elements that embody business knowledge, such as knowledge about events that took place, or are expected to take place, in the business. The business objects are easily created, and can be easily manipulated. Because business objects are knowledge elements, they can be easily managed, that is accessed, displayed, analysed, etc. Instantiating the second set of kitems may take place dynamically, during the use of the application. Applications can be built with a greater number of cascades or instantiations larger than two. Enquiries can be directed against kitems at any level of instantiation using a consultation process. The business objects may be accessed using a consultation process during use of the application. The kitems related to the type of business objects may be accessed for inspecting and managing the design of the application. The invention, in a second aspect, is an adaptable software application, comprising: at least two levels of instantiation, where the templates for the second level instantiations are the first level instantiations. The use of a two level instantiation, or cascading, design enables the first level to define the application without hard-coding, and the second to define the documents that are handled by the application. So, adaptable software transforms the definition of the application into specifying a knowledge application, and treats its documents (or business objects) as knowledge items inside the application. The application may involve adaptable, intelligent modules or agents that communicate with each other, where each module has its business logic defined and implemented using the adaptable architecture of the system. This means that each module can be quickly developed and that its functionality can evolve with the business and with experience. Each adaptable module implements the knowledge about the best way to run that module. These modules are used to facilitate development and to increase the functionality and adaptability of the solution. An adaptable meta-module may define and implement the logic between the other modules. In addition the meta-module may monitor a part of, or the whole chain of modules. This software is able to adapt itself automatically using the knowledge entered into it, without programming, to determine when and how it should adapt; that is, modify its behaviour. It may also adapt dynamically to the needs of users as software is being used. Adaptablability is a very important commercial feature for software. It holds the promise of speeding up software development, implementation, customization and flexibility. In addition to RAD (Rapid Application Development), adaptable software holds the promise of software that can be modified easily during all stages of its life-cycle, without major effort or redesign. Adaptable software also holds the promise of software that can adapt itself, that is customize itself and evolve as needs change while it is being used. Adaptable software has the potential to save, worldwide, enormous sums in development costs, and to reduce the total cost of ownership of software solutions over their lifetimes. Using adaptable software generic applications can be quickly produced. The frameworks for a variety of generic applications, for a variety of domains, can be quickly produced. An application framework is a knowledge system in which the knowledge elements express the knowledge about what the application is meant to do. Also client specific applications can be quickly produced. Knowledge in generic framework can be easily modified or added to, so as to represent the knowledge about a client's specific requirements.
Generic knowledge agents
This invention concerns a generic knowledge software agent, or kuark, existing within a knowledge environment. The kuark comprises: a defined domain of discourse within the environment which determines a source context of information detectable by the kuark, and a destination context of information identifiable by the kuark. The kuark has a defined source region, or pattern, within the and problems source context and a mapping between that source region and a defined destination region, or pattern, within the destination context. The kuark detects all environmental parameters compatible with the source region and source context so that when the source region is satisfied the mapping fires to determine the destination region in the destination context and makes it available to the environment where it can be observed by a user. It also concerns systems or kitems, made up of kuarks and a process for constructing them. It also concerns mult-kitem systems and searching processes for those systems. Finally it also concerns software agents and a knowledge management system comprising kitems.
1. A generic knowledge software agent, or kuark, existing within a knowledge environment, and comprising: a defined domain of discourse within the environment which determines a source context of information detectable by the kuark and a destination context of information identifiable by the kuark; the kuark having a defined source region, or pattern, within the source context and a mapping between that source region and a defined destination region, or pattern, within the destination context; and the kuark detects all environmental parameters compatible with the source region and source context so that when the source region is satisfied the mapping fires to determine the destination region in the destination context and makes it available to the environment where it can be observed by a user. 2. A software agent according to claim 1, and further comprising: a univeral id; a title; an explanation, which may be multimedia; a time and date created; an author; a time and date when last modified; a person who last modified agent; a version number; a status to an enquiry, such as available, candidate, rejected, definite; a description of the source context at time of last firing; a description of destination context after last firing; or, links to other kuarks. 3. A software agent according to claim 2, where the links are: the hierarchical parent's universal id, and more than one parents could be allowed; the id of the previous version kuark; the ids of the kuarks that are part of this kuark; or, the ids of the kuarks this kuark belongs to or is associated with. 4. A software agent according to claim 1, operating to: create a daughter/parent kuark; adopt an existing kuark as daughter or parent; display an internal state (contexts, regions, mapping); or make a copy each time it fires. 5. A software agent according to claim 1 where, depending on the the state of the environment with respect to the source context and source region, the kuark fires a relevance mapping that determines the relevance of the environment. 6. A software agent according to claim 1 where, the mapping is a program or procedure that takes as input some elements of the source context and produces, as output, a pattern that fits within the destination context, and. the mapping executes when the environment is compatible with the source context and the region. 7. A software agent according to claim 1, implemented as classes and objects using an object oriented language. 8. A system, or kitem, comprising more than one software agent according to claim 1, where the kitem has its own properties and methods in addition to those of its components 9. A process for constructing a system, or kitem, according to claim 8 from its components, comprising the steps of: concatenating their source contexts; concatenating their destination contexts; concatenating their source regions; concatenating their destination regions; concatenating their mappings; concatenating their explanations. 10. A process according to claim 9, comprising the additional step of building a list of the component kuarks or kitems. 11. A system, or kitem, according to claim 8, where all its components (kuarks or kitems) keep their individual contexts. 12. A system, or kitem, according to claim 8, where all its components have their individual contexts concatenated. 13. A system, or kitem, according to claim 8, expressed as an object containing another object. 14. A multi-kitem system comprising two or more systems, or kitems, according to claim 8, where the kitems are linked to one another. 15. A multi-kitem system according to claim 14, comprising a serial arrangement of kitems in which the destination context of one kitem is used as at least part of the source context of another. 16. A multi-kitem system according to claim 14, comprising a parallel arrangement of kitems in which the kitems that have independent (or disjointed), but can also have overlapping, source contexts. 17. A kitem according to claim 8, including within itself a multi-kitem system and having access to all the properties and methods of kitems contained within it, in addition to the properties that belong only to that kitem. 18. A kitem according to claim 15, comprising two kuarks, where the first kuark, depending on the the state of the environment with respect to the source context and source region, fires a relevance mapping that determines the relevance of the environment, and the second kuark uses the output of the first kuark as its source context and region, and a has a mapping which is a program or procedure that produces material to be displayed as output when the relevance mapping fires. 19. A kitem according to claim 15, comprising two kuarks, where the first kuark, depending on the the state of the environment with respect to the source context and source region, fires a relevance mapping that determines the relevance of the environment, and the second kuark uses the output of the first kuark as part of its source context and its source region, and a has a mapping which is a program or procedure that produces a destination region as output when the relevance mapping fires. 20. A searching process for identifying those kitems in a multi-kitem system according to claim 14, that have source regions compatible with an enquiry, the process comprising the step of: determining the ‘relevance status’ of a kitem with respect to the enquiry or consultation so far, where an available status is the default status of a kitem before a search takes place, a candidate kitem is one that is not incompatible with the enquiry so far, and a rejected kitem is one that is incompatible with the enquiry so far and a definite kitem is one for which the enquiry satisfies the source region. 21. A searching process according to claim 20, where the source context is organised as a tree structure, and each kitem is indexed against the context tree so that each object in the context possesses a list of the kitems it is associated with, and during the search questions are context objects and all the indexed kitems to the questions asked and answered so far in the consultation have their relevance status checked and modified if appropriate. 22. A searching process according to claim 21, where the tree structure is a modified context space kitem, and the enquiry process instantiates a consultation kitem at each iteration, which contains the list of the linked kitems affected by the enquiry, with their states. 23. A searching process according to claim 20, where the source context is organised as a tree structure in which each element is a kitem and the search process is also a kitem which flags the kitems in the tree structurre that correspond to the questions answered so far. 24. A searching process according to claim 23, where context tree element determines whether all the indexed elements have taken into account the values of the context tree element, and if so, it passes the value(s) to the indexed kitems and waits until all the responses have been received before the next step in the enquiry can proceed. 25. A searching process according to claim 24, where the tree structure is a modified context space kitem, and the enquiry process instantiates a consultation kitem at each iteration, which contains the list of the linked kitems affected by the enquiry, with their states. 26. A searching process according to claim 20, where the source region determines the state of the kitem's relevance status and, depending on its state, the mapping becomes applicable. 27. A searching process according to claim 20, where each kitem has a unique ID or address that determines its location in a distributed memory space or network, it also possess a set of applicability addresses that correspond to the kitems in its source region, and during processing, the enquiry process submits each answer to all the kitems which have, in their list of applicability addresses, the ID of the kitem that formed the question. 28. A searching process according to claim 27, where each kitem determines by itself whether it can fire or not: the enquiry process submits its answers to the network; the answers reach the kitems that have the corresponding addresses in their applicability list (the kitems that form the source region); the kitems update their relevance status with respect to this enquiry; if the relevance status is definite or candidate, the kitem sends its ID and its status to the enquiry process; and the enquiry process, when submitting, sends its (return) address, an enquiry number and an enquiry step number. These numbers are returned by the kitems that reply and this enables the enquiry process to know what replies correspond to what enquiry, or enquiry step. 29. An agent comprising a kitem according to claim 15, and having the following structure: Beliefs, Desires and Intentions are parallel kitems and their respective components are also kitems; and the Beliefs, Desires and Intentions kitems are organised in a serial architecture, that is the destination and output region of the Beliefs kitem map on the source and source region of the Desires kitem; the destination and output region of the Desires kitem map on the source and source region of the Intentions kitem. 30. A knowledge management system, comprising: a context element is a display kitem according to claim 18; a source or destination context kitem is a parallel display kitem, and its members comprise the list of all the context elements defined in the domain of discourse; a context executable kitem to edit a context element, edit a context space or display a context space as a tree; a source or destination context tree as an executable kitem, where the display kitem display the elements that belong to the source and destination kitems, its source context is the source or destination kitem and its destination context is the visual tree-like representation of the context; a knowledge editing kitem is an executable kitem that has: as source space: the source and destination contexts, as destination space: the source and destination contexts, as source region a subset of the source context that defines the applicability of the knowledge kitem, as mapping a link to a subset of the destination context, or a procedure or executable (sub-)kitem, as destination region a subset of the destination context that defines the outcome of the mapping, or the outcome of the procedure, and an explanation; a search kitem is an executable kitem with: as source context the source and destination contexts, as source region the enquiry as defined so far, as destination context the kitems in the domain of discourse (if known), as mapping: the search process that identifies the candidate and definite kitems with respect to the enquiry, as destination region the candidate and definite kitems ‘retrieved’ by the mapping; an enquiry kitem is an executable kitem with: as source space the enquiry (the consultation process, showing each step in the question-answer session), as destination space the status of the enquiry (answered, not answered, etc.), the ids of the definite kitems; a display kitem is an executable kitem with: as source space: the kitems to be displayed (itself another kitem); the display parameters; as destination space: the code that puts the kitems on the screen in a hierarchical way.
<SOH> BACKGROUND ART <EOH>The model for the agents uses the inventions described in International Patent Application No. PCT/AU99/00501 entitled ‘Generic Knowledge Management System’ (GKMS), and Australian provisional patent application no PR0852 entitled ‘Networked Knowledge Management And Learning’ (NKML) both of which are incorporated herein by reference. A summary of these knowledge models will now be given with reference to FIGS. 1 to 4 . A computerised generic knowledge management system, comprises: a multi-dimensional global space within computer memory defined by attributes, where each attribute defines a feature of the external world or the internal state of the system, or actions that can be taken to modify them, and each attribute is a dimension of the global space; a source space, within the global space, made up of selected ones of the attributes to define a context in which to state problems; a destination space, within the global space, made of selected ones of the attributes to define a context in which to provide answers to problems stated in the source space; mappings between defined parts of the source space which each represent one or more stated problems, to defined parts of the destination space which each represent one or more answers expressing and embodying knowledge supplied by experts appropriate to the respective problems stated in the part of the source space. Knowledge is mapped as regions (or patterns) in a source (or problem) space that are linked to regions (or patterns) in a destination (or solution) space, as illustrated in FIG. 1 . The interactions for defining the problem and solution spaces, and for defining the mappings, and the interactions for accessing the knowledge take place in a network of client-servers, as illustrated in FIG. 2 . FIGS. 3 and 4 show the decision-making process and the knowledge capture process, and how knowledge can be improved on an on-going basis. FIG. 4 is an extension of FIG. 3 in which the consequences of the decisions taken by a GKMS are evaluated and used to qualify the knowledge in the GKMS. Once again it involves signal exchanges between servers and clients. The accepted architecture for agents is summarized in FIGS. 5 and 6 . FIG. 5 illustrates the BDI model, that is agents have Beliefs, Desires and Intentions. FIG. 5 shows that in each part of the BDI model, agents need knowledge to be able to operate. They must have belief which is knowledge about what their role in existence is. Their desires or goals represent knowledge about what they are capable of and their intentions represent the knowledge about implementing the goals. Similarly, each of the DBI boxes in FIG. 5 is made of components, each representing a kind of knowledge in its category. All these different types of knowledge can be easily and conveniently represented using the GKMS model for knowledge. In addition, the use of that knowledge can be supported by the knowledge processing described in the GKMS patent. This means that the advantages of the GKMS model for knowledge representation and processing can become available to agents. It is also important to note that the use of the GKMS knowledge model for representation and processing is not restricted to the categories of knowledge shown in FIG. 5 , but to any knowledge that may be useful in agents. FIG. 6 shows how an agent situated in an environment interacts with this environment. Th interaction is very similar to FIGS. 3 and 4 which show how interaction with the environment and learning can take place in the GKMS model. Therefore the GKMS model is also suitable for dealing with agents interacting with their environments and learning in the process.
