text
stringlengths
0
1.67M
<SOH> SUMMARY OF THE INVENTION <EOH>In general, the invention provides self-encoding analytic chemical sensor arrays comprising a substrate with a surface comprising discrete sites and a population of microspheres comprising at least a first and a second subpopulation, wherein each subpopulation comprises at least one reporter dye. The reporting dye has a first characteristic optical response signature when subjected to excitation light energy in the presence of a reference analyte, and the microspheres are distributed on the surface. The beads may further comprise a bioactive agent. In an additional aspect, the invention provides methods of detecting a target analyte in a sample comprising contacting the sample with an sensor array. The sensor array comprises a substrate with a surface comprising discrete sites and a population of microspheres. The microspheres comprise at least a first and a second subpopulation, each subpopulation comprising a bioactive agent and at least one reporter dye. The reporting dye has a first characteristic optical response signature when subjected to excitation light energy in the presence of a reference analyte and the microspheres are distributed on the surface. The presence or absence (or quantity) of the analyte is then detected. The methods may further comprise identifying the location of each bioactive agent on said substrate by adding the reference analyte. In a further aspect, the invention provides methods for reducing the signal-to-noise ratio in the characteristic optical response signature of a sensor array having a subpopulations of array elements. The methods comprise decoding the array so as to identify the location of each sensor element within each sensor subpopulation within the array and measuring the characteristic optical response signature of each sensor element in the array. The baseline of the optical response signature is then adjusted for each sensor element in said array, and the baseline-adjusted characteristic optical response signature of all sensor elements within each of the sensor subpopulations is summed. The characteristic optical response signature of each sensor subpopulation as a summation of said baseline-adjusted characteristic optical response signatures of all sensor elements within each of said subpopulations is then reported. In an additional aspect, the invention provides methods for amplifying the characteristic optical response signature of a sensor array having subpopulations of array elements. The methods comprise decoding the array so as to identify the location of each sensor element within each sensor subpopulation within the array and measuring a characteristic optical response signature of each sensor element in the array. The baseline of the optical response signature for each sensor element in said array is then adjusted. The baseline-adjusted characteristic optical response signature of all sensor elements within each of the sensor subpopulations is then summed and the characteristic optical response signature of each sensor subpopulation as a summation of the baseline-adjusted characteristic optical response signatures of all sensor elements within each of the subpopulations is reported. The above and other features of the invention including various novel details of construction and combinations of parts, and other advantages, will now be more particularly described with reference to the accompanying drawings and pointed out in the claims. It will be understood that the particular method and device embodying the invention are shown by way of illustration and not as a limitation of the invention. The principles and features of this invention may be employed in various and numerous embodiments without departing from the scope of the invention. In an additional aspect the invention provides a method of detecting a target analyte comprising providing a first classifier for the response of a first population of sensors from a first pool of sensors to a first target analyte and distributing a second population of sensors from the first pool of sensors on an array. The method further includes and determining the response of the second population of sensors to a sample, wherein the response of the second population resembles the first classifier for the response of the first population to the first target analyte, thereby indicating the presence of the first target analyte in the sample. In addition the invention provides a method of detecting a target analyte comprising providing a first and second classifier for the response of a first and a second population of sensors from first and second pools of sensors, respectively, to a first target analyte and distributing third and fourth populations of sensors from the first and second pools of sensors, respectively, on an array. The method further includes determining the response of the third and fourth population of sensors to a sample, wherein the response of the third and fourth populations resembles the first and second classifiers for the response of the first and second populations, respectively, to the first target analyte, thereby indicating the presence of the first target analyte in the sample. In addition the invention provides a method of making an array comprising providing a population of microspheres, wherein the microspheres comprise an optical signature, contacting the microspheres with a sample comprising a target analytem recording the response of the microspheres to the target analyte, and generating a classifier for the response of the microspheres to the target analyte. The method further includes distributing the microspheres on a substrate with a surface comprising discrete sites.
Whole cell engineering by mutagenizing a substantial portion of a starting genome combining mutations and optionally repeating
This invention relates to the field of cellular and whole organism engineering. Specifically, this invention relates to a cellular transformation, directed evolution, and screening method for creating novel transgenic organisms having desirable properties. Thus in one aspect, this invention relates to a method of generating a transgenic organism, such as a microbe or a plant, having a plurality of traits that are diffenentially activatable.
1. A method for identifying proteins by differential labeling of peptides, the method comprising the following steps: (a) providing a sample comprising a polypeptide; (b) providing a plurality of labeling reagents which differ in molecular mass that can generate differential labeled peptides that do not differ in chromatographic retention properties and do not differ in ionization and detection properties in mass spectrographic analysis, wherein the differences in molecular mass are distinguishable by mass spectrographic analysis; (c) fragmenting the polypeptide into peptide fragments by enzymatic digestion or by non-enzymatic fragmentation; (d) contacting the labeling reagents of step (b) with the peptide fragments of step (c), thereby labeling the peptides with the differential labeling reagents; (e) separating the peptides by chromatography to generate an eluate; (f) feeding the eluate of step (e) into a mass spectrometer and quantifying the amount of each peptide and generating the sequence of each peptide by use of the mass spectrometer; (g) inputting the sequence to a computer program product which compares the inputted sequence to a database of polypeptide sequences to identify the polypeptide from which the sequenced peptide originated. 2. The method of claim 1, wherein the sample of step (a) comprises a cell or a cell extract. 3. The method of claim 1, further comprising providing two or more samples comprising a polypeptide. 4. The method of claim 3, wherein one sample is derived from a wild type cell and one sample is derived from an abnormal or a modified cell. 5. The method of claim 4, wherein the abnormal cell is a cancer cell. 6. The method of claim 1, further comprising purifying or fractionating the polypeptide before the fragmenting of step (c). 7. The method of claim 1, further comprising purifying or fractionating the polypeptide before the labeling of step (d). 8. The method of claim 1, further comprising purifying or fractionating the labeled peptide before the chromatography of step (e). 9. The method of claim 6, claim 8 or claim 8, wherein the purifying or fractionating comprises a method selected from the group consisting of size exclusion chromatography, size exclusion chromatography, HPLC, reverse phase HPLC and affinity purification. 10. The method of claim 1, further comprising contacting the polypeptide with a labeling reagent of step (b) before the fragmenting of step (c). 11. The method of claim 1, wherein the labeling reagent of step (b) comprises the general formulae selected from the group consisting of: i. ZAOH and ZBOH, to esterify peptide C-terminals and/or Glu and Asp side chains; ii. ZANH2 and ZBNH2, to form amide bond with peptide C-terminals and/or Glu and Asp side chains; and iii. ZACO2H and ZBCO2H. to form amide bond with peptide N-terminals and/or Lys and Arg side chains; wherein ZA and ZB independently of one another comprise the general formula R-Z1-A1-Z2-A2-Z3-A3-Z4-A4-, Z1, Z2, Z3, and Z4 independently of one another, are selected from the group consisting of nothing, 0, OC(O), OC(S), OC(O)O, OC(O)NR, OC(S)NR, OSiRR1, S, SC(O), SC(S), SS, S(O), S(O2), NR, NRR1+, C(O), C(O)O, C(S), C(S)O, C(O)S, C(O)NR, C(S)NR, SiRR1, (Si(RR1)O)n, SnRR1, Sn(RR1)O, BR(OR1), BRR1, B(OR)(OR1), OBR(OR1, OBRR1, and OB(OR)(OR1), and R and R1 is an alkyl group, A1, A2, A3, and A4 independently of one another, are selected from the group consisting of nothing or (CRR1)n, wherein R, R1, independently from other R and R1 in Z1 to Z4 and independently from other R and R1 in A1 to A4, are selected from the group consisting of a hydrogen atom, a halogen atom and an alkyl group; n in Z1to Z4, independent of n in A1 to A4, is an integer having a value selected from the group consisting of 0 to about 51; 0 to about 41; 0 to about 31; 0 to about 21, 0 to about 11 and 0 to about 6. 12. The method of claim 11, wherein the alkyl group is selected from the group consisting of an alkenyl, an alkynyl and an aryl group. 13. The method of claim 11, wherein one or more C—C bonds from (CRR1)n are replaced with a double or a triple bond, 14. The method of claim 13, wherein an R or an R1 group is deleted. 15. The method of claim 13, wherein (CRR1)n is selected from the group consisting of an o-arylene, an m-arylene and a p-arylene, wherein each group has none or up to 6 substituents. 16. The method of claim 13, wherein (CRR1)n is selected from the group consisting of a carbocyclic, a bicyclic and a tricyclic fragment, wherein the fragment has up to 8 atoms in the cycle with or without a heteroatom selected from the group consisting of an O atom, a N atom and an S atom. 17. The method of claim 1, wherein two or more labeling reagents have the same structure but a different isotope composition. 18. The method of claim 11, wherein ZA has the same structure as ZB, but ZA has a different isotope composition than ZB. 19. The method of claim 17, wherein the isotope is boron-10 and boron-11. 20. The method of claim 17, wherein the isotope is carbon-12 and carbon-13. 21. The method of claim 17, wherein the isotope is nitrogen-14 and nitrogen-15. 22. The method of claim 17, wherein the isotope is sulfur-32 and sulfur-34. 23. The method of claim 17, wherein, where the isotope with the lower mass is x and the isotope with the higher mass is y, and x and y are integers, x is greater than y. 24. The method of claim 17, wherein x and y are between 1 and about 11, between 1 and about 21, between 1 and about 31, between 1 and about 41, or between 1 and about 51. 25. The method of claim 1, wherein the labeling reagent of step (b) comprises the general formulae selected from the group consisting of: i. CD3(CD2)nOH/CH3(CH2)nOH, to esterify peptide C-terminals, where n=0, 1, 2 or y; ii. CD3(CD2)nNH2 CH3(CH2)nNH2, to form amide bond with peptide C-terminals, where n=0, 1, 2 or y; and iii. D(CD2)nCO2H/H(CH2)nCO2H, to form amide bond with peptide N-terminals, where n=0, 1, 2 or y; wherein D is a deuteron atom, and y is an integer selected from the group consisting of about 51; about 41; about 31; about 21, about 11; about 6 and between about 5 and 51. 26. The method of claim 1, wherein the labeling reagent of step (b) comprises the general formulae selected from the group consisting of: i. ZAOH and ZBOH to esterify peptide C-terminals; ii. ZANH2/ZBNH2 to form an amide bond with peptide C-terminals; and iii. ZACO2H/ZBCO2H to form an amide bond with peptide N-terminals; wherein ZA and ZB have the general formula R-Z1-A1-Z2-A2-Z3-A3-Z4-A4-Z1, Z2, Z3, and Z4, independently of one another, are selected from the group consisting of nothing, 0, OC(O), OC(S), OC(O)O, OC(O)NR, OC(S)NR, OSiRR1, S, SC(O), SC(S), SS, S(O), S(O2), NR, NRR1+, C(O), C(O)O, C(S), C(S)O, C(O)S, C(O)NR, C(S)NR, SiRR1, (Si(RR1)O)n, SnRR1, Sn(RR1)O, BR(OR1), BRR1, B(OR)(OR1), OBR(OR1), OBRR1, and OB(OR)(OR1); A1, A2, A3, and A4, independently of one another, are selected from the group consisting of nothing and the general formulae (CRR1)n, and, R and R1 is an alkyl group. 27. The method of claim 26, wherein a single C—C bond in a (CRR1)n group is replaced with a double or a triple bond. 28. The method of claim 27, wherein R and R1 are absent. 29. The method of claim 27, wherein (CRR1.)n comprises a moiety selected from the group consisting of an o-arylene, an m-arylene and ap-arylene, wherein the group has none or up to 6 substituents. 30. The method of claim 27, wherein the group comprises a carbocyclic, a bicyclic, or a tricyclic fragments with up to 8 atoms in the cycle, with or without a heteroatom selected from the group consisting of an O atom, an N atom and an S atom. 31. The method of claim 26, wherein R, R1, independently from other R and R1 in Z1-Z4 and independently from other R and R1 in A1-A4, are selected from the group consisting of a hydrogen atom, a halogen and an alkyl group. 32. The method of claim 31, wherein the alkyl group is selected from the group consisting of an alkenyl, an alkynyl and an aryl group. 33. The method of claim 26, wherein n in Z1-Z4 is independent of n in A1-A4 and is an integer selected from the group consisting of about 51; about 41; about 31; about 21, about 11 and about 6. 34. The method of claim 26, wherein ZA has the same structure a ZB but ZA further comprises x number of —CH2— fragment(s) in one or more A1-A4 fragments, wherein x is an integer. 35. The method of claim 26, wherein ZA has the same structure a ZB but ZA further comprises x number of —CF2— fragment(s) in one or more A1-A4 fragments, wherein x is an integer. 36. The method of claim 26, wherein ZA comprises x number of protons and ZB comprises y number of halogens in the place of protons, wherein x and y are integers. 37. The method of claim 26, wherein ZA contains x number of protons and ZB contains y number of halogens, and there are x−y number of protons remaining in one or more A1-A4 fragments, wherein x and y are integers 38. The method of claim 26, wherein ZA further comprises x number of-O— fragment(s) in one or more A1-A4 fragments, wherein x is an integer. 39. The method of claim 26, wherein ZA further comprises x number of —S— fragment(s) in one or more A1-A4 fragments, wherein x is an integer. 40. The method of claim 26, wherein ZA further comprises x number of —O— fragment(s) and ZB further comprises y number of —S— fragment(s) in the place of —O— fragment(s), wherein x and y are integers. 41. The method of claim 26, wherein ZA further comprises x−y number of —O— fragment(s) in one or more A1-A4 fragments, wherein x and y are integers. 42. The method of claim 37, claim 40 or claim 41, wherein x and y are integers selected from the group consisting of between 1 about 51; between 1 about 41; between 1 about 31; between 1 about 21, between 1 about 11 and between 1 about 6, wherein x is greater than y. 43. The method of claim 1, wherein the labeling reagent of step (b) comprises the general formulae selected from the group consisting of: i. CH3(CH2)nOH/CH3(CH2)n+mOH, to esterify peptide C-terminals, where n=0, 1, 2, . . . , y; m=1, 2, . . . y; ii. CH3(CH2)n NH2/CH3(CH2)n+mNH2, to form amide bond with peptide C-terminals, where n=0, 1, 2, . . . , y; m=1, 2, . . . , y; and, iii. H(CH2)nCO2H/H(CH2)n+mCO2H, to form amide bond with peptide N-terminals, where n=0, 1, 2, . . . , y; m=1, 2, . . . , y; wherein n, m and y are integers. 44. The method of claim 43, wherein n, m and y are integers selected from the group consisting of about 51; about 41; about 31; about 21, about 11; about 6 and between about 5 and 51. 45. The method of claim 1, wherein the separating of step (e) comprises a liquid chromatography system. 46. The method of claim 1, wherein the liquid chromatography system comprises a multidimensional liquid chromatography. 47. The method of claim 1, wherein the mass spectrometer comprises a tandem mass spectrometry device. 48. The method of claim 1, further comprising quantifying the amount of each polypeptide. 49. The method of claim 1, further comprising quantifying the amount of each peptide. 50. A method for defining the expressed proteins associated with a given cellular state, the method comprising the following steps: (a) providing a sample comprising a cell in the desired cellular state; (b) providing a plurality of labeling reagents which differ in molecular mass but do not differ in chromatographic retention properties and do not differ in ionization and detection properties in mass spectrographic analysis, wherein the differences in molecular mass are distinguishable by mass spectrographic analysis; (c) fragmenting polypeptides derived from the cell into peptide fragments by enzymatic digestion or by non-enzymatic fragmentation; (d) contacting the labeling reagents of step (b) with the peptide fragments of step (c), thereby labeling the peptides with the differential labeling reagents; (e) separating the peptides by chromatography to generate an eluate; (f) feeding the eluate of step (e) into a mass spectrometer and quantifying the amount of each peptide and generating the sequence of each peptide by use of the mass spectrometer; (g) inputting the sequence to a computer program product which compares the inputted sequence to a database of polypeptide sequences to identify the polypeptide from which the sequenced peptide originated, thereby defining the expressed proteins associated with the cellular state. 51. A method for quantifying changes in protein expression between at least two cellular states, the method comprising the following steps: state; (b) providing a plurality of labeling reagents which differ in molecular mass but do not differ in chromatographic retention properties and do not differ in ionization and detection properties in mass spectrographic analysis, wherein the differences in molecular mass are distinguishable by mass spectrographic analysis; (c) fragmenting polypeptides derived from the cells into peptide fragments by enzymatic digestion or by non-enzymatic fragmentation; (d) contacting the labeling reagents of step (b) with the peptide fragments of step (c), thereby labeling the peptides with the differential labeling reagents, wherein the labels used in one same are different from the labels used in other samples; (e) separating the peptides by chromatography to generate an eluate; (f) feeding the eluate of step (e) into a mass spectrometer and quantifying the amount of each peptide and generating the sequence of each peptide by use of the mass spectrometer; (g) inputting the sequence to a computer program product which identifies from which sample each peptide was derived, compares the inputted sequence to a database of polypeptide sequences to identify the polypeptide from which the sequenced peptide originated, and compares the amount of each polypeptide in each sample, thereby quantifying changes in protein expression between at least two cellular states. 52. A method for identifying proteins by differential labeling of peptides, the method comprising the following steps: (a) providing a sample comprising a polypeptide; (b) providing a plurality of labeling reagents which differ in molecular mass but do not differ in chromatographic retention properties and do not differ in ionization and detection properties in mass spectrographic analysis, wherein the differences in molecular mass are distinguishable by mass spectrographic analysis; (c) fragmenting the polypeptide into peptide fragments by enzymatic digestion or by non-enzymatic fragmentation; (d) contacting the labeling reagents of step (b) with the peptide fragments of step (c), thereby labeling the peptides with the differential labeling reagents; (e) separating the peptides by multidimensional liquid chromatography to generate an eluate; (f) feeding the eluate of step (e) into a tandem mass spectrometer and quantifying the amount of each peptide and generating the sequence of each peptide by use of the mass spectrometer; (g) inputting the sequence to a computer program product which compares the inputted sequence to a database of polypeptide sequences to identify the polypeptide from which the sequenced peptide originated. 53. A chimeric labeling reagent comprising (a) a first domain comprising a biotin; and (b) a second domain comprising a reactive group capable of covalently binding to an amino acid, wherein the chimeric labeling reagent comprises at least one isotope. 54. The chimeric labeling reagent of claim 53, wherein the isotope is in the first domain. 55. The chimeric labeling reagent of claim 54, wherein the isotope is in the biotin. 56. The chimeric labeling reagent of claim 53, wherein the isotope is in the second domain. 57. The chimeric labeling reagent of claim 53, wherein the isotope is selected from the group consisting of a deuterium isotope, a boron-10 or boron-11 isotope, a carbon-12 or a carbon-13 isotope, a nitrogen-14 or a nitrogen-15 isotope and a sulfur-32 or a sulfur-34 isotope. 58. The chimeric labeling reagent of claim 53 comprising two or more isotopes. 59. The chimeric labeling reagent of claim 53, wherein the reactive group capable of covalently binding to an amino acid is selected from the group consisting of a succimide group, an isothiocyanate group and an isocyanate group. 60. The chimeric labeling reagent of claim 53, wherein the reactive group capable of covalently binding to an amino acid binds to a lysine or a cysteine. 61. The chimeric labeling reagent of claim 53, further comprising a linker moiety linking the biotin group and the reactive group. 62. The chimeric labeling reagent of claim 53; wherein the linker moiety comprises at least one isotope. 63. The chimeric labeling reagent of claim 53, wherein the linker is a cleavable moiety. 64. The chimeric labeling reagent of claim 53, wherein the linker can be cleaved by enzymatic digest. 65. The chimeric labeling reagent of claim 53, wherein the linker can be cleaved by reduction. 66. A method of comparing relative protein concentrations in a sample comprising (a) providing a plurality of differential small molecule tags, wherein the small molecule tags are structurally identical but differ in their isotope composition, and the small molecules comprise reactive groups that covalently bind to cysteine or lysine residues or both; (b) providing at least two samples comprising polypeptides; (c) attaching covalently the differential small molecule tags to amino acids of the polypeptides; (d) determining the protein concentrations of each sample in a tandem mass spectrometer; and, (d) comparing relative protein concentrations of each sample. 67. The method of claim 66, wherein the sample comprises a complete or a fractionated cellular sample. 68. The method of claim 66, wherein differential small molecule tags comprise a chimeric labeling reagent comprising (a) a first domain comprising a biotin; and, (b) a second domain comprising a reactive group capable of covalently binding to an amino acid, wherein the chimeric labeling reagent comprises at least one isotope. 69. The method of claim 68, wherein the isotope is selected from the group consisting of a deuterium isotope, a boron-10 or boron-lI 1 isotope, a carbon-12 or a carbon-13 isotope, a nitrogen-14 or a nitrogen-15 isotope and a sulfur-32 or a sulfur-34 isotope. 70. The method of claim 68, wherein the chimeric labeling reagent comprises two or more isotopes. 71. The method of claim 68, wherein the reactive group capable of covalently binding to an amino acid is selected from the group consisting of a succimide group, an isothiocyanate group and an isocyanate group. 72. A method of comparing relative protein concentrations in a sample comprising (a) providing a plurality of differential small molecule tags, wherein the differential small molecule tags comprise a chimeric labeling reagent comprising (i) a first domain comprising a biotin; and, (ii) a second domain comprising a reactive group capable of covalently binding to an amino acid, wherein the chimeric labeling reagent comprises at least one isotope; (b) providing at least two samples comprising polypeptides; (c) attaching covalently the differential small molecule tags to amino acids of the polypeptides; (d) isolating the tagged polypeptides on a biotin-binding column by binding tagged polypeptides to the column, washing non-bound materials off the column, and eluting tagged polypeptides off the column; (e) determining the protein concentrations of each sample in a tandem mass spectrometer; and, (f) comparing relative protein concentrations of each sample. 73. A method of producing an improved organism having a desirable trait comprising: a) obtaining an initial population of organisms, b) generating a set of mutagenized organisms, such that when all the genetic mutations in the set of mutagenized organisms are taken as a whole, there is represented a set of substantial genetic mutations, and c) detecting the presence of said improved organism. 74. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of a knocking out of at least 15 different genes. 75. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of a knocking out of at least 50 different genes. 76. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of a knocking out of at least 100 different genes. 77. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of an introduction of at least 15 different genes. 78. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of an introduction of at least 50 different genes. 79. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of an introduction of at least 100 different genes. 80. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of an alteration in the expression of at least 15 different genes. 81. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of an alteration in the expression of at least 50 different genes. 82. The method of claim 73, wherein the set of substantial genetic mutations in step b) is comprised of an alteration in the expression of at least 100 different genes. 83. A method of producing an improved organism having a desirable trait comprising: a) obtaining an initial population of organisms, b) generating a set of mutagenized organisms each having at least one genetic mutation, such that when all the genetic mutations in the set of mutagenized organisms are taken as a whole, there is represented a set of substantial genetic mutations c) detecting the manifestation of at least two genetic mutations, d) introducing at least two detected genetic mutations into one organism, and e) optionally repeating any of steps a), b), c), and d). 84. The method of claim 83, wherein step d) is comprised of a knocking out of at least 15 different genes in one organism. 85. The method of claim 83, wherein step d) is comprised of a knocking out of at least 50 different genes in one organism. 86. The method of claim 83, wherein step d) is comprised of a knocking out of at least 100 different genes in one organism. 87. The method of claim 83, wherein step d) is comprised of an introduction of at least 15 different genes into one organism. 88. The method of claim 83, wherein step d) is comprised of an introduction of at least 50 different genes into one organism. 89. The method of claim 83, wherein step d) is comprised of an introduction of at least 100 different genes into one organism. 90. The method of claim 83, wherein step d) is comprised of an alteration in the expression of at least 15 different genes in one organism. 91. The method of claim 83, wherein step d) is comprised of an alteration in the expression of at least 50 different genes in one organism. 92. The method of claim 83, wherein step d) is comprised of an alteration in the expression of at least 100 different genes in one organism. 93. A method for identifying a gene that alters a trait of an organism, comprising: a) obtaining an initial population of organisms, b) generating a set of mutagenized organisms, such that when all the genetic mutations in the set of mutagenized organisms are taken as a whole, there is represented a set of substantial genetic mutations, and c) detecting the presence an organism having said altered trait, and d) determining the nucleotide sequence of a gene that has been mutagenized in the organism having the altered trait. 94. A method for producing an organism with an improved trait, comprising: a) functionally knocking out an enogenous gene in a substantially clonal population of organisms; b) transferring a library of altered genes into the substantially clonal population of organisms, wherein each altered gene differs from the endogenous gene at only one codon; c) detecting a mutagenized organism having an improved trait; and d) determining the nucleotide sequence of an gene that has been transferred into the detected organism. 95. A method of introducing differentially activatable stacked traits into a transgenic cell or organism, which method is comprised of the following steps: a) obtaining an initial cell or organism; b) introducing into the working cell or organism a plurality of traits (stacked traits), including selectively and differentially activatable traits, whereby serviceable traits for this purpose include traits conferred by genes and traits conferred by gene pathways; c) analyzing the information obtained from steps a) and b), and d) optionally repeating any number or all of the steps of a), b), c), and d); 96. The method of claim 95, wherein step a) also includes holistic monitoring of the strain or organism whereby holistic monitoring can include the detection and/or measurement of all detectable functions and physical parameters (such as but not limited to morphology, behavior, growth, responsiveness to stimuli [e.g., antibiotics, different environment, etc.], and profiles of all detectable molecules, including molecules that are chemically at least in part a nucleic acids, proteins, carbohydrates, proteoglycans, glycoproteins, or lipids) 97. The method of claim 95, wherein step d) also includes holistic monitoring of the strain or organism whereby holistic monitoring can include the detection and/or measurement of all detectable functions and physical parameters (such as but not limited to morphology, behavior, growth, responsiveness to stimuli [e.g., antibiotics, different environment, etc.], and profiles of all detectable molecules, including molecules that are chemically at least in part a nucleic acids, proteins, carbohydrates, proteoglycans, glycoproteins, or lipids) 98. The method of claim 95, wherein step a) and d) include holistic monitoring of the strain or organism whereby holistic monitoring can include the detection and/or measurement of all detectable functions and physical parameters (such as but not limited to morphology, behavior, growth, responsiveness to stimuli [e.g., antibiotics, different environment, etc.], and profiles of all detectable molecules, including molecules that are chemically at least in part a nucleic acids, proteins, carbohydrates, proteoglycans, glycoproteins, or lipids) 99. The method of claim 95, wherein step b) includes the introduction of at least 15 stacked traits 100. The method of claim 95, wherein step b) includes the introduction of at least 50 stacked traits 101. The method of claim 95, wherein step b) includes the introduction of at least 100 stacked traits 102. The method of claim 96, wherein step a) includes screening cellular characteristics by utilizing one or any combination of the following methods: a) genomics; b) transcriptome characterization or RNA profiling; c) proteomics; d) metabolomics or the analysis of metabolites; e) lipidomics or lipid profiling. 103. A method of claim 102, wherein proteomics specifically includes the use of amino acid reactive tags 104. A method of claim 97, wherein step d) includes screening cellular characteristics by utilizing one or any combination of the following methods: f) genomics; g) transcriptome characterization or RNA profiling; h) proteomics; i) metabolomics or the analysis of metabolites; j) lipidomics or lipid profiling. 105. A method of claim 104, wherein proteomics specifically includes the use of amino acid reactive tags 106. A method of claim 98, wherein steps a) and d) include screening cellular characteristics by utilizing one or any combination of the following methods: k) genomics; l) transcriptome characterization or RNA profiling; m) proteomics; n) metabolomics or the analysis of metabolites; o) lipidomics or lipid profiling. P) 107. A method of claim 106, wherein proteomics specifically includes the use of amino acid reactive tags 108. A method of claim 73, wherein step c) includes screening cellular characteristics by utilizing one or any combination of the following methods: q) genomics; r) transcriptome characterization or RNA profiling; s) proteomics; t) metabolomics or the analysis of metabolites; u) lipidomics or lipid profiling. 109. A method of claim 108, wherein proteomics specifically includes the use of amino acid reactive tags 110. A method of claim 93, wherein step c) includes screening cellular characteristics by utilizing one or any combination of the following methods: v) genomics; w) transcriptome characterization or RNA profiling; x) proteomics; y) metabolomics or the analysis of metabolites; z) lipidomics or lipid profiling. 111. A method of claim 110, wherein proteomics specifically includes the use of amino acid reactive tags 112. A method of claim 94, wherein step c) includes screening cellular characteristics by utilizing one or any combination of the following methods: aa) genomics; bb) transcriptome characterization or RNA profiling; cc) proteomics; dd) metabolomics or the analysis of metabolites; ee) lipidomics or lipid profiling. 113. A method of claim 112, wherein proteomics specifically includes the use of amino acid reactive tags 114. A method for whole cell engineering of new or modified phenotypes by using real-time metabolic flux analysis, the method comprising the following steps: (a) making a modified cell by modifying the genetic composition of a cell; (b) culturing the modified cell to generate a plurality of modified cells; (c) measuring at least one metabolic parameter of the cell by monitoring the cell culture of step (b) in real time; and, (d) analyzing the data of step (c) to determine if the measured parameter differs from a comparable measurement in an unmodified cell under similar conditions, thereby identifyng an engineered phenotype in the cell using real-time metabolic flux analysis. 115. The method of claim 114, wherein the genetic composition of the cell is modified by a method comprising addition of a nucleic acid to the cell. 116. The method of claim 115, wherein the nucleic acid comprises a nucleic acid heterologous to the cell. 117. The method of claim 115, wherein the nucleic acid comprises a nucleic acid homologous to the cell. 118. The method of claim 117, wherein the homologous nucleic acid comprises a modified homologous nucleic acid. 119. The method of claim 118, wherein the homologous nucleic acid comprises a modified homologous gene. 120. The method of claim 114, wherein the genetic composition of the cell is modified by a method comprising deletion of a sequence or modification of a sequence in the cell. 121. The method of claim 114, wherein the genetic composition of the cell is modified by a method comprising modifying or knocking out the expression of a gene. 122. The method of claim 114, further comprising selecting a cell comprising a newly engineered phenotype. 123. The method of claim 122, further comprising culturing the selected cell, thereby generating a new cell strain comprising a newly engineered phenotype. 124. The method of claim 122, wherein the newly engineered phenotype is selected from the group consisting of an increased or decreased expression or amount of a polypeptide, an increased or decreased amount of an mRNA transcript, an increased or decreased expression of a gene, an increased or decreased resistance or sensitivity to a toxin, an increased or decreased resistance use or production of a metabolite, an increased or decreased uptake of a compound by the cell, an increased or decreased rate of metabolism, and an increased or decreased growth rate. 125. The method of claim 114, further comprising isolating a cell comprising a newly engineered phenotype. 126. The method of claim 114, wherein the newly engineered phenotype is a stable phenotype. 127. The method of claim 126, wherein modifying the genetic composition of a cell comprises insertion of a construct into the cell, wherein construct comprises a nucleic acid operably linked to a constitutively active promoter. 128. The method of claim 114, wherein the newly engineered phenotype is an inducible phenotype. 129. The method of claim 128, wherein modifying the genetic composition of a cell comprises insertion of a construct into the cell, wherein construct comprises a nucleic acid operably linked to an inducible promoter. 130. The method of claim 115, wherein nucleic acid added to the cell in step (a) is stably inserted into the genome of the cell. 131. The method of claim 115, wherein nucleic acid added to the cell in step (a) propagates as an episome in the cell. 132. The method of claim 115, wherein nucleic acid added to the cell in step (a) encodes a polypeptide. 133. The method of claim 132, wherein the polypeptide comprises a modified homologous polypeptide. 134. The method of claim 132, wherein the polypeptide comprises a heterologous polypeptide. 135. The method of claim 115, wherein the nucleic acid added to the cell in step (a) encodes a transcript comprising a sequence that is antisense to a homologous transcript. 136. The method of claim 114, wherein modifying the genetic composition of the cell in step (a) comprises increasing or decreasing the expression of an mRNA transcript. 137. The method of claim 114, wherein modifying the genetic composition of the cell in step (a) comprises increasing or decreasing the expression of a polypeptide. 138. The method of claim 114, wherein modifying the homologous gene in step (a) comprises knocking out expression of the homologous gene. 139. The method of claim 114, wherein modifying the homologous gene in step (a) comprises increasing the expression of the homologous gene. 140. The method of claim 114, wherein the heterologous gene in step (a) comprises a sequence-modified homologous gene, wherein the sequence modification is made by a method comprising the following steps: (a) providing a template polynucleotide, wherein the template polynucleotide comprises a homologous gene of the cell; (b) providing a plurality of oligonucleotides, wherein each oligonucleotide comprises a sequence homologous to the template polynucleotide, thereby targeting a specific sequence of the template polynucleotide, and a sequence that is a variant of the homologous gene; (c) generating progeny polynucleotides comprising non-stochastic sequence variations by replicating the template polynucleotide of step (a) with the oligonucleotides of step (b), thereby generating polynucleotides comprising homologous gene sequence variations. 141. The method of claim 114, wherein the heterologous gene in step (a) comprises a sequence-modified homologous gene, wherein the sequence modification is made by a method comprising the following steps: (a) providing a template polynucleotide, wherein the template polynucleotide comprises sequence encoding a homologous gene; (b) providing a plurality of building block polynucleotides, wherein the building block polynucleotides are designed to cross-over reassemble with the template polynucleotide at a predetermined sequence, and a building block polynucleotide comprises a sequence that is a variant of the homologous gene and a sequence homologous to the template polynucleotide flanking the variant sequence; (c) combining a building block polynucleotide with a template polynucleotide such that the building block polynucleotide cross-over reassembles with the template polynucleotide to generate polynucleotides comprising homologous gene sequence variations. 142. The method of claim 114, wherein the cell is a prokaryotic cell. 143. The method of claim 142, wherein the prokaryotic cell is a bacterial cell. 144. The method of claim 114, wherein the cell is a selected from the group consisting of a fungal cell, a yeast cell, a plant cell and an insect cell. 145. The method of claim 114, wherein the cell is a eukaryotic cell. 146. The method of claim 145, wherein the cell is a mammalian cell. 147. The method of claim 146, wherein the mammalian cell is a human cell. 148. The method of claim 114, wherein the measured metabolic parameter comprises rate of cell growth. 149. The method of claim 148, wherein the rate of cell growth is measured by a change in optical density of the culture. 150. The method of claim 114, wherein the measured metabolic parameter comprises a change in the expression of a polypeptide. 151. The method of claim 150, wherein the change in the expression of the polypeptide is measured by a method selected from the group consisting of a one-dimensional gel electrophoresis, a two-dimensional gel electrophoresis, a tandem mass spectography, an RIA, an ELISA, an immunoprecipitation and a Western blot. 152. The method of claim 114, wherein the measured metabolic parameter comprises a change in expression of at least one transcript, or, the expression of a transcript of a newly introduced gene. 153. The method of claim 152, wherein the change in expression of the transcript is measured by a method selected from the group consisting of a hybridization, a quantitative amplification and a Northern blot. 154. The method of claim 153, wherein transcript expression is measured by hybridization of a sample comprising transcripts of a cell or nucleic acid representative of or complementary to transcripts of a cell by hybridization to immobilized nucleic acids on an array. 155. The method of claim 114, wherein the measured metabolic parameter comprises an increase or a decrease in a secondary metabolite. 156. The method of claim 155, wherein secondary metabolite is selected from the group consisting of a glycerol and a methanol. 157. The method of claim 114, wherein the measured metabolic parameter comprises an increase or a decrease in an organic acid. 158. The method of claim 157, wherein the organic acid is selected from the group consisting of an acetate, a butyrate, a succinate and an oxaloacetate. 159. The method of claim 114, wherein the measured metabolic parameter comprises an increase or a decrease in intracellular pH. 160. The method of claim 159, wherein the increase or a decrease in intracellular pH is measured by intracellular application of a dye, and the change in fluorescence of the dye is measured over time. 161. The method of claim 114, wherein the measured metabolic parameter comprises an increase or a decrease in synthesis of DNA over time. 162. The method of claim 161, wherein the increase or a decrease in synthesis of DNA over time is measured by intracellular application of a dye, and the change in fluorescence of the dye is measured over time. 163. The method of claim 114, wherein the measured metabolic parameter comprises an increase or a decrease in uptake of a composition. 164. The method of claim 163, wherein the composition is a metabolite. 165. The method of claim 164, wherein the metabolite is selected from the group consisting of a monosaccharide, a disaccharide, a polysaccharide, a lipid, a nucleic acid, an amino acid and a polypeptide. 166. The method of claim 165, wherein the saccharide, disaccharide or polysaccharide comprises a glucose or a sucrose. 167. The method of claim 163, wherein the composition is selected from the group consisting of an antibiotic, a metal, a steroid and an antibody. 168. The method of claim 114, wherein the measured metabolic parameter comprises an increase or a decrease in the secretion of a byproduct or a secreted composition of a cell. 169. The method of claim 168, wherein the byproduct or secreted composition is selected from the group consisting of a toxin, a lymphokine, a polysaccharide, a lipid, a nucleic acid, an amino acid, a polypeptide and an antibody. 170. The method of claim 114, wherein the real time monitoring simultaneously measures a plurality of metabolic parameters. 171. The method of claim 170, wherein real time monitoring of a plurality of metabolic parameters comprises use of a Cell Growth Monitor device. 172. The method of claim 171, wherein the Cell Growth Monitor device is a Wedgewood Technology, Inc., Cell Growth Monitor model 652. 173. The method of claim 171, wherein the real time simultaneous monitoring measures uptake of substrates, levels of intracellular organic acids and levels of intracellular amino acids. 174. The method of claim 171, wherein the real time simultaneous monitoring measures: uptake of glucose; levels of acetate, butyrate, succinate or oxaloacetate; and, levels of intracellular natural amino acids. 175. The method of claim 171, further comprising use of a computer-implemented program to real time monitor the change in measured metabolic parameters over time. 176. The method of claim 175, wherein the computer-implemented program comprises a computer-implemented method as set forth in FIG. 28. 177. The method of claim 176, wherein the computer-implemented method comprises metabolic network equations. 178. The method of claim 176, wherein the computer-implemented method comprises a pathway analysis. 179. The method of claim 176, wherein the computer-implemented program comprises a preprocessing unit to filter out the errors for the measurement before the metabolic flux analysis.
<SOH> B—BACKGROUND <EOH>
<SOH> C—SUMMARY OF THE INVENTION <EOH>This invention relates generally to the field of cellular and whole organism engineering. Specifically, this invention relates to a cellular transformation, directed evolution, and screening method for creating novel transgenic organisms having desirable properties. Thus in one aspect, this invention relates to a method of generating a transgenic organism, such as a microbe or a plant, having a plurality of traits that are differentially activatable. In one embodiment, this invention is directed to a method of producing an improved organism having a desirable trait to by: a) obtaining an initial population of organisms, b) generating a set of mutagenized organisms, such that when all the genetic mutations in the set of mutagenized organisms are taken as a whole, there is represented a set of substantial genetic mutations, and c) detecting the presence of said improved organism. This invention provides that any of steps a), b), and c) can be further repeated in any particular order and any number of times; accordingly, this invention specifically provides methods comprised of any iterative combination of steps a), b), and c), with a number of iterations. In another embodiment, this invention is directed to a method of producing an improved organism having a desirable trait to by: a) obtaining an initial population of organisms, which can be a clonal population or otherwise, b) generating a set of mutagenized organisms each having at least one genetic mutation, such that when all the genetic mutations in the set of mutagenized organisms are taken as a whole, there is represented a set of substantial genetic mutations c) detecting the manifestation of at least two genetic mutations, and d) introducing at least two detected genetic mutations into one organism. Additionally, this invention provides that any of steps a), b), c), and d) can be further repeated in any particular order and any number of times; accordingly, this invention specifically provides methods comprised of any iterative combination of steps a), b), c), and d), with a total number of iterations can be from one up to one million, including specifically every integer value in between. In a preferred aspect of embodiments specified herein the step of b) generating a second set of mutagenized organisms is comprised of generating a plurality of organisms, each of which organisms has a particular transgenic mutation. As used herein, “generating a set of mutagenized organisms having genetic mutations” can be achieved by any means known in the art to mutagenized including any radiation known to mutagenized, such as ionizing and ultra violet. Further examples of serviceable mutagenizing methods include site-saturation mutagenesis, transposon-based methods, and homologous recombination. “Combining” means incorporating a plurality of different genetic mutations in the genetic makeup (e.g. the genome) of the same organism; and methods to achieve this combining” step including sexual recombination, homologous recombination, and transposon-based methods. As used herein, an “initial population of organisms” means a “Working population of organisms”, which refers simply to a population of organisms with which one is working, and which is comprised of at least one organism. An “initial population of organisms” which can be a clonal population or otherwise. Accordingly, in step 1) an “initial population of organisms” may be a population of multicellular organisms or of unicellular organisms or of both. An “initial population of organisms” may be comprised of unicellular organisms or multicellular organisms or both. An “initial population of organisms” may be comprised of prokaryotic organisms or eukaryotic organisms or both. This invention provides that an “initial population of organisms” is comprised of at least one organism, and preferred embodiments include at least that. By “organism” is meant any biological form or thing that is capable of self replication or replication in a host. Examples of “organisms” include the following kinds of organisms (which kinds are not necessarily mutually-exclusive): animals, plants, insects, cyanobacteria, microorganisms, fungi, bacteria, eukaryotes, prokaryotes, mycoplasma, viral organisms (including DNA viruses, RNA viruses), and prions. Non-limiting particularly preferred examples of kinds of “organisms” also include Archaea (archaebacteria) and Bacteria (eubacteria). Non-limiting examples of Archaea (archaebacteria) include Crenarchaeota, Euryarchaeota, and Korarchaeota. Non-limiting examples Bacteria (eubacteria) include Aquificales, CFB/Green sulfur bacteria group, Chlamydiales/Verrucomicrobia group, Chrysiogenes group, Coprothermobacter group, Cyanobacteria & chloroplasts, Cytophaga/Flexibacter/Bacteriods group, Dictyoglomus group, Fibrobacter/Acidobacteria group, Firmicutes, Flexistipes group, Fusobacteria, Green non-sulfur bacteria, Nitrospira group, Planctomycetales, Proteobacteria, Spirochaetales, Synergistes group, Thermodesulfobacterium group, Thermotogales, Thermus/Deinococcus group. As non-limiting examples, particularly preferred kinds of organisms include Aquifex, Aspergillus, Bacillus, Clostridium, E. coli, Lactobacillus, Mycobacterium, Pseudomonas, Streptomyces , and Thermotoga . As additional non-limiting examples, particularly preferred organisms include cultivated organisms such as CHO, VERO, BHK, HeLa, COS, MDCK, Jurkat, HEK-293, and WI38. Particularly preferred non-limiting examples of organisms further include host organisms that are serviceable for the expression of recombinant molecules. Organisms further include primary cultures (e.g. cells from harvested mammalian tissues), immortalized cells, all cultivated and culturable cells and multicellular organisms, and all uncultivated and uculturable cells and multicellular organisms. In a preferred embodiment, knowledge of genomic information is useful for performing the claimed methods; thus, this invention provides the following as preferred but non-limiting examples of organisms that are particularly serviceable for this invention, because there is a significant amount of—if not complete—genomic sequence information (in terms of primary sequence &/or annotation) for these organisms: Human, Insect (e.g. Drosophila melanogaster ), Higher plants (e.g. Arabidopsis thaliana ), Protozoan (e.g. Plasmodium falciparum ), Nematode (e.g. Caenorhabditis elegans ), Fungi (e.g. Saccharomyces cerevisiae ), Proteobacteria gamma subdivision (e.g. Escherichia coli K-12, Haemophilus influenzae Rd, Xylella fastidiosa 9a5c, Vibrio cholerae E1 Tor N16961, Pseudomonas aeruginosa PA01, Buchnera sp. APS), Proteobacteria beta subdivision (e.g. Neisseria meningitidis MC58 (serogroup B), Neisseria meningitidis Z2491 (serogroup A)), Proteobacteria other subdivisions (e.g. Helicobacter pylori 26695, Helicobacter pylori J99, Campylobacter jejuni NCTCI 11168, Rickettsia prowazekii ), Gram-positive bacteria (e.g. Bacillus subtilis, Mycoplasma genitalium, Mycoplasma pneumoniae, Ureaplasma urealyticum, Mycobacterium tuberculosis H37Rv), Chlamydia (e.g. Chlamydia trachomatisserovar D, Chlamydia muridarum ( Chlamydia trachomatis MoPn), Chlamydia pneumoniae CWL029 , Chlamydia pneumoniae AR39 , Chlamydia pneumoniae J138), Spirochete (e.g. Borrelia burgdorferi B31, Treponema pallidum ), Cyanobacteria (e.g. Synechocystis sp. PCC6803), Radioresistant bacteria (e.g. Deinococcus radiodurans R1), Hyperthermophilic bacteria (e.g. Aquifex aeolicus VF5 , Thermotoga marilima MSB8), and Archaea (e.g. Methanococcus jannaschii, Methanobacterium thermoautotrophicum deltaH, Archaeoglobus fulgidus, Pyrococcus horikoshii OT3, Pyrococcus abyssi, Aeropyrum pernix K1). Non-limiting particularly preferred examples of kinds of plant “organisms” include those listed in Table 1. TABLE 1 Non-limiting examples of plant organisms and sources of transgenic molecules (e.g. nucleic acids & nucleic acid products) 1. Alfalfa 2. Amelanchier laevis 3. Apple 4. Arab. thaliana 5. Arabidopsis 6. Aspergillus flavus 7. Barley 8. Beet 9. Belladonna 10. Brassica oleracea 11. Carrot 12. Chrysanthemum 13. Cichorium intybus 14. Clavibacter 15. Clavibacter xyli 16. Coffee 17. Corn 18. Cotton 19. Cranberry 20. Creeping bentgrass 21. Cryphonectria parasitica 22. Eggplant 23. Festuca arundinacea 24. Fusarium graminearum 25. Fusarium moniliforme 26. Fusarium sporotrichioides 27. Gladiolus 28. Grape 29. Heterorhabditis bacteriophora 30. Kentucky bluegrass 31. Lettuce 32. Melon 33. Oat 34. Onion 35. Papaya 36. Pea 37. Peanut 38. Pelargonium 39. Pepper 40. Persimmon 41. Petunia 42. Pine 43. Pineapple 44. Pink bollworm 45. Plum 46. Poplar 47. Potato 48. Pseudomonas 49. Pseudomonas putida 50. Pseudomonas syringae 51. Rapeseed 52. Rhizobium 53. Rhizobium etli 54. Rhizobium fredii 55. Rhizobium leguminosarum 56. Rhizobium meliloti 57. Rice 58. Rubus idaeus 59. Spruce 60. Soybean 61. Squash 62. Squash-cucumber 63. Squash- cucurbita texana 64. Strawberry 65. Sugarcane 66. Sunflower 67. Sweet potato 68. Sweetgum 69. TMV 70. Tobacco 71. Tomato 72. Walnut 73. Watermelon 74. Wheat 75. Xanthomonas 76. Xanthomonas campestris As used herein, the meaning of “generating a set of mutagenized organisms having genetic mutations” includes the steps of substituting, deleting, as well as introducing a nucleotide sequence into organism; and this invention provides a nucleotide sequence that serviceable for this purpose may be a single-stranded or double-stranded and the fact that its length may be from one nucleotide up to 10,000,000,000 nucleotides in length including specifically every integer value in between. A mutation in an organism includes any alteration in the structure of one or more molecules that encode the organism. These molecules include nucleic acid, DNA, RNA, prionic molecules, and may be exemplified by a variety of molecules in an organism such as a DNA that is genomic, episomal, or nucleic, or by a nucleic acid that is vectoral (e.g. viral, cosmid, phage, phagemid). In one aspect, as used herein, a “set of substantial genetic mutations” is preferably a disruption (e.g. a functional knock-out) of at least about 15 to about 150,000 genomic locations or nucleotide sequences (e.g. genes, promoters, regulatory sequences, codons etc.), including specifically every integer value in between. In another aspect, as used herein, a “set of substantial genetic mutations” is preferably an alteration in an expression level (e.g. decreased or increased expression level) or an alteration in the expression pattern (e.g. throughout a period of time) of at least about 15 to about 150,000 genes, including specifically every integer value in between. Corresponding to another aspect, as used herein, a “set of substantial genetic mutations” is preferably an alteration in an expression level (e.g. decreased or increased expression level) or an alteration in the expression pattern (e.g. throughout a period of time) of at least about 15 to about 150,000 gene products &/or phenotypes &/or traits, including specifically every integer value in between. In another aspect, as used herein, a “set of substantial genetic mutations” with respect to an organism (or type of organism) is preferably a disruption (e.g. a functional knock-out) of at least about 1% to about 100% of genomic locations or nucleotide sequences (e.g. genes, promoters, regulatory sequences, codons etc.) in the organism (or type of organism), including specifically percentages of every integer value in between. In another aspect, as used herein, a “set of substantial genetic mutations” is preferably an alteration in an expression level (e.g. decreased or increased expression level) or an alteration in the expression pattern (e.g. throughout a period of time) of at least about 1% to about 100% of genes in an organism (or type of organism), including specifically percentages of every integer value in between. Corresponding to another aspect, as used herein, a “set of substantial genetic mutations” is preferably an alteration in an expression level (e.g. decreased or increased expression level) or an alteration in the expression pattern (e.g. throughout a period of time) of at least about 1% to about 100% of the gene products &/or phenotypes &/or traits of an organism (or type of organism), including specifically every integer value in between. In yet another aspect, as used herein, a “set of substantial genetic mutations” is preferably an introduction or deletion of at least about 15 to 150,000 genes promoters or other nucleotide sequences (where each sequence is from 1 base to 10,000,000 bases), including specifically every integer value in between. For example, one can introduce a library of at least about 15 to 150,000 nucleotides (genes or promoters) produced by “site-saturation mutagenesis” &/or by “ligation reassembly” (including any specific aspect thereof provided herein) into an “initial population of organisms”. It is provided that wherever the manipulation of a plurality of “genes” is mentioned herein, gene pathways (e.g. that ultimately lead to the production of small molecules) are also included. It is appreciated herein that knocking-out, altering expression level, and altering expression pattern can be achieved, by non-limiting exemplification, by mutagenizing a nucleotide sequence corresponding gene as well as a corresponding promoter that affects the expression of the gene. As used herein, a “mutagenized organism” includes any organism that has been altered by a genetic mutation. A “genetic mutation” can be, by way of non-limiting and non-mutually exclusive exemplification, and change in the nucleotide sequence (DNA or RNA) with respect to genomic, extra-genomic, episomal, mitochondrial, and any nucleotide sequence associated with (e.g. contained within or considered part of) an organism. According to this invention, detecting the manifestation of a “genetic mutation” means “detecting the manifestation of a detectable parameter”, including but not limited to a change in the genomic sequence. Accordingly, this invention provides that a step of sequencing (&/or annotating) of and organism's genomic DNA is necessary for some methods of this invention, and exemplary but non-limiting aspects of this sequencing (&/or annotating) step are provided herein. A detectable “trait”, as used herein, is any detectable parameter associated with the organism. Accordingly, such a detectable “parameter” includes, by way of non-limiting exemplification, any detectable “nucleotide knock-in”, any detectable “nucleotide knock-outs”, any detectable “phenotype”, and any detectable “genotype”. By way of further illustration, a “trait” includes any substance produced or not produced by the organism. Accordingly, a “trait” includes viability or non-viability, behavior, growth rate, size, morphology. “Trait” includes increased (or alternatively decreased) expression of a gene product or gene pathway product. “Trait” also includes small molecule production (including vitamins, antibiotics), herbicide resistance, drought resistance, pest resistance, production of any recombinant biomolecule (ie.g. vaccines, enzymes, protein therapeutics, chiral enzymes). Additional examples of serviceable traits for this invention are shown in Table 2. TABLE 2 Non-limiting examples of serviceable genes, gene products, phenotypes, or traits according to the methods of this invention (e.g. knockouts, knockins, increased or decreased expression level, increased or decreased expression pattern) Table 2 - Part 1. Non-limiting examples of genes or gene products 1. 