<SOH> SUMMARY OF THE INVENTION <EOH>The invention is a generic knowledge software agent, or kuark, existing within a knowledge environment, and comprising: a defined domain of discourse within the environment which determines a source context of information detectable by the kuark and a destination context of information identifiable by the kuark; the kuark having a defined source region, or pattern, within the source context and a mapping between that source region and a defined destination region, or pattern, within the destination context; and the kuark detects all environmental parameters compatible with the source region and source context so that when the source region is satisfied the mapping fires to determine the destination region in the destination context and makes it available to the environment where it can be observed by a user. So the kuark makes predetermined information automatically available to a user when input criteria are met. The kuark is only able to detect parameters that fall within its defined source context and when the environment satisfies the source region the mapping is triggered. The source region may be identicle to the source context, or it may be a subset within it. A kuark may comprise many other elements, such as: a univeral id; a title; an explanation, which may be multimedia; a time and date created; an author; a time and date when last modified; a person who last modified agent; a version number; a status to an enquiry, such as available, candidate, rejected, definite; a description of the source context at time of last firing; a description of destination context after last firing; and, links to other kuarks. The links implement relationships to other kuarks. The are four links: The hierarchical parent's universal id, and more than one parents could be allowed; The id of the previous version kuark; The ids of the kuarks that are part of this kuark; The ids of the kuarks this kuark belongs to or is associated with. This last link may be implemented as separate kuark(s) designed to contain the IDs of the kuarks the kuark in question relates (or belongs) to. The kuark may also operate to do the following: Create a daughter/parent kuark; Adopt an existing kuark as daughter or parent; Display an internal state (contexts, regions, mapping); and Make a copy each time it fires. It is possible to implement these methods outside the current kuark, in a separate class or subroutine for example. A first special kuark, known as a ‘relevance kuark’, determines the relevance of the environment, and depending on the the state of the environment with respect to the source context and source region, the kuark fires a relevance mapping that determines (calculates) the relevance of the environment. The relevance can be expressed as a probability. The destination context may be made of the different relevance values that the environment can take with respect to the source context and region. For example, the values could be ‘definitely relevant’, ‘possibly relevant’, ‘relevant’, ‘likely not relevant’, ‘not relevant’. Another special kuark is known as a ‘procedure kuark’, and contains as a mapping a (usually small) program or procedure that takes as input some elements of the source context and produces, as output, a pattern that fits within the destination context. This kuark executes its mapping when the environment is compatible with the source context and the region. That is, the input to the procedure is the source region in the source context. The destination context describes the range of outputs the procedure can produce when run, and the destination region or pattern is the actual result of the execution of the mapping, using an instance of the environment. Kuarks may be implemented as classes and objects using an object oriented language such as Java. A kuark is a class in OOP and some of its properties are static, final, etc. A kuark that is given values for its source and destination contexts and regions are objects or instances of the corresponding kuark class. A system made of several kuarks is still a kuark with all the allowed properties and methods, but is known as a ‘kitem’. In a kitem all the component kuark and kitems may keep their identities. The new kitem may have its own properties and methods in addition to those of its component kitems. The new kitem may have its own title and explanation, and may also have its own source and destination contexts and regions, and its own mapping. A kitem may be constructed form kuarks and kitems by the following process: concatenating the source contexts; concatenating the destination contexts; concatenating the source regions; concatenating the destination regions; concatenating the mappings; concatenating the explanations. Here concatenates means ‘take the union’ of the entities in question; for mappings, it is the juxtaposition of the individual mappings. The kitem construction process may also build the list of kuarks or kitems that are part of the new kitem. Typically, when a kitem is built, all its components (kuarks or kitems) keep their individual contexts. Alternatively the kitem construction process may be set so that all components get the same source and destination contexts, that is the concatenation of the individual contexts of the kitem components. A kuark or kitem can be represented as an object using an object oriented language such as Java. A kitem can be expressed as an object containing another object (or a class with sub-classes) which preserves all the features for knowledge items disclosed in the GKMS patent. This means that kitem objects can be generated without programming. The other features disclosed in the GKMS patent also apply to kitems implemented as classes. Kitems can be linked. A link has a source or origin kitem and a destination kitem. The links can be stored inside the kitem or kuark, as a property of the kitem. An alternative is to store inside the kitem the address of the ‘link kitem’, that is, the kitem that is designed to contain the links that relate to a source kitem. A multi-kuark or multi-kitem system may comprise a serial arrangement, in which the destination context of one kitem is used as the source context (or part of the source context) of another. More than two kitems can be chained. Alternatively, a parallel kitem is made of kitems that have independent (or disjointed), but can also have overlapping, source contexts. Inside a parallel kitem, the component kitems are treated as peers, and links are defined between peers. An application or knowledge base may comprise a set of knowledge items, kitems, defined with respect to a domain of discourse, where the kitems are arranged in parallel. Some of the kitems may be linked in series or in parallel. A kitem may be a class that supports the creation of objects, or knowledge elements, where the kitem has access to all the properties and methods of other kitems contained within it, in addition to the properties that belong only to that kitem. Such a kitem is referred to as a ‘meta-kitem’ and is able to express meta-knowledge. A display kitem is a serial kitem comprising two kuarks. The first kuark is a relevance kuark. It uses its region and the state of the environment to determines the relevance of the display kitem. The second kuark is a procedure kuark and has the relevance (the output of the first kuark) as source context and region. The material to be displayed comprises the destination region and the explanation. An executable kitem is a serial kitem comprising two kuarks. The first kuark is a relevance kuark. It uses its region and the state of the environment to determines the relevance of the executable kitem. The second kuark is a procudure kuark. It has the relevance (the output of the first kuark) as region and as part of its source context. When the relevance is not ‘null’, the second kuark executes its mapping to produce the destination region. The inputs to the executable mapping are elements of the second kuark's source context that can be additional to the relevance context. A knowledge item is a kitem comprising a source context, a source region, a destination context, a destination region, a mapping and an explanation. Searching involves identifying those kitems that have source regions compatible with the enquiry. It means determining the ‘relevance status’(available, candidate, rejected or definite) of a kitem with respect to the enquiry or consultation so far. Searches may be synchronous or asynchronous. An available status is the default status of a kitem before a search takes place; a candidate kitem is one that is not incompatible with the enquiry so far; a rejected kitem is one that is incompatible with the enquiry so far and a definite kitem is one for which the enquiry satisfies the source region. The source context may be organised as a tree structure. Each kitem may be indexed against the context tree. This means that each object in the context possesses a list of the kitems it is associated with. During processing, all the indexed kitems to the questions (context objects) asked and answered so far in the consultation have their relevance status checked and modified if appropriate. Kitems which become definite can be presented to the user, candidates remain contenders for definite status depending on future answers, and rejected items will not be considered any longer. At the implementation level, the context tree is a modified context space kitem. The enquiry process instantiates a consultation kitem at each iteration, which contains the list of the linked kitems affected by the enquiry, with their states. These kitems affected are the available or candidate kitems in the knowledge base after the previous iteration, or all the kitems in the knowledge base is the enquiry searches the whole database at each iteration. The search process updates the status of the kitems according to the enquiry. Alternatively, the search process (also a kitem) may flag the elements in the context tree that correspond to the questions answered so far. Each context tree element is itself a kitem and its role is to determine whether all the indexed elements have taken into account the values of the context tree element. If they do this, it passes the value(s) to the indexed kitems and waits until all the responses have been received. The search process waits until all the tree elements corresponding to the enquiry so far have replied. When all have replied, the enquiry has been processed (the knowledge elements have determined their status based on the enquiry so far). Synchronisation is assured and the next step in the enquiry (presenting the results to the user and waiting for its input) can proceed. At the implementation level, the context tree is a modified context space kitem. The enquiry process instantiates a consultation kitem at each iteration, which contains the list of the linked kitems affected by the enquiry, with their states. These kitems affected are the available or candidate kitems in the knowledge base after the previous iteration, or all the kitems in the knowledge base is the enquiry searches the whole database at each iteration. Each kitem updates its status according to the enquiry and informs the searc process. Alternatively, in asynchronous searches, the source region may determine whether the outcome of the kitem becomes applicable. That is, the source region determines the state of the kitem's relevance status and, depending on its state, the mapping becomes applicable. The state of the relevance region determines what happens to the application region. Where each kitem has a unique ID or address that determines its location in a distributed memory space or network, it may also possess a set of applicability addresses that correspond to the kitems in its source region. During processing, the enquiry process may submit each answer to all the kitems which have, in their list of applicability addresses, the ID of the kitem that formed the question. In this model, each kitem determines by itself whether it can fire or not: the enquiry process submits its answers to the network; the answers reach the kitems that have the corresponding addresses in their applicability list (the kitems that form the source region); the kitems update their relevance status with respect to this enquiry; if the relevance status is definite or candidate, the kitem sends its ID and its status to the enquiry process. The enquiry process, when submitting, sends its (return) address, an enquiry number and an enquiry step number. These numbers are returned by the kitems that reply and this enables the enquiry process to know what replies correspond to what enquiry (or enquiry step). Each kitem can decide whether to answer more than once to an enquiry from the same origin or process. It is up to the enquiry process to manage all the replies it gets from the network. The solution outlined here is applicable to distributed processing. Another model for asynchronous searches is to give even more responsibility to the distributed kitems, and each kitem has the address of each of the context objects in its context and in its region (a subset of the context). The kitem interrogates the network at regular interval to determine whether the status of the region, and hence of the applicability of the kitem, has changed. Cascading refers to a knowledge element having, as part of its destination region, an object that belongs to the source region of another knowledge element. When the first knowledge element fires, it may change its source region in a way that impact on the applicability of the second knowledge element. The second knowledge element may have to fire, not as a direct effect of the enquiry but as an indirect effect, that is, a consequence of another knowledge element having changed its state. This process can extend to any arbitrary number of knowledge elements, called cascading knowledge elements. In order to ensure that all the cascaded elements update their state if appropriate, it is necessary to a) link the cascaded elements, and b) have the search process update the relevance status of these cascaded knowledge elements (synchronous search) or c) to communicate the state of the environment (outputs of previously fired knowledge elements) to the linked cascaded knowledge elements (asynchronous search). In a multi-kitem system, knowledge processing involves running a question-answer session. The system asks questions, get answers and, on the basis of these answers, asks further questions until the correct answers are found or no answers exist. The steps involved are: identifying the most important questions (the importance is determined by the ability of the answers to the question to lead to the correct answers as quickly as possible); asking these questions; on the basis of the answers to the questions, determining whether any answers can be found, and the next best questions to ask. Identifying the most promising questions may involve a synchronous search through a context tree with processing delegated to a search agent. As the context tree is updated each time a question is considered, the most important questions to ask next depend on the consultation so far; that is, on the state of the context tree after the previous enquiry has been processed. Therefore the questions are selected dynamically. Alternatively the search may have processing delegated to each kitem. Context tree search with processing delegated to each kitem As explained above, the search process waits until all the tree elements corresponding to the enquiry so far have replied. It is the responsibility of the enquiry process to collect these replies and to count the number that come from each of the tree context element: it waits for answers to get back from the network; it counts the number of candidate kitems from each context tree element—the ones with the highest number of candidate kitems correspond to the best questions to ask. In addition, the enquiry process may perform the following operations: it can pass the kitems it received to the display kitem (that will display them according to their relerance status), this process can also be asynchronous it can build its own operational domain of discourse (source and destination context spaces) by concatenating the context spaces of all the answers received so far. In an asynchronous searches the enquiry process (that sends the enquiry over the network for kitems to process it) may perform several tasks: it waits for answers to get back from the network; it counts the number of candidate kitems from each context tree element—the ones with the highest number of candidate kitems correspond to the best questions to ask. In addition, the enquiry process may perform the following operations: it can pass the kitems it received to the display kitem (that will display them according to their relevance status), this process can be asynchronous; it can build its own operational domain of discourse (source and destination context spaces) by concatenating the context spaces of all the answers received so far. In situations where the domain of discourse of a body of knowledge is known, one can identify the kitems with the largest number of indexed kitems with relevance status available or candidate in order to find hidden knowledge. The process of producing a context tree is a compilation of the kitems in an application or domain of discourse. Compilation keeps the identity of the kitems and all the data and information in these kitems that pertain to their execution. Keeping the identity of these kitems enables the non-executable parts of the kitems to be retrieved at execution time on a ‘as needed’ basis. There are two types of compilation: Just-in-time compilation imposes the compiled kitem to take the latest version of each kitem in its process, in particular the latest version of the source and destination regions; or Permanent compilation accepts that the kitems do not change (between compilations) and that their attributes and methods can be used without checking them. Therefore they are included in the compiled kitem. This is faster than the partial compilation. An agent may be represented as a kitem with the following structure: Beliefs 61 , Desires 62 and Intentions 63 are parallel kitems (their respective components are also kitems); the Beliefs 61 , Desires 62 and Intentions 63 kitems are organised in a serial architecture. That is, the agent kitem is a serial kitem; the destination and output region of the Beliefs kitem map on the source and source region of the Desires kitem; the destination and output region of the Desires kitem map on the source and source region of the Intentions kitem. The kitems needed to produce a workable implementation for knowledge management are shown below: A context element is a display kitem (a serial kitem made of two kuarks). A source or destination context kitem is a parallel display kitem. Its members comprise the list of all the context elements defined in the domain of discourse. Context executable kitems may edit a context element, edit a context space or display a context space as a tree. A source or destination context tree is an executable kitem. The display kitem may display the elements that belong to the source and destination kitems. Its source context is the source or destination kitem and its destination context is the visual tree-like representation of the context. A knowledge editing kitem is an executable kitem that has: As source space: the source and destination contexts. As destination space: the source and destination contexts. As source region: a subset of the source context that defines the applicability of the knowledge kitem. As mapping: a link to a subset of the destination context, or a procedure or executable (sub-)kitem. As destination region: a subset of the destination context that defines the outcome of the mapping, or the outcome of the procedure. An explanation. A search kitem is an executable kitem with: as source context: the source and destination contexts; as source region: the enquiry as defined so far; as destination context: the kitems in the domain of discourse (if known); as mapping: the search process that identifies the candidate and definite kitems with respect to the enquiry; as destination region: the candidate and definite kitems ‘retrieved’ by the mapping. An enquiry kitem is an executable kitem with: as source space: the enquiry (the consultation process, showing each step in the question-answer session); as destination space: the status of the enquiry (answered, not answered, etc.), the ids of the definite kitems. A display kitem is an executable kitem with: as source space: the kitems to be displayed (itself another kitem); the display parameters; as destination space: the code that puts the kitems on the screen in a hierarchical way. This technology supports rapid application development (RAD). RAD is a very important commercial feature for software. It holds the promise of speeding up software development, implementation and customization. RAD, or any significant progress in this area, has the potential to save, worldwide, enormous sums (billion of dollars) in development, and to reduce the total cost of ownership of software solutions over their lifetimes. RAD is also a very serious technical challenge for designers. Agents hold the promise of autonomous and intelligent software that is capable of co-operating with other agents (and human beings) towards achieving a goal. In addition agents, with proper architecture, should scale up. That is, they could provide a promising platform for building complex, adaptable systems that could have knoweldge and perhaps exhibit intelligent behaviour. Currently agents are difficult to design and buid. This is because agents draw on several disciplines, each one bringing its own theoretical and practical difficulties to design and development. They are: control theory, cognitive psychology and artificial intelligence.
Lugged cap forming system
Transfer apparatus and method are provided from first (15A, 16A) to second (15B, 16B) stations of tooling in a cap (11) making press. This cap is biased against the first upper tools by a first airstream (50) introduced under the cap and moves upward with the first station punch (45). As the punch approaches its top location, a transfer airstream (52) begins while the first airstream is still on, and moves the cap out through a transfer chute (18) to the second station. The cap departs the chute and passes detents (67) on a pair of closed retention fingers (60) which define an extension of the transfer path from the chute into the open second station tools. A vacuum (85) applied to a port in the second station punch then holds the cup against the rising upper tools. When the punch clears the closing fingers and approaches its top location, an ejection airstream (87) commences to propel the finished cup via a discharge chute (19).
1. A system for forming container caps, comprising: a first station including first station tools comprising a blank punch tool and a cooperating compound die tool adapted for mounting in a reciprocating press for cyclic movement between an open position in which said tools are separated and a closed position in which said tools are closely spaced to form a cap from a metal blank separated from a supply of thin sheet metal during the closing motion of said tools and then to draw the blank into a cup shaped part with a top panel and a side wall extending from said top panel; a second station aligned with and spaced from said first station, said second station including second station tools for receiving parts from said first station and performing further operations thereon; a chute extending between said first and second stations at the open position of said first station tools to guide parts in transfer from the first station into the second station; means projecting a first airstream into the part to hold the top panel against said blank punch tool during separating motion of said first station tools upon forming of a part; means projecting a second airstream across said blank punch tool at its open position and into said chute so as to propel cap parts into said second station tools; and control means coordinated with the operation of the press for switching on said first and second airstream projecting means to (a) initiate the first airstream at the beginning of separation of said first station tools and maintaining the first airstream at least until said first station tools are fully separated, and (b) initiate the second airstream before said first station tools are fully separated and maintaining the second airstream sufficiently to propel the part through said chute. 2. A system as defined in claim 1, wherein said chute terminates adjacent the interface of the second station tools; a set of spaced apart guide fingers for forming a continuation of said chute, said fingers being movably supported on opposite sides of the second station tools for movement perpendicular to said second station tools such as to provide for closing of the second station tools with said fingers moved apart and for closing motion of said fingers into an extension of said chute when the second station tools are open. 3. A system as defined in claim 2, wherein said fingers have complementary tracks formed thereon to receive and guide a part exiting said chute, said tracks having inwardly curved end sections which align with the center of the second station tools to provide a termination of the path of a cap part entering the second station tools, and cam means operated by the press in coordination with opening and closing of the second section tools to move the fingers apart when the tools are closing and to close the fingers into a chute extension when the tools are opening. 4. A system as defined in claim 3, wherein said finger tracks include detents spaced from said curved end sections to define a centered position of the incoming parts. 5. A system as defined in claim 1 wherein said second station tools are constructed to form lugs on a rolled rim of the cap parts. 6. A system for forming container caps, comprising: a first station including first station tools comprising a blank punch tool and a cooperating compound die tool adapted for mounting in a reciprocating press for cyclic movement between an open position in which said tools are separated and a closed interface position in which said tools are closely spaced (a) to form a cap from a metal blank separated from a supply of thin sheet metal during the closing motion of said tools and (b) then to draw the blank into a cup shaped part with a top panel and a side wall extending from said top panel, said tools also form an outward rolled rim on the side wall; a second station aligned with and spaced from said first station, said second station including second station tools for receiving parts from said first station and performing a lug shaping operation on the rolled rim; a chute extending between said first and second stations at the open position of said first station tools to guide parts in transfer from the first station into the second station, said chute terminating adjacent the interface of the second station tools; a set of spaced apart guide fingers for forming a continuation of said chute, said fingers being pivotally supported on opposite sides of the second station tools for opening and closing motion perpendicular to said second station tools such as to provide for closing of the second station tools with said fingers moved apart and for closing motion of said fingers into an extension of said chute when the second station tools are open; said fingers have complementary tracks formed thereon to receive and guide a part exiting said cute, said tracks having inwardly curved end sections which align with the center of the second station tools, said finger tracks include detents spaced from said curved end sections to define a centered position of the incoming parts and to provide a termination of the path of a cap part entering the second station tools; cam means operated by the press in coordination with opening and closing of the second section tools to move the fingers apart when the tools are closing and to close the fingers into a chute extension when the tools are opening; means projecting a first airstream into the part to hold the top panel against said blank punch tool during separating motion of said first station tools upon forming of a part; means projecting a second airstream across said blank punch tool at its open position and into said chute so as to propel cap parts into said second station; and control means coordinated with the operation of the press for switching on said first and second airstream projecting means to (a) initiate the first airstream at the beginning of separation of said first station tools and maintaining the first airstream at least until said first station tools are fully separated, and (b) initiate the second airstream before said first station tools are fully separated and maintaining the second airstream sufficiently to propel the part through said chute, said control means including an input pulse generator driven in synchronism with the cyclic operation of the press to generate a train of control pulses related to the angular position of the press crankshaft as it rotates to open and close the tools, whereby the initiation and termination of said airstream projecting means is synchronized with cyclic operation of the tools. 7. A method for discharging and transferring a cup shaped object from a work station, having upper and lower tooling which is opened and closed to form the object, along a transfer path, the open position of the upper tooling defining a ready position, comprising the steps of: (a) locating the object within the work station in the ready position by directing a first stream of pressurized gas against the object to cause the object to remain with the upper tooling upon opening of the tooling whereby the object is supported by the first stream against the upper tooling; (b) prior to locating the object in said ready position, initiating a second flow of pressurized gas through orifice means located adjacent to and directed across said ready position, thereby causing the transfer of said object from the work station when the object is released at the ready position; and (c) discontinuing the second flow of pressurized gas through said orifice means after the object has exited the open tooling. 8. The method of claim 7 including the additional steps of: (d) receiving the object in a chute extending to a second work station which defines the transfer path to adjacent second upper and lower tools of the second work station, (e) directing the object into an extension of the transfer path defined by pivoting fingers located at opposite sides of the second tools, and (f) opening the fingers when the second tools are closing, and closing the fingers when the second tools are opening such as to guide the object into a centered position between the second station tools.
<SOH> BACKGROUND OF THE INVENTION <EOH>U.S. Pat. Nos. 6,082,944 and 6,015,062, assigned to the assignee of this application, disclose closure constructions for reclosable containers (e.g a can body) wherein a domed container end with a neck portion having a pour opening is provided with a reclosable lugged type of cap. That invention provides a unique and versatile container for fluids, particularly for beverages, wherein various standard can bodies are provided with a two part end including a neck with a pour opening, a lug formation on the neck below the pour opening, a reclosable cap having a lug formation which can interlock with the lug formation on the neck and including a seal surrounding the pour opening, and thus capable of maintaining product under pressure. The two part end is affixed to a can body by conventional double rolled seam attachment between the bottom of the neck and the rim of the can body. However, it is possible to affix the domed end to a can body without a cap, fill the can though the pour opening, and then apply the cap. With the possibility of expanded markets for these lugged caps, which are also useful on various jars and bottles, there is a need for a system (method and tooling) for producing lugged cap members in a single machine, e.g. a reciprocating press fitted with appropriate tooling, which is capable of precise high speed (e.g. in the range of 135 to 150 strokes/minute) to achieve acceptable commercial production of the cap. To develop such production speeds it is desirable to divide the progressive tooling operations into more than one step, and this in turn requires a rapid and precise transfer system to move the partially completed caps from a first station to a second station, and precisely register the caps in the second station. Prior art transfer systems are known for moving and registering can end shells, such as in U.S. Pat. Nos. 4,770,022 and 4,895,012, however, the end shells are relatively flat disc-like objects with a quite small height to diameter ratio, whereas the caps made by the present invention have a substantially greater height with respect to their diameter. Thus, the physical dimensions of the caps involved in this invention are quite different from can shells or easy self-opening can ends. This in turn introduces needs not required or anticipated in shell transfer systems, for example with regard to tipping of the caps during high speed transfer operations. Also, because of the relatively high press cycling, and need for precision in cap positioning and deceleration immediately after each transfer to another press station, there is a requirement for precise transfer of each cap from a first press station through high acceleration, very rapid transfer to the next press station, and high deceleration to a precisely defined stationary location at that next station.
<SOH> SUMMARY OF THE INVENTION <EOH>The present invention provides a transfer apparatus and method for a first to a second station of progressive tooling in a cap making press. The caps are of a type having substantial height with respect to their diameter. Thus a first station punch and die form a cap of generally inverted cup shape, with an outward curled rim, and a second station punch and die form lugs into that rim, requiring just two strokes of the press to sever a disc from a supply sheet, form it, and discharge a completed cap. Thus, in the first station, a cap is formed except for lugs which are added to the cap in the second station. The formed cap in the first station, which is in the nature of an inverted cup, has an outward curl formed on its lower edge during the initial up stoke of the press. This cup is then biased against the upper forming punch, by a first airstream introduced into the cavity within the underside of the cap, and moves upward in contact with the first station punch. As the punch approaches its top dead center location (tooling fully open), a second airstream begins, before the first station punch is fully raised and while the first airstream is still on. This second airstream moves the cap off the raised first station punch, and into and through a transfer chute directed toward the second station. At the second station, the cap departs the chute and passes detents on a pair of closed retention fingers which, with the tooling open, define an extension of the transfer path from the chute into the open second station tools. As the second station tools begin to close and the partially finished cap is located, the fingers are opened and the tooling operates to form lugs into the cap rim. As the second station tooling opens, a vacuum applied to a port in the second station punch begins to hold the finished cup against that punch tool, and when that punch clears the closing fingers and approaches its top dead center location, an ejection airstream commences to propel the finished cup from the tooling via a discharge chute.
Diving aid
Diving aid for divers, at least comprising a power supply, alarm sound-generating means (3) and activating means (2) for activating the sound-generating means (3), which activating means (2) are designed in such a way that when the alarm device is in use said activating means can be activated by an uncontrolled shock load, and that the device comprises means (6) for attaching to the divers's body or the divers's equipment in an unmistakable orientation. The diving aid according to the invention is preferably designed in the form to a wristwatch.
1. Diving aid for reducing the dive stress of divers, in particular pre-dive stress, at least comprising a power supply, alarm sound-generating means and activating means for activating the alarm sound-generating means, which activating means are designed in such a way that when the alarm device is in use said activating means can be activated by an uncontrolled shock load, and which device also comprises means for attaching to the diver's body or the diver's equipment in an unmistakable orientation. 2. Diving aid according to claim 1, wherein the diving aid also comprises light-signal-generating means that are connected to the activating means. 3. Diving aid according to claim 1, wherein the activating means comprise a push-button. 4. Diving aid according to claim 1, wherein the activating means comprise an acceleration detector. 5. Diving aid according claim 1, wherein the diving aid comprises water detection means by means of which the device can be activated. 6. Diving aid according to claim 1, wherein the diving aid is designed in the form of a wristwatch. 7. Diving aid according to claim 1, wherein the means for attaching the diving aid to the diver's body or the diver's equipment comprise a clip. 8. Diving aid according to claim 1, wherein the diving aid forms part of an ordinary diving computer.
Composition comprising an oxidizing and complexing compound
The present invention is related to a composition comprising an oxidizing compound and a complexing compound with the chemical formula wherein R1, R2, R3 and R4 are selected from the group consisting of H and any organic side chain. The oxidizing compound can be in the form of an aqueous solution. The complexing compound is for complexing metal ions. Metal ions can be present in the solution or in an external medium being contacted with the solution. The present invention can be used for cleaning a semiconductor substrate.
1. A composition comprising an aqueous solution, said aqueous solution comprising an oxidizing compound and a complexing compound, said complexing compound comprising a chemical formula wherein R1, R2, R3, and R4 are each independently selected from the group consisting of H and an organic side chain. 2. The composition as recited in claim 1, further comprising an alkaline compound. 3. The composition as recited in claim 1, wherein said organic side chain is selected from the group consisting of an aliphatic side chain, a heterocyclic side chain, and an aromatic side chain. 4. The composition as recited in claim 1, wherein R3 and R4 each comprise hydrogen and R1 and R2 each comprise a functionalized aliphatic side chain. 5. The composition as recited in claim 1, wherein said complexing compound is selected from the group consisting of 1,2-diethyl-3-hydroxy-4(1H)-pyridinone, 1-ethyl-2-methyl-3-hydroxy-4(1H)-pyridinone, 1-methyl-2-ethyl-3-hydroxy-4(1H)-pyridinone 1,2-dimethyl-3-hydroxy-4(1H)pyridinone, 1-propyl-2-ethyl-3-hydroxy-4(1H)-pyridinone, 1-propyl-2-methyl-3-hydroxy-4(1H)-pyridinone, 1-(2-carboxyethyl)-2-methyl-3-hydroxy-4( 1H)-pyridinone, and 1-(2-carboxyethyl)-2-ethyl-3-hydroxy-4(1H)-pyridinone. 6. The composition as recited in claim 1, wherein said oxidizing compound comprises hydrogen peroxide. 7. The composition as recited in recited in claim 2, wherein said alkaline compound comprises an inorganic basic compound or an organic basic compound. 8. The composition as recited in claim 7, wherein said alkaline compound is selected from the group consisting of ammonia and organic amine compounds. 9. The composition as recited in claim 2, wherein an amount of oxidizing compound comprises from 0.001 to 30 weight %. 10. The composition as recited in claim 1, wherein an amount of the complexing agent comprises from 0.1 to 1000 ppm of said solution. 11. The composition as recited in claim 7, wherein an amount of the inorganic basic compound or the organic basic compound comprises from 0.001 to 30 weight %. 12. The composition as recited in claim 1, wherein an amount of said oxidizing compound comprises from 0.001 to 95 weight %. 13. A method of treating a semiconductor substrate, wherein said method comprises treating a semiconductor substrate with a composition comprising an aqueous solution, said aqueous solution comprising a complexing compound comprising the chemical formula wherein R1, R2, R3, and R4 are each independently selected from the group consisting of H and an organic side chain. 14. The method as recited in claim 13, wherein said composition further comprises an oxidizing compound. 15. The method as recited in claim 13, wherein said composition further comprises an alkaline compound. 16. The method as recited in claim 13, wherein said organic side chain is selected from the group consisting of an aliphatic side chain, a heterocyclic side chain, and an aromatic side chain. 17. The method as recited in claim 13, wherein R3 and R4 each comprise hydrogen and R1 and R2 each comprise a functionalized aliphatic side chain. 18. The method as recited in claim 13, wherein said complexing compound is selected from the group consisting of 1,2-diethyl-3-hydroxy-4(1H)-pyridinone, 1-ethyl-2-methyl-3-hydroxy-4(1H)-pyridinone, 1-methyl-2-ethyl-3-hydroxy-4(1H)-pyridinone, 1,2-dimethyl-3-hydroxy-4(1H)pyridinone, 1-propyl-2-ethyl-3-hydroxy-4(1H)-pyridinone, 1-propyl-2-methyl-3-hydroxy-4(1H)-pyridinone, 1-(2-carboxyethyl)-2-methyl-3-hydroxy-4(1H)-pyridinone, and 1-(2-carboxyethyl)-2-ethyl-3-hydroxy-4(1H)-pyridinone. 19. The method as recited in claim 14, wherein said oxidizing compound comprises hydrogen peroxide. 20. The method as recited in claim 15, wherein the alkaline compound comprises an inorganic basic compound or an organic basic compound. 21. The method as recited in claim 20, wherein said inorganic basic compound or said organic basic compound is selected from the group consisting of ammonia and organic amine compounds. 22. The method as recited in claim 14, wherein an amount of the oxidizing compound comprises from 0.001 to 30 weight %. 23. The method as recited in claim 15, wherein an amount of the alkaline compound comprises from 0.001 to 30 weight %. 24. The method as recited in claim 13, wherein an amount of said complexing compound comprises from 0.1 ppm to 1000 ppm. 25. Use of a composition as recited in claim 1 in a field selected from the group consisting of soil remediation, metal etch bath, denture bleaching, bleaching of fats, bleaching of oils, bleaching of waxes, wastewater treatment, paper pulp bleaching, and textile bleaching. 26. The composition as recited in claim 8, wherein said organic amine is selected from the group consisting of tetraalkylammoniumhydroxide compounds, alkanolamine compounds, choline(hydroxyltrialkylammoniumhydroxide) compounds, and guanidine compounds. 27. The method as recited in claim 21, wherein said organic amine is selected from the group consisting of tetraalkylammoniumhydroxide compounds, alkanolamine compounds, choline(hydroxyltrialkylammoniumhydroxide) compounds, and guanidine compounds.
<SOH> FIELD OF THE INVENTION <EOH>This invention is situated in the field of products and methods for the stabilizing a composition comprising an oxidizing compound.
<SOH> SUMMARY OF THE INVENTION <EOH>In a first aspect of this invention, a composition is disclosed comprising an oxidizing compound and a complexing compound with the chemical formula wherein R1, R2, R3 and R4 are selected from the group consisting of H and any organic side chain. The oxidizing compound can be in the form of an aqueous solution. The complexing compound is for complexing metal ions. Metal ions can be present in the solution or in an external medium being contacted with the solution. Depending on the metal ion being complexed, one or more complexing molecules/metal ion are required. In an embodiment of this first aspect, said composition as recited in the first aspect of this invention can further comprise an alkaline compound. In a further embodiment of this first aspect, said organic side chain can comprise an aliphatic side chain, a heterocyclic side chain or an aromatic side chain. In a further embodiment of the first aspect of this invention, R3 and R4 can be hydrogen while R1 and R2 can be a functionalized aliphatic side chain. Preferably, said complexing compound is one of the group consisting of DEHP, 1-EMHP, 2-EMHP, DMHP, PEPH, PMHP, ECMHP and ECEHP. In a further embodiment of this first aspect, a solution as recited in the first aspect of this invention is disclosed characterized in that said oxidizing compound is hydrogen peroxide. Hydrogen peroxide will be stabilized by the addition of the complexing compound, such that decomposition is substantially inhibited. In a further embodiment of this first aspect of the invention, a composition as disclosed wherein said oxidizing compound is hydrogen peroxide. In a further embodiment of this first aspect, a composition as recited in the first embodiment of the first aspect of the invention is disclosed wherein alkaline compound comprises an inorganic or organic basic compound. Said alkaline compound can be ammonia or an organic amine, preferably an organic amine chosen from the group consisting of tetraalkylammoniumhydroxide, alkanolamine, choline(hydroxyltrialkylammoniumhydroxide) and guanidine compounds. In a second aspect of this invention, a method for treating a semiconductor substrate is disclosed. Said semicondcutor substrate is treated with a composition comprising an aqueous solution of a complexing compound with the chemical formula wherein R1, R2, R3 and R4 are selected from the group consisting of H and any organic side chain. Said composition can further comprise an oxidizing compound. Said composition can be used for treating a substrate, such that particles are oxidized and metallic contamination is removed. The complexing molecule is for complexing metallic residues being present on the substrate and in the solution. Additionally, the lifetime of the solution is increased since de decomposition of the oxidizing compound is substantially inhibited. Said solution is particularly suitable for cleaning a semiconductor substrate. Said composition for cleaning a semiconductor substrate can be any composition described in the first aspect of this invention. In an embodiment of this second aspect of this invention, said R1, R2, R3 and R4 are selected from the group consisting of H and any organic side chain. Said organic side chain can comprise an aliphatic side chain, a heterocyclic side chain or an aromatic side chain. In a further embodiment of this second aspect of the invention, R3 and R4 can be hydrogen while R1 and R1 can be a functionalized aliphatic side chain. In a further embodiment, said complexing compound is one of the group consisting of DEHP, 1-EMHP, 2-EMHP, DMHP, PEPH, PMHP, ECMHP and ECEHP. In another embodiment of the second aspect of the present invention, said oxidizing compound can be hydrogen peroxide. In an embodiment of the second aspect of this invention, said composition can further comprise an alkaline compound. Said alkaline compound can comprise an inorganic or organic basic compound. Said basic compound can be chosen from the group consisting of ammonia and organic amine, preferably an organic amine chosen from the group consisting of tetraalkylammoniumhydroxide, alkanolamine, choline(hydroxyltrialkylammoniumhydroxide) and guanidine compounds. In an embodiment of the second aspect of this invention, the method as recited in second aspect of the invention is disclosed wherein the concentration of hydrogen peroxide in the solution lies between 0.001 to 30 weight %. In an embodiment of the second aspect of this invention, the method as recited in second aspect of the invention is disclosed in which the amount of the complexing agent lies between 0.1 and 1000 ppm of said solution. In an embodiment of the second aspect of this invention, the method as recited in second aspect of the invention is disclosed wherein the amount of the alkaline compound lies between 0.001 and 30 weight %. detailed-description description="Detailed Description" end="lead"?
Level sensor
A level sensor (2) comprising a support (6) with a bearing (11) for a lever arm (3) with a float (4) and a guide rail (16) pointing in the direction of the bearing (11). The lever arm (3) has a stirrup (5) made of plastic and a lever wire (7) clipped thereto. The stirrup has two guide elements (12,13) which are arranged next to each other and which engage with the guide rail (16), thereby preventing the dual contact (10) of the stirrup (5) from rising above the slideways (9) of a potentiometer (8) which are disposed on the support (6).
1. A filling-level sensor for generating electrical signals as a function of a pivoting angle of a float-carrying lever arm for a tank of a motor vehicle, comprising: a carrier for fastening to a lateral wall in the tank; a yoke made of plastic and pivotable in a mounting of the carrier; a lever wire holding a float and fastened to the yoke; and a guide rail, arranged on the carrier, for guiding a guide element of the yoke in an intended axial position in relation to the carrier, wherein the guide rail is arranged so as to point in the direction of the mounting, and the yoke has a fork-shaped guide element which is open at an end facing away from the mounting and which is in engagement with the guide rail. 2. The filling-level sensor as claimed in claim 1, wherein the guide rail has a web-shaped configuration and is arranged parallel to the carrier. 3. The filling-level sensor as claimed in claim 1, wherein the fork-shaped guide element is arranged at the end of the yoke which faces away from the mounting. 4. The filling-level sensor as claimed in claim 1, wherein the yoke is designed to engage around at least part of the carrier in the region of the mounting. 5. The filling-level sensor as claimed in claim 1, wherein two fork-shaped guide elements are arranged next to one another. 6. The filling-level sensor as claimed in claim 1, further comprising a spring elements prestressing a wiper contact against a wiping track of a potentiometer fastened between the guide elements arranged next to one another. 7. The filling-level sensor as claimed in claim 1, wherein the guide rail has a projecting edge. 8. The filling-level sensor as claimed in claim 1, wherein the guide rail is at a greater or equal distance from the carrier at its end facing away from the mounting than at its end facing the mounting, and in that surfaces of the carrier and of the guide rail which face one another in each case have a plane configuration. 9. The filling-level sensor as claimed in claim 1, wherein the guide rail is held at its ends by holding parts in front of the carrier.
<SOH> BACKGROUND OF THE INVENTION <EOH>Prior art filling-level sensors usually have a potentiometer with a wiping track arranged on the carrier and with wiper contacts, fastened to the yoke, for generating the electrical signals. Alternatively, the electrical signals are often also generated by means of a magnetically active position sensor. Magnetically active position sensors mostly have a resistance network which is to be fastened to the carrier and which is switchable by a magnet arranged on the yoke. The tank may be, for example, a fuel tank or a washing-water tank of a windshield cleaning system. The guide rail of the known filling-level sensor is designed as part of the carrier having a plane surface and is oriented away from the mounting. A U-shaped guide element arranged on the yoke engages behind the guide rail. This, in the case of lateral forces acting on the yoke and the lever wire, prevents the yoke from being bent away from the carrier and therefore the wiper contacts from being lifted off from the wiping track or the magnet from being removed from the resistance network. One disadvantage of the known filling-level sensor is that, because of the guide element, the yoke has very large dimensions and is therefore heavy. As a result, the center of gravity of the yoke is at a very great distance from the mounting, and therefore the filling-level sensor is very slow-acting. Consequently, minor variations in the filling level are detected by the potentiometer only with a delay or abruptly. Moreover, because of the high weight of the yoke, the latter requires a large quantity of material.
<SOH> SUMMARY OF THE INVENTION <EOH>The invention is directed a filling-level sensor, with a carrier provided for fastening to a lateral wall in the tank, with a yoke made from plastic and pivotable in the mounting of the carrier, with a lever wire holding the float and fastened to the yoke, and with a guide rail, arranged on the carrier, for guiding a guide element of the yoke in an intended axial position in relation to the carrier. The filling-level sensor according to the invention detects a variation in the filling level particularly quickly and is constructed cost-effectively. According to an aspect of the invention, the guide rail is arranged so as to point in the direction of the mounting, and the yoke has a fork-shaped guide element which is open at its end facing away from the mounting and engages with a guide rail. By virtue of this design, the yoke has very small dimensions and a particularly low weight. Moreover, the center of gravity of the yoke is arranged very near the mounting, so that the yoke has particularly low inertia. The yoke can therefore follow a variation in the filling level in the tank particularly quickly. Due to the low weight of the yoke, the latter necessitates the use of a correspondingly small amount of material, so that it can be produced particularly cost-effectively. The guide rail could, for example, have a round cross section. However, according to an aspect of the invention, the carrier can be manufactured at a particularly low outlay in one piece with the guide rail from plastic by the injection molding method if the guide rail has a web-shaped configuration and is arranged parallel to the carrier. By virtue of this configuration, the guide rail has a sliding track for the guide element on each of the two sides. The fork-shaped guide element could, for example, be arranged in a middle region of the yoke. However, a contribution to further reducing the dimensions of the yoke is made when the fork-shaped guide element is arranged at that end of the yoke which faces away from the mounting. The yoke is particularly reliably mounted on the carrier axially and can nevertheless be mounted on the carrier very simply when it is designed to engage around at least part of the carrier in the region of the mounting. For mounting, the yoke can simply be pushed over the carrier from the mounting. The yoke is subsequently guided axially in the region of the mounting and on the guide rail. To prevent the yoke from tilting, the guide element may be made particularly wide. However, because of this, the guide element has a high weight and a large bearing surface on the guide rail. The large bearing surface leads to undesirable frictional losses during the pivoting movement of the yoke. According to another aspect of the invention, a tilting of the yoke can be prevented in a simple way by two fork-shaped guide elements arranged next to one another, without the weight of the yoke being appreciably increased thereby. According to another aspect of the invention, the yoke is mounted in a particularly simple way when a spring element prestressing the wiper contact against the wiping track of the potentiometer is fastened between the guide elements arranged next to one another. A deformation of the guide rail could lead to the wiper contact being lifted off from the wiping track or to a rubbing of the guide element. According to another aspect of the invention, the guide rail has very high stability when it has a projecting edge. The filling-level sensor according to the invention is manufactured particularly cost-effectively when the guide rail is at a greater or equal distance from the carrier at its end facing away from the mounting than at its end facing the mounting, and when surfaces of the carrier and of the guide rail which face one another in each case have a plane configuration. By virtue of this configuration, the carrier can be manufactured, together with the guide rail, in a very simple way from plastic by the injection molding method in an injection mold from which removal can easily take place axially. According to another aspect of the invention, the carrier requires the use of a particularly small amount of material when the guide rail is held at its ends by holding parts in front of the carrier.
Map kinase integrin-binding domain
Dislosed is the integrin-binding domain of MAP kinase. Interaction between MAP kinase and integrin via this domain activates MAP kinase and initiates cellular activity. Methods of modulating cellular activity by inhibiting direct MAP kinase-integrin binding are provided. These methods have particular use in inhibiting growth of cancer cells.