17 kDa protein 2. 3-hydroxy-3-methylglutaryl CoenzymeA reductase 3. 4-Coumarate: CoA ligase knockout 4. 60 kDa protein 5. Ac transposable element 6. ACC deaminase 7. ACC oxidase knockout 8. ACC synthase 9. ACC synthase knockout 10. Acetohydroxyacid synthase variant 11. Acetolactate synthase 12. Acetyl CoA carboxylase 13. ACP acyl-ACP thioesterase 14. ACP thioesterase 15. Acyl CoA reductase 16. Acyl-ACP knockout 17. Acyl-ACP desaturase 18. Acyl-ACP desaturase knockout 19. Acyl-ACP thioesterase 20. ADP glucose pyrophosphorylase 21. ADP glucose pyrophosphorylase knockout 22. Agglutinin 23. Aleurone 1 24. Alpha hordothinonin 25. Alpha-amylase 26. Alpha-hemoglobin 27. Aminoglycoside 3′-adenylytransferase 28. Amylase 29. Anionic peroxidase 30. Antibody 31. Antifungal protein 32. Antithrombin 33. Antitrypsin 34. Antiviral protein 35. Aspartokinase 36. Attacin E 37. B1 regulatory gene 38. B-1,3-glucanase knockout 39. B-1,4-endoglucanase knockout 40. Bacteropsin 41. Barnase 42. Barstar 43. Beta-hemoglobin 44. B-glucuronidase 45. C1 knockout 46. C1 regulatory gene 47. C2 knockout 48. C3 knockout 49. Caffeate O-methylthransferase 50. Caffeate O-methyltransferase knockout 51. Caffeoyl CoA O-methyltransferase knockout 52. Casein 53. Cecropin 54. Cecropin B 55. Cellulose binding protein 56. Chalcone synthase knockout 57. Chitinase 58. Chitobiosidase 59. Chloramphenicol acetyltransferase 60. Cholera toxin B 61. Choline oxidase 62. Cinnamate 4-hydroxylase 63. Cinnamate 4-hydroxylase knockout 64. Coat protein 65. Coat protein knockout 66. Conglycinin 67. CryIA 68. CryIAb 69. CryIAc 70. CryIB 71. CryIIA 72. CryIIIA 73. CryVIA 74. Cyclin dependent kinase 75. Cyclodexlrin glycosyltransferase 76. Cylindrical inclusion protein 77. Cystathionine synthase 78. Delta-12 desaturase 79. Delta-12 desaturase knockout 80. Delta-12 saturase 81. Delta-12 saturase knockout 82. Delta-15 desaturase 83. Delta-15 desaturase knockout 84. Delta-9 desaturase 85. Delta-9 desturase knockout 86. Deoxyhypusine synthase (DHS) 87. Deoxyhypusine synthase knockout 88. Diacylglycerol acetyl tansferase 89. Dihydrodipicolinate synthase 90. Dihydrofolate reductase 91. Diptheria toxin A 92. Disease resistance response gene 49 93. Double stranded ribonuclease 94. Ds transposable element 95. Elongase 96. EPSPS 97. Ethylene forming enzyme knockout 98. Ethylene receptor protein 99. Ethylene receptor protein knockout 100. Fatty acid elongase 101. Fluorescent protein 102. G glycoprotein 103. Galactanase 104. Galanthus nivalis agglutinin 105. Genome-linked protein 106. Glucanase 107. Glucanase knockout 108. Glucose oxidase 109. Glutamate dehydrogenase 110. Glutamine binding protein 111. Glutamine synthetase 112. Glutenin 113. Glycerol-3-phosphate acetyl transferase 114. Glyphosate exidoreductase 115. Glyphosate oxidoreductase 116. Green fluorescent protein 117. Helper component 118. Hemicellulase 119. Hup locus 120. Hygromycin phosphotransferase 121. Hyoscamine 6B-hydroxylase 122. IAA monooxygenase 123. Invertase 124. Invertase knockout 125. Isopentenyl transferase 126. Ketoacyl-ACP synthase 127. Ketoacyl-ACP synthase knockout 128. Larval serum protein 129. Leafy homeotic regulatory gene 130. Lectin 131. Lignin peroxidase 132. Luciferase 133. Lysine-2 gene 134. Lysophosphatidic acid acetyl transferase 135. Lysozyme 136. Mabinlin 137. Male sterility protein 138. Metallothionein 139. Modified ethylene receptor protein 140. Modified ethylene receptor protein knockout 141. Monooxygenase 142. Movement protein 143. Movement protein nonfunctional 144. N gene for TMV resistance 145. N-acetyl glucosidase 146. Nitrilase 147. Nopaline synthase 148. Notch 149. NptII 150. Nuclear inclusion protein a 151. Nuclear inclusion protein b 152. Nucleocapsid 153. Nucleoprotein 154. O-acyl transferase 155. Oleayl-ACP thioesterase 156. Omega 3 desaturase 157. Omega 3 desaturease knockout 158. Omega 6 desaturase 159. Omega 6 desaturase knockout 160. O-methyltransferase 161. Osmotin 162. Oxalate oxidase 163. Par locus 164. Pathogenesis protein 1a 165. Pectate lyase 166. Pectin esterase 167. Pectin esterase knockout 168. Pectin methylesterase 169. Pectin methylesterase knockout 170. Pentenlypyrophosphate isomerase 171. Phosphinothricin 172. Phosphinothricin acetyl transferase 173. Phytochrome A 174. Phytoene synthase 175. Phleomycin binding protein 176. Polygalacturonase 177. Polygalacturonase knockout 178. Polygalacturonase inhibitor protein 179. Prf regulatory gene 180. Prosystemin 181. Protease 182. Protein A 183. Protein kinase 184. Proteinase inhibitor 1 185. Pti5 transcription factor 186. R regulatory gene 187. Receptor kinase 188. Recombinase 189. Reductase 190. Replicase 191. Resveratrol synthase 192. Ribonuclease 193. ro1c 194. Rol hormone gene 195. S-adenosylmethione decarboxylase 196. S-adenosylmethione hydrolase 197. S-adenosylmethionine transferase 198. Salicylate hydroxylase 199. Satellite RNA 200. Seed storage protein 201. Serine-threonine protein kinase 202. Serum albumin 203. Shrunken 2 204. Sorbitol dehydrogenase 205. Sorbitol synthase 206. Stilbene synthase 207. Storage protein 208. Sucrose phosphate synthase 209. Systemic acquired resistance gene 8.2 210. Tetracycline binding protein 211. Thioesterase (×2) 212. Thiolase 213. TobRB7 214. Transcriptional activator 215. Transposon Tn5 216. Trehalase 217. Trehalase knockout 218. Trichodiene synthase 219. Trichosanthin 220. Trifolitoxin 221. Trypsin inhibitor 222. T-URF13 mitochondrial 223. UDP glucose glucosyltransferase 224. Violaxanthin de-epoxidase 225. Violaxanthin de-epoxidase knockout 226. Wheat germ agglutinin 227. Xanthosine-N7-methyltransferase knockout 228. Zein storage protein Table 2 - Part 2. Non-limiting examples of input traits/phenotypes 1. 2,4-D tolerant 2. Alernaria resistant 3. Altered amino acid composition 4. Alternaria solani resistant 5. Ammonium assimilation increased 6. AMV resistant 7. Aphid resistant 8. Apple scab resistant 9. Aspergillus resistant 10. B-1,4-endoglucanase 11. Bacterial leaf blight resistant 12. Bacterial speck resistant 13. BCTV resistant 14. Blackspot bruise resistant 15. BLRV resistant 16. BNYVV Resistant 17. Botrytis cinerea resistant 18. Botrytis resistant 19. BPMV resistant 20. Bromoxynil tolerant 21. BYDV resistant 22. BYMV resistant 23. Carbohydrate metabolism altered 24. Cell wall altered 25. Chlorsulfuron tolerant 26. Clavibacter resistant 27. CLRV resistant 28. CMV resistant 29. Cold tolerant 30. Coleopteran resistant 31. Colletotrichum resistant 32. Colorado potato beetle resistant 33. Constitutive expression of glutamine synthetase 34. Corynebacterium sepedonicum resistant 35. Cottonwood leaf beetle resistant 36. Crown gall resistant 37. Crown rot resistant 38. Cucumovirus resistant 39. Cutting rootability increased 40. Downy mildew resistant 41. Drought tolerant 42. Erwinia carotovora resistant 43. Ethylene production reduced 44. European Corn Borer resistant 45. Female sterile 46. Fenthion susceptible 47. Fertility altered 48. Fire blight resistant 49. Flower and fruit abscission reduced 50. Flower and fruit set altered 51. Flowering altered 52. Flowering time altered 53. Frogeye leaf spot resistant 54. Fruit ripening altered 55. Fruit ripening delayed 56. Fruit rot resistant 57. Fruit solids increased 58. Fruit sweetness increased 59. Fungal post-harvest resistant 60. Fungal resistant 61. Fungal resistant general 62. Fusarium resistant 63. Glyphosate tolerant 64. Growth rate altered 65. Growth rate reduced 66. Heat stable glucanase produced 67. Hordothionin produced 68. Imidazolinone tolerant 69. Insect resistant general 70. Kanamycin resistant 71. Lepidopteran resistant 72. Lesser cornstalk borer resistant 73. LMV resistant 74. Loss of systemic resistance 75. Male sterile 76. Marssonina resistant 77. MCDV resistant 78. MCMV resistant 79. MDMV resistant 80. MDMV-B resistant 81. Mealybug wilt virus resistant 82. Melamtsora resistant 83. Melodgyne resistant 84. Methotrexate resistant 85. Mexican Rice Borer resistant 86. Nucleocapsid protein produced 87. Oblique banded leafroller resistant 88. PEMV resistant 89. PeSV resistant 90. Phoma resistant 91. Phosphinothricin tolerant 92. Phratora leaf beetle resistant 93. Phytophthora resistant 94. PLRV resistant 95. Polyamine metabolism altered 96. Potyvirus resistant 97. Powdery mildew resistant 98. PPV resistant 99. Pratylenchus vulnus resistant 100. Proteinase inhibitors level constitutive 101. PRSV resistant 102. PRV resistant 103. PSbMV resistant 104. Pseudomonas syringae resistant 105. PStV resistant 106. PVX resistant 107. PVY resistant 108. RBDV resistant 109. Rhizoctonia resistant 110. Rhizoctonia solani resistant 111. Ring rot resistance 112. Root-knot nematode resistant 113. SbMV resistant 114. Sclerotinia resistant 115. SCMV resistant 116. SCYLV resistant 117. Secondary metabolite increased 118. Seed set reduced 119. Selectable marker 120. Senescence altered 121. Septoria resistant 122. Shorter stems 123. Soft rot fungal resistant 124. Soft rot resistant 125. SqMV resistant 126. SrMV resistant 127. Storage protein altered 128. Streptomyces scabies resistant 129. Sulfonylurea tolerant 130. Tetracycline binding protein produced 131. TEV resistant 132. Thelaviopsis resistant 133. TMV resistant 134. Tobamovirus resistant 135. ToMoV resistant 136. ToMV resistant 137. Transposon activator 138. Transposon inserted 139. TRV resistant 140. TSWV resistant 141. TVMV resistant 142. TYLCV resistant 143. Tyrosine level increased 144. Venturia resistant 145. Verticillium dahliae resistant 146. Verticillium resistant 147. Visual marker 148. WMV2 resistant 149. WSMV resistant 150. Yield increased 151. ZYMV resistant Table 2 - Part 3. Non-limiting examples of output traits/phenotypes 1. ACC oxidase level decreased 2. Altered lignin biosynthesis 3. B-1,4-endoglucanase 4. Botrytis resistant 5. Carbohydrate metabolism altered 6. Carotenoid content altered 7. Cell wall altered 8. CMV resistant 9. Coleopteran resistant 10. Dry matter content increased 11. Ethylene production reduced 12. Ethylene synthesis reduced 13. Fatty acid metabolism altered 14. Fire blight resistant 15. Flower and fruit abscission reduced 16. Flower and fruit set altered 17. Flowering time altered 18. Fruit firmness increased 19. Fruit pectin esterase levels decreased 20. Fruit ripening altered 21. Fruit ripening delayed 22. Fruit solids increased 23. Fruit sugar profile altered 24. Fruit sweetness increased 25. Glucuronidase expressing 26. Heat stable glucanase produced 27. Heavy metals sequestered 28. Hordothionin produced 29. Improved fruit quality 30. Industrial enzyme produced 31. Lepidopteran resistant 32. Lysine level increased 33. Mealybug wilt virus resistant 34. Methionine level increased 35. Nucleocapsid protein produced 36. Oil profile altered 37. Pectin esterase level reduced 38. Pharmaceutical proteins produced 39. Phosphinothricin tolerant 40. Phytoene synthase activity increased 41. Pigment metabolism altered 42. Polygalacturonase level reduced 43. Processing characteristics altered 44. Prolonged shelf life 45. Protein altered 46. Protein quality altered 47. PRSV resistant 48. Root-knot nematode resistant 49. Sclerotinia resistant 50. Seed composition altered 51. Seed methionine storage increased 52. Seed set reduced 53. Seed storage protein 54. Senescence altered (e.g. Shelf life increased) 55. Shorter stems 56. Solids increased 57. SqMV resistant 58. Starch level increased 59. Starch metabolism altered 60. Starch reduced 61. Sterols increased 62. Storage protein altered 63. Sugar alcohol levels increased 64. Telracycline binding protein produced 65. Tyrosine level increased 66. Verticillium resistant 67. Visual marker 68. WMV2 resistant 69. Yield increased 70. ZYMV resistant Table 2 - Part 4. Non-limiting examples of traits/phenotypes with agronomic properties 1. ACC oxidase level decreased 2. Altered amino acid composition 3. Altered lignin biosynthesis 4. Altered maturing 5. Altered plant development 6. Aluminum tolerant 7. Ammonium assimilation increased 8. Anthocyanin produced in seed 9. B-1,4-endoglucanase 10. Calmodulin level altered 11. Carbohydrate metabolism altered 12. Carotenoid content altered 13. Cell wall altered 14. Cold tolerant 15. Constitutive expression of glutamine synthetase 16. Cutting root ability increased 17. Development altered 18. Drought tolerant 19. Dry matter content increased 20. Environmental stress reduced 21. Ethylene metabolism altered 22. Ethylene production reduced 23. Ethylene synthesis reduced 24. Fatty acid metabolism altered 25. Female sterile 26. Fenthion susceptible 27. Fertility altered 28. Fiber quality altered 29. Flower and fruit abscission reduced 30. Flower and fruit set altered 31. Flowering altered 32. Flower color altered 33. Flowering time altered 34. Fruit firmness increased 35. Fruit pectin esterase and levels decreased 36. Fruit polygalacturonase level decreased 37. Fruit ripening altered 38. Fruit ripening delayed 39. Fruit solids increased 40. Fruit sugar profile altered 41. Fruit sweetness increased 42. Glucuronidase expressing 43. Growth rate altered 44. Growth rate increased 45. Growth rate reduced 46. Heat stable glucanase produced 47. Heat tolerant 48. Heavy metals sequestered 49. Hordothionin produced 50. Improved fruit quality 51. Increased phosphorus 52. Increased stalk strength 53. Industrial enzyme produced 54. Lignin levels decreased 55. Lipase expressed in seeds 56. Lysine level increased 57. Male sterile 58. Male sterile reversible 59. Methionine level increased 60. Modified growth characteristics 61. Mycotoxin degradation 62. Nitrogen metabolism altered 63. Nucleocapsid protein produced 64. Oil profile altered 65. Oil quality altered 66. Oxidative stress tolerant 67. Pectin esterase level reduced 68. Pharmaceutical proteins produced 69. Photosynthesis enhanced 70. Phytoene synthase activity increased 71. Pigment metabolism altered 72. Polyamine metabolism altered 73. Polygalacturonase level reduced 74. Pratylenchus vulnus resistant 75. Processing characteristics altered 76. Prolonged shelf life 77. Protein altered 78. Protein lysine level increased 79. Protein quality altered 80. Proteinase inhibitors level constitutive 81. Salt tolerance increased 82. Seed composition altered 83. Seed methionine storage increased 84. Seed set reduced 85. Selectable marker 86. Senescence altered 87. Shorter stems 88. Solids increased 89. Starch level increased 90. Starch metabolism altered 91. Starch reduced 92. Sterols increased 93. Storage protein altered 94. Stress tolerant 95. Sugar alcohol levels increased 96. Tetracycline binding protein produced 97. Thermostable protein produced 98. Transposon activator 99. Transposon inserted 100. Tyrosine level increased 101. Visual marker 102. Vivipary increased 103. Yield increased Table 2 - Part 5. Non-limiting examples of traits/phenotypes with product quality properties 1. 2,4-D tolerant 2. ACC oxidase level decreased 3. Altered amino acid composition 4. Altered lignin biosynthesis 5. Anthocyanin produced in seed 6. Antioxidant enzyme increased 7. Auxin metabolism and increased tuber solids 8. B-1,4-endoglucanase 9. Blackspot bruise resistant 10. Brown spot resistant 11. Bruising reduced 12. Caffeine levels reduced 13. Carbohydrate metabolism altered 14. Carotenoid content altered 15. Cell wall altered 16. Cold tolerant 17. Delayed softening 18. Disulfides reduced in endosperm 19. Dry matter content increased 20. Ear mold resistant 21. Ethylene production reduced 22. Ethylene synthesis reduced 23. Extended flower life 24. Fatty acid metabolism altered 25. Fiber quality altered 26. Fiber strength altered 27. Flavor enhancer 28. Flower and fruit abscission reduced 29. Fruit firmness increased 30. Fruit invertase level decreased 31. Fruit polygalacturonase level decreased 32. Fruit ripening altered 33. Fruit ripening delayed 34. Fruit solids increased 35. Fruit sugar profile altered 36. Fruit sweetness increased 37. Glyphosate tolerant 38. Heat stable glucanase produced 39. Improved fruit quality 40. Increased phosphorus 41. Increased protein levels 42. Lignin levels decreased 43. Lysine level increased 44. Male sterile 45. Melanin produced in cotton fibers 46. Metabolism altered 47. Methionine level increased 48. Mycotoxin degradation 49. Mycotoxin production inhibited 50. Nicotine levels reduced 51. Nitrogen metabolism altered 52. Novel protein produced 53. Nutritional quality altered 54. Oil profile altered 55. Oil quality altered 56. Pectin esterase level reduced 57. Photosynthesis enhanced 58. Phytoene synthase activity increased 59. Pigment metabolism altered 60. Polyamine metabolism altered 61. Polygalacturonase level reduced 62. Processing characteristics altered 63. Prolonged shelf life 64. Protein altered 65. Protein lysine level increased 66. Protein quality altered 67. Proteinase inhibitors level constitutive 68. Rust resistant 69. Seed composition altered 70. Seed methionine storage increased 71. Seed number increased 72. Seed quality altered 73. Seed set reduced 74. Seed weight increased 75. Senescence altered 76. Solids increased 77. Starch level increased 78. Starch metabolism altered 79. Starch reduced 80. Steroidal glycoalkaloids reduced 81. Sterols increased 82. Storage protein altered 83. Sugar alcohol levels increased 84. Thermostable protein produced 85. Tryptophan level increased 86. Tuber solids increased 87. Yield increased Table 2 - Part 6. Non-limiting examples of traits/phenotypes with herbicide tolerance properties 1. 2,4-D tolerant 2. Chloroacetanilide tolerant 3. Fertility altered 4. Protein altered 5. Lignin levels decreased 6. Methionine level increased 7. Bromoxynil tolerant 8. Metabolism altered 9. Imidazole tolerant 10. Imidazolinone tolerant 11. Sulfonylurea tolerant 12. Northern corn leaf blight resistant 13. Herbicide tolerant 14. Isoxazole tolerant 15. Chlorsulfuron tolerant 16. Glyphosate tolerant 17. Lepidopteran resistant 18. Phosphinothricin tolerant 19. Sulfonylurea tolerant Table 2 - Part 7. Non-limiting examples of traits/phenotypes with pest resistance properties 1. Agrobacterium resistant - BR 2. Alternaria resistant - FR 3. Alternaria daucii resistant - FR 4. Alternaria solani resistant - FR 5. AMV resistant - VR 6. Anthracnose resistant - FR 7. Aphid resistant - IR 8. Apple scab resistant - FR 9. Aspergillus resistant - FR 10. Bacterial leaf blight resistant - BR 11. Bacterial resistant - BR 12. Bacterial soft rot resistant - BR 13. Bacterial soft rot resistant - VR 14. Bacterial speck resistant - BR 15. BCTV resistant - VR 16. Black shank resistant - FR 17. BLRV resistant - VR 18. BNYVV resistant - VR 19. Botrytis cinerea resistant - FR 20. Botrytis resistant - FR 21. BPMV resistant - VR 22. Brown spot resistant - FR 23. BYDV resistant - VR 24. BYMV resistant - VR 25. CaMV resistant - VR 26. Cercospora resistant - FR 27. Clavibacter resistant - BR 28. Closteroviurs resistant - BR 29. CLRV resistant - VR 30. CMV resistant - FR 31. Coleopteran resistant - IR 32. Colletotrichum resistant - FR 33. Colorado potato beetle resistant - IR 34. Corn earworm resistant - IR 35. Corynebacterium sepedonicum resistant - BR 36. Cottonwood leaf beetle resistant - IR 37. Criconnemella resistant - NR 38. Crown gall resistant - BR 39. Cucumovirus resistant - VR 40. Cylindrosporium resistant - FR 41. Disease resistant general - FR 42. Dollar spot resistant - FR 43. Downy mildew resistant - FR 44. Ear mold resistant - FR 45. Erwinia carotovora resistant - BR 46. European Corn Borer resistant - IR 47. Eyespot resistant - FR 48. Fall armyworm resistant - IR 49. Fire blight resistant - BR 50. Frogeye leaf spot resistanT - FR 51. Fruit rot resistant - FR 52. Fungal post-harvest resistant - FR 53. Fungal resistant - FR 54. Fungal resistant general - FR 55. Fusarium dehlae resistant - FR 56. Fusarium resistant - FR 57. Geminivirus resistant - VR 58. Gray lead spot resistant - FR 59. Helminthosporium resistant - FR 60. Hordothionin produced - BR 61. Insect predator resistant - IR 62. Insect resistant general - IR 63. Late blight resistant - FR 64. Leaf blight resistant - FR 65. Leaf spot resistant - FR 66. Lepidopteran resistant - IR 67. Lesser cornstalk borer resistant - IR 68. LMV resistant - VR 69. Loss of systemic resistance - VR 70. Marssonina resistant - FR 71. MCDV resistant - VR 72. MCMV resistant - VR 73. MDMV resistant - VR 74. MDMV-B resistant - VR 75. Mealybug wilt virus resistant - VR 76. Melamtsora resistant - FR 77. Melodgyne resistant - NR 78. Meloidogyne resistant - NR 79. Mexican Rice Borer resistant - IR 80. Mycotoxin degradation - FR 81. Nepovirus resistant - VR 82. Northern corn leaf blight resistant - IR 83. Nucleocapsid protein produced - VR 84. Oblique banded leafroller resistant - IR 85. Oomycete resistant - FR 86. Pathogenesis related proteins level increased - FR 87. PEMV resistant - VR 88. PeSV Resistant - VR 89. Phatora leaf beetle resistant - IR 90. Phoma resistant - FR 91. Phytophthora resistant - FR 92. PLRV resistant - VR 93. Potyvirus resistant - VR 94. Powdery mildew resistant - FR 95. PPV resistant - VR 96. Pralylenchus vulnus resistant - NR 97. PRSV resistant - VR 98. PRV resistant - VR 99. PSbMV resistant - VR 100. Pseudomonas syringae resistant - BR 101. PStV resistant - VR 102. PVX resistant - VR 103. PVY resistant - VR 104. RBDV resistant - VR 105. Rhizoctonia resistant - FR 106. Rhizoctonia solani resistant - FR 107. Ring rot resistance - BR 108. Root-knot nematode resistant - NR 109. Rust resistant - FR 110. SbMV resistant - VR 111. Sclerotinia resistant - FR 112. SCMV resistant - VR 113. SCYLV resistant - VR 114. Septoria resistant - FR 115. Smut resistant - FR 116. SMV resistant - VR 117. Sod web worm resistant - IR 118. Soft rot fungal resistant - FR 119. Soft rot resistant - BR 120. Southwestern corn borer resistant- IR 121. SPFMV resistant - VR 122. Sphaeropsis fruit rot resistant - FR 123. SqMV resistant - VR 124. SrMV resistant - VR 125. Streptomyces scabie s resistant - BR 126. Sugar cane borer resistant - IR 127. TEV resistant - VR 128. Thelaviopsis resistant - FR 129. TMV resistant - FR 130. Tobamovirus resistant - VR 131. ToMoV resistant - VR 132. ToMV resistant - VR 133. TRV resistant - VR 134. TSWV resistant - VR 135. TVMV resistant - VR 136. TYLCV resistant - VR 137. Venturia resistant - FR 138. Verticillium dahliae resistant - FR 139. Verticillium resistant - FR 140. Western corn root worm resistant - IR 141. WMV2 resistant - VR 142. WSMV resistant - VR 143. ZYMV resistant - VR Table 2 - Part 8. Non-limiting examples of miscellaneous traits/ phenotypes with properties 1. Antibiotic produced 2. Antiprotease producing 3. Capable of growth on defined synthetic media 4. Carbohydrate metabolism altered 5. Cell wall altered 6. Cold tolerant 7. Coleopteran resistant 8. Color altered 9. Color sectors in seeds 10. Colored sectors in leaves 11. Constitutive expression of glutaminc synthetase 12. Cre recombinase produced 13. Dalapon tolerant 14. Development altered 15. Disease resistant general 16. Ethylene metabolism altered 17. Expression optimization 18. Fenthion susceptible 19. Glucuronidase expressing 20. Glyphosate tolerant 21. Growth rate reduced 22. Heavy metals sequestered 23. Hygromycin tolerant 24. Inducible DNA modification 25. Industrial enzyme produced 26. Kanamycin resistant 27. Lipase expressed in seeds 28. Methotrexate resistant 29. Modified growth characteristics 30. Mycotoxin deficient 31. Mycotoxin production inhibited 32. Mycotoxin restored 33. Non-lesion forming mutant 34. Novel protein produced 35. Oil quality altered 36. Peroxidase levels increased 37. Pharmaceutical proteins produced 38. Phosphinothricin tolerant 39. Pigment metabolism altered 40. Pollen visual marker 41. Polyamine metablosim altered 42. Polymer produced 43. Recombinase produced 44. Secondary metabolite increased 45. Seed color altered 46. Seed weight increased 47. Selectable marker 48. Spectromycin resistant 49. Sterile 50. Sterols increased 51. Sulfonylurea susceptible 52. Syringomycin deficient 53. Transposon activator 54. Transposon elements inserted 55. Transposon inserted 56. Trifolitoxin producing 57. Trifolitoxin resistant 58. Virulence reduced 59. Visual marker 60. Visual marker inactive Legend BR—Bacterial Resistant FR—Fungal Resistant IR—Insent Resistant NR—Nematode Resistant VR—Viral Resistant In a particular examplification, “producing an organism having a desirable trait” includes an organism that is with respect to an organ or a part of an organ but not necessarily altered anywhere else. By “trait” is meant any detectable parameter associated with an organism under a set of conditions. Examples of “detectable parameters” include the ability to produce a substance, the ability to not produce a substance, an altered pattern of (such as an increased or a decreased) ability to produce a substance, viability, non-viability, behaviour, growth rate, size, morphology or morphological characteristic, In another embodiment, this invention is directed to a method of producing an organism having a desirable trait or a desirable improvement in a trait by: a) obtaining an initial population of organisms comprised of at least one starting organism, b) mutagenizing the population such that mutations occur throughout a substantial part of the genome of at least one initial organism, c) selecting at least one mutagenized organism having a desirable trait or a desirable improvement in a trait, and d) optionally repeating the method by subjecting one or more mutagenized organisms to a repetition of the method. A mutagenized organism having a desirable trait or a desirable improvement in a trait can be referred to as an “up-mutant”, and the associated mutation(s) contained in an up-mutant organism can be referred to as up-mutation(s). In one embodiment, step c) is comprised of selecting at least two different mutagenized organisms, each having a different mutagenized genome, and the method of producing an organism having a desirable trait or a desirable improvement in a trait is comprised of a) obtaining a starting population of organisms comprised of at least one starting organism, b) mutagenizing the population such that mutations occur throughout a substantial part of the genome of at least one starting organism, c) selecting at least two mutagenized organism having a desirable trait or a desirable improvement in a trait, d) creating combinations of the mutations of the two or more mutagenized organisms, e) selecting at least one mutagenized organism having a desirable trait or a desirable improvement in a trait, and f) optionally repeating the method by subjecting one or more mutagenized organisms to a repetition of the method. In one embodiment, the method is repeated. Thus, for example, an up-mutant organism can serve as a starting organism for the above method. Also, for example, an up mutant organism having a combination of two or more up-mutations in its genome can serve as a starting organism for the above method. Thus, in one embodiment, this invention is directed to a method of producing an organism having a desirable trait or a desirable improvement in a trait by: a) obtaining a starting population of organisms comprised of at least one starting organism, b) mutagenizing the population such that mutations occur throughout a substantial part of the genome of at least one starting organism, c) selecting at least one mutagenized organism having a desirable trait or a desirable improvement in a trait, and d) optionally repeating the method by subjecting one or more mutagenized organisms to a repetition of the method. A mutagenized organism having a desirable trait or a desirable improvement in a trait can be referred to as an “up-mutant”, and the associated mutation(s) contained in an up-mutant organism can be referred to as up-mutation(s). Mutagenizing a starting population such that mutations occur throughout a substantial part of the genome of at least one starting organism refers to mutagenizing at least approximately 1% of the genes of a genome, or at least approximately 10% of the genes of a genome, or at least approximately 20% of the genes of a genome, or at least approximately 30% of the genes of a genome, or at least approximately 40% of the genes of a genome, or at least approximately 50% of the genes of a genome, or at least approximately 60% of the genes of a genome, or at least approximately 70% of the genes of a genome, or at least approximately 80% of the genes of a genome, or at least approximately 90% of the genes of a genome, or at least approximately 95% of the genes of a genome, or at least approximately 98% of the genes of a genome. In a particular embodiment, this invention provides a method of producing an organism having a desirable trait or a desirable improvement in a trait by: a) obtaining sequence information of a genome; b) annotating the genomic sequence obtained; c) mutagenizing a substantial part of the genome the genome; d) selecting at least one mutagenized genome having a desirable trait or a desirable improvement in a trait; and e) optionally repeating the method by subjecting one or more mutagenized genomes to a repetition of the method. Thus in one aspect, this invention provides a process comprised of: 1.) Subjecting a working cell or organism to holistic monitoring (which can include the detection and/or measurement of all detectable functions and physical parameters). Examples of such parameters include morphology, behavior, growth, responsiveness to stimuli (e.g., antibiotics, different environment, etc.). Additional examples include all measurable molecules, including molecules that are chemically at least in part a nucleic acids, proteins, carbohydrates, proteoglycans, glycoproteins, or lipids. In a particular aspect, performing holistic monitoring is comprised of using a microarray-based method. In another aspect, performing holistic monitoring is comprised of sequencing a substantial portion of the genome, i.e. for example at least approximately 10% of the genome, or for example at least approximately 20% of the genome, or for example at least approximately 30% of the genome, or for example at least approximately 40% of the genome, or for example at least approximately 50% of the genome, or for example at least approximately 60% of the genome, or for example at least approximately 70% of the genome, or for example at least approximately 80% of the genome, or for example at least approximately 90% of the genome, or for example at least approximately 95% of the genome, or for example at least approximately 98% of the genome. 2) Introducing into the working cell or organism a plurality of traits (stacked traits), including selectively and differentially activatable traits. Serviceable traits for this purpose include traits conferred by genes and traits conferred by gene pathways. 3) Subjecting the working cell or organism to holistic monitoring. 4) Compiling the information obtained from steps 1) and 3), and processing &/or analyzing it to better understand the changes introduced into the working cell or organisms. Such data processing includes identifying correlations between and/or among the measured parameters. 