1. A fragment of a MAP kinase wherein the fragment consists of a binding domain of the MAP kinase for an integrin, or an analog or derivative thereof capable of binding to the integrin. 2. A fragment of a MAP kinase wherein the fragment comprises a binding domain of the MAP kinase for an integrin or a partial amino acid sequence of the binding domain that is capable of binding to the integrin and has a length of 100 amino acids or less, or an analog or derivative of the fragment that is capable of binding to the integrin. 3. A fragment according to claim 2 wherein the fragment has a length of about 50 amino acids or less. 4. A fragment according to claim 3 wherein the fragment has a length of about 40 amino acids or less. 5. A fragment according to claim 4 wherein the fragment has a length of about 30 amino acids or less. 6. A fragment according to claim 5 wherein the fragment has a length of about 20 amino acids or less. 7. A fragment according to claim 2 comprising the amino acid sequence HRDLKPSNLLLNTTCDLKICDFGLAR or a contiguous amino acid sequence thereof. 8. A fragment according to claim 2 comprising the amino acid sequence PSNLLLNTTCDLKIC or a contiguous amino acid sequence thereof. 9. A fragment according to claim 1 wherein the MAP kinase is an ERK or a JNK family member. 10. A fragment according to claim 9 wherein the MAP kinase is ERK2. 11. A fragment according to claim 1 wherein the integrin is selected from the group consisting of β3, β5 and β6. 12. An isolated or purified polypeptide capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin, or an analog or derivative of the polypeptide that is capable of binding to the binding domain of the integrin, wherein the polypeptide is other than a MAP kinase or a fragment of a MAP kinase. 13. A polypeptide according to claim 12 comprising a core amino acid sequence of a binding domain of the MAP kinase for the integrin. 14. A polypeptide according to claim 12 wherein the MAP kinase is a member of the ERK family or the JNK family. 15. A polypeptide according to claim 14 wherein the MAP kinase is ERK2. 16. A polypeptide according to claim 12 wherein the integrin is selected from the group consisting of β3, β5 and β6. 17. A fusion protein incorporating a binding moiety capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin. 18. A fusion protein according to claim 17 adapted for passing across a cell membrane. 19. A fusion protein according to claim 18 further incorporating a carrier moiety for facilitating passage across the cell membrane. 20. A fusion protein according to claim 17 wherein the binding moiety comprises a fragment of a MAP kinase consisting of a binding domain of the MAP kinase for an integrin, or an analog or derivative thereof capable of binding to the integrin or a polypeptide capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin, or an analog or derivative of the polypeptide that is capable of binding to the binding domain of the integrin, wherein the polypeptide is other than a MAP kinase or a fragment of a MAP kinase. 21. A fusion protein according to claim 18 adapted for releasing the binding moiety after passing across the cell membrane. 22. An agent for inhibiting binding of a MAP kinase to an integrin, comprising: a targeting moiety for targeting a cell expressing the integrin; a binding moiety for binding to a binding domain of the integrin for the MAP kinase to thereby inhibit the binding of the MAP kinase to the integrin; and a carrier moiety for facilitating passage of the binding moiety across the cell membrane of the cell; wherein the binding moiety is other than an antibody or a binding fragment thereof. 23. An agent according to claim 22 wherein the binding and carrier moieties are capable of being released from the targeting moiety at the cell for passage of the carrier moiety and the binding moiety across the cell membrane of the cell. 24. An agent according to claim 23 wherein the agent incorporates an enzyme cleavage site for being cleaved to thereby release the carrier moiety and the inhibitor moiety at the cell. 25. An agent according to claim 24 wherein the enzyme cleavage site is a cleavage site for matrix-metalloproteinase-9 (MMP-9). 26. An agent according to claim 22 adapted for release of the inhibitor moiety from the carrier moiety after passage of the inhibitor moiety across the cell membrane of the cell. 27. An agent according to claim 22 wherein the targeting moiety is an antibody or a binding fragment of an antibody. 28. An agent according to claim 27 wherein the antibody or the binding fragment is specific for an extracellular region of the integrin. 29. An agent according to claim 22 wherein the targeting moiety is an integrin receptor targeted peptide for binding to the integrin. 30. An agent according to claim 22 wherein the binding moiety is capable of binding to a binding domain of the integrin for the MAP kinase. 31. An agent according to claim 30 wherein the binding moiety comprises a fragment consisting of a binding domain of the MAP kinase for an integrin, or an analog or derivative thereof capable of binding to the integrin or a polypeptide capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin, or an analog or derivative of the polypeptide that is capable of binding to the binding domain of the integrin, wherein the polypeptide is other than a MAP kinase or a fragment of a MAP kinase, or an analog or derivative of the fragment or polypeptide that is capable of binding to the binding domain of the integrin for the MAP kinase. 32. An agent according to claim 22 wherein the binding moiety is an analog of a binding domain of a MAP kinase for an integrin. 33. An agent capable of inhibiting binding of a MAP kinase to an integrin, wherein the agent comprises an analog of a binding domain of the MAP kinase for the integrin. 34. An agent according to claim 33 wherein the analog comprises a mimetic of the binding domain of the MAP kinase. 35. An isolated nucleic acid sequence encoding a fragment as defined in claim 1 or an analog or derivative thereof capable of binding to the binding domain of the integrin for the MAP kinase. 36. An isolated nucleic acid sequence encoding a polypeptide as defined in claim 12 or an analog or derivative thereof capable of binding to the binding domain of the integrin for the MAP kinase. 37. An isolated nucleic acid sequence encoding a fusion protein as defined in claim 17. 38. An isolated antisense nucleic acid sequence complementary to a nucleic acid sequence as defined in claim 35. 39. An isolated antisense nucleic acid sequence according to claim 38 wherein the antisense nucleic acid sequence is labelled. 40. An oligonucleotide comprising from 14 to 45 contiguous nucleotides of a nucleic acid sequence as defined in claim 35. 41. An oligonucleotide comprising from 14 to 45 nucleotides and which is capable of hybridising with a nucleic acid sequence as defined in claim 35. 42. A vector incorporating a nucleic acid sequence as defined in claim 35. 43. An expression vector incorporating a nucleic acid sequence as defined in claim 35. 44. A host cell transformed with a vector as defined in claim 42. 45. A host cell transformed according to claim 44 wherein the host cell is selected from the group consisting of a mammalian cell, an epithelial cell, a neoplastic cell and a cancer cell. 46. A host cell according to claim 45 wherein the host cell is a colon cancer cell. 47. A pharmaceutical composition comprising a fragment as defined in claim 1 or an analog or derivative thereof capable of binding to the binding domain of the integrin for the MAP kinase, together with a pharmaceutically acceptable carrier or diluent. 48. A pharmaceutical composition comprising a polypeptide as defined in claim 12 or an analog or derivative thereof capable of binding to the binding domain of the integrin of the MAP kinase, together with a pharmaceutically acceptable carrier or diluent. 49. A pharmaceutical composition comprising fusion protein as defined in claim 17 together with a pharmaceutically acceptable carrier or diluent. 50. A pharmaceutical composition comprising an agent for inhibiting binding of a MAP kinase with an integrin as defined in claim 22, together with a pharmaceutically acceptable carrier or diluent. 51. A pharmaceutical composition comprising a vector as defined in claim 43 together with a pharmaceutically acceptable carrier or diluent. 52. A pharmaceutical composition comprising an antisense nucleic acid sequence as defined in claim 38 together with a pharmaceutically acceptable carrier or diluent. 53. A pharmaceutical composition comprising an oligonucleotide as defined in claim 40 together with a pharmaceutically acceptable carrier or diluent. 54. An antibody specific for a binding domain of a MAP kinase for an integrin, or a binding fragment of the antibody. 55. An antibody specific for a fragment as defined in claim 1 or an analog or derivative of the fragment that is capable of binding to the binding domain of the integrin for the MAP kinase. 56. An antibody specific for a polypeptide as defined in claim 12 or an analog or derivative of the polypeptide that is capable of binding to the binding domain of the integrin in the MAP kinase. 57. An antibody according to claim 54 wherein the antibody is a monoclonal antibody. 58. A method of screening for an agent capable of binding to a binding domain of a MAP kinase for an integrin comprising: (a) testing a number of agents for ability to bind to the binding domain of the MAP kinase for the integrin; and (b) determining if any said agent is capable of binding to the binding domain of the MAP kinase on the basis of the testing. 59. A method of screening for an agent capable of binding to a binding domain of a MAP kinase for an integrin, comprising: (a) testing a number of agents for ability to bind to the MAP kinase; (b) selecting an agent or agents identified as being able to bind to the MAP kinase on the basis of the testing; and (c) utilising the selected said agent or agents in an assay for indicating whether the or any of the selected said agents is capable of binding to the binding domain of the MAP kinase for the integrin. 60. A method of evaluating whether an agent is capable of binding to a binding domain of a MAP kinase for an integrin, comprising: (a) testing the agent for ability to bind to the binding domain of the MAP kinase for the integrin; and (b) determining if the agent is capable of binding to the binding domain of the MAP kinase on the basis of the testing. 61. A method according to claim 60 wherein the integrin is selected from the group consisting of β3, β5 and β6. 62. A method according to claim 61 wherein the MAP kinase is an ERK family member or a JNK family member. 63. A method according to claim 62 wherein the MAP kinase is ERK2. 64. An agent identified to be capable of binding to a binding domain of a MAP kinase for an integrin by a method as defined in claim 61. 65. A method of isolating an agent from a sample utilizing a molecule immobilised on a solid support and which is capable of binding to a binding domain of an integrin for a MAP kinase, comprising: (a) contacting the molecule immobilised on the solid support with the sample under conditions suitable for binding of the agent to the molecule; (b) eluting the agent from the solid support; and (c) collecting the eluted agent; wherein the molecule is other than an antibody or binding fragment of an antibody. 66. A method according to claim 65 wherein the molecule is a MAP kinase or fragment thereof, or an analog or derivative of a fragment of a MAP kinase. 67. A method according to claim 65 wherein the molecule is a fragment consisting of a binding domain of the MAP kinase for an integrin, or an analog or derivative thereof capable of binding to the integrin, or an analog or derivative thereof. 68. A method according to claim 65 wherein the molecule is a polypeptide capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin, or an analog or derivative of the polypeptide that is capable of binding to the binding domain of the integrin, wherein the polypeptide is other than a MAP kinase or a fragment of a MAP kinase, or an analog or derivative thereof. 69. A method according to claim 65 wherein the integrin is selected from the group consisting of β3, β5 and β6. 70. A method according to claim 65 wherein the MAP kinase is an ERK family member or a JNK family member. 71. A method according to claim 70 wherein the MAP kinase is ERK2. 72. An agent isolated from a sample by a method as defined in claim 61. 73. A method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: treating the cell with an antisense oligonucleotide for hybridising with at least part of a nucleic acid sequence coding for the binding domain of the MAP kinase for the integrin to thereby cause expression of the MAP kinase to be down-regulated. 74. A method according to claim 73 wherein the oligonucleotide is about 50 nucleotides or less in length. 75. A method according to claim 74 wherein the oligonucleotide is 45 nucleotides in length or less. 76. A method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising treating the cell with an antisense oligonucleotide for hybridising with at least part of a nucleic acid sequence coding for the binding domain of the MAP kinase for the integrin to thereby cause expression of the MAP kinase to be down-regulated, wherein the antisense oligonucleotide is an oligonucleotide as defined in claim 38. 77. A method according to claim 73 wherein the cell is transfected with the antisense oligonucleotide or the antisense oligonucleotide is transcribed from a vector introduced into the cell. 78. A method according to claim 73 wherein the cell is a cancer cell. 79. A method according to claim 73 wherein the MAP kinase is an ERK family member or a JNK family member. 80. A method according to claim 79 wherein the MAP kinase is ERK2. 81. A method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: transfecting the cell with an expression vector incorporating a nucleic acid sequence encoding a MAP kinase of a fragment thereof, or a homolog, analog or variant of the MAP kinase or fragment, with a mutagenised binding domain for the integrin or in which the binding domain has been deleted whereby binding to the integrin is inhibited or prevented. 82. A method according to claim 81 wherein the binding domain is mutagenised. 83. A method according to claim 81 wherein the MAP kinase is an ERK family member of a JNK family member. 84. A method according to claim 81 wherein the MAP kinase is ERK2. 85. A method according to claim 81 wherein the integrin subunit is selected from the group consisting of β3, β5 and β6. 86. A method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: treating the cell with an effective amount of an agent capable of binding to the binding domain of the integrin for the MAP kinase, wherein the agent is other than an antibody or a binding fragment thereof. 87. A method according to claim 86 wherein the agent is a fragment consisting of a binding domain of the MAP kinase for an integrin, or an analog or derivative thereof capable of binding to the integrin, or an analog or derivative thereof capable of binding to the binding domain of the integrin. 88. A method according to claim 86 wherein the agent is a polypeptide capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin, or an analog or derivative of the polypeptide that is capable of binding to the binding domain of the integrin, wherein the polypeptide is other than a MAP kinase or a fragment of a MAP kinase, or an analog or derivative thereof capable of binding to the binding domain of the integrin. 89. A method according to claim 86 wherein the agent is a fusion protein incorporating a binding moiety capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin. 90. A method according to claim 86 wherein the agent is an agent comprising: a targeting moiety for targeting a cell expressing the integrin; a binding moiety for binding to a binding domain of the integrin for the MAP kinase to thereby inhibit the binding of the MAP kinase to the integrin; and a carrier moiety for facilitating passage of the binding moiety across the cell membrane of the cell; wherein the binding moiety is other than an antibody or a binding fragment thereof. 91. A method according to claim 86 wherein the activity of the cell is growth, migration, spreading, invasion, matrix-degrading enzyme secretion, differentiation and/or apoptosis of the cell. 92. A method according to claim 86 wherein the cell is a cancer cell. 93. A method according to claim 92 wherein the cancer cell is a colon cancer cell. 94. A method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: treating the cell with an effective amount of an agent capable of binding to the binding domain of the MAP kinase, wherein the agent is other than an agent comprising a binding domain of the integrin for the MAP kinase or an analog or derivative thereof. 95. A method according to claim 94 wherein the agent comprises an antibody or binding fragment thereof. 96. A method according to claim 94 wherein the activity of the cell is growth, migration, spreading, invasion, matrix-degrading enzyme secretion, differentiation and/or apoptosis. 97. A method according to claim 94 wherein the cell is a cancer cell. 98. A method according to claim 94 wherein the cancer cell is a colon cancer cell. 99. A method according to claim 73 comprising treating a mammal in need of such treatment. 100. A method according to claim 73 wherein the method comprises treatment or prophylaxis of cancer or a condition associated with a predisposition to cancer. 101. A method according to claim 100 wherein the cancer is selected from the group consisting of cancer of the lip, tongue, salivary glands, gums, floor and other areas of the mouth, oropharynx, nasopharynx, hypopharynx and other oral cavities, oesophagus, stomach, small intestine, duodenum colon, rectum, gallbladder, pancreas, larynx, trachea, bronchus, lung, breast, uterus, cervix, ovary, vagina, vulva, prostate, testes, penis, bladder, kidney, thyroid and skin. 102. A method according to claim 101 wherein the cancer is colon cancer. 103. A method of prophylaxis or treatment of cancer in a mammal, comprising: treating the mammal with an effective amount of an antisense nucleic acid for hybridising with a nucleic acid encoding a MAP kinase to cause expression of the MAP kinase to be down-regulated wherein the MAP kinase has a binding domain for an integrin. 104. A method according to claim 103 comprising administering an effective amount of the antisense nucleic acid to the mammal. 105. A method according to claim 103 comprising administering to the mammal an effective amount of a vector incorporating an nucleic acid sequence complementary to the antisense nucleic acid for generation of the antisense nucleic sequence in vivo. 106. A method according to claim 103 wherein the antisense nucleic acid is an antisense oligonucleotide for hybridising with at least part of the sequence of the nucleic acid encoding the binding domain of the MAP kinase. 107. A method according to claim 106 wherein the oligonucleotide has a length of 150 nucleotides or less. 108. A method according to claim 107 wherein the oligonucleotide has a length of 90 nucleotides or less. 109. A method according to claim 108 wherein the oligonucleotide has a length of 50 nucleotides or less. 110. A method according to claim 109 wherein the oligonucleotide has a length of from 15 to 30 nucleotides. 111. A method according to claim 103 wherein the integrin subunit is selected from the group consisting of β3, β5 and β6. 112. A method according to claim 103 wherein the cancer is colon cancer. 113. A fragment according to claim 9 wherein the integrin is selected from the group consisting of β3, β5 and β6. 114. A polypeptide according to claim 15 wherein the integrin is selected from the group consisting of β3, β5 and β6. 115. A vector incorporating an oligonucleotide as defined in claim 40. 116. An expression vector incorporating an oligonucleotide as defined in claim 41 for being expressed in a cell. 117. A host cell transformed with a vector as defined in claim 43. 118. A pharmaceutical composition comprising an antisense nucleic acid sequence as defined in claim 39 together with a pharmaceutically acceptable carrier or diluent. 119. A pharmaceutical composition comprising an oligonucleotide as defined in claim 41 together with a pharmaceutically acceptable carrier or diluent. 120. An antibody according to claim 55 wherein the antibody is a monoclonal antibody. 121. An antibody according to claim 56 wherein the antibody is a monoclonal antibody. 122. A method according to claim 69 wherein the MAP kinase is an ERK family member or a JNK family member. 123. A method according to claim 86 comprising treating a mammal in need of such treatment. 124. A method according to claim 86 wherein the method comprises treatment or prophylaxis of cancer or a condition associated with a predisposition to cancer.