5) Repeating any number or all of steps 2), 3), and 4). This invention provides that molecules serviceable for introducing transgenic traits into a plant include all known genes and nucleic acids. By way of non-limiting exemplification, this invention specifically names any number &/or combination of genes listed herein or listed in any reference incorporated herein by reference. Furthermore, by way of non-limiting exemplification, this invention specifically names any number &/or combination of genes & gene pathways listed herein as well as in any reference incorporated by reference herein. This invention provides that molecules serviceable as detectable parameters include molecule, any enzyme, substrate thereof, product thereof, and any gene or gene pathway listed herein including in any figure or table herein as well as in any reference incorporated by reference herein. This invention also relates generally to the field of nucleic acid engineering and correspondingly encoded recombinant protein engineering. More particularly, the invention relates to the directed evolution of nucleic acids and screening of clones containing the evolved nucleic acids for resultant activity(ies) of interest, such nucleic acid activity(ies) &/or specified protein, particularly enzyme, activity(ies) of interest. Mutagenized molecules provided by this invention may have chimeric molecules and molecules with point mutations, including biological molecules that contain a carbohydrate, a lipid, a nucleic acid, &/or a protein component, and specific but non-limiting examples of these include antibiotics, antibodies, enzymes, and steroidal and non-steroidal hormones. This invention relates generally to a method of: 1) preparing a progeny generation of molecule(s) (including a molecule that is comprised of a polynucleotide sequence, a molecule that is comprised of a polypeptide sequence, and a molecules that is comprised in part of a polynucleotide sequence and in part of a polypeptide sequence), that is mutagenized to achieve at least one point mutation, addition, deletion, &/or chimerization, from one or more ancestral or parental generation template(s); 2) screening the progeny generation molecule(s)—preferably using a high throughput method—for at least one property of interest (such as an improvement in an enzyme activity or an increase in stability or a novel chemotherapeutic effect); 3) optionally obtaining &/or cataloguing structural &/or and functional information regarding the parental &/or progeny generation molecules; and 4) optionally repeating any of steps 1) to 3). In a preferred embodiment, there is generated (e.g. from a parent polynucleotide template)—in what is termed “codon site-saturation mutagenesis”—a progeny generation of polynucleotides, each having at least one set of up to three contiguous point mutations (i.e. different bases comprising a new codon), such that every codon (or every family of degenerate codons encoding the same amino acid) is represented at each codon position. Corresponding to—and encoded by—this progeny generation of polynucleotides, there is also generated a set of progeny polypeptides, each having at least one single amino acid point mutation. In a preferred aspect, there is generated—in what is termed “amino acid site-saturation mutagenesis”—one such mutant polypeptide for each of the 19 naturally encoded polypeptide-forming alpha-amino acid substitutions at each and every amino acid position along the polypeptide. This yields—for each and every amino acid position along the parental polypeptide—a total of 20 distinct progeny polypeptides including the original amino acid, or potentially more than 21 distinct progeny polypeptides if additional amino acids are used either instead of or in addition to the 20 naturally encoded amino acids. Thus, in another aspect, this approach is also serviceable for generating mutants containing—in addition to &/or in combination with the 20 naturally encoded polypeptide-forming alpha-amino acids—other rare &/or not naturally-encoded amino acids and amino acid derivatives. In yet another aspect, this approach is also serviceable for generating mutants by the use of—in addition to &/or in combination with natural or unaltered codon recognition systems of suitable hosts—altered, mutagenized, &/or designer codon recognition systems (such as in a host cell with one or more altered tRNA molecules). In yet another aspect, this invention relates to recombination and more specifically to a method for preparing polynucleotides encoding a polypeptide by a method of in vivo re-assortment of polynucleotide sequences containing regions of partial homology, assembling the polynucleotides to form at least one polynucleotide and screening the polynucleotides for the production of polypeptide(s) having a useful property. In yet another preferred embodiment, this invention is serviceable for analyzing and cataloguing—with respect to any molecular property (e.g. an enzymatic activity) or combination of properties allowed by current technology—the effects of any mutational change achieved (including particularly saturation mutagenesis). Thus, a comprehensive method is provided for determining the effect of changing each amino acid in a parental polypeptide into each of at least 19 possible substitutions. This allows each amino acid in a parental polypeptide to be characterized and catalogued according to its spectrum of potential effects on a measurable property of the polypeptide. In another aspect, the method of the present invention utilizes the natural property of cells to recombine molecules and/or to mediate reductive processes that reduce the complexity of sequences and extent of repeated or consecutive sequences possessing regions of homology. It is an object of the present invention to provide a method for generating hybrid polynucleotides encoding biologically active hybrid polypeptides with enhanced activities. In accomplishing these and other objects, there has been provided, in accordance with one aspect of the invention, a method for introducing polynucleotides into a suitable host cell and growing the host cell under conditions that produce a hybrid polynucleotide. In another aspect of the invention, the invention provides a method for screening for biologically active hybrid polypeptides encoded by hybrid polynucleotides. The present method allows for the identification of biologically active hybrid polypeptides with enhanced biological activities. Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. In yet another aspect, this invention relates to a method of discovering which phenotype corresponds to a gene by disrupting every gene in the organism. Accordingly, this invention provides a method for determining a gene that alters a characteristic of an organism, comprising: a) obtaining an initial population of organisms, b) generating a set of mutagenized organisms, such that when all the genetic mutations in the set of mutagenized organisms are taken as a whole, there is represented a set of substantial genetic mutations, and c) detecting the presence an organism having an altered trait, and d) determining the nucleotide sequence of a gene that has been mutagenized in the organism having the altered trait. In yet another aspect, this invention relates to a method of improving a trait in an organism by functionally knocking out a particular gene in the organism, and then transferring a library of genes, which only vary from the wild-type at one codon position, into the organism. Accordingly, this invention provides a method method for producing an organism with an improved trait, comprising: a) functionally knocking out an enogenous gene in a substantially clonal population of organisms; b) transferring the set of altered genes into the clonal population of organisms, wherein each altered gene differs from the endogenous gene at only one codon; and c) detecting a mutagenized organism having an improved trait; and d) determining the nucleotide sequence of a gene that has been transferred into the detected organism.
Apparatus and method for detecting transmitting rate of turbo decoder
The present invention relates to an apparatus and method for detecting a data rate in a turbo decoder for a mobile communication system. When a rate selector selects one data rate among a plurality of data rates, a turbo decoder repeatedly decodes an input data frame within a predetermined repetition limit number using the selected data rate and outputs the decoded data. A CRC detector performs CRC check on the decoded data and outputs the CRC check result, and a decoding state measurer measures decoding quality depending on the decoded data and outputs decoding state information. A controller then sets the repetition limit number to a predetermined minimum value, controls the repetition limit number according to the decoding state information, controls the rate selector and determines a data rate of the input data depending on the CRC check result.
1. A method for decoding coded data in one frame transmitted from a transmitter at one data rate of a plurality of data rates by a turbo decoder of a receiver having no information on a data rate at which the coded data is transmitted, and detecting a data rate of the coded data, comprising the steps of: (a) decoding the coded data in the frame at a selected data rate of the plurality of the data rates by the turbo decoder and calculating a current decoding state value indicating an average of absolute values of log likelihood ratio (llr) values being identical to decoded values of the coded data, output from the turbo decoder; (b) calculating a current under-decoding state value defined as a difference between the current decoding state value and a previous decoding state value; (c) performing CRC (Cyclic Redundancy Check) checking on the decoded data, if the current decoding state value is larger than a first threshold or the current under-decoding state value is larger than a second threshold; and (d) determining the selected data rate as a data rate of the coded data transmitted from the transmitter, if the CRC checking result is good. 2. The method as claimed in claim further comprising the step of (e) repeating the steps (a) to (d) within an iteration limit number until the selected data rate is determined as a data rate of the coded data, if the CRC checkingy result is not good. 3. The method as claimed in claim 2, further comprising the step of repeating the steps (a) to (e) until another selected data rate of the plurality of the data rates is determined as a data rate of the coded data, if a data rate of the coded data is not detected from the selected data rate. 4. The method as claimed in claim 3, wherein the iteration limit number is changed based on the current decoding state value and the current under-decoding state value during every decoding. 5. The method as claimed in claim 4, wherein the step (a) comprises the step of setting the iteration limit number to a predetermined, minimum value. 6. The method as claimed in claim 5, wherein the iteration limit number is set to a predetermined maximum value, if the current decoding state value is larger than the first threshold or the current under-decoding state value is larger than the second threshold. 7. The method as claimed in claim 5, wherein the iteration limit number is set to a predetermined maximum value, if the current decoding state value is larger than the first threshold and the current under-decoding state value is less than or equal to the second threshold. 8. The method as claimed in claim 7, wherein the iteration limit number is an intermediate value between the minimum value and the maximum value, if the current decoding state value is less than or equal to the first threshold and the current under-decoding state value is larger than the second threshold. 9. The method as claimed in claim 6 or 8, further comprising the step of returning to the step (a) without performing CRC checking on the decoded data, if the current decoding state value is less than or equal to the first threshold and the current under-decoding state value is less than or equal to the second threshold. 10. The method as claimed in any of claims 1 to 3, wherein the decoding state value is calculated by m =  llr ⁡ ( a )  +  llr ⁡ ( a + 1 )  +  llr ⁡ ( a + 2 )  ⁢ ⋯ ⁢  llr ⁡ ( b )  b - ( a - 1 ) where m represents the decoding state value, ‘a’ and ‘b’ represent constants, wherein 0≦a<b<FL where FL represents a frame length associated with the selected data rate, and llr(n) represents a soft output value of an nth bit from the turbo decoder for the FL. 11. A method for decoding coded data in one frame transmitted from a transmitter at one data rate of a plurality of data rates by a turbo decoder of a receiver having no information on a data rate at which the coded data is transmitted, and detecting a data rate of the coded data, comprising the steps of: (a) decoding the data coded at a selected data rate of the plurality of the data rates and outputting decoded data; (b) controlling an iteration limit number depending on decoding state information measured for the decoded data, if a number of decoding performed at the selected data rate is less than the iteration limit number; (c) performing CRC checking on the decoded data, if the number of decoding is larger than or equal to the iteration limit number; (d) determining the selected data rate as a data rate of the coded data transmitted from the transmitter, if the CRC checking result is good; (e) repeating the steps (a) to (d) within the iteration limit number until the selected data rate is determined as the data of the coded data, if the CRC checking result is not good; and (f) repeating the steps (a) to (e) until another selected data rate of the plurality of the data rates is determined as the data rate of the coded data, if the selected data rate is not determined as the data rate of the coded data. 12. The method as claimed in claim 11, wherein the decoding state information includes a current decoding state value indicating an average of absolute values of log likelihood ratio (llr) values being identical to soft output values of the decoded data, and also includes a current under-decoding state value defined as a difference between a current decoding state value and a previous decoding state value. 13. The method as claimed in claim 12, wherein the decoding state value is calculated by m =  llr ⁡ ( a )  +  llr ⁡ ( a + 1 )  +  llr ⁡ ( a + 2 )  ⁢ ⋯ ⁢  llr ⁡ ( b )  b - ( a - 1 ) where m represents the decoding state value, ‘a’ and ‘b’ represent constants, wherein 0≦a<b<FL where FL represents a frame length associated with the selected data rate, and llr(n) represents a soft output value of an nth bit from the turbo decoder for the FL. 14. The method as claimed in claim 12, wherein the step (a) comprises the step of setting the iteration limit number to a predetermined minimum value. 15. The method as claimed in claim 14, wherein in the step (b), the iteration limit number is set to a predetermined maximum value, if the current decoding state value is larger than a first threshold or the current under-decoding state value is larger than a second threshold. 16. The method as claimed in claim 14, wherein in the step (b), the iteration limit number is set to a predetermined maximum value, if the current decoding state value is larger than a first threshold and the current under-decoding state value is less than or equal to a second threshold. 17. The method as claimed in claim 16, wherein in the step (b), the iteration limit number is set to an intermediate vague between the minimum value and the maximum value, if the current decoding state value is less than or equal to the first threshold and the current under-decoding state value is larger than the second threshold. 18. The method as claimed in claim 15 or 17, further comprising the step of returning to the step (a) without performing CRC checking on the decoded data, if the current decoding state value is less than or equal to a first threshold and the current under-decoding state value is less than or equal to a second threshold. 19. An apparatus for decoding coded data in one frame transmitted from a transmitter at one data rate of a plurality of data rates by a turbo decoder of a receiver having no information on a data rate at which the coded data is transmitted, and detecting a data rate of the coded data, the apparatus comprising: a data rate determiner for selecting a data rate from a plurality of data rates; a turbo decoder for iteratively decoding an input data frame within an iteration limit number using the selected data rate, and outputting decoded data; a CRC detector for performing CRC checking on the decoded data and outputting a CRC checking result; a decoding state measurer for measuring decoding quality using the decoded data and outputting decoding state information; and a controller for first determining the iteration limit number as a minimum value, controlling the iteration limit number based on the decoding state information, controlling the data rate determiner, and determining a data rate of the input data based on the CRC checking result. 20. The apparatus as claimed in claim 19, wherein the decoding state information includes a current decoding state value indicating an average of absolute values of log likelihood ratio (llr) values being identical to soft output values of the decoded data, and also includes a current under-decoding state value defined as a difference between a current decoding state value and a previous decoding state value. 21. The apparatus as claimed in claim 20, wherein the decoding state value is calculated by m =  llr ⁡ ( a )  +  llr ⁡ ( a + 1 )  +  llr ⁡ ( a + 2 )  ⁢ ⋯ ⁢  llr ⁡ ( b )  b - ( a - 1 ) where in represents the decoding state value, ‘a’ and ‘b’ represent constants, wherein 0≦a<b<FL where FL represents a frame length associated with the selected data rate, and llr(n) represents a soft output value of an nth bit from the turbo decoder for the FL. 22. The apparatus as claimed in claim 20, wherein the controller sets the iteration limit number to a predetermined maximum value, if the current decoding state value is larger than a first threshold or the current under-decoding state value is larger than a second threshold. 23. The apparatus as claimed in claim 20, wherein the controller sets the iteration limit number to a predetermined maximum value if the current decoding state value is larger than a first threshold and the current under-decoding state value is less than or equal to a second threshold. 24. The apparatus as claimed in, claim 23, wherein the controller sets the iteration limit member to an intermediate value between the minimum value and the maximum value, if the current decoding state value is less than or equal to the first threshold and the current under-decoding state value is larger than the second threshold.
<SOH> BACKGROUND OF THE INVENTION <EOH>1. Field of the Invention The present invention relates generally to an apparatus and method for detecting a data rate, and in particular, to an apparatus and method for detecting a data rate of a turbo decoder in a mobile communication system. 2. Description of the Related Art In general, an encoder and a decoder are used in a digital mobile communication system to correct an error of a forward channel. The digital mobile communication system transmits and receives data in a radio environment, so it employs a coding technique to prevent generation of noises in a transmission channel. The mobile communication system typically uses a turbo coding technique for the coding technique. 3GPP (3 rd Generation Partnership Projection) or 3GPP2 (3 rd Generation Partnership Projection 2), an ongoing standardization work on the mobile communication system, specifies that transmission data can be transmitted at different data rates. Here, the data rate depends upon a length (or size) of a frame decoded by a turbo decoder. For example, a data rate of 2.4 Kbps corresponds to a decoded frame length of 24 bits/frame, and a data rate of 4.8 Kbps corresponds to a decoded frame length of 48 bits/frame. When transmitting data at various data rates, a transmitter generally transmits the data to a receiver along with information on a data rate of the currently transmitted data. However, transmitting the data along with the data rate of the transmission data causes a waste of transmission power, especially when the transmission data has a low data rate. Therefore, there is a demand for a method for transmitting data without information on its data rate. There is a known blind rate detection (BRD) technique, in which a transmitter transmits data without information on its data rate and a receiver detects the data rate based on only the received data. When data is transmitted in the BRD mode after being subject to turbo encoding the results of CRC (Cyclic Redundancy Code) checking performed on an output of a turbo decoder are typically used to detect a data rate. That is, the transmitter adds CRC information to transmission data before transmission. Then, a channel decoder of the receiver decodes the received data at all of its available data rates and determines whether the decoded data includes noises, through CRC checking. If it is determined through the CRC checking performed at a specific data rate that the frame is not damaged, the receiver detects the corresponding data rate. A procedure for detecting a data rate by the receiver using the CRC checking will be described in detail with reference to FIG. 1 . FIG. 1 illustrates a procedure for detecting a data rate by a receiver through CRC checking of the BRD technique in a mobile communication system according to the prior art. A detailed description of the conventional procedure for detecting a data rate will now be made with reference to FIG. 1 . In step 10 , the receiver performing the turbo decoding receives frame data over a radio channel. In this procedure, a description of a radio processing process and a channel decoding process will not be provided. After receiving the frame data, the receiver sets count values i and j to ‘0’, in step 12 . Here, i represents a value for counting a decoding iteration number (or decoding frequency) to perform turbo decoding on one frame at a selected data rate. Further, j represents a value for counting the number of types of the data rates. That is, if the turbo decoding is iteratively performed 6 times, the i value represents a decoding iteration count value for limiting the turbo decoding to be performed up to 6 times, and if the number of the types of the data rates is 7, the j value represents a data rate type count value for performing the decoding at up to 7 data rates. After the step 12 , the receiver sets an iteration limit number to its maximum value in step 14 . Here, the “iteration limit number” refers to the maximum number of iterative decoding at a specific data rate, i.e., a value for setting a threshold for the iteration count value ‘i’ of the turbo decoder. In step 16 , the receiver performs turbo decoding at a data rate set to the j value. For example, if decoding on voice data is performed at 2 Kbps, 4 Kbps and 8 Kbps, then the receiver performs the decoding on the data with a frame length FL(j) associated with 2 Kbps or 8 Kbps, whichever determined first. That is, the receiver performs the decoding at one initial data rate. After the decoding, the receiver analyzes the result of CRC checking performed on the decoded data in step 18 . As the result of the analysis, if it is determined that the CRC is ‘good’, the receiver performs a data rate detection process for the current j value in step 20 . Otherwise, the receiver determines whether the decoding iteration count value i is less than the iteration limit number, in order to iterate the decoding up to the iteration limit number. As the result of the determination, if the decoding iteration count value i is less than the iteration limit number, the receiver proceeds to step 24 , and otherwise, proceeds to step 26 . In the step 24 , the receiver increases the decoding iteration count value i by 1, and then returns to the step 16 . The reason for iteratively performing the decoding at one data rate will be described with reference to FIGS. 2 and 3 . FIG. 2 illustrates distribution of a decoding state value m(i) indicating the quality of a data frame when data with a frame size 60 transmitted over a radio channel is subject to iterative turbo decoding by a turbo decoder with a frame size 60 . FIG. 3 illustrates distribution of an m(i) value when data with a frame size 40 transmitted over a radio channel is subject to iterative turbo decoding by a turbo decoder with a frame size 60 . In FIG. 2 , a part represented by a solid line represents a part having an m(i) value of correctly decoded frame, while a part represented by dots represents an incorrectly decoded part. When performing the decoding for second time, the receiver iteratively performs the decoding for restoration, using the incorrectly decoded frames represented by the dots. Then, the incorrectly decoded frames are divided again into restored frames and non-restored frames, as shown in a second graph of FIG. 2 . If the process is iterated several times, the frame data will be more correctly decoded, thus increasing a probability that the CRC will be detected in a ‘good’ state. However, in the case where the data is decoded with a frame length associated with another data rate, even though the decoding is performed several times, the decoding results will continuously show an error state as shown in FIG. 3 . Turning back to FIG. 1 , the iterative limit number is typically set to 6. If the CRC is not ‘good’ after iterating the decoding six times at one data rate, the receiver determines in step 26 whether the data rate type count value j is larger than the number N of the data rates. As the result of the determination, if the data rate type count value j is not less than the number N, e.g., 3, of the data rates, the receiver proceeds to step 28 to perform a data rate detection failure process, considering that the decoding and the CRC checking have been completed for all of the available data rates. However, if the count value j indicates that the decoding has not yet performed at all of the data rates, i.e., if the j value is less than N, the receiver increases the count value j by 1 and sets the decoding iteration count value i to 0 in step 30 , and then returns to the step 16 to repeat the steps 16 to 30 . However, the turbo decoding technique for detecting a data rate through the above process causes a waste of time in detecting the data rate, especially when there is a great difference between a data rate at which the data frame has been transmitted and the initial data rate at which the turbo decoding is performed. That is, for example, if it is assumed that the number of the data rates to be detected is N and the iteration number is 8, the decoding is performed (N−1)*8 times at the worst. In addition, an increase in the number of the data rates for the transmission data causes an increase in the detection time and also causes an increase in power consumption for detecting the data rate.
<SOH> SUMMARY OF THE INVENTION <EOH>It is, therefore, an object of the present invention to provide an apparatus and method for controlling an iteration limit number for turbo decoding when detecting a data rate of a turbo-coded data in a BRD mode. It is another object of the present invention to provide an apparatus and method for reducing a delay time and power consumption when detecting a data rate of turbo-coded data in a BRD mode. According to one aspect of the present invention, A method for decoding coded data in one frame transmitted from a transmitter at one data rate of a plurality of data rates by a turbo decoder of a receiver having no information on a data rate at which the coded data is transmitted, and detecting a data rate of the coded data, comprising the steps of: (a) decoding the coded data in the frame at a selected data rate of the plurality of the data rates by the turbo decoder and calculating a current decoding state value indicating an average of absolute values of log likelihood ratio (llr) values being identical to decoded values of the coded data, output from the turbo decoder; (b) calculating a current under-decoding state value defined as a difference between the current decoding state value and a previous decoding state value; (c) performing CRC (Cyclic Redundancy Check) checking on the decoded data, if the current decoding state value it larger than a first threshold or the current under-decoding state value is larger than a second threshold; and (d) determining the selected data rate as a data rate of the coded data transmitted from the transmitter, if the CRC checking result is good. According to a second aspect of the present invention, a method for decoding coded data in one frame transmitted from a transmitter at one data rate of a plurality of data rates by a turbo decoder of a receiver having no information on a data rate at which the coded data is transmitted, and detecting a data rate of the coded data, comprising the steps of: (a) decoding the data coded at a selected data rate of the plurality of the data rates and outputting decoded data; (b) controlling an iteration limit number depending on decoding state information measured for the decoded data, if a number of decoding performed at the selected data rate is less than the iteration limit number; (c) performing CRC checking on the decoded data, if the number of decoding is larger than or equal to the iteration limit number; (d) determining the selected data rate as a data rate of the coded data transmitted from the transmitter, if the CRC checking result is good; (e) repeating the steps (a) to (d) within the iteration limit number until the selected data rate is determined as the data of the coded data, if the CRC checking result is not good; and (f) repeating the steps (a) to (e) until another selected data rate of the plurality of the data rates is determined as the data rate of the coded data, if the selected data rate is not determined as the data rate of the coded data. According to a third aspect of the present invention, an apparatus for decoding coded data in one frame transmitted from a transmitter at one data rate of a plurality of data rates by a turbo decoder of a receiver having no information on a data rate at which the coded data is transmitted., and detecting a data rate of the coded data, the apparatus comprising: a data rate determiner for selecting a data rate from a plurality of data rates; a turbo decoder for iteratively decoding an input data frame within an iteration limit number using the selected data rate, and outputting decoded data; a CRC detector for performing CRC checking on the decoded data and outputting a CRC checking result; a decoding state measurer for measuring decoding quality using the decoded data and outputting decoding state information; and a controller for first determining the iteration limit number as a minimum value, controlling the iteration limit number based on the decoding state information, controlling the data rate determiner, and determining a data rate of the input data based on the CRC checking result.