<SOH> BACKGROUND OF THE INVENTION <EOH>Colorectal cancer is the commonest internal malignancy affecting men and women in Australia. About 4% of individuals develop this disease during the course of their lifetime and it was responsible for 14% of cancer deaths in that country in 1990. In 1995 there were 10,615 cases of colorectal cancer and 4508 deaths in Australia. Worldwide, an estimated 875,000 cases of colorectal cancer occurred in 1996, accounting for 8.5% of all new cases of cancer. Incidence rates vary approximately 20-fold around the world, with the highest rates seen in the developed world and lowest in India. Australian incidence rates are towards the higher end of the scale internationally alongside those for North America and New Zealand. Five-year survival following diagnosis of colon cancer is around 55% in the developed world and has altered little during the past few decades despite advances in chemo-, immuno- and radiotherapy. Colorectal cancer is a malignant tumour that starts in the bowel wall and is confined locally for a relatively long period before spreading through the bowel wall and metastasising to lymph nodes and other parts of the body. Survival rates are significantly improved where the disease is detected and treated early. The aetiology of colorectal cancer is complex and appears to involve interactions between inherited susceptibility and environmental factors. Recognition of the genetic component of colorectal cancer is growing. Mutations are present as inherited germline defects or arise in somatic cells secondary to environmental insult. There are two main inherited predisposition syndromes: Familial Adenomatous Polyposis (FAP) and Hereditary Non-Polyposis Colorectal Cancer (HNPCC); the remaining cases are attributed to so-called sporadic colorectal cancer. FAP and HNPCC contribute to approximately 1% and 4%, respectively, of all colorectal cancers and a strong family history of bowel cancer in first-degree relatives is obtained in another 10-15% of patients. However, in the vast majority of patients the aetiology of large bowel cancer remains unknown. Most colon cancer arises within pre-existing benign precursor lesions or adenomas. Adenomas are classified by histological architecture as tubular, tubulovillous or villous. Villous change is associated with a higher malignant potential, as are large and high-grade epithelial dysplasia. Environmental risk factors for development of colorectal cancer include diets low in fibre and vegetables and high in fat, red meat and alcohol and cigarette smoking which may induce mutations in somatic cells. Studies have shown that persistent genetic instability and accumulation of mutations in several genes that are mainly concerned with cell growth or DNA repair, may be critical for the development of all colorectal cancers. For example, a normal mucosal cell with inactivation of tumour suppressor genes such as the Adenomatous Polyposis Coli (APC) gene or Mutated in Colon Cancer (MCC) gene can proliferate and become a small adenomatous polyp. Mutations in oncogenes such as k ras and in tumour suppressor genes such as p53 and the Deleted in Colon Cancer gene (DCC) may then occur and lead to the transformation of the polyp into a large adenoma, from which a carcinoma can eventually arise. The uncontrolled cell growth that leads to the development of neoplasia is believed to result, therefore, from a series of inherited and acquired accumulated genetic changes. This multistep process confers on cells the capacity to survive and proliferate in a manner freed from the constraints imposed on normal cell growth. Spread of cancer cells involves tumour cell migration through the extracellular matrix scaffold, invasion of basement membranes, arrest of circulating tumour cells, and tumour cell extravasation and proliferation at metastatic sites. Detachment of cells from the primary tumour mass and modification of the peri-cellular environment aid penetration of tumour cells into blood and lymphatic vessels. It is the invasive and metastatic potential of tumour cells that ultimately dictates the fate of most patients suffering from malignant diseases. Hence, tumourigenesis can be viewed as a tissue remodelling process that reflects the ability of cancer cells to proliferate and digest surrounding matrix barriers. These events are thought to be regulated, at least in part, by cell adhesion molecules and matrix-degrading enzymes. Cell adhesion receptors on the surface of colon cancer cells are involved in complex cell signalling which may regulate cell proliferation, migration, invasion and metastasis and several families of adhesion molecules have now been identified including integrins, cadherins, the immunoglobulin superfamily, hyaluronate receptors, and mucins (Agrez, 1996.) In general, these cell surface molecules mediate both cell-cell and cell-matrix binding, the latter involving attachment of tumour cells to extracellular scaffolding molecules such as collagen, fibronectin and laminin. It is now clear that multiple and varied cell adhesion receptors exist on colon cancer cells at any one time and an understanding of the role of individual receptors in promoting growth and spread of colon cancer is only just beginning to be elucidated. Of all the families of cell adhesion molecules, the best-characterised at the present time is the family known as integrins. Integrins are involved in several fundamental processes including leucocyte recruitment, immune activation, thrombosis, wound healing, embryogenesis, virus internalisation and tumourigenesis. Integrins are transmembrane glycoproteins consisting of an alpha (α) and beta (β) chain in close association that provide a structural and functional bridge between extracellular matrix molecules and cytoskeletal components with the cell. The integrin family comprises 17 different α and 8β subunits and the αβ combinations are subsumed under 3 subfamilies. Excluding the leucocyte integrin subfamily that is designated by the β2 nomenclature, the remaining integrins are arranged into two major subgroups, designated β1 and αv based on sharing common chains. In the β1 subfamily, the β1 chain combines with any one of nine a chain members (α1-9), and the a chain which associates with β1 determines the matrix-binding specificity of that receptor. For example, α2β1 binds collagen and laminin, α3β1 binds collagen, laminin and fibronectin, and α5β1 binds fibronectin. In the αv subfamily of receptors, the abundant and promiscuous αv chain combines with any one of five β chains, and a distinguishing feature of αv integrins is that they all recognise and bind with high affinity to arginine-glycine-aspartate (RGD) sequences present in the matrix molecules to which they adhere (Hynes, 1992). The current picture of integrins is that the N-terminal domains of α and β subunits combine to form a ligand-binding head on each integrin. This head, containing the cation binding domains, is connected by two stalks representing both subunits, to the membrane-spanning segments and thus to the two cytoplasmic domains. The β subunits all show considerable similarity at the amino acid level (Loftus et al, 1994). All have a molecular mass between 90 and 110 kDa, with the exception of β4 which is larger at 210 kDa. Similarly, they all contain 56 conserved cysteine residues, except for β4 which has 48. These cysteines are arranged in four repeating patterns which are thought to be linked internally by disulphide bonds. The α-subunits have a molecular mass ranging from 150-200 kDa. They exhibit a lower degree of similarity than the β chains, although all contain seven repeating amino acid sequences interspaced with non-repeating domains. The β subunit cytoplasmic domain is required for linking integrins to the cytoskeleton (Hynes, 1992). In many cases, this linkage is reflected in localisation to focal contacts, which is believed to lead to the assembly of signalling complexes that include α-actinin, talin, and focal adhesion kinase (FAK) (Otey et al, 1990; Guan and Shalloway, 1992; Kornberg et al, 1992). At least three different regions that are required for focal contact localisation of β1 integrins have been delineated (Reszka et al, 1992). These regions contain conserved sequences that are also found in the cytoplasmic domains of the β2, β3, β5, β6 and β7 integrin subunits. The functional differences between these cytoplasmic domains with regard to their signalling capacity have not yet been established. Ligation of integrins by their extracellular matrix protein ligands induces a cascade of intracellular signals that include tyrosine phosphorylation of focal adhesion kinase, increases in intracellular Ca 2+ levels, inositol lipid synthesis, synthesis of cyclins and expression of immediate early genes. In contrast, prevention of integrin-ligand interactions suppresses cellular growth or induces apoptotic cell death (Meredith et al, 1993; Montgomery et al, 1994; Brooks et al, 1994; Varner et al, 1995; Boudreau et al, 1995). Thus, integrins play roles in a number of cellular processes that impact on the development of tumours, including the regulation of proliferation and apoptosis. The integrin β6 subunit was first identified in cultured epithelial cells as part of the αvβ6 heterodimer, and the αvβ6 complex was shown to bind fibronectin in an arginine-glycine-aspartate (RGD)-dependent manner in human pancreatic carcinoma cells (Sheppard et al, 1990; Busk et al, 1992). The β6 subunit is composed of 788 amino acids and shares 34-51% sequence homology with other β integrin subunits β1-β5. The β6 subunit also contains 9 potential glycosylation sites on the extracellular domain (Sheppard et al, 1990). The cytoplasmic domain differs from other β subunits in that it is composed of a 41 amino acid region that is highly conserved among integrin β subunits, and a unique 11 amino acid carboxy-terminal extension. The 11 amino acid extension has been shown not to be necessary for localisation of β6 to focal contacts; in fact, its removal appears to increase receptor localisation. However, removal of any of the three conserved regions previously identified as important for the localisation of β1 integrins to focal contacts (Reszka et al, 1992) has been shown to eliminate recruitment of β6 to focal contacts (Cone et al, 1994). The integrin αvβ6 has previously been shown to enhance growth of colon cancer cells in vitro and in vivo, and this growth-enhancing effect is due, at least in part, to αvβ6-mediated gelatinase B secretion (Agrez et al, 1994; Niu et al, 1998 Agrez et al, 1999). What makes this epithelial-restricted integrin of particular interest in cancer is that it is not expressed on normal epithelial cells but is highly expressed during would healing and tumourigenesis, particularly at the invading edge of tumour cell islands (Breuss et al, 1995; Agrez et al, 1996). Further more, αvβ6 has been found to induce its own expression in an autocrine manner with cell crowding and a self-perpetuating model of colon cancer progression regulated by αvβ6-mediated gelatinase B secretion has been proposed (Niu et al, 2000). Invasion of the extracellular matrix and metastatic spread of colon cancer is also likely to reflect the ability of tumour cells to digest their surrounding matrix scaffold through secretion of matrix-degrading enzymes such as matrix metalloproteinases (MMPs). The mechanisms whereby human colon cancer cells escape the constraints on growth imposed on normal cells by cell crowding and dense pericellular matrices is unclear. However, even colon cancer cells are subject to relative growth inhibition in vitro in a dense extracellular matrix environment (Agrez, 1989). Integrins can signal through the cell membrane in either direction. The extracellular binding activity of integrins can be regulated from the cell interior as, for example, by phosphorylation of integrin cytolasmic domains (inside-out signalling), while the binding of the extracellular matrix (ECM) elicits signals that are transmitted into the cell (outside-in signalling) (Giancotti and Ruoslahti, 1999). Outside-in signalling can be roughly divided into two descriptive categories. The first is ‘direct signalling’ in which ligation and clustering of integrins is the only extracellular stimulus. Thus, adhesion to ECM proteins can activate cytoplasmic tyrosine kinases (e.g. focal adhesion kinase FAK) and serine/threonine kinases (such as those in the mitogen-activated protein kinase (MAPK) cascade) and stimulate lipid metabolism (eg phosphatidylinositol-4,5-biphosphate (P 1 P 2 ) synthesis. The second category of integrin signalling is ‘collaborative signalling’, in which integrin-mediated cell adhesion modulates signalling events initiated through other types of receptors, particularly receptor tyrosine kinases that are activated by polypeptide growth factors (Howe et al, 1998). In all cases, however, integrin-mediated adhesion seems to be required for efficient transduction of signals into the cytosol or nucleus. MAP kinases behave as a convergence point for diverse receptor-initiated signalling events at the plasma membrane. The core unit of MAP kinase pathways is a three-member protein kinase cascade in which MAP kinases are phosphorylated by MAP kinase kinases (MEKs) which are in turn phosphorylated by MAP kinase kinases (e.g. Raf-1) (Garrington and Johnson, 1999). Amongst the 12 member proteins of the MAP kinase family are the extracellular signal-regulated kinases (ERKs) (Boulton et al, 1991) activated by phosphorylation of tyrosine and threonine residues (Payne et al, 1991) which is the type of activation common to all known MAP kinase isoforms. ERK 1/2 (44 kD and 42 kD MAPks, respectively) share 90% amino acid identity and are ubiquitous components of signal transduction pathways (Boulton et al, 1991). These serine/threonine kinases phosphorylate and modulate the function of many proteins with regulatory functions including other protein kinases (such as p90 rsk ) cytoskeletal proteins (such as microtubule-associated phospholipase A 2 ), upstream regulators (such as the epidermal growth factor receptor and Ras exchange factor) and transcription factors (such as C-Myc and Elk-1). ERKs play a major role in growth-promoting events, especially when the concentration of growth factors available to a cell is limited (Giancotti and Ruoslahti, 1999). MAP kinases can be activated through non-receptor tyrosine kinases such as focal adhesion kinase (FAK), cytoplasmic tyrosine kinase (pp60 c-srk) (Schlaepher and Hunter, 1998), and growth factors acting through membrane-associated receptor tyrosine kinases. The FAK pathway is activated by most integrins. In addition to activating FAK, some β1 and αv integrins also activate the tyrosine kinase Fynu and through it, the adaptor protein Shc (Wang et al, 1996). It is likely that both FAK and Shc contribute to the activation of Ras and thence to the downstream kinase cascade of Raf-1, MEK, and MAP kinases (Schlaepfer et al, 1994; 1997). It is now generally accepted that the activation of ERK in response to integrin ligation requires Ras signalling (Wary et al, 1996; Schlaepfer and Hunter, 1997). The laminin receptor α6β4, the laminin/collagen receptor α1β1, the fibronectin receptor α5β1 and the RGD binding receptor αvβ3 are linked to the Ras-Raf-MEK-ERK signalling pathway and control of immediate early gene expression (Wary et al, 1996; Maniero et al, 1995; 1997). The ability of integrins to activate ERK may be especially important when the concentration of growth factors available to the cell is limited. In this setting, proliferation is likely to require co-stimulation of ERK through integrins and growth factor receptors (Giancotti and Ruoslahti, 1999). Moreover, activation of ERK in response to integrin ligation may play a role in regulating cell migration (Klemke et al, 1997) possibly by initiating matrix-degrading enzyme secretion. While there is a good deal of evidence in support of a key role for FAK and the phosphotyrosine-domain-containing adaptor protein Shc (Howe et al, 1998; Giancotti & Ruoslahti, 1999) in the Ras-Raf MEK-MAP kinase activation pathway there are also data implicating alternate pathways independent of MEKs. For example, MEK-independent regulation of MAP kinase activation in NIH3T3 fibroblasts has been shown to be mediated by phosphatidylinositol-3-kinases and the conventional protein kinase C (PKC) isoforms and is thought to be due to inactivation of a MAP kinase inhibitor (Grammer and Blenis, 1997). Although the mechanism by which PKC regulates integrin function is not known, PKC has been shown to regulate integrin-induced activation of the MAP kinase pathway upstream of Shc. For example, PKC inhibition has been shown to inhibit ERK2 activation by fibronectin receptors without any effect on integrin-induced FAK or paxillin tyrosine phosphorylation (Miranti et al, 1999). Hence, MAP kinase activation is more complicated than a simple linear pathway, and the mechanistic basis for the commonly observed integrin-mediated activation of MAP kinases remains controversial. The importance of the MAP kinase pathway in promoting colon cancer growth in vivo is now no longer in question. Although relatively little is known about the role of MAP kinase activation in for instance, colon tumour progression, MAP kinase pathways have been shown to be highly activated during the late progression of colorectal cancer (Licato et al, 1997). In a recent breakthrough in this field, a highly potent inhibitor of MAP kinase activation has been identified which is capable of inhibiting human colon cancer growth in immune-deficient mice raising hops for the clinical application of MAP kinase inhibitors in the treatment of colon cancer (Sebolt-Leopold et at, 1999). Various intracellular proteins may be linked directly or spatially to integrin cytoplasmic domains. Direct interactions have been identified between cytoskeletal proteins such as α-actinin and talin and β1 and β3 integrin tails (Horwitz et al, 1986; Otey et al, 1990; Knezevic et al, 1996; Pfaff et al, 1998). A direct association between FAK and the β1 integrin tail has been suggested based on in vitro β1 peptide studies, but this remains to be confirmed (Schaller et al, 1995). More recently, the cytoplasmic domain of the α4 subunit has been found to be physically associated with the signalling adaptor protein paxillin in Jurkat T cells, and this binding event regulates the kinetics of FAK tyrosine phosphorylation (Liu et al, 1999). Direct integrin links with the intracellular calcium-binding protein, calreticulin, and integrin-linked kinase (ILK) (Hannigan et al, 1996) have been shown to regulate “inside-out” integrin signalling. For example, calreticulin has been shown to bind to a chain cytoplasmic domains (Rojiani et al, 1991) and modify α2β1 integrin activation by phorbol esters and anti-integrin antibodies (Coppolino et al, 1995). Newly identified integrin-binding molecules include the serine/threonine integrin-linked kinase, ILK, which can associate with the β1, β2 and β3 subunits. When over-expressed, ILK has been shown to reduce anchorage-independent growth and tumourigenicity in nude mice (Hannigan et al, 1996). Co-immunoprecipitation strategies have also demonstrated interactions between integrins and the integral plasma membrane protein IAP, and members of the four transmembrane domain protein family (tetraspans). The extracellular Ig region of the IAP molecule mediates association with αvβ3 and is required for cell binding to vitronectin-coated particles (Lindberg et al, 1996). An emerging model for tetraspans is that they recruit signalling enzymes such as phosphatidylinositol-4-kinase and PKC into complexes with integrins (Hemler, 1998). Integrins have also been shown to be physically linked with matrix-degrading enzymes and growth factors. For example, the integrin αvβ6 has been shown to bind and activate latent TGFβ1 in keratinocytes (Munger et al, 1999) which is thought to be important in modulating the inflammatory process following epithelial injury. In melanoma cells, αvβ3 binds activated gelatinase A (Brooks et al, 1996), and both insulin and platelet-derived growth factor (PDGF) co-immunoprecipitate with this integrin in NIH3T3 mouse fibroblasts (Schneller et al, 1997). Synergism between integrin-mediated signalling processes and growth factor responses is now well-recognised and Schneller et al (1997) showed that a small subset of each of the insulin receptor and PDGF β-receptor is tyrosine phosphorylated upon growth factor stimulation. Interestingly, this subset can associate with the αvβ3 integrin, and PDGF activity is enhanced in association with increased MAP kinase activity in cells plated on the αvβ3 ligand, vitronectin.
<SOH> SUMMARY OF THE INVENTION <EOH>Broadly stated, the present invention relates to the surprising finding that members of the mitogen activated protein (MAP) kinase family can associate with the cytoplasmic domain of an integrin molecule. It is believed that no member of the MAP kinase family has previously been found to directly associate with any integrin. The identification of this functional relationship permits the rational design of agents for therapeutically or prophylactically modulating cellular activity mediated by the MAP kinase and integrin interaction. In an aspect of the present invention there is provided an agent capable of binding to a binding domain of an integrin for a MAP kinase. In another aspect of the present invention there is provided an isolated or purified polypeptide capable of binding to a binding domain of an integrin for a MAP kinase or a homolog, analog, variant or derivative of the polypeptide. The polypeptide may consist of or comprise the binding domain of the MAP kinase to which the integrin binds or sufficient core amino acid sequence of the binding domain of the MAP kinase to enable binding of the polypeptide to the integrin. Preferably, the polypeptide will be a fragment of a MAP kinase. Most preferably, the polypeptide will comprise the amino acid sequence HRDLKPSNLLLNTTCDLKICDFGLAR or PSNLLLNTTCDLKIC or a region or regions of such sequences. Accordingly, in a further aspect of the present invention there is provided a fragment of a MAP kinase wherein the fragment is capable of binding with an integrin, or a homolog, analog, variant or derivative of the fragment. In another aspect of the present invention there is provided a fragment of a MAP kinase wherein the fragment consists of a binding domain of the MAP kinase for an integrin, or an analog or derivative thereof capable of binding to the integrin. In another aspect of the present invention there is provided an isolated or purified polypeptide capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin, or an analog or derivative of the polypeptide that is capable of binding to the binding domain of the integrin, wherein the polypeptide is other than a MAP kinase or a fragment of a MAP kinase. Preferably, a polypeptide or fragment of the invention will have a length of about 150 amino acids or less, more preferably about 100 or 50 amino acids or less and more usually about 40 amino acids or less. Typically, the length will be between about 5 to about 30 amino acids. In yet another aspect of the present invention there is provided a fusion protein a fusion protein incorporating a binding moiety capable of binding to a binding domain of an integrin for a MAP kinase and thereby inhibiting binding of the MAP kinase to the integrin. Preferably, the binding moiety will comprise a fragment or polypeptide of the invention. Most preferably, the fusion protein will incorporate a carrier moiety for facilitating passage across the cell membrane. The carrier moiety may be penetratin. In still another aspect of the present invention there is provided an agent for inhibiting binding of a MAP kinase to an integrin, comprising: a targeting moiety for targeting a cell expressing the integrin; a binding moiety for binding to a binding domain of the integrin for the MAP kinase to thereby inhibit the binding of the MAP kinase to the integrin; and a carrier moiety for facilitating passage of the binding moiety across the cell membrane of the cell; wherein the binding moiety is other than an antibody or an binding fragment thereof. In another aspect of the present invention there is provided a MAP kinase or a fragment thereof with a mutagenised binding domain for an integrin or in which the binding domain is deleted, or a homolog, analog or variant of the MAP kinase or fragment, wherein capability to bind with the integrin is thereby reduced. In another aspect of the present invention there is provided an isolated nucleic acid sequence encoding a fragment or polypeptide of the invention or a homolog, analog, variant or derivative of the fragment or polypeptide. In another aspect of the invention there is provided a nucleic acid sequence coding for a MAP kinase or fragment thereof having a mutagenised binding domain for an integrin or in which the binding domain is deleted or a homolog, analog or variant of the MAP kinase or fragment, wherein capability to bind with the integrin is thereby reduced. In a still further aspect there is provided an isolated nucleic acid sequence encoding a fusion protein or agent of the invention. There are also provided antisense nucleic acid sequences complimentary to the sense nucleic sequences of the invention. Such antisense sequences find application in antisense therapy of cells in which down regulation of cellular activity is desired, and include oligonucleotides. Sense oligonucleotides coding for all or part of the binding domain of a MAP kinase for an integrin, or that of a homolog, analog or variant thereof, and complimentary antisense oligonucleotides find particular application as primers or probes. A nucleic acid primer or probe of the invention may be labelled with a suitable reporter molecule for enabling detection of hybridisation of the primer or probe to a target nucleic acid sequence. In yet another aspect of the present invention there is provided a vector incorporating a nucleic acid sequence of the invention. Typically, the vector will be an expression vector and the nucleic acid sequence will be capable of being transcribed. In a further aspect of the present invention there is provided a host cell transformed with a vector of the invention. Preferably, the host cell will be selected from the group consisting of a mammalian cell, an epithelial cell, a neoplastic cell, and a cancer cell. Preferably, the host cell will be a mammalian cell and most preferably, a colon cancer cell. In yet another aspect of the present invention there is provided an antibody generated with the use of a polypeptide, fragment, agent or fusion protein of the invention. In a still further aspect of the invention there is provided an antibody and binding fragment thereof capable of binding to a binding domain of a MAP kinase for an integrin. The antibody may be a polyclonal or monoclonal antibody. Preferably, the antibody is a monoclonal antibody. In still another aspect of the present invention there is provided a method of screening for an agent capable of binding to a binding domain of a MAP kinase for an integrin, comprising: (a) testing a number of agents for ability to bind to the binding domain of the MAP kinase for the integrin; and (b) determining if any said agent is capable of binding to the binding domain of the MAP kinase on the basis of the testing. In yet another aspect of the invention there is provided a method of screening for an agent capable of binding to a binding domain of a MAP kinase for an integrin, comprising: (a) testing a number of agents for ability to bind to the MAP kinase; (b) selecting an agent or agents identified as being able to bind to the MAP kinase on the basis of the testing; and (c) utilising the selected said agent or agents in an assay for indicating whether the or any of the selected said agents is capable of binding to the binding domain of the MAP kinase for the integrin. In another aspect of the present invention there is provided a method of evaluating whether an agent is capable of binding to a binding domain of a MAP kinase for an integrin, comprising: (a) testing the agent for ability to bind to the binding domain of the MAP kinase for the integrin; and (b) determining if the agent is capable of binding to the binding domain on the basis of the testing. Preferably, a polypeptide or fragment of the invention comprising or consisting of the binding domain of the MAP kinase or core amino acid sequence of the binding domain or a homolog, analog, variant or derivative of the polypeptide or fragment or core amino acid sequence is used in the testing or assaying for whether an agent is capable of binding to the binding domain of the MAP kinase. Most preferably, the polypeptide or fragment will comprise or consist of the amino acid sequence HRDLKPSNLLLNTTCDLKICDFGLAR or PSNLLLNTTCDLKIC, core amino acid sequence of such sequences capable of binding to an integrin, or a homolog variant, analog or derivative of such sequences. Testing or assaying of an agent for ability to bind to the binding domain of the MAP kinase for the integrin and thereby inhibit binding of the MAP kinase to the integrin, may comprise exposing the MAP kinase to the agent(s) to enable binding of the agent(s) to the MAP kinase to occur either in the presence of the integrin or prior to the addition of the integrin. Rather than using the MAP kinase, a polypeptide, fragment of a MAP kinase or other molecule capable of binding to the binding site on the integrin for the MAP kinase may be used in such testing or assaying. Similarly, rather than utilising an intact integrin, a polypeptide or integrin fragment comprising or incorporating the binding domain of the integrin for the MAP kinase, or other molecule capable of binding with the binding domain on the MAP kinase for the integrin, may be utilised. In still another aspect of the present invention there is provided a method of isolating an agent from a sample utilising a molecule immobilised on a solid support and which is capable of binding to a binding domain of an integrin for a MAP kinase, comprising: (a) contacting the molecule immobilised on the solid support with the sample under conditions suitable for binding of the agent to the molecule; (b) eluting the agent from the solid support; and (c) collecting the eluted agent; wherein the molecule is other than an antibody or binding fragment of an antibody. The molecule may be the MAP kinase, or a fusion protein, a polypeptide, or a fragment of the invention to which the integrin is capable of binding, or for instance a homolog, analog, variant or derivative of such a polypeptide or fragment. In yet another aspect of the present invention there is provided the agent so isolated. An agent of the invention will usually be provided in the form of a pharmaceutical composition. Accordingly, in another aspect of the present invention there is provided a pharmaceutical composition comprising an agent of the invention capable of binding to a binding domain of an integrin for a MAP kinase, and a pharmaceutically acceptable carrier or diluent. The agent may or not be proteinaceous in nature. Preferably, the agent will comprise a fusion protein, or polypeptide. Most preferably, the agent will be coupled to a carrier molecule for facilitating entry of the agent into a cell. In a further aspect of the invention there is provided a pharmaceutical composition comprising a nucleic acid sequence of the invention and a pharmaceutically acceptable carrier or diluent. Preferably, the nucleic acid sequence is incorporated into a vector as described herein. Alternatively, the nucleic acid sequence may be joined to a carrier molecule such as penetratin for facilitating entry of the nucleic acid sequence into a target cell. In another aspect of the present invention there is provided a method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: transfecting the cell with an expression vector incorporating a nucleic acid sequence encoding a MAP kinase of a fragment thereof, or a homolog, analog or variant of the MAP kinase or fragment, for being expressed by the cell and having a mutagenised binding domain for the integrin or in which the binding domain has been deleted whereby binding to the integrin is inhibited or prevented. In still another aspect of the present invention there is provided a method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: transfecting the cell with a nucleic acid sequence encoding a fragment of a MAP kinase, or a homolog, analog or variant of the fragment, for being expressed by the cell and capable of binding to the binding domain of an integrin for the MAP kinase. In yet another aspect of the present invention there is provided a method of modulating activity of a cell, comprising: transfecting the cell with a nucleic acid encoding a polypeptide for being expressed by the cell wherein the polypeptide is capable of binding to a binding domain of an integrin for a MAP kinase. In still another aspect of the invention there is provided a method of modulating activity of a cell, comprising causing the expression of a MAP kinase to which an integrin is able to bind to be down-regulated. Preferably, down-regulation of the expression of the MAP kinase is achieved using an antisense nucleic acid sequence that inhibits expression of gene coding for the MAP kinase. The antisense nucleic acid sequence may be administered to the cell or generated within the cell. Preferably, the cell will be transformed with a vector of the invention for generation of the antisense nucleic acid sequence. Preferably, the antisense nucleic acid sequence will specifically hybridise with sense nucleic acid sequence coding for at least part of the binding domain of the MAP kinase for the integrin and/or intron sequence between such coding sequence. Accordingly, in another aspect of the present invention there is provided a method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: treating the cell with an antisense oligonucleotide for hybridising with at least part of a nucleic acid sequence coding for the binding domain of a MAP kinase for an integrin to thereby cause expression of the MAP kinase to be down-regulated. In another aspect of the present invention there is provided a method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: treating the cell with an effective amount of an agent capable of binding to the binding domain of the integrin for the MAP kinase, wherein the agent is other than an antibody or a binding fragment thereof. In a further aspect of the present invention there is provided a method of modulating activity of a cell expressing a MAP kinase with a binding domain for an integrin, comprising: treating the cell with an effective amount of an agent capable of binding to the binding domain of the MAP kinase, wherein the agent is other than an agent comprising a binding domain of the integrin for the MAP kinase or an analog or derivative thereof. In yet another aspect of the invention there is provided a method of modulating the activity of a cell, comprising: contacting the cell with an effective amount of a fusion protein, antibody or other molecule of the invention capable of binding to the binding domain of an integrin for a MAP kinase. Preferably, modulation of the activity of a cell by a method as described herein will comprise treating a mammal in need of such treatment. Accordingly, in a further aspect of the present invention there is provided a method of prophylaxis or treatment of a disease or condition in a mammal wherein modulation of cellular activity is desirable, comprising: administering to the mammal an effective amount of a nucleic acid sequence encoding a MAP kinase or a fragment thereof or a homolog, analog or variant of the MAP kinase or fragment, with a mutagenised binding domain for an integrin or in which the binding domain has been deleted and wherein the nucleic acid sequence is capable of being expressed. In another aspect of the invention there is provided a method of prophylaxis or treatment of a disease or condition in a mammal wherein modulation of cellular activity is desirable, comprising: administering to the mammal an effective amount of a nucleic acid capable of causing the expression of a MAP kinase to which an integrin is able to bind to be down regulated. In a further aspect of the present invention there is provided a method of prophylaxis or treatment of a disease or condition in a mammal wherein modulation of cellular activity is desirable, comprising: administering to the mammal a nucleic acid sequence encoding a fragment of a MAP kinase, or a homolog, analog or variant of the fragment, for being expressed by the cell and capable of binding to the binding domain of an integrin for the MAP kinase. In still another aspect of the present invention there is provided a method of prophylaxis or treatment of a disease or condition in a mammal wherein modulation of cellular activity is desirable, comprising: administering to the mammal an effective amount of a nucleic acid sequence for expression of a polypeptide or a fragment of a MAP kinase within the cell, wherein the polypeptide or fragment is capable of binding to a binding domain of an integrin for the MAP kinase to thereby modulate the cellular activity. In another aspect of the present invention there is provided a method of prophylaxis or treatment of a disease or condition in a mammal wherein modulation of cellular activity is desirable, comprising: administering to the mammal an effective amount of an agent capable of binding to the binding domain of an integrin for a MAP kinase, wherein the agent is other than an antibody or a binding fragment thereof. In another aspect of the present invention there is provided a method of prophylaxis or treatment of a disease or condition in a mammal wherein modulation of cellular activity is desirable, comprising: administering to the mammal an effective amount of an agent capable of binding to the binding domain of a MAP kinase for an integrin wherein the agent is other than an agent comprising the binding domain of the integrin for the MAP kinase or an analog or derivative thereof. In another aspect of there is provided a method of treatment or prophylaxis of a disease or condition in a mammal, wherein the disease or condition is responsive to an agent capable of binding to a binding domain of an integrin for a MAP kinase and wherein the agent comprises a polypeptide or fragment of the invention or a homolog, variant, analog or derivative of such a polypeptide or fragment, and the method comprises administering an effective amount of the agent to the mammal. In yet another aspect of the present invention there is provided a method of treatment or prophylaxis of a disease or condition in a mammal, wherein said condition is responsive to an agent capable of binding to a binding domain of an integrin for a MAP kinase wherein the agent is a fusion protein of the invention. In another aspect there is provided use of a nucleic acid of the invention in the manufacture of a medicament for administration to a mammal in the prophylaxis or treatment of a disease or condition in which modulation of cellular activity is desirable. In a still further aspect of the invention there is provided the use of an agent capable of binding to a binding domain of an integrin for a MAP kinase in the manufacture of a medicament for administration to a mammal for the prophylaxis or treatment of a disease or condition in which modulation of cellular activity is desirable, wherein the agent is other than an antibody or binding fragment thereof. The agent may for instance be a polypeptide, fragment or the like as described herein. In another aspect of the present invention there is provided the use of an agent capable of binding to the binding domain of a MAP kinase for an integrin wherein the agent is other than an agent comprising the binding domain of the integrin for the MAP kinase or an analog or derivative thereof. In another aspect there is provided the use of fusion protein of the invention in the manufacture of a medicament for administration to a mammal for the prophylaxis or treatment of a disease or condition in which modulation of cellular activity is desirable. Methods as described above will typically comprise treatment or prophylaxis of cancer or a condition associated with a predisposition to cancer. The cancer may for instance be selected from the group consisting of cancer of the lip, tongue, salivary glands, gums, floor and other areas of the mouth, oropharynx, nasopharynx, hypopharynx and other oral cavities, oesophagus, stomach, small intestine, duodenum, colon, rectum, gallbladder, pancreas, larynx, trachea, bronchus, lung, breast, uterus, cervix, ovary, vagina, vulva, prostate, testes, penis, bladder, kidney, thyroid and skin. In still another aspect of the present invention there is provided a method of causing an activity of a cell to be upregulated, comprising: contacting the cell with an effective amount of a nucleic acid capable of being expressed within the cell and coding for an agent capable of binding to a binding domain of a MAP kinase for an integrin or a binding domain of the integrin for the MAP kinase. In a still further aspect of the invention there is provided a method of causing an activity of a cell to be upregulated, comprising contacting the cell with an effective amount of an agent capable of binding to a binding domain of a MAP kinase for an integrin or a binding domain of the integrin for the MAP kinase. Typically, the agent will be a polypeptide or fragment of an integrin comprising or consisting of the binding domain of an integrin for a MAP kinase or comprising or consisting of the core amino acid sequence of the binding domain directly involved in the binding to the MAP kinase, or a homolog, variant, analog or derivative of such polypeptide or fragment. Preferably, the polypeptide or fragment will have a length of about 150 amino acids or less, more preferably about 100 or 50 amino acids or less and more usually about 40 amino acids or less. Typically, the length will be between about 5 to about 30 amino acids. Most preferably, the polypeptide will comprise or consist of the amino acid sequence RSKAKWQTGTNPLYR or RSKAKNPLYR. Up regulation of cellular activity is particularly desirable for instance in wound healing, re-endothelialisation within natural or artificial blood vessels, nerve growth and for instance induction of labour. The cellular activity desired to be modulated will typically but not exclusively, be cell growth or proliferation. Indeed, any activity mediated by MAP kinase signalling is expressly included within the scope of the invention. The cell may be any cell type in which functional activity arising from signalling mediated by a MAP kinase may occur. Preferably, the cell will be an epithelial cell and usually, a neoplastic cell. Usually, the MAP kinase will be selected from the group consisting of an extracellular signal-regulated kinase (ERK) and a JNK MAP kinase. Preferably, the MAP kinase is ERK2 or JNK-1. Most preferably, the MAP kinase is ERK2. The mammal may be any mammal treatable with a method of the invention. For instance, the mammal may be a member of the bovine, porcine, ovine or equine families, a laboratory test animal such as a mouse, rabbit, guinea pig, a cat or dog, a primate or a human being. Preferably, the mammal will be a human being. As will be understood, modulation of cellular activity within the context of the invention may be up regulation or down regulation of cellular activity. Reference to “down modulation” or “down-regulating” or the like should be understood to include preventing, reducing or otherwise inhibiting one or more aspects of the activity of the cell. Conversely, reference to “up regulation” or like terms should be understood to include enhancing or increasing one or more aspects of the activity of the cell. In the broadest sense, the term “integrin” unless otherwise specified, is to be taken to encompass an integrin family member or a homolog, derivative, variant or analog of an integrin subunit, or an integrin family member incorporating at least one such homolog, derivative, variant or analog of an integrin subunit. Usually, the integrin will be a member of the αv subfamily. Preferably, the integrin is or incorporates an integrin subunit selected from the group consisting of β3, β5 and β6. Most preferably, the integrin comprises β6. By “binding domain” is meant the minimum length of contiguous amino acid sequence required for binding. By “core amino acid sequence” is meant regions or amino acids of the binding domain that directly participate in the binding as distinct from any amino acids that do not directly participate in the binding interaction. Typically, the core amino acid sequence of the binding domain will comprise regions of the binding domain linked together by a number of intervening amino acids which do not directly participate in the binding. The term “homolog” is to be taken to mean a molecule that has amino acid sequence similarity. The homology between amino acid sequences can be determined by comparing amino acids at each position in the sequences when optimally aligned for the purpose of comparison. The sequences are considered homologous at a position if the amino acids at that position are the same. Typically, a homolog will have an overall amino acid sequence homology of at least about 30% more preferably at least about 50% or 70% and most preferably, greater than about 80%, 90% or 98% sequence homology. Homology with a binding domain may be greater than the overall amino acid sequence homology of the homolog, and will usually be greater than about 60% or 80%, and more usually greater than about 90%, 95% or 98%. The term “analog” is to be taken to mean a molecule that has one or more aspects of biological function characteristic of the molecule on which at least part of the analog is based or which was otherwise utilised in the design or preparation of the analog. An analog may have substantial overall structural similarity with the molecule or only structural similarity with one or more regions or domains thereof responsible for the desired characteristic biological function. By “structural” similarity is meant similarity in shape, conformation and/or other structural features responsible for the provision of the biological function or which otherwise have involvement in the provision of the biological function. Alternatively, it will be understood that with knowledge of the region(s) or domain(s) of a molecule that provide(s) the characteristic biological function, analogs may be designed that while differing in structure nevertheless possess such biological function. Indeed, it is not necessary that an analog have amino acid sequence homology, and an analog may not be proteinaceous at all. An analog may for instance be a mimetic of a molecule. By the term “variant” is meant an isoform, an allelic variant of a gene or region thereof, a naturally occurring mutant form of a gene or region thereof, or a polypeptide or fragment having an amino acid sequence that differs in one or more amino acids but which retains one or more aspects of desired characteristic biological function. This may be achieved by the addition of one or more amino acids to an amino acid sequence, deletion of one or more amino acids from an amino acid sequence and/or the substitution of one or more amino acids with another amino acid or amino acids. Inversion of amino acids and any other mutational change that results in alteration of an amino acid sequence are also encompassed. A variant may be prepared by introducing nucleotide changes in a nucleic acid sequence such that the desired amino acid changes are achieved upon expression of the mutagenised nucleic acid sequence, or for instance by synthesising an amino acid sequence incorporating the desired amino acid changes which possibility is well within the capability of the skilled addressee. Substitution of an amino acid may involve a conservative or non-conservative amino acid substitution. By conservative amino acid substitution is meant replacing an amino acid residue with another amino acid having similar stereochemical properties (eg. structure, charge, acidity or basicity characteristics) and which does not substantially effect conformation or the desired aspect or aspects of characteristic biological function. Preferred variants include ones having amino acid sequences in which one or more amino acids have been substituted with alanine or other neutrally charged amino acid residue(s), or to which one or more such residues have been added. A variant may also incorporate an amino acid or amino acids that are not encoded by the genetic code. By the term “derivative” is meant a molecule that is derived or obtained from another molecule and which retains one or more aspects of characteristic biological function of that molecule. A derivative may for instance arise as a result of the cleavage of the parent molecule, cyclisation and/or coupling with one or more additional moieties that improve solubility, lipophilic characteristics to enhance uptake by cells, stability or biological half-life, decreased cellular toxicity, or for instance to act as a label for subsequent detection or the like. A derivative may also result from post-translational or post-synthesis modification such as the attachment of carbohydrate moieties or chemical reaction(s) resulting in structural modification(s) such as the alkylation or acetylation of amino acid residues or other changes involving the formation of chemical bonds. The term “polypeptide” is used interchangeably herein with “peptide” and encompasses amino acid sequences incorporating only a few amino acid residues or many amino acid residues coupled by peptide bonds. The term “neoplastic cell” is to be taken to mean a cell exhibiting abnormal growth and may or may not be a malignant cell. “Growth” is to be taken in its broadest sense and includes proliferation of the cell. In this regard, an example of abnormal cell growth is the uncontrolled proliferation of a cell. Unless the context clearly requires otherwise, throughout the description and the claims, the words ‘comprise’, ‘comprising’, and the like are to be construed in an inclusive sense as opposed to an exclusive or exhaustive sense; that is to say, in the sense of “including, but not limited to”. The features and advantages of the present invention will become further apparent from the following detailed description of preferred embodiments and the accompanying drawings.