Methods and composition for visualizing and interfering with chromosomal tethering of extrachromosomal molecules
The invention provides methods and compositions for visualizing and interfering with chromosomal tethering and segregation of extrachromosomal molecules.
1. A preselected nucleic acid molecule comprising an extrachromosomal molecule operably linked to a tag. 2. The preselected molecule of claim 1, wherein the extrachromosomal molecule is a double minute chromosome. 3. The preselected molecule of claim 1, wherein the extrachromosomal molecule is a viral nucleic acid sequence. 4. The vector of claim 3, wherein the viral genome is maintained as an episome within a cell. 5. The preselected molecule of claim 3, wherein the viral nucleic acid sequence is from an RNA virus. 6. The preselected molecule of claim 3, wherein the viral nucleic acid sequence is from a DNA virus. 7. The preselected molecule of claim 3, wherein the viral nucleic acid sequence is from Flaviviridae, Retroviridae, Hepadnaviridae, Papovaviridae, Adenoviridae, Herpesviridae, Poxviridae, a Hepatitis C virus, a Papillomavirus, an Epstein-Barr virus, an Influenza virus or a Polyomavirus. 8. The preselected molecule of claim 1, wherein the extrachromosomal molecule comprises an oncogene. 9. The preselected molecule of claim 8, wherein the oncogene is selected from sis, erbB, fins, sea, kit, ros, mpl, eyk, erbA, H-ras, K-ras, crk, src, abl, fps, fes, fgr, yes, mos, raf, mil, akt, jun, fos, myc, myb, ets, rel, mat, ski or qin. 10. The preselected molecule of claim 1, wherein the tag is a reporter gene. 11. The preselected molecule of claim 10, wherein the reporter gene is green fluorescent protein, cyan fluorescent protein, red fluorescent or yellow fluorescent protein. 12. The preselected molecule of claim 1, wherein the tag is a selection marker. 13. The preselected molecule of claim 12, wherein the selection marker is resistance to chloramphenicol, rifampicin, ampicillin or blasticidin. 14. The preselected molecule of claim 13, wherein the selection marker is a blasticidin resistance gene (bsr) driven by an SRα promoter. 15. The preselected molecule of claim 1, wherein the tag is a binding site for a detectable trans-acting element that binds to the tag. 16. The preselected molecule of claim 1, comprising a vector that integrates into an extrachromosomal molecule. 17. The preselected molecule of claim 16, wherein the vector comprises Epstein-Barr virus (EBV), bovine papillomavirus (BPV), or Kaposi's sarcoma associated herpesvirus (KSHV) vector sequences. 18. The preselected molecule of claim 16, comprising EBNA-1 sequences and an oriP sequence, wherein the oriP sequence has a plurality of EBNA-1 binding sites. 19. The preselected molecule of claim 18, wherein the EBNA-1 binding sites are located at two distinct regions within the vector. 20. The preselected molecule of claim 18, wherein the EBNA-1 binding sites have dyad symmetry within the vector. 21. The preselected molecule of claim 13, comprising an FR element from oriP. 22. The preselected molecule of claim 18, comprising a plurality of tandem repeats of a lac operator (lacO). 23. The preselected molecule of claim 13, further comprising a nucleotide sequence that interferes with chromosomal tethering. 24. The preselected molecule of claim 13, further comprising a polynucleotide that encodes a lac repressor (lacR)-GFP fusion protein that binds to the lac operator. 25. A preselected molecule comprising a reporter gene fused to a lac repressor-nuclear localization signal. 26. The preselected molecule of claim 25, wherein the reporter gene is green fluorescent protein, cyan fluorescent protein, red fluorescent or yellow fluorescent protein. 27. The preselected molecule of claim 26, further comprising retroviral sequences. 28. The preselected molecule of claim 26, further comprising plasmid sequences. 29. A vector comprising a histone H2B gene fused to a reporter gene. 30. The vector of claim 29, wherein the reporter gene is green fluorescent protein, cyan fluorescent protein, red fluorescent or yellow fluorescent protein. 31. The vector of claim 29, further comprising retroviral sequences. 32. The vector of claim 29, further comprising plasmid sequences. 33. A recombinant chromosomal tethering polypeptide. 34. The polypeptide of claim 33, operably linked to a cellular chromatid and an extrachromosomal molecule. 35. The polypeptide of claim 34, wherein the extrachromosomal molecule is a double minute chromosome. 36. The polypeptide of claim 33, operably linked to a cellular chromatid and to an oriP-containing vector. 37. The polypeptide of claim 33, wherein the polypeptide comprises a cellular polypeptide. 38. The polypeptide of claim 33, wherein the polypeptide comprises a viral polypeptide. 39. A cell comprising the preselected molecule of claim 1, the vector of claim 29 or the polypeptide of claim 33. 40. The cell of claim 39, wherein the cell is a COLO320DM cell. 41. A method of visualizing chromosomal tethering of an extrachromosomal molecule comprising contacting the vector of claims 13, 25, or 29, with a cell suspected of containing an extrachromosomal molecule. 42. The method of claim 41, wherein the extrachromosomal molecule is a double minute chromosome. 43. The method of claim 41, wherein the extrachromosomal molecule is a virus. 44. A method of interfering with chromosomal tethering of extrachromosomal molecule by contacting the vector of claims 13, 25, or 29, with a cell suspected of containing an extrachromosomal molecule. 45. The method of claim 44, wherein the extrachromosomal molecule is a double minute chromosome. 46. The method of claim 44, wherein the extrachromosomal molecule is a virus. 47. A method to identify at least one agent that modulates chromosomal tethering of an extrachromosomal molecule comprising: (a) contacting a cell that contains an extrachromosomal molecule with a test agent; and (b) determining if the agent causes an increase or decrease in segregation of the extrachromosomal molecule when the cell divides. 48. The method of claim 47, wherein the extrachromosomal molecule is operably linked to a tag. 49. The method of claim 48, wherein the tag is a reporter gene. 50. The method of claim 49, wherein the reporter gene is green fluorescent protein, cyan fluorescent protein, red fluorescent or yellow fluorescent protein. 51. The method of claim 47, wherein the extrachromosomal molecule is operably linked to a selection marker. 52. The method of claim 51, wherein the selection marker is resistance to chloramphenicol, rifampicin, ampicillin or blasticidin. 53. The method of claim 47, wherein the cell is the cell of claim 39. 54. The method of claim 47, wherein the cell comprises an extrachromosomal molecule into which a vector of claim 13 has integrated. 55. The method of claim 47, wherein the extrachromosomal molecule is a double minute chromosome or a virus. 56. A method of treating cancer comprising administering a pharmaceutical composition comprising a compound that inhibits tethering of an extrachromosomal molecule to a chromosome. 57. The method of claim 56, wherein the extrachromosomal molecule is a virus or a double minute chromosome. 58. A method of treating a viral infection comprising administering a pharmaceutical composition comprising a compound that inhibits tethering of a viral extrachromosomal molecule to a cellular chromatid. 59. A method for identifying an antiviral agent, comprising: (a) contacting a cell comprising a viral acentric extrachromosomal molecule with a test compound; and (b) identifying a compound that inhibits association of the viral acentric extrachromosomal molecule with a cellular chromatid. 60. The method according to claim 59, further comprising (c) determining if the compound inhibits the association of a tethering polypeptide with the cellular chromatid or with the viral acentric extrachromosomal molecule. 61. The method of claim 60, wherein the tethering polypeptide is Epstein-Barr nuclear antigen or herpesvirus latent nuclear antigen. 62. A method for identifying an anticancer agent, comprising: (a) contacting a cell comprising an extrachromosomal molecule with a test compound; and (b) identifying a compound that inhibits association of the extrachromosomal molecule with a cellular chromatid. 63. The method according to claim 62, further comprising (c) determining if the compound inhibits association of a tethering polypeptide with the cellular chromatid or with the extrachromosomal molecule. 64. The method according to claim 63, wherein the polypeptide is Epstein-Barr nuclear antigen or herpesvirus latent nuclear antigen. 65. A chromosomally integrating vector that specifically labels double-minute chromosomes (DMs). 66. The vector of claim 65, wherein the vector is an Epstein-Barr virus (EBV), bovine papillomavirus (BPV), or Kaposi's sarcoma associated herpesvirus (KSHV) vector sequences. 67. The vector of claim 65, wherein the vector comprises an EBNA-1 gene and an oriP sequence, wherein the oriP sequence has a plurality of EBNA-1 binding sites in two distinct regions. 68. The vector of claim 65, wherein the vector comprises a plurality of tandem repeats of a lac operator (lacO). 69. The vector of claim 65, wherein the vector comprises a reporter gene. 70. The vector of claim 69, wherein the reporter gene is GFP or YFP. 71. The vector of claim 65, wherein the vector comprises a selection marker. 72. The vector of claim 71, wherein the selection marker is a blasticidin resistance gene (bsr) driven by SR promoter. 73. The vector of claim 65, wherein the vector further comprises a nucleotide sequence that interferes with chromosomal tethering. 74. The vector of claim 65, wherein the vector further encodes a lac repressor (lacR)-GFP fusion protein that binds with high affinity to the lacO. 75. The vector of claim 65, wherein the vector is a modified virus. 76. The vector of claim 65, wherein the vector is a modified animal virus. 77. The vector of claim 65, wherein the vector is a DNA virus. 78. The vector of claim 65, wherein the vector is a member of the Herpesviridae, Papovaviridae or Adenoviridae. 79. A plasmid vector comprising retroviral vector, a gene encoding GFP fused to lac repressor-nuclear localization signal. 80. A plasmid vector comprising retroviral vector, a gene encoding YFP fused to lac repressor-nuclear localization signal. 81. A plasmid vector comprising a histone H2B gene and a CFP gene.
<SOH> BACKGROUND OF THE INVENTION <EOH>The replication and subsequent faithful segregation of duplicated chromosomes are crucial for the proper transmission of the cellular genome to daughter cells. In higher eukaryotes, the nuclear membrane breaks down at the beginning of mitosis, and subsequently spindle microtubules attach to centromeric kinetochores to assure the even distribution of sister chromatids. At the end of mitosis, the nuclear membrane reassembles around each group of chromosomes to form two daughter nuclei. Therefore, it would be reasonable to assume that acentric DNA molecules should not be maintained stably in nuclei as they do not attach to microtubules, and that acentric DNA molecules should be dispersed throughout the cytoplasm subsequent to nuclear membrane breakdown. In many cases, however, acentric DNA molecules, lacking functional centromeres, exhibit a surprisingly high stability in dividing cells. Examples of such stably-transmitted acentric DNA molecules in human cells include cellular acentric chromosomes called double minute chromosomes (DMs) and extrachromosomally replicating viral DNAs. DMs are cancer specific genomic anomalies known to harbor amplified oncogenes and drug resistance genes (Alitalo and Schwab, 1986; Hahn, 1993; Wahl, 1989). They are autonomously replicating, acentric, atelomeric, circular chromatin bodies, and usually 1-2 megabase pairs in size. Although they apparently lack functional centromeres (Barker and Hsu, 1978; Levan and Levan, 1978), their segregation efficiency is much higher than expected (Kimmel et al., 1992; Pauletti et al., 1990). Clues to the mechanisms underlying the efficient segregation came from light and electron microscopic observations showing that DMs frequently associated with mitotic chromosomes (Barker and Hsu, 1978; Hamkalo et al., 1985; Jack et al., 1987; Levan and Levan, 1978). These observations were extended using a fusion protein of human histone H2B and Aequorea victoria green fluorescent protein (H2B-GFP) to reveal DM clusters tethered to segregating daughter chromosomes in living cancer cells (Kanda et al., 1998). Time-lapse microscopy demonstrated that DMs could ‘hitchhike’ on segregating chromosomes from anaphase to telophase, indicating how chromosome tethering could contribute to increased segregation efficiency. There is a continuing need for improved visualization of viral and cellular acentric extrachromosomal molecules that tether to chromosomes. There is a further need to identify compounds that effectively interfere with the chromosomal tethering of these viral and cellular acentric extrachromosomal molecules. Abbreviations: Epstein-Barr virus (EBV); lac operator (lacO); Epstein-Barr nuclear antigen-1 (EBNA-1); green fluorescent protein (GFP); yellow fluorescent protein (YFP); red fluorescent protein (RFP); cyan fluorescent protein (CFP); Chinese hamster ovary cells (CHO); double minute chromosome (DM); lac repressor (lacR); fluorescent in situ hybridization (FISH); ampicillin (Amp); family of repeats (FR).
<SOH> SUMMARY OF THE INVENTION <EOH>The invention provides a preselected nucleic acid molecule comprising an extrachromosomal molecule operably linked to a tag. The extrachromosomal molecule can be any molecule that segregates with cellular chromosomes during cell division. For example, the extrachromosomal molecule may be a double minute chromosome or a viral nucleic acid sequence from a DNA or RNA virus. The viral genome may be maintained as an episome within a cell. Examples of sources of the viral nucleic acid include, but are not limited to Flaviviridae, Retroviridae, Hepadnaviridae, Papovaviridae, Adenoviridae, Herpesviridae, Poxviridae, a Hepatitis C virus, a Papillomavirus, an Epstein-Barr virus, an Influenza virus or a Polyomavirus. Alternatively, the extrachromosomal molecule may be an oncogene, such as sis, erbB, fins, sea, kit, ros, mpl, eyk, erbA, H-ras, K-ras, crk, src, abl, fps, fes, fgr, yes, mos, raf, mil, akt, jun, fos, myc, myb, ets, rel, maf, ski or qin. The tag includes any nucleic acid sequence that encodes a selection marker (such as a blasticidin resistance gene (bsr), ampicillin, rifampicin, chloramphenicol or other art recognized drug resistance selection marker gene). Such selection markers may be driven by any operably linked promoter, such as the SRα promoter. The tag may be a reporter gene (such as green fluorescent protein, yellow fluorescent protein, red fluorescent protein or cyan fluorescent protein) that allows the extrachromosomal molecule to be detected. The tag may also be a binding site for a detectable trans-acting element that binds to the tag and thereby allows detection of the extrachromosomal molecule. The tag can be operably linked to the extrachromosomal molecule through integration of an integrating vector, into the extrachromosomal molecule. The preselected nucleic acid may further contain an integrating vector that specifically labels double-minute chromosomes (DMs). The vector may contain Epstein-Barr virus (EBV), bovine papillomavirus (BPV), or Kaposi's sarcoma associated herpesvirus (KSHV) sequences. The vector may contain an EBNA-1 gene and an oriP sequence, wherein the oriP sequence has a plurality of EBNA-1 binding sites in two distinct regions. The vector may contain a plurality of tandem repeats of a lac operator (lacO). The vector may contain a nucleotide sequence that interferes with chromosomal tethering such as an antisense message to a tethering protein, such as the Epstein-barr nuclear antigen. The vector may encode a fusion protein that binds to an extrachromosomal molecule such as a lac repressor (lacR)-GFP fusion protein that binds with high affinity to the lacO that may be integrated into the extrachromosomal molecule. Examples of the present invention include, but are not limited to, a plasmid vector containing a retroviral vector, a gene encoding GFP fused to lac repressor-nuclear localization signal, or a vector containing a gene encoding YFP fused to lac repressor-nuclear localization signal, or a histone H2B gene fused to a CFP gene. The present invention further provides a chromosomal tethering polypeptide. The polypeptide can operably link a cellular chromatid and an extracellular molecule. Such an extracellular molecule may be an oriP-containing vector. This polypeptide may be of cellular or viral origin. The tethering protein may also be a peptidomimetic or a fusion polypeptide having a chromatid binding domain and a domain that binds to an extrachromosomal molecule. The present invention further provides a method of visualizing chromosomal tethering of extrachromosomal molecules, such as viral acentric extrachromosomal molecules (DAE) or double-minute chromosomes (DM), by contacting a vector of the invention with a cell suspected of containing a DM or a DAE. The extrachromosomal molecule, such as the DM or DAE, may associate with a cellular chromatid through the action of a tethering polypeptide. The present invention provides a method of interfering with chromosomal tethering of extrachromosomal molecules, such as viral or cellular acentric extrachromosomal molecules, by administering a vector that specifically labels the extrachromosomal molecule, such as a double-minute chromosome (DM). Additionally, the vector may produce a product that inhibits the expression or function of a tethering polypeptide, such as an antisense message to the Epstein-Barr nuclear antigen or the herpesvirus latent nuclear antigen. The present invention also provides a method to identify an agent that modulates segregation of extrachromosomal molecules into daughter cells following division of a parent cell. Such methods can be used to identify agents that are useful for treating cancer, viral infections or other afflictions that involve extrachromosomal molecules. The present invention also provides a method of treating cancer or viral infections by administering a pharmaceutical composition containing a compound that inhibits the tethering of viral or cellular acentric extrachromosomal molecules to a chromosome. The present invention further comprises a chromosomally integrating vector that specifically labels DMs. The vector may be an Epstein-Barr virus (EBV), bovine papillomavirus (BPV), or Kaposi's sarcoma associated herpesvirus (KSHV) vector sequences. The vector may contain an EBNA-1 gene and an oriP sequence, wherein the oriP sequence has a plurality of EBNA-1 binding sites in two distinct regions. Further, the vector may contain a plurality of tandem repeats of a lac operator (lacO). The vector may contain a reporter gene, such as GFP or YFP, and/or may contain a selection marker, such as a blasticidin resistance gene (bsr) driven by SR promoter. The vector may further contain a nucleotide sequence that interferes with chromosomal tethering. The vector may contain a lac repressor (lacR)-GFP fusion protein that binds with high affinity to the lacO. The vector may be a modified virus, such as a modified animal virus. It may be a DNA virus, such as a member of the Herpesviridae, Papovaviridae or Adenoviridae. The present invention provides a plasmid vector comprising retroviral vector, a gene encoding GFP fused to lac repressor-nuclear localization signal. It also provides a plasmid vector comprising retroviral vector, a gene encoding YFP fused to lac repressor-nuclear localization signal. Moreover, the present invention provides a plasmid vector comprising a histone H2B gene and a CFP gene.
Process for cracking an olefin-rich hydrocarbon feedstock
A process for cracking an olefin containing hydrocarbon feedstock which is selective towards light olefins in the effluent, the process comprising passing a hydrocarbon feedstock containing one or more olefins through a moving bed reactor containing a crystalline silicate catalyst selected from an MFI-type crystalline silicate having a silicontalurniniumn atomic ratio of a least 180 and an MEL-type crystalline silicate having a silicon/aluminium atomic ration of from 150 to 800 which has been subjected to a steaming step, at an inlet temperature of from 500 to 600° C., at an olefin partial pressure of from 0.1 to 2 bars and the feedstock being passed over the catalyst at an LHSV of from 5 to 30 h−1 to produce an effluent with an olefin content of lower molecular weight than that of the feedstock, intermittently removing a first fraction of the catalyst from the moving bed reactor, regenerating the first fraction of the catalyst in a regenerator and intermittently feeding into the moving bed reactor a second fraction of the catalyst which has been regenerated in the regenerator, the catalyst regeneration rate being controlled whereby the propylene purity is maintained constant at a value corresponding to the average value observed in a fixed bed reactor using the same feedstock:, catalyst and cracking conditions, for example at least 94 wt %.
1. A process for cracking an olefin-containing hydrocarbon feedstock which is selective towards light olefins in the effluent, the process comprising passing a hydrocarbon feedstock containing one or more olefins through a moving bed reactor containing a crystalline silicate catalyst selected from an MFI-type crystalline silicate having a silicon/aluminum atomic ratio of at least 180 and an MEL-type crystalline silicate having a silicon/aluminum atomic ratio from 150 to 800 which has been subjected to steaming step, at an inlet temperature of from 500 to 600° C., at an olefin partial pressure of from 0.1 to 2 bars and the feedstock being passed over the catalyst at an LHSV of from 5 to 30 h−1 to produce an effluent containing propylene and with an olefin content of lower molecular weight than the olefin content of the feedstock with concomittant deactivation of said catalyst, removing a first fraction of the deactivated catalyst from the moving bed reactor and transferring said deactivated catalyst to a regenerator, regenerating said deactivated catalyst in said regenerator to produce a second fraction of regenerated catalyst and recycling said regenerated catalyst to the moving bed reactor, continuing the transfer of deactivated catalyst and the recycle of regenerated catalyst while carrying out the cracking of the olefin-containing hydrocarbon feedstock, the catalyst regeneration and recycle rate being controlled to maintain the propylene purity at a relatively constant value corresponding to the average value observed in a fixed bed reactor using the same feedstock, catalyst and cracking conditions. 2. The process of claim 1 wherein the catalyst regeneration and recycle rate is controlled to provide an ethylene yield in the effluent on an olefin basis which is less than 10 wt %. 3. The process of claim 1, wherein the effluent has a propylene purity of at least 94 wt % propylene based upon the total C3 content of the effluent. 4. The process of claim 1, wherein the olefin content of the effluent is within ±15 wt % of the olefin content of the feed stock. 5. The process of claim 1, wherein said first fraction of the catalyst is intermittently removed from said moving bed reactor. 6. The process of claim 5, wherein said second fraction of the regenerated catalyst is intermittently supplied from said regenerator to said moving bed reactor. 7. The process of claim 1, wherein said catalyst is regenerated in said regenerator by supplying an oxidizing gas containing oxygen in amount within the range of 0.2 to 2 volume percent of said oxidizing gas. 8. The method of claim 1, wherein the regeneration of catalyst in said regenerator involves a supply of an initial oxygen-containing gas to the regenerator and a supply of a second oxygen-containing gas to the regenerator at a point downstream of the introduction of said initial oxygen-containing gas, said second oxygen-containing gas having a higher oxygen content than said initial oxygen-containing gas. 9. The process of claim 8, wherein said second oxygen-containing gas contains from 5 to 21 volume percent oxygen. 10. The process of claim 1, wherein said moving bed reactor comprises a first stage reactor and a second stage reactor connected in series with said first stage reactor, wherein the effluent from the first stage reactor is heated and then supplied to the inlet of said second stage reactor. 11. The process of claim 10, wherein the contact time of the reaction mixture with the catalyst in the second reactor is greater than the contact time of the reaction mixture with the catalyst in the first stage reactor. 12. A process for cracking an olefin-containing hydrocarbon feedstock which is selective towards light olefins in the effluent, the process comprising passing a hydrocarbon feedstock containing one or more olefins through a moving bed reactor containing a crystalline silicate catalyst selected from an MFI-type crystalline silicate having a silicon/aluminum atomic ratio of at least 180 and an MEL-type crystalline silicate having a silicon/aluminum atomic ratio from 150 to 800 which has been subjected to steaming step, at an inlet temperature of from 500 to 600° C., at an olefin partial pressure of from 0.1 to 2 bars and the feedstock being passed over the catalyst at an LHSV of from 5 to 30 h−1 to produce an effluent with an olefin content of lower molecular weight than that of the feedstock with concomittant deactivation of said catalyst, removing a first fraction of the deactivated catalyst from the moving bed reactor and transferring said deactivated catalyst to a regenerator, regenerating said deactivated catalyst in said regenerator to produce a second fraction of regenerated catalyst and recycling said regenerated catalyst to the moving bed reactor, continuing the transfer of deactivated catalyst and the recycle of regenerated catalyst while carrying out the cracking of the olefin-containing hydrocarbon feedstock, the catalyst regeneration and recycle rate being controlled to provide that all of the catalyst in the moving bed reactor is regenerated in a period of from 20 to 240 hours. 13. The process of claim 12 wherein the propylene purity is maintained at a relative constant value corresponding to the average value obtained in a fixed bed reactor using the same feedstock, catalyst and cracking conditions. 14. The process of claim 12 where the regeneration and recycle rate is controlled to proceed on ethylene yield on an olefin basis which is less than 10 wt %. 15. The process of claim 12, wherein the propylene yield of said process is within the range of 30 to 50 wt % propylene with a selectivity to propylene of at least 92 wt % of the total amount of propylene and propane in the effluent. 16. The process of claim 15, wherein the olefin content of the effluent is within the range of ±10 wt % of the olefin content of the feed stock. 17. The method of claim 12, wherein the regeneration of catalyst in said regenerator involves a supply of an initial oxygen-containing gas to the regenerator and a supply of a second oxygen-containing gas to the regenerator at a point downstream of the introduction of said initial oxygen-containing gas, said second oxygen-containing gas having a higher oxygen content than said initial oxygen-containing gas. 18. The process of claim 17, wherein said second oxygen-containing gas contains from 5 to 21 volume percent oxygen. 19. The process of claim 12, wherein said moving bed reactor comprises a first stage reactor and a second stage reactor connected in series with said first stage reactor, wherein the effluent from the first stage reactor is heated and then supplied to the inlet of said second stage reactor. 20. The process of claim 17, wherein the contact time of the reaction mixture with the catalyst in the second stage reactor is greater than the contact time of the reaction mixture with the catalyst in the first stage reactor. 21. A process for cracking an olefin-containing hydrocarbon feedstock which is selective towards light olefins in the effluent, the process comprising passing a hydrocarbon feedstock containing one or more olefins through a moving bed reactor containing a crystalline silicate catalyst to produce an effluent with an olefin content of lower molecular weight than that of the feedstock with concomittant deactivation of said catalyst, removing a deactivated catalyst from the moving bed reactor and regenerating said deactivated catalyst to produce a regenerated catalyst which is recycled to said moving bed reactor, continuing the removal of deactivated catalyst and the recycle of regenerated catalyst while carrying out the cracking of the olefin-containing hydrocarbon feedstock, the catalyst regeneration and recycle rate being controlled to provide a propylene content in the effluent that is maintained relatively constant at a value corresponding to the initial value observed in the effluent from a fixed bed reactor during an initial period of an olefin cracking process using the same feedstock, catalyst and cracking conditions. 22. The process of claim 21, wherein the initial period of the fixed bed reaction is the first 10 to 40 hours of said reaction. 23. The process according to claim 21, wherein said moving bed reactor comprises a first stage reactor and a second stage reactor connected in series with said first stage reactor, wherein the effluent from the first stage reactor is heated and then supplied to the inlet of said second stage reactor. 24. The process of claim 23, wherein the contact time of the reaction mixture with the catalyst in the second reactor is greater than the contact time of the reaction mixture with the catalyst in the first stage reactor. 25. The process of claim 21, wherein fresh makeup catalyst is supplied to said moving bed reactor along with said regenerated catalyst recycled to said moving bed reactor.
Personal message delivery system
The present invention provides for a personal message system comprised of a plurality of interfaces configured to interface with a plurality of subscribers communication devices using a plurality of formats. A group services module is provided configured to maintain communications among groups of the subscribers. A platform conversion module is also provided and is coupled to the plurality of interfaces and the group services modules configured to connect each of the plurality of subscribers within a group, regardless of the communication protocols used by the subscribers.
1. A personal message system comprising: a plurality of interfaces configured to interface with a plurality of subscribers communication devices using a plurality of formats; a group services module configured to maintain communications among groups of said subscribers; and a platform conversion module coupled to said plurality of interfaces and said group services modules configured to connect each of Said plurality of subscribers within a group, regardless of the communication protocols used by said subscribers. 2. A personal message system as claimed in claim 1, wherein one of said plurality of interfaces is an Hypertext Transfer Protocol/Wireless Application Protocol interface. 3. A personal message system as claimed in claim 2, wherein one of said plurality of interfaces is a Simple Mail Transfer Protocol interface. 4. A personal message system as claimed in claim 3, wherein one of said plurality of interfaces is a Short Message Service/2-way paging protocol interface. 5. A personal message system as claimed in claim 4, wherein one of said plurality of interfaces is an Interactive Voice Response protocol interface. 6. A personal message system as claimed in claim 1, further comprising a personal home page service module configured to provide personal mobile home pages for subscribers on said messaging system. 7. A personal message system as claimed in claim 1, wherein said group service module further comprises an alert module configured to provide alerts to said plurality of subscribers so as to alert them to incoming messages. 8. A personal message system as claimed in claim 1, wherein said group service module further comprises a send message module such that said plurality of subscribers can formulate messages to be sent through said messaging system to other said subscribers. 9. A personal message system as claimed in claim 1, wherein said group service module further comprises a group module configured to allow said subscribers to create, and send messages to and receive messages from subscribers of said groups. 10. A personal message system as claimed in claim 9, further comprising a group main page utility configured to display of a list of said groups. 11. A personal message deliver system as claimed in claim 9, further comprising a group details utility configured to list the details of said groups including number of members, number of messages per day. 12. A personal message system as claimed in claim 9, further comprising group administration utility configured to provide administration function for said group including allowing in new subscribers, removing existing subscribers and setting parameters for group actions. 13. A personal message system as claimed in claim 9, further comprising a group history page configured to store the history of messages sent by said subscribers to said group. 14. A personal message system as claimed in claim 10, further comprising a group search results utility configured to provide search results for searches conducted from said group main page utility. 15. A personal message system as claimed in claim 9, further comprising a create group utility configured to allow said subscribers to create a new group for said messaging system. 16. A personal message system as claimed in claim 1, wherein said group service module further comprising a group membership module configured to allow said subscribers with the necessary utilities-to facilitate the membership functions of said groups. 17. A personal message system as claimed in claim 16, further comprising a request to join utility configured to allow a subscriber to request to join one of said groups. 18. A personal message system as claimed in claim 16, further comprising an invite to group utility configured to allow a subscriber of one of said groups to invite another non-member subscriber to said group. 19. A personal message system as claimed in claim 16, further comprising an accept invite utility configured to allow a subscriber who receives an invite to one of said groups to accept or reject the invitation. 20. A personal message system as claimed in claim 16, further comprising a member directory utility configured to provide a list of the member subscribers to said groups. 21. A personal message system as claimed in claim 1, further comprising a user database configured to store information relating to the accounts of said plurality of subscribers. 22. A personal message system as claimed in claim 1, further comprising a multimedia database configured to store information relating to multimedia data for delivery to said plurality of subscribers. 23. A personal message system comprising: a plurality of interfaces configured to interface with a plurality of subscribers communication devices using a plurality of formats; a channel services module configured to maintain communications from a content provider to said subscribers; and a platform conversion module coupled to said plurality of interfaces and said channel services modules configured to connect each of said plurality of subscribers with said content providers, regardless of the communication protocols used by said subscribers. 24. A personal message system as claimed in claim 23, wherein one of said plurality of interfaces is an Hypertext Transfer Protocol/Wireless Application Protocol interface. 25. A personal message system as claimed in claim 24, wherein one of said plurality of interfaces is a Simple Mail Transfer Protocol interface. 26. A personal message system as claimed in claim 25, wherein one of said plurality of interfaces is a Short Message Service/2-way paging protocol interface. 27. A personal message system as claimed in claim 26, wherein one of said plurality of interfaces is an Interactive Voice Response protocol interface. 28. A personal message system as claimed in claim 23, further comprising a personal home page service module configured to provide personal mobile home pages for subscribers on said messaging system. 29. A personal message system as claimed in claim 23, wherein said channel service module further comprises an alert module configured to provide alerts to said plurality of subscribers so as to alert them to incoming messages. 30. A personal message system as claimed in claim 23, wherein said channel service module further comprises a send message module such that said content provider can formulate messages to be sent through said messaging system to said subscribers. 31. A personal message system as claimed in claim 23, wherein said channel service module further comprises a poll module configured to provide said content provider with the ability to send polls to said channel subscribers. 32. A personal message system as claimed in claim 23, wherein said channel service module further comprises a channel module configured to allow said content providers create and send messages to said subscribers of said channel. 33. A personal message system as claimed in claim 32, further comprising a channel main page utility configured to display of a list of said channels. 34. A personal message system as claimed in claim 32, further comprising a channel details utility configured to list the details of said channels including the number of messages per day and a description of the content of those channels. 35. A personal message system as claimed in claim 32, further comprising a channel categories utility configured to list said channels into categories based on their content. 36. A personal message system as claimed in claim 32, further comprising a channel history utility configured to store the history of messages sent by said content provider to said channel. 37. A personal message system as claimed in claim 32, further comprising a channel configuration utility configured to allow said channel subscribers to control the content that they receive from said channel. 38. A personal message system as claimed in claim 23, wherein said channel service module further comprising a channel administration module configured to allow said content providers administrate said channels. 39. A personal message system as claimed in claim 38, further comprising a channel text message utility configured to allow said content provider to generate a text message to be sent to said member subscribers of said channel. 40. A personal message system as claimed in claim 38, further comprising a channel voice message utility configured to allow said content provider to generate a voice message to be sent to said member subscribers of said channel. 41. A personal message system as claimed in claim 38, further comprising a channel toolset utility configured to allow said content provider to administrate the content of said channel. 42. A personal message system as claimed in claim 38, further comprising a channel message scheduling utility configured to allow a content provider to pre-schedule the deliver of messages to said member subscribers of said channel in advance. 43. A personal message system as claimed in claim 38, further comprising a channel poll utility configured to allow said channel provider to enter the information necessary to conduct a poll. 44. A personal message system as claimed in claim 23, further comprising a user database configured to store information relating to the accounts of said plurality of subscribers. 44. A personal message system as claimed in claim 23, further comprising a multimedia database configured to store information relating to multimedia data of said content providers for delivery to said plurality of subscribers. 45 A personal message system comprising: a plurality of interfaces configured to interface with a plurality of subscribers communication devices using a plurality of formats; a group services module configured to maintain communications among groups of said subscribers; a platform conversion module coupled to said plurality of interfaces and said group services modules configured to connect each of said plurality of subscribers within a group, regardless of the communication protocols used by said subscribers; and a message delivery subsystem coupled to one of said plurality of interfaces configured to create a virtual number based on a the devices of both said receiving and sending said subscriber and a randomly generated virtual number which are stored in a call completion data table. 46. A personal message system as claimed in claim 45, wherein said call completion data table maintains a receiving subscriber device field configured to store the wireless number associated with the device of the call receiving subscriber. 47. A personal message system as claimed in claim 45, wherein said call completion data table maintains a virtual number field configured to store the virtual number associated with the call between said devices of the call receiving and call sender subscribers. 48. A personal message system as claimed in claim 45, wherein said call completion data table maintains a sending subscriber device field configured to store the wireless number associated with the device of the call sending subscriber 49. A personal message system comprising: a plurality of interfaces configured to interface with a plurality of subscribers communication devices using a plurality of formats; a group services module configured to maintain communications among groups of said subscribers; a platform conversion module coupled to said plurality of interfaces and said group services modules configured to connect each of said plurality of subscribers within a group, regardless of the communication protocols used by said subscribers; and a wireless application protocol—short message service/interactive voice response handler module coupled to said platform conversion module configured to provide interactive voice response links into a wireless application protocol sessions such that a subscriber in said wireless application protocol sessions can directly link to an interactive voice response session so as to directly download a multimedia file. 50. A personal message system as claimed in claim 49, further comprises an interactive voice response subsystem handler module, coupled to said wireless application protocol—short message service/interactive voice response handler module configured to provide an external interactive voice response session from an independent multimedia content provider. 51. A personal message system as claimed in claim 50, further comprises an interactive voice response multimedia database, coupled to said interactive voice response subsystem handler module configured to store multimedia content to be delivered by said interactive voice response subsystem handler module and said wireless application protocol—short message service/interactive voice response handler module. 52. A personal message system comprising: a plurality of interfaces configured to interface with a plurality of subscribers communication devices using a plurality of formats; a group services module configured to maintain communications among groups of said subscribers; a platform conversion module coupled to said plurality of interfaces and said group services modules configured to connect each of said plurality of subscribers within a group, regardless of the communication protocols used by said subscribers; and a subscriber information table configured to store the information necessary for said platform conversion module to send messages to said subscribers in the proper format. 53. A personal message system as claimed in claim 52, further comprising a username field configured to store information pertaining to said subscribers username, used to identify said subscriber to said other subscribers on said message system. 54. A personal message system as claimed in claim 52, further comprising a personal information/e-mail field configured to store information pertaining to said subscribers personal information such as real name, mailing address, phone number and e-mail address for use by said message system in contacting said subscriber. 55. A personal message system as claimed in claim 52, further comprising a Personal Identification Number (PIN) field configured to store information pertaining to said subscribers personal identification number used by said message system to restrict access to said subscribers account. 56. A personal message system as claimed in claim 52, further comprising a validation list field configured to store information pertaining to said subscribers personal communication devices which are recognized by said message system. 57. A personal message system as claimed in claim 52, further comprising a preferred voice field configured to store information pertaining to said subscribers preferred device for receiving voice messages from said message system. 58. A personal message system as claimed in claim 52, further comprising a preferred text field configured to store information pertaining to said subscribers preferred device for receiving text messages from said message system. 59. A personal message system as claimed in claim 52, further comprising a groups field configured to store information pertaining to said subscribers group membership record. 60. A personal message system as claimed in claim 52, further comprising a channel field configured to store information pertaining to said subscribers channel membership record. 61. A personal message system as claimed in claim 52, flier comprising a addresses field configured to store information pertaining to said subscribers address book of all other subscribers on said message system that said subscriber has previously contacted. 62. A personal message system as claimed in claim 52, further comprising a history field configured to store information pertaining to said subscribers recently received messages from said message system 63. A method for sending a message on a personal messaging system having a plurality of interfaces, a group services module and a platform conversion module, said method comprising the steps of: sending a message to said message system via any one of said plurality of interfaces; sending said message to said group services module for sending said message to said subscriber members of said group; reformatting said message in said platform conversion module into various formats corresponding to said group member subscriber preferences; and delivering said message through said plurality of interfaces to said members of said group. 64. A method for delivering a message on a personal messaging system having a plurality of interfaces, a channel services module and a platform conversion module, said method comprising the steps of: creating a channel message on said message system; sending said message to said channel services module for sending said message to said subscribers members of said channel; reformatting said message in said platform conversion module into various formats corresponding to said channel member subscriber preferences; and delivering said message through said plurality of interfaces to said members of said channel.
<SOH> BACKGROUND OF THE INVENTION <EOH>Many systems are currently in place which interconnect people via electronic messaging services. These services include phones (mobile and fixed-line), pager services and text paging (SMS), wireless palm communicators, wireless internet and e-mail, all of which are designed -to provide mobile communication services to users. However, each particular system both wireless and standard (e-mail via PC and fixed-line phone) require different protocols to deliver the messages. The drawback to these systems is that they are not well integrated As such, communications within a group are limited to the subscribers of a particular service provider. Subscribers of various service providers do not have the ability to interact For example, there is currently no ability if for people desire to form a group where, each communicates via different modes of operation; such as, one by cell phone, one by e-mail, one by paging, and one by paging with SMS. Therefore, there exists a need for developing an enhanced personal message delivery system that allows people with various communication devices supported by various service providers to communicate in groups of their own design.
<SOH> SUMMARY OF THE INVENTION <EOH>In accordance with one embodiment of the present invention, a system is provided that fully integrates all personal communication forms including but not limited to mobile and fixed-line voice, SMS (mobile-terminated and mobile-originated), text paging (SMS), standard paging, web, e-mail, WAP & (Phone).com and instant messaging, such that a single message intended to reach a group of users is entered only once in any available format whereby it is automatically converted into all of the other necessary formats and delivered to all members of a group. Furthermore, the system comprises integrated components and navigation architecture to allow smooth cross platform transitions during system sessions.
Intravascular ultrasonic catheter arrangements
Apparatus for intravascular ultrasonic imaging comprises a catheter having an ultrasonic transducer array fabricated at least in part from an electrostrictive material, wherein the electrostrictive material is non-polymeric.
1. An apparatus for intravascular ultrasonic imaging, the apparatus comprising: a catheter having an ultrasonic transducer array fabricated at least in part from non-polymeric electrostrictive material; and a high permitivity ceramic member or layer that couples a common radiofrequency source to elements of the array. 2. The apparatus of claim 1 further comprising controls for energizing the array and processing signals derived therefrom, the controls being adapted to apply a bias voltage to elements of the array to render those elements substantially piezoelectric. 3. The apparatus of claim 1 wherein the electrostrictive material is a relaxor ferroelectric material. 4. The apparatus of claim 3, wherein the electrostrictive material comprises lead magnesium niobate. 5. The apparatus of claim 2, wherein the controls are adapted to transmit and receive signals only while the bias voltage is applied. 6. The apparatus of claim 1 further comprising a high permitivity ceramic member or layer that couples a common radiofrequency source to the elements of the array. 7. The apparatus of claim 1 further comprising a multiplexer arrangement through which the transducer array is energized and through which echo signals received by the transducer array are passed to the proximal end of the catheter, when in use. 8. The apparatus of claim 7 wherein the multiplexer arrangement comprises a plurality of integrated circuits. 9. The apparatus as claimed in claim 8 wherein the integrated circuits are arranged in substantially cylindrical configuration. 10. The apparatus of claim 8 wherein adjacent integrated circuits are spaced from one another by a slot. 11. The apparatus of claim 8, wherein the integrated circuits are flip-chip bonded to an electrical circuit. 12. The apparatus of claim 11 wherein the electrical circuit is a printed circuit. 13. A method of manufacturing a catheter for intravascular ultrasonic imaging having an ultrasonic transducer array fabricated at least in part from a non-polymeric, electrostrictive material, the method comprising: fabricating the transducer in-the-flat; the transducer array comprising a high permitivity ceramic member or layer that couples a common radiofrequency source to elements of the array; and reconfiguring the array into a substantially cylindrical configuration. 14. The method of claim 13 further comprising electrically coupling controls to the array for energizing the array and processing signals derived therefrom, the controls being adapted to apply a bias voltage to elements of the array to render those elements substantially piezoelectric. 15. The method of claim 13, wherein fabricating the transducer array in-the-flat further comprises: providing a substrate; forming electrically conductive tracks on the substrate; bonding a piezoelectric block to the conductive tracks; and forming slots in the piezoelectric block to form a plurality of discrete transducer elements. 16. The method of claim 15, further comprising coupling at least one multiplexer circuit to the conductive tracks. 17. The method of claim 14, wherein the electrostrictive material comprises a relaxor ferroelectric material. 18. The method of claims 16, wherein the relaxor ferroelectric material comprises lead magnesium niobate. 19. The method of claim 15, further comprising forming slot bottoms in the substrate adjacent the electrically conductive tracks prior to bonding the piezoelectric block to the conductive tracks. 20. The method of claim 19, wherein forming slot bottoms comprises forming slot bottoms with a laser. 21. The method of claim 19, further comprising performing a photoresist technique within the slot bottoms.
Vehicle signaling device
A signalling aapparatus for a motor vehicle is provided with communication means for receiving information coming from another motor vehicle and for broadcasting information intended for a nearby motor vehicle, and with sensor means for providing inforation about the status or status change of thr respective motor vehicle with regard to at least the safety of this vehicle. A computer is present for determining, on the ground of the information coming from the sensor means and/or on the ground of the received information, information to be broadcast and further action signals. In addition, means are present to enable, on the ground of the action signals, realization of a status change of the respective motor vehicle, which means are formed by a signalling element for making observable a signalling knowable for the driver of the vehicle or of a nearby vehicle.
1. A signalling apparatus for a motor vehicle, provided with communication means for receiving information coming from another motor vehicle and for broadcasting information with regard to at least the safety of the first-mentioned motor vehicle to a further nearby vehicle, characterized in that sensor means are present for providing information about the status or a status change of the respective motor vehicle with regard to at least the safety of this vehicle, as well as a computer which is arranged for generating action signals and information to be broadcast, on the ground of a combination of the information provided by the sensor means and information received from a nearby vehicle, and -means to enable, on the ground of these action signals, initiation of a status change of the respective vehicle or of a nearby vehicle, which means comprise a signalling element for making observable a signalling knowable for the driver of the respective vehicle or the nearby vehicle. 2. A signalling apparatus according to claim 1, characterized in that the computer is arranged to generate combined information for the information to be broadcast by weakened transfer of an extent of exceptionality of the information received from the nearby vehicle and/or enhancement of the extent of exceptionality of the combined information depending on the information provided by the sensor means. 3. A signalling apparatus according to claim 1, characterized in that the communication means are arranged on the front side and rear side of the vehicle. 4. A signalling apparatus according to claim 1, characterized in that the communication means comprise at least one infrared transmitter and infrared receiver. 5. A signalling apparatus according to claim 1, characterized in that the communication means are formed by connecting means which enable a line-of-sight connection between two vehicles driving within view of each other, as well as radio connecting means, wherein, when according to the line-of-sight a connection between two vehicles driving within view of each other has been established, a data transmission by the broadcasting vehicle to the receiving vehicle is realized with the aid of the radio connection. 6. A signalling apparatus according to claim 1, characterized in that the information to be broadcast is formed by a single safety status signal. 7. A signalling apparatus according to claim 6, characterized in that in the vehicle a display is present for displaying, on the ground of a received safety status signal, a warning signal for the driver of this vehicle. 8. A signalling apparatus according to claim 6, characterized in that at the back of the vehicle, a display is present for displaying, on the ground of a received signal and on the ground of signals produced by the sensor means, a warning signal for the driver of a following vehicle. 9. A signalling apparatus according to claim 6, characterized in that the vehicle is provided with brake lights for displaying, on the ground of a received safety status signal, a warning signal for the driver of a following vehicle. 10. A signalling apparatus according to claim 7, characterized in that the warning signal is in the form of a warning triangle which lights up to an increasing extent as to its dimensions according as the safety status signal represents a greater danger. 11. A signalling apparatus according to claim 6, characterized in that the means for enabling a status change of the vehicle or of a nearby vehicle are formed by brake energization means controlled by the safety status signal. 12. A signalling apparatus according to claim 1, characterized in that the sensor means comprise means indicating the position of the brake pedal. 13. A signalling apparatus according to claim 1, characterized in that the sensor means comprise means determining the speed and/or the acceleration of the vehicle. 14. A signalling apparatus according to claim 1, characterized in that the sensor means comprise a global positioning system, which functions as sensor for measuring at least the position of the respective vehicle.
Storage medium convey apparatus and convey method and recording and/or reproduction apparatus
Apparatus that carries or transfers a memory medium having a solid-state memory over the inside and outside portions of a recording and/or reproducing apparatus is adapted so that, by selective engagement with respect to a chassis or a holder of a fluctuation lever the memory medium is inserted and held. The memory medium is moved in a manner bridging between an eject position where insertion/withdrawal of the memory medium is permitted and a loading position. A first biasing member moves and biases the holder in an eject direction opposite to a memory medium insertion direction is caused to store biasing force in the eject direction. A second biasing member biases the holder in the memory medium insertion direction and is caused to be operative to carry out switching between movement in the memory medium insertion direction of the holder and movement in the eject direction.
1. A carrying apparatus for a memory medium, comprising: a holder adapted to have the memory medium inserted therein including a connection terminal connected to a contact provided at a front end side of the inserted memory medium; a lock mechanism provided on the holder and locked with respect to a chassis of the holder so that the holder is movably supported, the lock mechanism serving to limit movement with respect to the chassis of the holder, so that when the contact of the memory medium inserted into the holder is connected to the connection terminal of the holder, the lock mechanism is pressed by the memory medium to thereby release a locking state with respect to the chassis; a fluctuation lever engaged with the holder and moved when the lock mechanism is released, the fluctuation lever moved in an insertion direction of the memory medium as one with the holder; first biasing means for biasing the fluctuation lever in an eject direction opposite to the insertion direction of the memory medium; and second biasing means for biasing the holder in the insertion direction of the memory medium, wherein the fluctuation lever is moved in the insertion direction of the memory medium as one with the holder to thereby allow the first biasing means to store a biasing force that moves the fluctuation lever in the eject direction, and engagement with respect to the holder is released by movement in the insertion direction of the memory medium of the fluctuation lever and only the holder into which the memory medium is inserted is drawn in the insertion direction of the memory medium by the second biasing means in accordance with engagement with respect to the chassis. 2. The carrying apparatus for a memory medium as set forth in claim 1, wherein the fluctuation lever is rotationally biased by the first biasing means in a direction where engagement with respect to the holder is released. 3. The carrying apparatus for memory medium as set forth in claim 2, wherein when the fluctuation lever is moved in the insertion direction of the memory medium as one with the holder, release of engagement with respect to the holder is limited by rotation limiting means provided at a side of the chassis that movably supports the holder. 4. The carrying apparatus for a memory medium as set forth in claim 1, wherein the lock mechanism includes a pressing operation portion caused to undergo a pressing operation by a lock piece engaged with the chassis and the memory medium inserted into the holder and a lock lever rotatably supported by the holder and rotationally biased by a biasing member in a direction to engage the lock piece with the chassis. 5. The carrying apparatus for a memory medium as set forth in claim 1, wherein a cover body rotatably attached to the chassis opens and closes a memory medium insertion/withdrawal opening adapted so that the memory medium to be inserted into the holder is inserted thereinto or is withdrawn therefrom, the cover body being rotationally biased in a direction to close the memory medium insertion/withdrawal opening by a biasing member at all times. 6. The carrying apparatus for a memory medium as set forth in claim 1, wherein the apparatus further comprises a slider movably supported by the chassis in the insertion direction with respect to the holder of the memory medium, and adapted so that the fluctuation lever is rotatably supported, wherein the first biasing means includes a first biasing member provided in a manner bridging between the slider and the chassis and biases the holder in the eject direction opposite to the insertion direction of the memory medium, wherein the second biasing means includes a second biasing member provided in a manner bridging between the slider and the holder and biases the holder in the insertion direction of the memory medium, and wherein the fluctuation lever is adapted so that the locking state with respect to the chassis by the lock mechanism is released and is moved in the insertion direction of the memory medium in one body with the holder moved in the insertion direction of the memory medium to allow the first biasing member to store a biasing force that moves the slider in the eject direction, whereby when a fluctuation lever lock mechanism provided at the fluctuation lever is moved to a position opposite to a cut portion provided at the chassis side, the fluctuation lever lock mechanism is inserted into the cut portion and engagement between the engagement portion and the holder is released by a rotational force produced as a result of a force in the insertion direction of the memory medium by the holder and a biasing force in the eject direction by the first biasing member with respect to the fluctuation lever act with respect to an engagement portion of the fluctuation lever formed at a position engaged with the holder, and the fluctuation lever and the slider are locked with respect to the chassis and only the holder into which the memory medium is inserted is drawn in the insertion direction of the memory medium by the biasing force of the second biasing member. 7. The carrying apparatus for a memory medium as set forth in claim 1, wherein the apparatus further comprises eject operation means for causing the holder moved in the insertion direction of the memory medium by the second biasing means to undergo movement operation in the eject direction against a biasing force of the second biasing means, and wherein when only the holder into which the memory medium is inserted is drawn in the insertion direction of the memory medium by the second biasing means so that it is held at a loading position of the memory medium, the holder is moved in the eject direction against the biasing force of the second biasing means through the eject operation means to thereby release engagement with respect to the chassis of the fluctuation lever to receive the biasing force of the first biasing means to move the holder in the eject direction as one with the fluctuation lever to move the holder to an initial position. 8. The carrying apparatus for a memory medium as set forth in claim 7, wherein the eject operation means comprises an eject operation piece moved as one with the holder. 9. The carrying apparatus for a memory medium as set forth in claim 7, wherein the apparatus further comprises a slider movably supported by the chassis in the insertion direction with respect to the holder of the memory medium and adapted so that the fluctuation lever is rotatably supported, wherein the first biasing means includes a first biasing member provided in a manner bridging between the slider and the chassis for biasing the slider in the eject direction opposite to the insertion direction of the memory medium, wherein the second biasing means includes a second biasing member provided in a manner bridging between the slider and the holder for biasing the holder in the insertion direction of the memory medium, wherein the fluctuation lever is adapted so that the locking state with respect to the chassis by the lock mechanism is released and the fluctuation lever is moved in the insertion direction of the memory medium as one with the holder moved in the insertion direction of the memory medium to thereby allow the first biasing member to store the biasing force that moves the slider in the eject direction, and when a fluctuation lever lock mechanism provided at the fluctuation lever is moved to a position opposite to a cut portion provided at the chassis side, the fluctuation lever lock mechanism is inserted into the cut portion and engagement between the engagement portion and the holder is released by a rotational force produced as a result of a force in the insertion direction of the memory medium by the holder and the biasing force in the eject direction by the first biasing member with respect to the fluctuation lever act with respect to engagement portion of the fluctuation lever formed ay a position engaged with the holder, and the fluctuation lever and the slider are locked with respect to the chassis and only the holder into which the memory medium is inserted is drawn in the insertion direction of the memory medium by a biasing force of the second biasing means, and wherein the holder is moved in the eject direction against the biasing force of the second biasing member through the eject operation means, and the fluctuation lever is rotated by the biasing force by the first biasing member to release the locking state with respect to the chassis by the fluctuation lever lock mechanism to receive the biasing force of the first biasing member to move the holder in the eject direction as one with the fluctuation lever to move the holder to an initial position. 10. A recording and/or reproducing apparatus, comprising: a holder adapted to have a memory medium inserted therein including a connection terminal connected to a contact provided at a front end side of the inserted memory medium; recording and/or reproducing means for carrying out recording and/or reproduction of data with respect to the memory medium having the contact connected to the connection terminal of the holder; a lock mechanism provided on the holder and locked by a chassis of the holder adapted so that the holder is movably supported, the lock mechanism serving to limit movement with respect to the chassis of the holder, so that when the contact of the memory medium inserted into the holder is connected to the connection terminal of the holder, the lock mechanism is pressed by the memory medium to thereby release a locking state with respect to the chassis; a fluctuation lever engaged with the holder and moved when the lock mechanism is released in an insertion direction of the memory medium as one with the holder; first biasing means for biasing the fluctuation lever in an eject direction opposite to the insertion direction of the memory medium; and second biasing means for biasing the holder in the insertion direction of the memory medium, wherein the fluctuation lever is moved in the insertion direction of the memory medium as one with the holder to thereby allow the first a biasing means to store biasing force that moves the fluctuation lever in the eject direction, and engagement with respect to the holder is released by movement in the insertion direction of the memory medium of the fluctuation lever and only the holder into which the memory medium is inserted is drawn in the insertion direction of the memory medium by the second biasing means in accordance with engagement with respect to the chassis. 11. The recording and/or reproducing apparatus as set forth in claim 10, wherein the recording and/or reproducing means carries out recording and/or reproduction of data with respect to the memory medium when the contact is connected to the connection terminal of the holder at a reproduction position where the memory medium is located as a result of only the holder into which the memory medium is inserted is drawn in the insertion direction of the memory medium by the second biasing means. 12. A carrying method for a memory medium comprising: a step of inserting a memory medium into a holder; a step of electrically connecting a contact of the memory medium and a connection terminal within the holder; a step of pressing lock means that locks the holder with respect to a chassis of the holder by the memory means after the contact of the memory medium and the connection terminal within the holder have been electrically connected to thereby release a locking state by the lock means; a step of further moving the holder in an insertion direction along with the memory medium; a step of engaging a lever moved in the insertion direction as one with the holder as a result of the lever being engaged with the holder, and of releasing engagement with respect to the holder; a step of storing a biasing force in an eject direction opposite to the insertion direction at first biasing means for a time period during which the lever is moved in the insertion direction where engagement with the holder is released; and a step of drawing only the holder in which engagement by the lever has been released in the insertion direction along with the memory medium by second biasing means. 13. The carrying method for memory medium as set forth in claim 12, wherein the method further comprises: a step of drawing only the holder in which engagement by the lever has been released in the insertion direction along with the memory medium by the second biasing means to hold the holder at a loading position; and a step of moving the holder held at the loading position in the eject direction against a biasing force of the second biasing means to thereby release engagement with respect to the chassis of the fluctuation lever to receive a biasing force of the first biasing means to move the holder in the eject direction as one with the fluctuation lever to move the holder to an initial position.