Modified tachykinin receptors
The invention provides a modified tachykinin receptor in which the three amino acids of the DRY sequence that occurs adjacent to the junction of the TM3 domain with intracellular loop 2 are replaced with amino acids whose side chains are neither lipophilic nor contain charged groups. The receptor exhibits similar ligand binding characteristics to the wild type receptor but is incapable or substantially incapable of initiating an endogenous signal. Thus the ligand exhibits no or substantially no intra-cellular coupling of the receptor to the G protein, whereby there is substantially no transduction of ligand binding signals to the cell, The invention also includes fragments of the receptor containing the modified DRY sequence, and polynucleotides that encode the modified receptor as aforesaid. Therapeutic and diagnostic uses for the receptor are disclosed.
1. A mutant tachykinin receptor in which the three amino acids of the DRY sequence that occurs adjacent to the junction of the TM3 domain with 5 intracellular loop 2 are replaced with amino acids whose side chains are neither lipophilic nor contain charged groups, said receptor exhibiting similar ligand binding characteristics to the wild type receptor but exhibiting substantially no intra-cellular coupling of the receptor to the G-protein, whereby there is substantially no transduction of ligand binding 10 signals to the cell; or a fragment of said receptor containing said modified DRY sequence; or an isolated protein or polypeptide containing an amino acid sequence at least 80% identical to the above sequence; or a variant thereof with sequential amino acid deletions from either the C terminus or the N-terminus; or an allelic variant, heterospecific homologue or biologically active proteolytic or other fragment thereof containing said modified DRY sequence. 2. A non-human mammalian receptor according to claim 1. 3. A rat or mouse receptor according to claim 1. 4. A human receptor according to claim 1. 5. A mutant NIL-1 receptor according to claim 1. 6. A mutant NK-2 receptor according to claim 1. 7. A mutant NK-3 receptor according to claim 1. 8. A receptor according to claim 1, wherein the replacement amino acids are selected from G and A. 9. The receptor of claim 8, wherein DRY is replaced by GGA. 10. A tachykinin receptor of SEQ ID No5, or a receptor having at least 80% amino acid identity with the receptor of SEQ ID No5, and that is capable of binding to substance P but is substantially incapable of initiating its endogenous signal, or a fragment of said receptor. 11. An isolated cell membrane incorporating a tachykinin receptor as defined in claim 1. 12. Any of the following: (a) an isolated nucleic acid molecule comprising a polynucleotide that encodes a tachykinin receptor as claimed in claim 1; (b) an isolated nucleic acid molecule comprising a sequence that is hybridizable to the above sequence; (c) a gene which is the result of extending the above sequence or any sequence that is hybridizable to the above sequence; (d) a sequence or gene that is functionally equivalent to the above sequence or to a gene that is an extension of the above sequence, i.e. that is not identical to the sequence or gene referred to but functions biologically as equivalent to the sequence or gene referred to, including any allelic variants and heterospecific mammalian homologues, including artificial or recombinant sequences created from cDNA or genomic DNA; (e) a recombinant vector comprising the above gene sequence; and (f) a host cell transformed with the vector. 13. Any of the following: (a) an isolated nucleic acid molecule having the nucleotide sequence of SEQ ID No6; (b) an isolated nucleic acid molecule comprising a sequence that is hybridizable to the above sequence; (c) a gene which is the result of extending the above sequence or any sequence that is hybridizable to the above sequence; (d) a sequence or gene that is functionally equivalent to the above sequence or to a gene that is an extension of the above sequence, i.e. that is not identical to the sequence or gene referred to but functions biologically as equivalent to the sequence or gene referred to, including any allelic variants and heterospecific mammalian homologues, including artificial or recombinant sequences created from cDNA or genomic DNA; (e) a recombinant vector comprising the above gene sequence; and (f) a host cell transformed with the vector. 14. A method for producing a receptor protein having an amino acid sequenceas defined in claim 1, which method comprises the steps of: (a) inserting said nucleic acid sequence into an appropriate vector; (b) culturing, in an a culture medium, a host cell previously transformed or transfected with the recombinant vector of step (a); (c) harvesting cells containing the receptor protein obtained from step (b); and (d) separating or purifying, from said culture medium or from said host cell, the thus-produced receptor protein. 15. A pharmaceutical composition comprising an effective amount of a modified tachykinin ligand as defined in claim 1 or a nucleic acid sequence encoding said ligand and a pharmaceutically and pharmacologically acceptable carrier. 16. Use of a modified tachykinin receptor as defined in claim 1 in the preparation of a medicament for the treatment or prophylaxis of a condition associated with substance P or other tachykinin (neurokinin) receptor-binding ligand; 17. A method for the treatment or prevention of a condition associated with over-expression or inappropriate expression of an endogenous tachykinin ligand, which method comprises administration to a patient in need thereof of a non-toxic, effective amount of such a modified tachykinin ligand as defined in claim 1. 18. A method for screening for therapeutically active compounds, said method comprising the following steps: (a) providing a cell line expressing a modified tachykinin receptor as defined in claim 1; (b) adding test sample to a solution containing labeled tachykinin ligand and the cell line from step (a); (c) incubating the cell line, test sample and labeled ligand mixture from step (b) to allow binding of said ligand and test sample to the modified tachykinin receptor; (d) optionally, separating the non-bound labeled ligand from the labeled ligand bound to the modified tachykinin receptor; and, if desired, (e) measuring the amount of labeled ligand that is bound to the modified tachykinin receptor. 19. Use of a modified tachykinin receptor as defined in claim 1 as a substitute in an assay to identify and/or evaluate entities that bind to the wild type tachykinin receptor. 20. Use of a modified tachykinin receptor as defined in claim 1 as a substitute in an assay in order to determine the concentration of ligand in body fluids in patients with arthritis, pain, migraine, anxiety, schizophrenia, asthma, rheumatoid arthritis, and in gastrointestinal disorders and diseases of the GI tract. 21. An assay procedure comprising the following steps: (a) providing a cell line is provided that expresses a modified tachykinin receptor as defined in claim 1; (b) labeling the cell line; (c) adding the test sample and labeled cells to a matrix binding SP or other ligand; (d) incubating the labeled cells, test sample and matrix-bound SP or other ligand to allow binding of SP or other ligand and test sample to the expressed modified tachykinin receptor; (e) separating the labelled non-bound cells from the SP or other ligand bound cells; and, if desired, (f) measuring the amount of labelled cells containing the modified tachykinin receptor that has bound to SF or other ligand. 22. Use of a modified tachykinin receptor as defined in claim 1 in protein therapy to reduce the effects of an excess of or inappropriately produced endogenous ligand. 23. A method for treatment of a patient in need thereof, which comprises administering to said patient a composition in the form of an aerosol that comprises a modified tachykinin receptor as defined in claim 1. 24. A method for gene therapy treatment of a patient in need thereof, which comprises administering to said patient a nucleic acid sequence, virus or plasmid encoding a modified tachykinin receptor as defined in claim 1.
<SOH> BACKGROUND TO THE INVENTION <EOH>Tachykinins are important in the mediation of many physiological and pathological processes including inflammation, pain, migraine, headache and allergy induced asthma. They belong to an evolutionary conserved family of peptide neurotransmitters that have an established role in neurotransmission. They share the C-terminal sequence Phe-Xaa-Gly-Leu-Met-NH 2 (SEQ ID NO 12) in which Xaa represents a hydrophobic residue. That sequence is characteristic of tachykinins and believed to be mainly responsible for their biological activity at neurokinin receptors. Mammalian tachykinins include substance P (SP), neurokinin A (NKA) and neurokinin B (NKB) which exert their effects by binding to specific receptors. SP is the most prominent member of the tachykinergic system and is released from sensory nerve endings throughout the body. Its amino acid sequence is: in-line-formulae description="In-line Formulae" end="lead"? H-Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly-Leu-Met-NH 2 (SEQ ID N o 1) in-line-formulae description="In-line Formulae" end="tail"? The amino acid sequence for NKA is: in-line-formulae description="In-line Formulae" end="lead"? H-His-Lys-Thr-Asp-Ser-Phe-Val-Gly-Leu-Met-NH 2 (SEQ ID N o 2) in-line-formulae description="In-line Formulae" end="tail"? The amino acid sequence for NKB is: in-line-formulae description="In-line Formulae" end="lead"? H-Asp-Met-His-Asp-Phe-Phe-Val-Gly-Leu-Met-NH 2 (SEQ ID N o 3) in-line-formulae description="In-line Formulae" end="tail"? SP has been implicated in the pathology of numerous diseases. For example, it has been shown to be involved in the transmission of pain, in conditions associated with vasodilation, smooth muscle contraction, bronchoconstriction, activation of the immune system and neurogenic inflammation. It has been implicated in migraine, as well as in disorders of the central nervous system, such as anxiety and schizophrenia; in respiratory and inflammatory diseases, such as asthma and rheumatoid arthritis; and in gastrointestinal (GI) disorders and diseases of the GI tract, such as ulcerative colitis and Crohn's disease. Tachykinin receptors include NK-1, NK-2 and NK-3 and are membrane proteins of the super-family of guanine nucleotide-binding protein (G-protein)-coupled receptors (GPCR). They have extra-cellular binding sites that have preferential affinities for the ligands SP, NKA and NKB respectively. Like other G-protein coupled receptors, they possess an extra-cellular N-terminus, an intra-cellular C-terminus and seven trans-membrane (TM) α-helices of 20-30 amino acids connected by first, second and third extra-cellular loops and by first, second and third intra-cellular or cytoplasmic loops. The greatest homology is found in the membrane-spanning α-helices of trans-membrane domains TM1-TM7 while the N- and C-termini show greater diversity between the three types of neurokinin receptor. Cloned NK-1 receptors have been reported, including those for the Rana catesbeina , (Simmons et al., Neuroscience, 79: 1219-1229 (1997)), Mus musculus , (Sundelin et al, Eur. J. Biochem. 203: 625-631 (1992)), Rattus norvegicus , (Hershey et al, J. Biol Chem., 266: 4366-4374 (1991) and Yokota et al, J. Biol. Chem., 264: 17649-17652 (1989)), Cavia porcellus , (Gorbulev et al., Biochem. Biophys. Acta 1131: 99-102 (1992)), and Homo sapiens (human), (Takeda et al, Biochem. Biophys. Res. Comm. 179: 1232-1240 (1991) and Fong et al U.S. Pat. No. 5,525,712 and U.S. Pat. No. 5,584,886). Cloned rat and bovine neurokinin-2 receptors have been reported (Y. Sasi et al., Biochem. Biophys. Res. Comm., 165: 695 (1989), and Y. Masu, et al., Nature 329: 836 (1987)). Cloned rat neurokinin-3 receptor has also been reported (R. Shigemoto, et al., J. Biol. Chem., 265:623 (1990)). All three receptors share the signal transduction mechanisms of a G-protein coupled receptor. The receptor is in an OFF state when no ligand is present, but, is triggered into an ON state when an agonist ligand binds to the receptor. The G-protein when in its ON state triggers a downstream signaling pathway or signal cascade. The G-protein comprises α-, β- and γ-units that are bound together in the OFF state, the α-subunit then having GDP bound to it. In the ON state GTP replaces the GDP bound to the α-subunit. The α-subunit becomes dissociated from the β- and γ-subunits and becomes available for activating the signal cascade. After a short period, the GTP becomes hydrolyzed to GDP and the G-protein returns to its non activated OFF state. Hydrolysis provides a negative feedback mechanism that ensures that the G protein is only in its activated ON state for a short period. Various studies have been undertaken, involving different G-protein receptors, to determine how the various regions of the protein structure affect intra-cellular coupling of the receptor to the G-protein and consequential transduction of ligand binding signals to the cell. G-protein receptors have a well-conserved sequence in the second intracellular loop where the loop joins the third trans-membrane domain that is known as the DRY sequence. It has the residues in-line-formulae description="In-line Formulae" end="lead"? 5′-DRYXXV(P)XXPL-3′  (SEQ ID N o 4) in-line-formulae description="In-line Formulae" end="tail"? in which L represents Leu, Ile, Val, Met or Phe and X represents any amino acid. It has been suggested that the DRY sequence contributes to the efficient binding and activation of G-proteins. Fraser et al., Proc. Natl Acad Sci. USA, 85: 5478-5482 (1988) report a change of Asp to Asn at position 130 of the human β-adrenergic receptor (i.e. the D of the DRY sequence) resulting in human β-adrenergic receptor that exhibits high affinity binding of agonist whilst being unable to interact effectively with G-protein. However, a second paper from the same laboratory reports that the previous very high agonist binding efficiency in human β-adrenergic receptor mutated at position 130, upon which the above mentioned conclusion had been based, had not been reproduced (Wang et al., Mol Pharmacol 40(2): 168-79 (1991)). In a review article Savarese and Fraser said that this locus may be important for coupling to some, but not all, G-proteins ( Biochem J., 283: 1-19 (1992)). Moro et al made mutants of the Hm1 muscarinic cholinergic receptor with changes towards the 5′-end of the DRY sequence and found that replacing L at position 131 with A gave the strongest reduction in coupling efficiency ( J. Biol. Chem. 268: 22273-22278 (1993)). Subsequently, Shibata et al made a mutant of the angiotensin II receptor type I in which DRY at positions 125-127 is replaced by GGA and M at position 134 is replaced by A, resulting in uncoupling of the mutant A receptor from G-proteins ( Biochem. Biophys. Res. Com. 218: 383-389 (1996)). The authors concluded that DRY sequence as a whole including the final lipophilic amino acid L serves as a general site for G-protein coupling but they did not go on to consider what effects might be obtained by change confined to the DRY portion of the sequence. Comparing the binding affinities of the two known isoforms of the human NK-1 receptor shows the importance of the cytoplasmic tail. The long form (407 amino acids) and short form (311 amino acids) differ in the length of the C-terminus. The long form has similar substance P binding characteristics to the rat NK-1 receptor, while the short form of the receptor has an apparent substance P binding affinity 10-fold less than the rat NK-1 receptor. Furthermore, studies on these and other receptors have shown that the effect of mutations is unpredictable and specific to certain families of receptors. Hence, a change in one G-protein coupling family will not necessarily have the same effect on another. Other attempts to distinguish protein binding and signaling effects bear this out.
<SOH> SUMMARY OF THE INVENTION <EOH>The invention provides a mutant tachykinin receptor in which the three amino acids of the DRY sequence that occurs adjacent to the junction of the TM3 domain with intracellular loop 2 are replaced with amino acids whose side chains are neither lipophilic nor contain charged groups, said receptor exhibiting similar ligand binding characteristics to the wild type receptor but being incapable or substantially incapable of initiating an endogenous signal. Thus the ligand exhibits no or substantially no intra-cellular coupling of the receptor to the G-protein, whereby there is substantially no transduction of ligand binding signals to the cell. The way in which tachykinin receptors attach to cell membranes, the trans-membrane domains extra-cellular and cytoplasmic loops and the place where the DRY sequence referred to above occurs are apparent by inspection of FIG. 5 of the accompanying drawings. The invention further provides any of the following: a fragment of said mutant tachykinin receptor containing said modified DRY sequence; an isolated protein or polypeptide containing an amino acid sequence at least 95% identical to the above sequence; a variant thereof with sequential amino acid deletions from either the C terminus or the N-terminus; and an allelic variant, heterospecific homologue or biologically active proteolytic or other fragment thereof containing said modified DRY sequence. The invention also comprises an isolated cell membrane in which a modified tachykinin receptor as aforesaid is incorporated as membrane protein. Such cell membrane material finds utility for research and in particular for screening for therapeutically useful compounds as described below. The invention yet further provides any of the following: (a) an isolated nucleic acid molecule comprising a polynucleotide that encodes a modified tachykinin receptor as aforesaid; (b) an isolated nucleic acid molecule comprising a sequence that is hybridizable to the above sequence; (c) a gene which is the result of extending the above sequence or any sequence that is hybridizable to the above sequence; (d) a sequence or gene that is functionally equivalent to the above sequence or to a gene that is an extension of the above sequence, i.e. that is not identical to the sequence or gene referred to but functions biologically as equivalent to the sequence or gene referred to, including any allelic variants and heterospecific mammalian homologues, including artificial or recombinant sequences created from cDNA or genomic DNA; (e) a recombinant vector comprising the above gene sequence; and (f) a host cell transformed with the vector. The invention also provides a method for producing one of the amino acid sequences described herein and especially the receptor protein defined by SEQ ID N o 5, which method comprises the steps of: (a) inserting said nucleic acid sequence into an appropriate vector; (b) culturing in a culture medium a host cell previously transformed or transfected with the recombinant vector of step (a); (c) harvesting cells containing the receptor protein obtained from step (b); and (d) separating or purifying from said culture medium or from said host cells the thus-produced receptor protein. In step (d) of the above method, the receptor protein may be obtained either from the culture medium and/or by lysing the host cell, for example by sonication or osmotic shock. The invention yet further provides a method for screening for therapeutically active compounds, said method comprising the following steps: (a) providing a cell line expressing a modified tachykinin receptor as aforesaid; (b) adding test sample to a solution containing labeled SP or other tachykinin ligand and the cell line from step (a); (c) incubating the cell line, test sample and labeled SP or other ligand mixture from step (b) to allow binding of SP or other ligand and test sample to the modified tachykinin receptor; (d) optionally separating the non-bound labeled SP or other ligand from the labeled SP or other ligand bound to the modified tachykinin receptor and, if desired, (e) measuring the amount of labeled SP or other ligand that is bound to the modified tachykinin receptor. The invention further provides: (a) the use of a modified tachykinin receptor according to the invention in the preparation of a medicament for the treatment or prophylaxis of a condition associated with substance P or other tachykinin (neurokinin) receptor-binding ligand; (b) the use of such a modified tachykinin receptor in therapy; (c) a method for the treatment or prevention of a condition associated with over-expression of an endogenous tachykinin ligand, which method comprises administration to a patient in need thereof of a non-toxic, effective amount of such a modified tackykinin ligand as described above; (d) a composition comprising a modified tachykinin ligand as described above in association with a pharmaceutically and pharmacologically acceptable carrier therefor; and (e) a use, method or composition according to any one of (a) to (d) above, in which the modified tachykinin ligand becomes generated in vivo from a nucleic acid sequence encoding such a ligand.