<SOH> BACKGROUND ART <EOH>Hitherto, memory medium using a solid-state memory as a memory element is used as a memory medium for information signal. Since the memory medium using the solid-state memory can be formed compact while ensuring large memory capacity, further miniaturization of the recording and/or reproducing apparatus using such memory medium can be realized.
<SOH> BRIEF DESCRIPTION OF THE DRAWINGS <EOH>FIG. 1 is a perspective view showing plane side of a memory medium used in the present invention. FIG. 2 is a perspective view showing bottom face side of the memory medium shown in FIG. 1 . FIG. 3 is a perspective view showing an example of a recording/reproducing apparatus to which the present invention is applied. FIG. 4 is a perspective view showing another example of the recording/reproducing apparatus to which the present invention is applied. FIG. 5 is an exploded perspective view showing loading unit to which carrying apparatus according to the present invention is applied. FIG. 6 is a bottom view showing connector to which memory medium is connected along with the memory medium. FIG. 7 is a bottom view showing the state where memory medium is connected to connector. FIG. 8 is a plan view showing the state where holder is moved to initial position. FIG. 9 is a side view showing the side where loading control mechanism including fluctuation lever is provided. FIG. 10 is a side view showing the side where lock mechanism which locks holder at chassis is provided. FIG. 11 is a side view showing loading control mechanism when holder is moved to initial position. FIG. 12 is a plan view showing the state where memory medium is inserted into holder so that contact portion of the memory medium is connected to connector. FIG. 13 is a side view showing the state where memory medium is inserted into holder so that contact portion of the memory medium is connected to connector. FIG. 14 is a plan view showing the state where fluctuation lever is moved by holder in memory medium insertion direction and is then fluctuated so that it is engaged with chassis. FIG. 15 is a side view showing the state where engagement between fluctuation lever and holder is released so that the fluctuation lever is engaged with chassis. FIG. 16 is a side view of loading control mechanism showing the state where engagement between fluctuation lever and holder is released so that the fluctuation lever is engaged with chassis. FIG. 17 is a plan view showing the state where memory medium inserted into holder is caused to undergo loading at loading position. FIG. 18 is a side view showing the state where holder is caused to undergo biasing force of second extension coil spring so that it is moved to memory medium loading position. FIG. 19 is a side view showing loading control mechanism when holder is caused to undergo biasing force of second extension coil spring so that the holder is placed in the state where it is moved to memory medium loading portion. FIG. 20 is a plan view showing the state where memory medium loaded into holder is ejected. FIG. 21 is a side view showing the state where memory medium loaded into holder is ejected. detailed-description description="Detailed Description" end="lead"?
Apparatus and method for hydroforming a tubular part
A method and apparatus for shaping a raw tube into a formed part. The part can be configured within a die assembly including a hydroforming die structure and a pair of tube-engaging punches. The punches are inserted into the ends of the raw tube to shape the ends into the desired configuration. The middle portion of the raw tube is shaped into the desired configuration by hydroforming. Thus, the method and apparatus can shape the raw tube along its entire length, leaving no remnants of the raw tube that must be trimmed away.
1. A hydroforming die assembly 10 for hydroforming a part from a tubular blank 40, said part having a desired configuration different from a configuration of said blank and including a desired cross section at one end thereof, said die assembly 10 including a die structure having interior surfaces defining a die cavity 52, said die cavity 52 having a cross sectional configuration conforming generally to the desired exterior shape of said part, said die assembly 10 further comprising: a pair of tube-end engaging structures 81 disposed at opposite ends of said die cavity 52 and constructed and arranged to engage opposite ends of said tubular blank 40, said tube-end engaging structures 81 being constructed and arranged to seal said opposite ends of said tubular blank 40 and to pressurize hydroforming fluid within said tubular blank 40 for expanding said tubular blank 40 into conformity with said interior surfaces of said die cavity 52, a first of said tube-end engaging structures 81 having an outer cross-sectional configuration corresponding to said desired cross section at said one end of said part, said first of said tube-end engaging structures 81 being movable into forced engagement with one end of said tubular blank 40 to conform said one end of said tubular blank 40 to said outer cross-sectional configuration of said first of said tube-engaging structures 81 and hence said predetermined cross section at said one end of said part. 2. A hydroforming die assembly 10 according to claim 1, wherein said first tube-end engaging structure 81 has an exterior surface including a beveled portion 82 which is partially inserted into said one end of said tubular blank 40, said first tube-end engaging structure 81 being moved further into said one end of said tubular blank 40 so as to cause inner surface portions of said one end of said tubular blank 40 to slide in forced relation along said beveled portion 82 and cause said one end of said tubular blank 40 to be deformed i) over said first tube-end engaging structure 81 and ii) into conformity with said exterior surface of said first tube-end engaging structure 81, and wherein one end of said die cavity 52 receives said first tube-engaging structure 81, said interior surfaces of said die cavity 52 at said one end of said die cavity 52 having a configuration conforming to an exterior surface configuration of said part at said one end of said part. 3. A method of producing a hydroformed part using a hydroforming die assembly 10 for hydroforming a part from a tubular blank 40, the part having a desired configuration different from a configuration of the blank and including a desired cross section at one end of the part, the die assembly 10 including a die structure having interior surfaces defining a die cavity 52, the die cavity 52 having a cross sectional configuration conforming to the desired cross section of the part, the method comprising the steps of: providing and a pair of tube-end engaging structures 81 disposed at opposite ends of the die cavity 52 and constructed and arranged to engage opposite ends of the tubular blank 40, the tube-end engaging structures 81 being constructed and arranged to seal the opposite ends of the tubular blank 40 and to pressurize hydroforming fluid within the tubular blank 40 for expanding the tubular blank 40 into conformity with the interior surfaces of the die cavity 52, a first of the tube-end engaging structures 81 having an outer cross-sectional configuration corresponding to the desired cross section at one end of the part; moving the first of the tube-engaging structures 81 into forced engagement with one end of the tubular blank 40 to conform the one end of said tubular blank 40 to the outer cross-sectional configuration of the first of the tube-engaging structures 81 and hence the predetermined cross section at the one end of the part; and applying pressure within the tubular blank 40 to form the tubular blank 40 in to the desired configuration of the part. 4. A method according to claim 3, further comprising the step of: incorporating the part into a product without cutting off the one end of the part formed by the forced engagement of the first of the tube-engaging structures 81. 5. A assembly according to claim 1, wherein a first of said tube-engaging structures 81 has a forward end 33 and a lateral shoulder 88 protruding from a multifaceted portion 84 such that the first of said tube engaging structures 81 can be inserted into said tubular blank 40 continuously from the forward end 33 of said tube-engaging structure to said lateral shoulder 88 along said multifaceted portion 84 until said lateral shoulder 88 abuts said tube-engaging structure 81 and halts further insertion of said first of said tube-engaging structures 81 relative to said tubular blank 40. 6. A method according to claim 3, wherein the moving of the first of the tube-engaging structures 81 includes inserting the first of the tube-engaging structures 81 into the tubular blank 40 continuously from a forward end 33 of the first of the tube-engaging structures 81, along a multifaceted portion 84 of the first of the tube-engaging structures 81, and to a lateral shoulder 88 of the first of the tube-engaging structures 81 that protrudes from the multifaceted portion 84, until the lateral shoulder 88 abuts the end of tubular blank 40 and halts further insertion of the tube-engaging structure 81 relative to the tubular blank 40. 7. A method according to claim 3, wherein the moving of the first of the tube-engaging structures 81 into forced engagement with one end of the tubular blank 40 is a complete insertion of the first of the tube-engaging structures 81 into the tubular blank 40 and a complete reconfiguration of the one end of the tubular blank 40 into the final, desired configuration of the part by fully forcing the one end of the tubular blank 40 to conform to the outer surface of the first of the tube-engaging structures 81 and to the inner surface of the die cavity 52.
<SOH> BACKGROUND OF THE INVENTION <EOH>Typically, to form a tubular part by hydroforming, a raw tube is positioned within a hydroforming tool and the tube is secured at its ends. The middle portion of the raw tube is then subjected to hydroforming, leaving a transitional zone between the ends of the raw tube and the hydroformed middle portion. The hydroformed part is then finished by having the two transition zones removed from the tube, leaving only the fully hydroformed middle portion. The ends of the tube can be secured by tip portions being generally wedge-shaped as disclosed in EP 1022073A1. Hydroforming is also disclosed in the U.S. Pat. Nos. 5,987,950 to Horton and U.S. Pat. No. 6,014,950 to Jaekel et al. Removing the ends of the hydroformed part creates inefficiencies. For example, the cut away ends become wasted raw material. Also, cutting away ends requires additional cutting tools, which complicates the apparatus needed to create the finished part. Further, time is wasted performing the added step of cutting off the transitional zones at each end.
<SOH> SUMMARY OF THE INVENTION <EOH>One object of the present invention is to provide an improved apparatus and method for forming a hollow part. Another object of the present invention is to provide an improved apparatus and method for efficiently and cost effectively shaping a hollow part by mechanically shaping at least one end of the part and by hydroforming a portion of the part. Still another object of the invention is to provide an apparatus and method for forming a part that uses a punch to secure each end of the part while the punch shapes the end so that each end has the same configuration as a hydroformed, middle portion. The forgoing objects are basically attained by providing a hydroforming die assembly for hydroforming a part from a tubular blank, the part having a desired configuration different from a configuration of the blank and including a desired cross section at one end thereof, the die assembly comprising: a die structure having interior surfaces defining a die cavity, the die cavity having a cross sectional configuration conforming to the desired cross section of the part; and a pair of tube-end engaging structures disposed at opposite ends of the die cavity and constructed and arranged to engage opposite ends of the tubular blank, the tube-end engaging structures being constructed and arranged to seal the opposite ends of the tubular blank and to pressurize hydroforming fluid within the tubular blank for expanding the tubular blank into conformity with the interior surfaces of the die cavity, a first of the tube-end engaging structures having an outer cross-sectional configuration corresponding to the desired cross section at one end of the part, the first of the tube-engaging structures being movable into forced engagement with one end of the tubular blank to conform the one end of the tubular blank to the outer cross-sectional configuration of the first of the tube-engaging structures and hence the predetermined cross section at the one end of the part. The forgoing objects are also attained by providing a method of forming a hydroformed part comprising the steps of: providing a hydroforming die assembly for hydroforming a part from a tubular blank, the part having a desired configuration different from a configuration of the blank and including a desired cross section at one end of the part, the die assembly including a die structure having interior surfaces defining a die cavity, the die cavity having a cross sectional configuration conforming to the desired cross section of the part, and a pair of tube-end engaging structures disposed at opposite ends of the die cavity and constructed and arranged to engage opposite ends of the tubular blank, the tube-end engaging structures being constructed and arranged to seal the opposite ends of the tubular blank and to pressurize hydroforming fluid within the tubular blank for expanding the tubular blank into conformity with the interior surfaces of the die cavity, a first of the tube-end engaging structures having an outer cross-sectional configuration corresponding to the desired cross section at one end of the part; moving the first of the tube-engaging structures into forced engagement with one end of the tubular blank to conform the one end of the tubular blank to the outer cross-sectional configuration of the first of the tube-engaging structures and hence the predetermined cross section at the one end of the part; and applying pressure within the tubular blank to form the tubular blank in to the desired configuration of the part. Other objects, advantages, and features of the invention will become apparent from the following detailed description, appended drawings, and claims.
Urine test for the diagnosis of prion diseases
The present invention relates to a method for detecting the presence of the abnormal isoform of prion protein (PrPSC) in a urine sample of a subject. The method of the invention comprising the steps of: (a) providing a urine sample of said subject; (b) isolating from said sample all proteins, preferably, isolating proteins having a molecular weight higher than about 8 Kda; (c) optionally, and preferably, subjecting the proteins obtained in step (b) to protease digestion, and isolating from the mixture obtained in step (c) any protease resistant proteins; and (d) detecting the presence of PrPSC in the protease resistant fraction obtained in step (c) by a suitable detection technique. Furthermore, the invention further relates to methods for diagnosing a prion disease in a subject and for screening donors of blood samples for the presence of prion diseases. The invention further provides for a diagnostic kit for diagnosing a prion disease in a subject.
1-46. (canceled) 47. A method for detecting the presence of the abnormal isoform of prion protein (PrPSC) in a urine sample of a subject, said method comprising the steps of: a. providing a urine sample of said subject; b. isolating from said sample proteins; and c. detecting the presence of PrPSC in the protein mixture obtained in step (b) by a suitable detection technique. 48. A method according to claim 47 further comprising the step of subjecting the proteins obtained in step (b) to protease digestion. 49. A method according to claim 48 for detecting the presence of the abnormal isoform of prion protein (PrPSC) in a urine sample of a subject, said method comprising the steps of: a. providing a urine sample of said subject; b. isolating from said sample all proteins having a molecular weight higher than 8 KDa; c. subjecting the proteins obtained in step (b) to protease digestion; d. isolating from the mixture obtained in step(c) any protease resistant proteins; and e. detecting the presence of PrPSC in the protease resistant fraction obtained in step (d) by a suitable detection technique. 50. A method according to claim 49 wherein in step (b) said proteins are isolated by subjecting the urine sample to dialysis and precipitating the proteins from the dialysate. 51. A method according to claim 50 wherein step (b) further comprises addition of a carrier to the dialysate, prior to the protein precipitation. 52. A method for detecting the presence of the abnormal isoform of prion protein (PrPSC) in a urine sample of a subject, said method comprising the steps of: a. providing a urine sample of said subject; b. isolating from said sample all proteins having a molecular weight higher than about 8 KDa by subjecting said sample to dialysis, wherein said dialysis is performed using a membrane having a pore range of from about 6 KDa to 8 KDa; c. precipitating said proteins by ultracentrifuging the dialysate; d. subjecting the proteins obtained in step (b) to protease digestion; e. isolating from the mixture obtained in step (c) any protease resistant proteins; and f. detecting the presence of PrPSC in the protease resistant fraction obtained in step (d) by a suitable detection technique. 53. A method for detecting the presence of the abnormal isoform of prion protein (PrPSC) in a urine sample of a subject, said method comprising the steps of: a. providing a urine sample of said subject; b. isolating from said sample all proteins having a ;molecular weight higher than about 8 KDa by subjecting said sample to dialysis, wherein said dialysis is performed using a membrane having a pore range of from about 6 KDa to 8 DKa; c. precipitating said proteins by ultracentrifuging the dialysate for 1 hour at 100,000×g at 4° C.; d. subjecting the proteins obtained in step (b) to protease digestion; e. isolating from the mixture obtained in step (c) any protease resistant proteins; and f. detecting the presence of PrPSC in the protease resistant fraction obtained in step (d) by a suitable detection technique. 54. A method according to claim 52 wherein the proteins are precipitated by any one of methanol and TCA. 55. A method according to claim 54 wherein the proteins are precipitated by methanol. 56. A method according to claim 52 wherein said protease is proteinase K. 57. A method according to claim 53 wherein said protease is proteinase K. 58. A method according to claim 56 wherein in step (e) the presence of the PrPSC protease-resistant core in said protease resistant fraction, is detected by immunoassay. 59. A method according to claim 58 wherein said immunoassay is by immunoblot SDS PAGE analysis. 60. A method according to claim 60 wherein said PrP antibodies are 3F4 or 6H4 monoclonal antibodies. 61. A method according to claim 60 wherein said PrP antibodies are 3F4 or 6H4 monoclonal antibodies. 62. A method for diagnosing a prion disease in a subject comprising the steps of: a. obtaining a urine sample of said subject; and b. detecting the presence of the abnormal isoform of prion protein (PrPSC) in said urine sample by the method of claim 52; whereby the presence the PrPSC protein in said sample indicates that said subject carries a prion disease. 63. A method for diagnosing a prion disease in a subject comprising the steps of: a. obtaining a urine sample of said subject; and b. detecting the presence of the abnormal isoform of prion protein (PrPSC) is said urine sample by the method of claim 53; whereby the presence the PrPSC protein in said sample indicates that said subject carries a prion disease. 64. A method of claim 62 wherein said prion disease is a TSE disease. 65. A method claim 64 wherein said subject is a human subject. 66. A method of claim 64 wherein said subject is a bovine animal. 67. A method of claim 66 wherein said prion disease is BSE. 68. A method according to claim 62 wherein diagnosing of said prion disease is prior to or after onset of clinical symptoms. 69. A method for detecting the presence of metabolites of the abnormal isoform of prion protein (PrPSC) in a urine sample of a human subject, said metabolites being unique for human prion disease carries, said method comprising the steps of: a. providing a urine sample of said subject; b. isolating from said sample all proteins having a molecular weight higher than 8 KDa; and c. detecting the presence of said metabolites of PrPSC in the protein sample obtained in step (b) by an immunoassay comprising the use of 6H4 monoclonal antibodies that specifically bind to the protease-resistant core of PrPSC found in the urine of human prion disease carriers. 70. A method according to claim 69 wherein step (b) said proteins are isolated by subjecting the urine sample to dialysis using a membrane having a pore range of from 6 Kda to 8 Kda, and precipitating the proteins from the dialysate by ultracentrifugation. 71. A method according to claim 70 wherein the precipitation is performed by ultracentrifuging the dialysate for about 1 hour at 100,000×g at 4° C. 72. A method according to claim 69 wherein the proteins are precipitated by any one of methanol and TCA. 73. A method according to claim 72 wherein the proteins are precipitated by methanol. 74. A method according to claim 69 wherein said human prion disease is CJD. 75. A method for diagnosing a human prion disease in a subject comprising the steps of: a. obtaining a urine sample of said subject; and b. detecting the presence of metabolites of the abnormal isoform of prion protein (PrPSC) that are unique for human prion disease patients in said urine sample by the method of claim 69; whereby the presence of said PrPSC protein metabolites in said sample indicates that said subject carries a human prion disease. 76. A method according to claim 75 wherein said human prion disease is CJD. 77. A diagnostic kit for detecting the presence of the abnormal isoform of prion protein (PrPSC) in a urine sample of a subject, said kit comprising: a. means for isolating from said urine sample proteins; b. optionally, suitable carrier for stabilizing the PrPSC in the urine sample. c. a protease for digesting the protein isolate obtained by (a) or (b); d. means for isolating from the digest by (c) any protease resistant proteins; e. means for detecting the presence of PrPSC in the protease resistant fraction obtained by (d) and f. instructions for carrying out the detection of the presence of PrPSc in the urine sample according to the method of claim 52. 78. A kit according to claim 77 wherein said means for isolating proteins is for isolating proteins having a molecular weight higher than 8 Kda. 79. A kit according to claim 78 wherein said protease is proteinase K. 80. A kit according to claim 77 wherein said means for detecting the presence of PrPSC comprise reagents for detecting PrPSC by immunoassay. 81. A kit according to claim 80 wherein said immunoassay reagents comprise antibodies that specifically react with the protease-resistant core of PrPSC. 82. A diagnostic kit for detecting the presence of metabolites of the abnormal isoform or prion protein (PrPSC) that are unique for human prion disease carriers in a urine sample of a human subject, said kit comprising: a. means for isolating from said urine sample all proteins having a molecular weight higher than about 8 KDa; b. means for detecting the presence of PrPSC metabolites that are unique for human prion disease carriers in the protein sample obtained by step (a) by immunoassay comprising antibodies that specifically react with the metabolites of PrPSC that are unique for human prion disease carriers; and c. instructions for carrying out the detection of the presence of PrPSC in the urine sample according to the method of claim 69. 83. A kit according to claim 82 wherein said human prion disease is CJD.
<SOH> BACKGROUND OF THE INVENTION <EOH>Prion diseases, also known as TSEs (transmissible spongiform encephalopathies), are a group of fatal neurodegenerative diseases of animals and humans. Among the animal diseases, the most prevalent today is BSE (bovine spongiform encephalopathy) also known as the “Mad Cow Disease”. Although less than 100 patients have been diagnosed to date to be BSE-infected, the number of individuals incubating the disease may be millions. Another animal prior disease is scrapie in sheep, which after transmission to rodents constitutes the main experimental prior animal model. In humans, the most prevalent prion disease is CJD (Creutzfeldt Jakob Discase), which can be manifested either sporadically (about 1 patent per year); genetically (via mutations in the prion protein PrP gene); or in transmissible form, as in the BSE affected cases. It is a well known experimental fact that the incubation of prior diseases in humans and large animals can last for decades. Prion diseases are believed to be caused by the accumulation in the brain of PrP SC , an abnormally folded isoform of PrP C , a GPI anchored protein of unknown function. It has been postulated that prion diseases propagate by the conversion of PrP C molecules into protease-resistant and insoluble PrP SC by an as yet unknown mechanism. The proteinase K (PK) resistant PrP in prion diseases was described by McKinley et al. [Cell 35(1):57-62 (1988)]. Immunoblotting of a Proteinase K-digested brain sample infected with a prion disease with an anti-PrP antibody, reveals a characteristic N-terminally truncated PrP protein (the protease resistant core of PrP SC , denominated PrP 27-30), which is not present in controls or in individuals affected with any other neurological disease. To date, the diagnosis of prion diseases was based on the presence of this characteristic protease-resistant PrP in brain biopsies, as well as on clinical criteria. Current methods for the conclusive identification of Prion diseases include mostly a post-mortem analysis of the patient's brain homogenate. Clinical symptoms of the disease can many times be misleading. Evidently, sampling brain tissue from the living patient involves a painful and risky surgical procedure and, moreover, does not give a definite answer since the distribution of PrP SC in the brain is not homogenous. All commercial tests used to date are based on brain presence of protease resistant PrP, for example the Prion-Test of Prionics AG, Switzerland (which company is in charge of Most European active surveillance for BSE cases), which is an immunological test for the detection of prions in brain and spinal cord tissue, and is mainly used for BSE and scrapie diagnostics. Since the incubation period in prion diseases is very long (years), it is possible that there is a large number of asymptomatic human and animal carriers. There exists therefore a need for developing a simple and readily available pre-clinical and clinical diagnostic test for the disease. The need for such an in-vivo test has been reinforced since the reports of the first cases of variant Creutzfeldt Jakob disease (vCJD) in 1996 [Zeidler, M., et al., Lancet 350(9082), 908-10 (1997); Bruce, M. E., et al., Nature 389(6650), 498-501 (1997); Ironside, J. W., et al., Histopathology 37(1), 1-9 (2000)]. vCJD is a fatal neurodegenerative disease believed to be caused by the consumption of BSE contaminated meat, and the incubation time between infection to clinical symptoms may be as long as decades [Bruce, M. E., et al., Nature ibid (1997)]. As opposed to cattle, the incubating individuals will be present for many years, donating blood and in some cases other organs to the non-affected population. Additionally, such test is important for the food industry, and would enable detecting BSE in bovine animals such as cows and sheep, and to prevent marketing of infected meat and diary products of these animals. Therefore, a major object of the present invention is the development of a reliable, non-invasive method for diagnosing prion diseases which will allow the pre-clinical and clinical diagnosis of the disease in humans and in animals. Since most urine proteins originate from blood, the present inventors speculated that some PrP SC , either from brain or from a peripheral organ, is released during the incubation period into the blood serum in a non-aggregated form, although at low and undetectable concentrations. Due to its protease resistance, PrP SC is not digested by blood proteases. However, since the MW of PrP is below the cutoff size for filtering through kidney cells (about 40 kDA) [Berne, R. M., and Levy, M. N. Physiology, 4th Ed (1998)], PrP may subsequently be secreted into the urine and thereby be concentrated, as other proteins, at about 120 folds of its concentration in blood [Kocisko. D. A., et al., Nature 370(6489), 471-4 (1994)]. The concentration by the kidney makes possible to detect PrP SC in urine more easily than in blood. Thus, as will become apparent as the description proceeds, the present inventors have identified a prion specific protease resistant PrP isoform in the urine of prion infected animals and humans (UPrP SC ), which may be used for the in-vivo early diagnosis of ill as well as seemingly healthy but prion infected individuals. Moreover, the present invention shows that this protease resistant isoform UPrP SC , can be detected, following a specific enrichment procedure, in the urine of scrapie-infected hamsters, BSE-infected cattle and humans suffering from CJD. This specific enrichment procedure, according to the present invention may include dialysis of the sample through membrane having a pore range of about 6 KDa to about 8 KDa. The present invention further shows that UPrP SC was also found in urine of hamsters inoculated with prions long before the appearance of clinical signs. These findings strongly indicate the possibility of using the method of the invention also for pre-clinical diagnosis. The theoretical possibility for diagnosis of prion diseases in variety of body fluids, such as urine, has been mentioned in several patent documents. EP 0854364, for example, discloses a diagnostic method for neuro-degenerative disorders such as Alzheimer's disease and prion diseases. This method is based on concentrating a protein associated with the specific neuro-degenerative disease (such as PrP in prion diseases and APP in Alzheimer's disease), in a sample (urine, for example). The concentration is carried out by contacting the sample with a solid, non-buoyant particulate material having free ionic valencies such as calcium phosphate. However, this patent exemplifies the detection of only the Alzheimer's disease associated peptide APP. WO 93/23432 discloses a diagnostic method for prion diseases in different body fluids such as CSF (cerebrospinal fluid) and theoretically, urine. Similarly to EP 0854364, this method is based on concentrating the prion protein by ammonium sulfate precipitation and affinity chromatography. This publication exemplifies CSF as a sample. However, contrary to the prior art methods, the present invention clearly demonstrates the detection of the aberrant protease resistant urine isoform UPrP SC in urine samples of prion infected animals and humans. Furthermore, as shown by the present invention, dialysis of the urine seems to improve the detection procedure. Therefore, the present inventors propose that UPrP SC is present in a semi-denatured form, probably due to the relative high concentrations of urine denaturing agents, and is subsequently re-natured for example by the dialysis step. Thus, the specific enrichment of the urine sample according to the present invention provides a novel and reliable method for the detection of different prion diseases by a non-invasive procedure.