Reformation of soft soil and system therefor
A system and a method for draining soft soil A bordered with soil B are disclosed. The system includes airtight sheet means 10 which covers the soft soil A to assist in vacuuming the soft soil A. The system further includes water gathering pipes 13 and water drain tank means 16 to receive water from the water gathering pipes 13. The water gathering pipes 13 and the water drain tank means 16 provide water passages and air passages separately so as to expedite the drainage operation.
1. A method for reforming target soft soil by vacuuming and draining the target soft soil covered with airtight sheet means, comprising separating water expelling routes and air expelling routes. 2. A method of claim 1, further comprising installing water drain tank means under water gathering pipes which are in fluid connection with horizontal drains and vertical drains so as to receive underground water from the water gathering pipes. 3. A method of claim 2, further comprising providing separator means which separates underground water coming from said water gathering pipes into water and air and sends the separated water into said water drain tank means. 4. A method of claim 2, further comprising providing water drain pump means to expel the water in said water drain tank means out of the target soft soil. 5. A method of claim 3, further comprising providing water drain pump means to expel the water in said water drain tank means out of the target soft soil. 6. A method of claim 1, further comprising providing first water drain tank means in connection with water gathering pipes, providing second water drain tank means in connection with said first water drain tank means such that water is received into said second water drain tank means from said first water drain tank means via connection pipes, providing said second water drain tank means with water drain pump means, and expelling water from said second water drain tank means with said water drain pump means. 7. A method of claim 2, wherein said horizontal drains and said water gathering pipes each have a water passage and an air passage separately. 8. A drain system for reforming target soft soil by vacuuming and draining the target soft soil, comprising separately provided water expelling routes and air expelling routes. 9. A drain system of claim 8, wherein the system comprises vertical drains installed in said target soft soil, horizontal drains laid on said target soft soil in contact with top portions of said vertical drains, water gathering pipes in contact with said horizontal drains, and water drain tank means installed below and in connection with said water gathering pipes, said water gathering pipes and said water drain tank means each providing a water passage and an air passage separately. 10. A drain system of claim 9, wherein said water gathering pipes and said water drain tank means are connected via separator means. 11. A drain system of claim 9, wherein said water drain tank means is provided with water drain pump means. 12. A drain system of claim 10, wherein said water drain tank means is provided with water drain pump means. 13. A drain system of claim 8, further comprising watertight type vacuum pump means having cooling water circulation tank means. 14. A drain system of claim 8, further comprising vertical pipes installed in the vicinity of the target soft soil to evaporate underground water therefrom. 15. A drain system of claim 14, wherein said vertical pipes are drain pipes. 16. A drain system of claim 15, wherein said drain pipes are provided with air blower means. 17. A drain system of claim 9, further comprising first water drain tank means in connection with said water gathering pipes and second water drain tank means in connection with said first water drain tank means such that water is received into said second water drain tank means from said first water drain tank means via connection pipes, wherein said second water drain tank means is provided with water drain pump means which expels water from said second water drain tank means. 18. A drain system of claim 9, wherein said horizontal drains and said water gathering pipes each have a water passage and an air passage separately.
<SOH> BACKGROUND OF THE INVENTION <EOH>1. Field of the Invention This invention generally relates to a method and a system for reforming soft soil such as muddy soil or swampy soil by draining underground water therefrom. More particularly, this invention relates to a method and a system for efficiently hardening soft soil by adequately separating air passages and water passages in the drainage routes. 2. Background Art JP Patent Application Laid-Open No.11-131465 teaches use of vertical drains which are laid vertically in target soft soil to vacuum the soil and drain underground water. FIG. 11 shows a conventional drain system comprised of vertical drains 1 , horizontal drains 2 laid in contact with the vertical drains 1 , water gathering pipes 3 laid in contact with the horizontal drains 2 , and airtight sheet means 6 which covers target soft soil A after the vertical drains 1 , the horizontal drains 2 and the water gathering pipes 3 have been installed in place. The drain system is further comprised of vacuum tank means 4 placed in connection with the water gathering pipes 3 and vacuum pump means 5 placed in connection with the vacuum tank means 4 . In use of the conventional drain system shown in FIG. 11 , the vacuum pump means 5 vacuums the vacuum tank means 4 . When a check valve (not shown) provided on the vacuum tank means 4 is opened, the water gathering pipes 3 are vacuumed. The vacuuming effect propagates to the horizontal drains 2 and the vertical drains 1 which are in connection with the horizontal drains 2 and reduces their respective inner pressures to below 0.4 atm. The target soft soil A is gradually evacuated from around the vertical drains 1 where air is drawn into the vertical drains 1 . The evacuated regions gradually spread throughout the soft soil A. Spread of the evacuated regions in the soft soil A directs underground water and underground air towards the vertical drains 1 and the water and air drawn into the vertical drains 1 travel up through the vertical drains 1 . The water and air are sucked into the horizontal drains 2 and then into the water gathering pipes 3 . The continued drainage of the target soft soil A further spreads the evacuated regions. The whole of the target soft soil A will eventually be vacuumed to around 0.4 atm, and underground water and air are eventually drained out of the soft soil A, leading to compaction of the soft soil A to a harder and stabler soil state. It is to be noted that in the conventional system the vacuuming routes and water drain routes are common. Therefore, sucked air and water flow together all through the common routes comprised of the vertical drains 1 , the horizontal drains 2 , the water gathering pipes 3 , the vacuum tank means 4 and the vacuum pump means 5 . Initially, underground water and air flow into the water gathering pipes 3 in large quantities from the horizontal drains 2 , which stuffs the water gathering pipes 3 and therefore impedes subsequent vacuuming of the target soft soil A as will be readily understood by those skilled in the art. In addition, as the compaction of the soft soil A progresses and the soft soil A sinks, the vertical distance from the underground water level to the vacuum pump means 5 widens, and efficiency of drainage degrades as will be readily understood by those with ordinary skills.
<SOH> BRIEF DESCRIPTION OF THE DRAWINGS <EOH>FIG. 1 is a schematic diagram showing a drain system of the present invention; FIG. 2 is a schematic diagram showing a vacuum tank of the present invention; FIG. 3 is a schematic diagram partially showing an assistant system of the present invention; FIG. 4 is a schematic diagram partially showing another assistant system of the present invention; FIG. 5 is a schematic diagram showing another drain system of the present invention; FIG. 6 is a schematic diagram showing a first drain tank of the drain system of FIG. 5 ; FIG. 7 is a schematic diagram showing a second drain tank of the drain system of FIG. 5 ; FIG. 8 is a schematic diagram partially showing a connection of a vertical drain and a horizontal drain of the present invention; FIG. 9 is a schematic diagram showing a sectional view of a water gathering pipe of the present invention; FIG. 10 is a schematic diagram showing a sectional view of another water gathering pipe of the present invention; and FIG. 11 is a schematic diagram showing a conventional drain system. detailed-description description="Detailed Description" end="lead"?
Method and apparatus for background segmentation based on motion localization
A system (1000) and method of detecting static background on a video sequence of images with moving foreground objects is described. The method includes localizing moving objects in each frame and training a background model using the rest of the image. The system is also capable of handling occasional background changes and camera movements.
1. A method of extracting a background image, comprising: localizing a moving object in a video sequence based on a change in the moving object over a plurality of frames of the video sequence, the moving object occupying frame areas of changing color; and training a background model for the plurality of frames outside of the frame areas of changing color. 2. The method of claim 1, wherein localizing comprises localizing the moving object using a change detection mask. 3. The method of claim 1, wherein localizing comprises: determining a boundary for the moving object that is of homogenous color; and constructing a hull around the moving object using the boundary. 4. The method of claim 3, wherein determining a boundary comprises: determining a maximum contour of a plurality of contours of the moving object, the maximum contour having the largest area of the plurality of contours; determining other contours of the moving object; and joining the maximum contour with the other contours. 5. The method of claim 4, further comprising: eliminating the smallest contour from joining with the maximum contour. 6. The method of claim 4, wherein joining comprises joining one of the other contours with the maximum contour if the distance between the maximum contour and the one of the other contours is less than a predetermined distance. 7. The method of claim 6, wherein the frames comprise a plurality of pixels and wherein the predetermined distance is based on a probability that a pixel of the plurality of pixels is considered moving given that it corresponds to the moving object. 8. The method of claim 7, wherein the predetermined distance is based on a probability that the pixel is considered moving given that it is static. 9. The method of claim 3, wherein the frames comprise a plurality of pixels and wherein the hull is constructed to contain only pixels of changing colors over consecutive frames. 10. The method of claim 3, wherein constructing the hull comprises: determining all connected components in the boundary, wherein each of the components has a contour having an area; filtering out a smallest area contour; selecting a maximum area contour; and joining the maximum area contour with other contours of the connected components. 11. The method of claim 1, wherein the frames comprise a plurality of pixels and wherein training comprises characterizing a pixel color at a given time with a value based on a state, each pixel corresponding to a state of a plurality of states. 12. The method of claim 11, wherein the plurality of states includes an untrained background state. 13. The method of claim 11, wherein the plurality of states includes a trained background state. 14. The method of claim 11, wherein the plurality of states includes a foreground state. 15. The method of claim 11, wherein the plurality of states includes an unknown background state. 16. The method of claim 11, wherein training comprises: training the background model for the pixel in a foreground; and changing the state of the pixel to an untrained background if the pixel represents a static behavior for a certain period of time. 17. The method of claim 16, further comprising changing the state to a trained background after a predetermined number of two frames. 18. The method of claim 1, wherein the video sequence is recorded with a video camera and wherein the method further comprises: detecting a motion of the video camera; and compensating for the motion of the video camera. 19. The method of claim 18, detecting the motion comprises: selecting a frame feature; and tracking the frame features over the plurality of frames. 20. The method of claim 19, wherein compensating comprises resetting the background model when the motion has stopped. 21. A machine readable medium having stored thereon instructions/which when executed by a processor, cause the processor to perform the following: localizing a moving object in a video sequence based on a change in the moving object over a plurality of frames of the video sequence/the moving object occupying frame areas of changing color; and training a background model for the plurality of frames outside of the frame areas of changing color. 22. The machine readable medium of claim 21, wherein localizing comprises localizing the moving object using a change detection mask. 23. The machine readable medium of claim 21, wherein localizing comprises: determining a boundary for the moving object that is of homogenous color; and constructing a hull around the moving object using the boundary. 24. The machine readable medium of claim 23, wherein determining a boundary comprises: determining a maximum contour of a plurality of contours of the moving object, the maximum contour having the largest area of the plurality of contours; determining other contours of the moving object; and joining the maximum contour with the other contours. 25. The machine readable medium of claim 24, wherein the processor further performs: determining a smallest contour of the plurality of contours; and eliminating the smallest contour from joining with the maximum contour. 26. The machine readable medium of claim 24, wherein joining comprises joining one of the other contours with the maximum contour if the distance between the maximum contour and the one of the other contours is less than a predetermined distance. 27. The machine readable medium of claim 23, wherein the processor performing constructing the hull comprises the processor performing: determining all connected components in the boundary, wherein each of the components has a contour having an area; filtering out a smallest area contour; selecting a maximum area contour; and joining the maximum area contour with other contours of the connected components. 28. The machine readable medium of claim 21, wherein the frames comprise a plurality of pixels and wherein the processor performing training, comprises the processor performing characterizing a pixel color at a given time with a value based on a state, each pixel corresponding to a state of a plurality of states. 29. The machine readable medium of claim 28, wherein the processor performing training comprises the processor performing: training the background model for the pixel in a foreground; and changing the state of the pixel to an untrained background if the pixel represents a static behavior for a certain period of time. 30. The machine readable medium of claim 21, wherein the video sequence is recorded with a video camera and wherein the processor further performs: detecting a motion of the video camera; and compensating for the motion of the video camera. 31. The machine readable medium of claim 30, wherein the processor performing detecting the motion comprises the processor performing the following: selecting a frame feature; and tracking the frame features over the plurality of frames. 32. The machine readable medium of claim 30, wherein the processor performing compensating comprises the processor performing the following: resetting the background model when the motion has stopped. 33. An apparatus for extracting a background image, comprising: means for localizing a moving object in a video sequence based on a change in the moving object over a plurality of frames of the video sequence, the moving object occupying frame areas of changing color; and means for training a background model for the plurality of frames outside of the frame areas of changing color. 34. The apparatus of claim 33, wherein the means for localizing comprises: means for determining a boundary for the moving object that is of homogenous color; and means for constructing a hull around the moving object using the boundary. 35. The apparatus of claim 33, wherein the video sequence is recorded with a video camera and wherein the apparatus further comprises: means for detecting a motion of the video camera; and means for compensating for the motion of the video camera. 36. An apparatus for extracting a background image, comprising: a processor to execute one or more routines to localize a moving object in a video sequence based on a change in the moving object over a plurality of frames of the video sequence, the moving object occupying frame areas of changing color, and to train a background model for the plurality of frames outside of the frame areas of changing color; and a storage device coupled with the processor, the storage device having stored therein the one or more routines to localize the moving object and train the background model. 37. The apparatus of claim 36, wherein the processor executes one or more routines to localize the moving object using a change detection mask. 38. The apparatus of claim 36, wherein the processor executes one or more routines to determine a boundary for the moving object that is of homogenous color and to construct a hull around the moving object using the boundary. 39. The apparatus of claim 36, further comprising a display coupled with the processor to display the plurality of frames of the video sequence. 40. The apparatus of claim 36, further comprising a camera coupled with the processor to record the plurality of frames of the video sequence. 41. The apparatus of claim 40, wherein the processor executes one or more routines to detect a motion of the video camera to compensate for the motion of the video camera.
<SOH> BACKGROUND OF THE INVENTION <EOH>Video conferencing and automatic video surveillance has been growing area driven by the increasing availability of lower priced systems and improvements in motion detection technology. Video display technology provides for the display of sequences of images through a display image rendering device such as a computer display. The sequence of images is time varying such that it can adequately represent motion in a scene. A frame is a single image in the sequence of images that is sent to the monitor. Each frame is composed of picture elements (pels or pixels) that are the basic unit of programming color in an image or frame. A pixel is the smallest area of a monitor's screen that can be turned on or off to help create the image with the physical size of a pixel depending on the resolution of the computer display. Pixels may be formed into rows and columns of a computer display in order to render a frame. If the frame contains a color image, each pixel may be turned on with a particular color in order to render the image. The specific color that a pixel describes is some blend of components of the color spectrum such as red, green, and blue. Video sequences may contain both stationary objects and moving objects. Stationary objects are those that remain stationary from one frame to another. As such, the pixels used to render a stationary object's colors remain substantially the same over consecutive frames. Frame regions containing objects with stationary color are referred to as background. Moving objects are those that change position in a frame with respect to a previous position within an earlier frame in the image sequence. If an object changes its position in a subsequent frame with respect to its position in a preceding frame, the pixels used to render the object's image will also change color over the consecutive frames. Such frame regions are referred to as foreground. Applications such as video display technology often rely on the detection of motion of objects in video sequences. In many systems, such detection of motion relies on the technique of background subtraction. Background subtraction is a simple and powerful method of identifying objects and events of interest in a video sequence. An essential stage of background subtraction is training a background model to learn the particular environment. Most often this implies acquiring a set of images of a background for subsequent comparison with test images where foreground objects might be present. However this approach experiences problems in applications where the background is not available or changes rapidly. Some prior art methods that deal with these problems are often referred to as background segmentation. The approaches to the task of background segmentation can be roughly classified into two stages: motion segmentation and background training. Motion segmentation is used to find regions in each frame of an image sequence that correspond to moving objects. Motion segmentation starts from a motion field obtained from optical flow calculated on two consecutive frames. The motion field is divided into two clusters using k-means. The largest group is considered a background. Background training trains background models on the rest of the image. Model-based background extraction extracts background from “museum-like” color images based on assumptions about image properties. This includes small numbers of objects on a background that is relatively smooth with spatial color variations and slight textures. The problem with these prior background segmentation solutions is that they propose pixel-based approaches to motion segmentation. A pixel-based approach analyses each pixel to make a decision whether it belongs to background or not. Hence, the time T of processing each pixel (T) is the sum of motion detection time (T 1 ) and background training time (T 2 ). If a frame consists of N pixels then the time of processing a single frame is T*N. Such an approach may be robust but it is very time-consuming.
<SOH> BRIEF DESCRIPTION OF THE DRAWINGS <EOH>The present invention is illustrated by way of example and not intended to be limited by the figures of the accompanying drawings. FIG. 1 illustrates one embodiment of a method for extracting a background image from a video sequence. FIG. 2A illustrates an exemplary frame from a video sequence. FIG. 2B illustrates another exemplary frame from the video sequence subsequent to the frame of FIG. 2A . FIG. 2C illustrates an exemplary embodiment of a change detection image. FIG. 2D illustrates an exemplary embodiment of the border contours of the change detection image of FIG. 2C . FIG. 2E illustrates an exemplary embodiment of hull construction. FIG. 3 illustrates one embodiment of an iterative construction of a hull. FIG. 4 illustrates one embodiment of a background training scheme. FIG. 5 illustrates an exemplary embodiment of the relative dispersion of running averages depending on a. FIG. 6 illustrates exemplary features to track on an exemplary frame background. FIG. 7 illustrates one embodiment of camera motion detection and compensation. FIG. 8 is an exemplary illustration of the percent of moving pixels segmented by a motion localization algorithm. FIG. 9 is an exemplary illustration of the percent of background pixels segmented as foreground obtained with a motion localization algorithm. FIG. 10 illustrates one embodiment of a computer system with a camera. detailed-description description="Detailed Description" end="lead"?