<SOH> SUMMARY OF THE INVENTION <EOH>The invention relates to a method for detecting the presence of the abnormal isoform of prion protein (PrP SC ) in a urine sample of a subject, said method comprising the steps of: (a) providing a urine sample of said subject; (b) isolating from said sample proteins; and (c) detecting the presence of PrP SC in the protein mixture obtained in step (b) by a suitable detection technique. A preferred embodiment relates to the method of the present invention, further comprising the step of subjecting the proteins obtained in step (b) to protease digestion. In a specifically preferred embodiment, the invention relates to a method for detecting the presence of the abnormal isoform of prion protein (PrP SC ) in a urine sample of a subject, said method comprising the steps of (a) providing a urine sample of said subject; (b) isolating from said sample all proteins having a molecular weight higher than about 8 KDa; (c) subjecting the proteins obtained in step (b) to protease digestion; (d) isolating from the mixture obtained in step (c) any protease resistant proteins; and (e) detecting the presence of PrP SC in the protease resistant fraction obtained in step (d) by a suitable detection technique. In step (b) of the method of the invention the proteins are preferably isolated by subjecting the urine sample to dialysis and precipitating the proteins from the dialysate. Optionally, prior to the protein precipitation, a carrier may be added to the dialysate for stabilizing the PrP SC . The dialysis is preferably performed using a membrane having a pore range of from about 6 KDa to about 8 KDa. The proteins may be precipitated from the dialysate by ultracentrifuging the same, for example for about 1 hour at 100,000×g at 4° C. Alternatively the proteins may be precipitated by any suitable protein precipitation technique. As a preferred embodiment proteins according to the invention may be precipitated by any one of methanol, TCA (Trichloracetic acid) or by any other precipitation method. Preferably, proteins may be precipitated by methanol, for example by the addition of methanol and freezing the sample to about −80° C. for about 1 hour, and subsequently centrifuging at 3000×rpm for about 30 minutes. The protein digestion is preferably performed by treating the sample with proteinase K, for example by adding proteinase K in concentration of up to 40 μg/ml and continuing digestion for about 30 min at 37° C. The presence of the PrP SC protease-resistant core in said non-digested fraction is preferably detected by immunoassay, for example by immunoblot SDS PAGE analysis, using monoclonal antibodies that specifically bind to the protease-resistant core of PrP SC , for example 3F4 or GH4 monoclonal antibodies. The invention also relates to a method for diagnosing a prion disease in a subject comprising the steps of (a) obtaining a urine sample of said subject; and (b) detecting the presence of the abnormal isoform of prion protein (PrP SC ) in said urine sample by the method of the invention, whereby the presence the PrP SC protein in said sample indicates that said subject carries a prion disease. In a preferred embodiment, said prion disease may be any TSE disease. The subject may be a human subject, for example a CJD, vCJD, GSS or FFI carrier or an individual infected with BSE. Alternatively the subject may be an animal infected with BSE, scrapie or any other TSE disease. The method of the invention further enables detection of different prion disease prior to or after onset of clinical symptoms. In yet a further embodiment that invention relates to a method for screening donors of blood samples for the presence of a prion disease in said donor comprising the steps of: (a) obtaining a urine sample from said donor; (b) detecting the presence of the abnormal isoform of prion protein (PrP SC ) in said urine sample by the method of the invention; and matching the results of the detection performed in step (b) to said blood sample. Still further, the invention relates to a method for detecting the presence of metabolites of the abnormal isoform which is probably a pathogenic isoform of prion protein (PrP SC ) in a urine sample of a subject, said metabolites being unique for human prion disease carriers. In a preferred embodiment such human prion disease may be CJD or vCJD. This method comprises the steps of: (a) providing a urine sample of said subject; (b) isolating from said sample all proteins having a molecular weight higher than about 8 KDa; and (c) detecting the presence of said metabolites of PrP SC in the protein sample obtained in step (b) by a suitable detection technique. In this embodiment, in step (b) said proteins may be isolated by subjecting the urine sample to dialysis and precipitating the proteins from the dialysate, for example by ultracentrifuging the dialysate, specifically for about 1 hour at 100,000×g at 4° C., or by any other suitable precipitation method. Preferred protein precipitation method may be methods such as methanol or TCA (Trichloracetic acid) precipitation. A specifically preferred technique for precipitation is methanol precipitation, specifically by the addition of methanol to the sample, freezing to about −80° C. for about 1 hour, and subsequently centrifuging at 3000×rpm (rounds per minute) for about 30 minutes. The detection of the presence of the said metabolites of PrP SC protease-resistant core in said protein sample is preferably by immunoassay, particularly SDS PAGE, using monoclonal antibodies that specifically bind to the specific metabolites of PrP SC found in urine of prion disease carriers, for example 6H4 monoclonal antibodies. The invention further relates to a method for diagnosing a prion disease in a subject comprising the steps of: (a) obtaining a urine sample of said subject; and (b) detecting the presence of metabolites of the abnormal isoform of prion protein (PrP SC ) that are unique for piron disease patients in said urine sample by a method of the invention; whereby the presence of said PrP SC protein metabolites in said sample indicates that said subject carries prion disease. According to a preferred embodiment, the method of the invention is intended for detection of the presence of metabolites unique for CJD and vCJD. Still further, the invention relates to a method for screening donors of blood samples for the presence of prion disease in said donor. This method comprises the steps of: (a) obtaining a urine sample from said donor; (b) detecting the presence of metabolites of the abnormal isoform of prion protein (PrP SC ) that are unique for prion disease patients in said urine sample by a method of the invention; and matching the results of the detection performed in step (b) to said blood sample. In another embodiment the invention relates to a diagnostic kit for detecting the presence of the abnormal isoform of prion protein (PrR SC ) in a urine sample of a subject, said kit comprising means for isolating from said urine sample all proteins; optionally, a carrier for stabilizing the PrP SC ; means for detecting the presence of PrP SC in the non-digested fraction; and instructions for carrying out the detection of the presence of PrP SC in the urine samples. In another preferred embodiment the invention relates to a diagnostic kit for detecting the presence of the abnormal isoform of prion protein (PrP SC ) in a urine sample of a subject, said kit comprising means for isolating from said urine sample all proteins having a molecular weight higher than about 8 KDa; optionally, a carrier for stabilizing the PrP SC in the dialysate; means for detecting the presence of PrP SC ; and instructions for carrying out the detection of the presence of PrP SC in the urine samples. In yet another specifically preferred embodiment the invention relates to a diagnostic kit for detecting the presence of the abnormal isoform of prion protein (PrP SC ) in a urine sample of a subject, said kit comprising means for isolating form said urine sample all proteins having a molecular weight higher than about 8 KDa; optionally, a carrier for stabilizing the PrP SC in the dialysate; a protease for digesting the protein isolate; means for isolating from the protein digest any protease digest any protease resistant proteins; means for detecting the presence of PrP SC in the protease resistant fraction; and instructions for carrying out the detection of the presence of PrP SC in the urine samples. In the kit of the invention, protease is preferably proteinase K and said means for detecting the presence of PrP SC comprise reagents for detecting PrP SC by immunoassay, such as antibodies that specifically react with the protease-resistant core of PrP SC . In another embodiment the invention relates to a diagnostic kit for detecting the presence of metabolites of the abnormal isoform of prion protein )PrP SC ) that are unique for human prion disease carriers, in a urine sample of a subject, said kit comprising: means for isolating from said urine sample all proteins having a molecular weight higher than about 8 KDa and means for detecting the presence of PrP SC metabolites that are unique for human prion disease carriers, preferably CJD and vCJD, in the obtained protein sample. The means for detecting the presence of said PrP SC metabolites preferably comprise reagents for detecting said PrP SC metabolites by immunoassay, for example antibodies that specifically react with the metabolites of PrP SC that are unique for human prion disease carriers. According to a preferred embodiment, said human prion disease may be CJD or vCJD.
Gel-type polymer electrolyte and use thereof
A gel-type polymer electrolyte, wherein said polymer comprises (A) an ethylene-unsaturated carboxylic acid copolymer or a derivative thereof and (B) a polyalkylene oxide having a hydroxyl group at one terminal thereof or a derivative thereof, which are bonded together by an ester bond. The gel-type polymer electrolyte has a high ionic conductivity, and makes it possible to provides a cell which has excellent charge/discharge characteristics at low temperatures as well as at high temperatures.
1. A gel-type polymer electrolyte, wherein said polymer comprises (A) an ethylene-unsaturated carboxylic acid copolymer or a derivative thereof and (B) a polyalkylene oxide having a hydroxyl group at one terminal thereof or a derivative thereof, which are bonded together by an ester bond. 2. A gel-type polymer electrolyte according to claim 1, wherein said ester bond is formed by the esterification of a carboxylic acid group of the ethylene-unsaturated carboxylic acid copolymer with the polyalkylene oxide having a hydroxyl group at one terminal thereof or with a derivative thereof. 3. A gel-type polymer electrolyte according to claim 1, wherein said ester bond is formed by the ester interchange reaction of an alkyl ester of the ethylene-unsaturated carboxylic acid copolymer or an alkyl ester derivative thereof with the polyalkylene oxide having a hydroxyl group at one terminal thereof or a derivative thereof. 4. A gel-type polymer electrolyte according to claim 1, wherein said polymer is formed by reacting the ethylene-unsaturated carboxylic acid copolymer or the derivative thereof (A) with the polyalkylene oxide having a hydroxyl group at one terminal thereof or the derivative thereof (B) at a-molar ratio expressed by the following formula, BHYD/ACAR wherein BHYD is a number of moles of hydroxyl groups of the polyalkylene oxide having a hydroxyl group at one terminal thereof or of the derivative thereof, and ACAR is a number of moles of carboxylic acid groups of the ethylene-unsaturated carboxylic acid copolymer or of the derivative thereof, of from 0.3 to 2.5. 5. A gel-type polymer electrolyte according to claim 1, wherein the remaining amount of the unreacted carboxylic acid groups in said polymer is not larger than 30 mol % on the basis of the carboxylic acid group of the ethylene-unsaturated carboxylic acid copolymer or the derivative thereof (A). 6. A gel-type polymer electrolyte according to claim 2, wherein said ethylene-unsaturated carboxylic acid copolymer or the derivative thereof (A) has a composition containing ethylene in an amount of from 50 to 98% by weight, an unsaturated carboxylic acid or an anhydride thereof in an amount of from 2 to 50% by weight, and other monomers in an amount of from 0 to 30% by weight. 7. A gel-type polymer electrolyte according to claim 3, wherein the alkyl ester of said ethylene-unsaturated carboxylic acid copolymer or the alkyl ester derivative thereof (A) has a composition containing ethylene in an amount of from 50 to 98% by weight, and an alkyl ester of an unsaturated carboxylic acid or an alkyl ester derivative thereof in an amount of from 2 to 50% by weight. 8. A gel-type polymer electrolyte according to claim 7, wherein the alkyl ester of said ethylene-unsaturated carboxylic acid copolymer is a methyl ester or an ethyl ester. 9. A gel-type polymer electrolyte according to claim 1, wherein said ethylene-unsaturated carboxylic acid copolymer or the derivative thereof (A) is an ionomer of which the carboxylic acid is partly neutralized with a monovalent metal or a multi-valent metal, the neutralization degree being in a range of from 0.5 to 60 mole %. 10. A gel-type polymer electrolyte according to claim 1, wherein said ethylene-unsaturated carboxylic acid copolymer or the derivative thereof (A) has a melt flow rate of from 0.1 to 500 g/10 min. at 190° C. under a load of 2160 g. 11. A gel-type polymer electrolyte according to claim 1, wherein the polyalkylene oxide having a hydroxyl group at one terminal thereof or the derivative thereof (B) has a number average molecular weight of from 200 to 100,000 and contains the ethylene oxide in an amount of from 30 to 100 mol %. 12. A gel-type polymer electrolyte according to claim 1, wherein a hydroxyl group at the other terminal of the polyalkylene oxide or the derivative thereof (B) is blocked by the etherification, esterification or by the reaction with a monoisocyanate. 13. A gel-type polymer electrolyte according to claim 2, wherein the esterification of said ethylene-unsaturated carboxylic acid copolymer or the derivative thereof (A) with the polyalkylene oxide having a hydroxyl group at one terminal thereof or the derivative thereof (B) is conducted in the presence of an acid catalyst. 14. A gel-type polymer electrolyte according to claim 3, wherein the formation of the ester bond by the ester interchange reaction is conducted in the presence of an organometal catalyst. 15. A gel-type polymer electrolyte according to claim 1, wherein said polymer is partly crosslinked in the presence of at least one kind of crosslinking agent selected from the group consisting of polyhydric alcohol, mono(meth)acrylic acid or an ester thereof, polyethylene glycol di(meth)acrylate, unsaturated higher fatty acid or an ester thereof and polyethylene glycol diglycidyl ether. 16. A gel-type polymer electrolyte according to claim 15, wherein said crosslinking agent is present in the reaction system in an amount of from 0.1 to 30% by weight. 17. A gel-type polymer electrolyte according to claim 1, wherein said gel-type polymer takes the form of a powder, a film or a sheet. 18. A gel-type polymer electrolyte according to claim 1, wherein said gel-type polymer is impregnated with an electrolytic solution comprising an electrolytic salt and a non-aqueous electrolytic solution. 19. A gel-type polymer electrolyte according to claim 18, wherein the solvent exists in the electrolytic solution at a ratio of from 30 to 95% by weight on the basis of the sum of said polymer and said electrolytic solution; 20. A gel-type polymer electrolyte according to claim 18, wherein the electrolyte exists in the electrolytic solution at a ratio of from 1 to 30% by weight on the basis of the sum of said polymer and said electrolytic solution; 21. A gel-type polymer electrolyte according to claim 18, wherein the electrolyte in the electrolytic solution is a lithium salt. 22. A gel-type polymer electrolyte according to claim 18, wherein the solvent in the electrolytic solution is a nonaqueous electrolytic solvent. 23. A polymer for polymer electrolyte comprising (A) an ethylene-unsaturated carboxylic acid copolymer or a derivative thereof and (B) a polyalkylene oxide having a hydroxyl group at one terminal thereof or a derivative thereof, the two being bonded together through an ester bond. 24. A secondary cell equipped with a layer of a gel-type polymer electrolyte of claim 1. 25. A secondary cell according to claim 24, wherein said secondary cell is a lithium cell. 26. A capacitor equipped with a layer of a gel-type polymer electrolyte of claim 1.
<SOH> TECHNICAL FIELD <EOH>The present invention relates to a gel-type polymer electrolyte comprising a non-halogen type polymer, and, particularly, to a polymer electrolyte that can be molded into a self-supported (self-erected) film and can, particularly, be used for polymer lithium cells suppressing the formation of lithium dendrite (tree-like traces) that occurs on the negative electrode surface when being electrically charged. The electrolyte can further be used for the capacitor.
<SOH> BRIEF DESCRIPTION OF DRAWINGS <EOH>FIG. 1 is a perspective view of a test cell for measuring the ionic conductivity and lithium ion transport number of the polymer electrolytes; FIG. 2 is a perspective view of a test cell for testing the charge/discharge characteristics; FIG. 3 is a graph showing the results of Example 1, i.e., showing changes in the weight of the polymers immersed in the electrolytic solution with the passage of time (Wo is the weight of the polymer only, and W is the weight of the polymer gel at that time); and FIGS. 4 a and 4 b are graphs showing the results of Example 23 (solid line) and results of Comparative Example 5 (broken line), wherein FIG. 4 ( a ) illustrates charge/discharge cycle characteristics and FIG. 4 ( b ) illustrates a charge/discharge curve at the eighth cycle. detailed-description description="Detailed Description" end="lead"?
Urotensin-II agonists and antagonists
The present invention features a novel class of cyclic polypeptides that have U-II antagonist and agonist activity. The invention also features methods for treating physiological or psychological conditions characterized by an excess or under expression of Ur4otensin-II.
1. A polypeptide or a variant thereof, said polypeptide having the formula: (R1)a-AA1-cyclo[AA2-AA3-AA4-AA5-AA6-Cys]-AA7-R2 wherein AA1 is the L isomer of an aromatic amino acid; AA2 is the L or D isomer of Cys; AA3 is an L isomer of an aromatic amino acid; AA4 is the L or D isomer of Trp; AA5 is the L or D isomer of Lys, N-Me-Lys, or Orn; AA6 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; AA7 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; R1 is H, lower alkyl, lower alkanoyl, or a lower acyl; a is 1 or 2; and R2 is OH, OR3, N(R3)2 or NHR3, where R3 is H, a lower alkyl, or arylalkyl; provided said peptide is not Cpa-c[D-Cys-Pal-D-Trp-Lys-Val-Cys]-Cpa-NH2; or a pharmaceutically acceptable salt of said polypeptide or variant. 2. The polypeptide of claim 1, wherein said aromatic amino acid has the formula: wherein X is H or a bond, and Ar represents a moiety selected from the group consisting of wherein n is 0, 1, 2, or 3 and each substituent Y independently represents NO2, CN, Cl, Br, I, F, Me, COR4, COOR4, or OR4, groups, where R4 is H or C1-C8 alkyl. 3. The polypeptide of claim 1, wherein AA3 is selected from the group consisting of Phe, Trp, Pal, His, β-Nal, 3-pyridyl-Ala, 4-pyridyl-Ala, 2,4-dichloro-phe, pentafluoro-Phe, p-Z-Phe, and o-Z-Phe, wherein Z is selected from the group consisting of Me, Cl, Br, F, OH, OMe, and NO2. 4. The polypeptide of claim 1, wherein AA4 is L-Trp. 5. The polypeptide of claim 1, wherein AA2 is D-Cys. 6. The polypeptide of claim 5, wherein AA3 is Phe, AA4 is Trp, AA5 is Lys, AA6 is Thr, AA7 is Val, and AA1 is Cpa. 7. The polypeptide of claim 6, wherein said polypeptide has the formula Cpa-c[D-Cys-Phe-Trp-Lys-Thr-Cys]-Val-NH2 (SEQ ID NO: 5). 8. A pharmaceutical composition comprising a polypeptide, or a variant thereof, and a pharmaceutically acceptable carrier, said polypeptide having the formula: (R1)a-AA1-cyclo[AA2-AA3-AA4-AA5-AA6-Cys]-AA7-R2 wherein AA1 is the L isomer of an aromatic amino acid; AA2 is the L or D isomer of Cys; AA3 is an L isomer of an aromatic amino acid; AA4 is the L or D isomer of Trp; AA5 is the L or D isomer of Lys, N-Me-Lys, or Orn; AA6 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; AA7 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; R1 is H, lower alkyl, lower alkanoyl, or a lower acyl; a is 1 or 2; and R2 is OH, OR3, N(R3)2 or NHR3, where R3 is H, a lower alkyl, or arylalkyl; provided said peptide is not Cpa-c[D-Cys-Pal-D-Trp-Lys-Val-Cys]-Cpa-NH2; or a pharmaceutically acceptable salt of said polypeptide or variant. 9. The pharmaceutical composition of claim 8, wherein said aromatic amino acid has the formula: wherein X is H or a bond, and Ar represents a moiety selected from the group consisting of wherein n is 0, 1, 2, or 3 and each substituent Y independently represents NO2, CN, Cl, Br, I, F, Me, COR4, COOR4, or OR4, groups, where R4 is H or C1-C8 alkyl. 10. The pharmaceutical composition of claim 8, wherein AA3 is selected from the group consisting of Phe, Trp, Pal, His, β-Nal, 3-pyridyl-Ala, 4-pyridyl-Ala, 2,4-dichloro-phe, pentafluoro-Phe, p-Z-Phe, and o-Z-Phe, wherein Z is selected from the group consisting of Me, Cl, Br, F, OH, OMe, and NO2. 11. The pharmaceutical composition of claim 8, wherein AA4 is L-Trp. 12. The pharmaceutical composition of claim 8, wherein AA2 is D-Cys. 13. The pharmaceutical composition of claim 12, wherein AA3 is Phe, AA4 is Trp, AA5 is Lys, AA6 is Thr, AA7 is Val, and AA1 is Cpa. 14. The pharmaceutical composition of claim 13, wherein said polypeptide has the formula Cpa-c[D-Cys-Phe-Tip-Lys-Thr-Cys]-Val-NH2, (SEQ ID NO: 5). 15. The pharmaceutical composition of claim 8, wherein said carrier is selected from the group consisting of saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. 16. A method of preventing or treating an abnormal condition characterized by an excess of Urotensin-II activity, said method comprising administering to a subject a therapeutically effective amount of a polypeptide, or variant thereof, said polypeptide having the formula: (R1)a-AA1-cyclo[AA2-AA3-AA4-AA5-AA6-Cys]-AA7-R2 wherein AA1 is the L isomer of an aromatic amino acid; AA2 is the L or D isomer of Cys; AA3 is an L isomer of an aromatic amino acid; AA4 is the L or D isomer of Trp; AA5 is the L or D isomer of Lys, N-Me-Lys, or Orn; AA6 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; AA7 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; R1 is H, lower alkyl, lower alkanoyl, or a lower acyl; a is 1 or 2; and R2 is OH, OR3, N(R3)2 or NHR3, where R3 is H, a lower alkyl, or arylalkyl; or a pharmaceutically acceptable salt thereof. 17. The method of claim 16, wherein said condition is selected from the group consisting of ischaemic heart disease, congestive heart failure, portal hypertension, variceal bleeding, hypotension, angina pectoris, myocardial infarction, ulcers, anxiety, schizophrenia, manic depression, delirium, dementia, mental retardation, and dyskinesias. 18. The method of claim 17, wherein said condition is ischaemic heart disease. 19. The method of claim 17, wherein said condition is congestive heart failure. 20. The method of claim 17, wherein said condition is portal hypertension 21. The method of claim 17, wherein said condition is variceal bleeding. 22. A method of modulating the effect of a Urotensin-II (U-II) peptide, said method comprising administering to a subject a polypeptide, or variant thereof, said polypeptide having the formula: (R1)a-AA1-cyclo[AA2-AA3-AA4-AA5-AA6-Cys]-AA7-R2 wherein AA1 is the L isomer of an aromatic amino acid; AA2 is the L or D isomer of Cys; AA3 is an L isomer of an aromatic amino acid; AA4 is the L or D isomer of Trp; AA5 is the L or D isomer of Lys, N-Me-Lys, or Orn; AA6 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; AA7 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; R1 is H, lower alkyl, lower alkanoyl, or a lower acyl; a is 1 or 2; and R2 is OH, OR3, N(R3)2 or NHR3, where R3 is H, a lower alkyl, or arylalkyl; or a pharmaceutically acceptable salt thereof. 23. The method of claim 22, wherein said modulating comprises decreasing the effect of said U-II peptide. 24. A urotensin II agonist polypeptide, or variant thereof, said polypeptide having the formula: Asp-c[Cys-Phe-Trp-Lys-Tyr-Cys)-Val-OH (SEQ ID NO: 3). 25. A method of modulating the effect of a Urotensin-II (U-II) peptide, said method comprising administering to a subject the polypeptide of claim 24. 26. The method of claim 25, wherein said modulating comprises increasing the effect of said U-II peptide. 27. A method of preventing or treating an abnormal condition characterized by an under expression of Urotensin-II activity, said method comprising administering to a subject a therapeutically effective amount of the polypeptide of claim 24.
<SOH> BACKGROUND OF THE INVENTION <EOH>Urotensin-II (U-II) is a cyclic neuropeptide with potent cardiovascular effects. Originally isolated from the caudal neurosecretory system of teleost fish, the primary structure of U-II has been established for several species of vertebrates, including various fish species, frogs, and humans. Sequence analysis of various U-II peptides from different species has revealed that, while the N-terminal region is highly variable, the C-terminal cyclic region of U-II is strongly conserved. Indeed, this cyclic region, which is responsible for the biological activity of U-II, is fully conserved from fish to humans (Coulouran, et al., Proc. Natl. Acad. Sci. USA (physiology), 95:15803-15808 (1998)). The fact that evolutionary pressure has acted to fully conserve the biologically active sequence of U-II suggests that this polypeptide plays an important role in human physiology. The cyclic region of U-U includes six amino acid residues (-Cys-Phe-Trp-Lys-Tyr-Cys-(SEQ ID NO: 1)) and is structurally similar to the biologically important central region of somatostatin-14 (-Phe-Trp-Lys-Thr-(SEQ ID NO: 2)). However, molecular cloning and sequence analysis of the carp preprourotensin II gene suggests that U-II and somatostatin are not derived from a common ancestor (Ohsako, S., et al., J. Neurosci., 6:2730-2735 (1986)). In fish, U-II peptides have been shown to exhibit several activities, including general smooth muscle contracting activity, although responses vary between species and vascular beds (Davenport, A., and Maquire, J., Trends in Pharmacological Sciences, 21:80-82 (2000); Bern, H. A., et al., Recent Prog. Horm. Res., 45:533-552 (1995)). Fish U-II has also been shown to possess constrictor activity in mammals, including major arteries in rats, but the receptor(s) mediating these peptide actions are not fully characterized. Recent studies have reported that an orphan human G-protein-coupled receptor, homologous to the rat GPR14 and expressed predominantly in cardiovascular tissue, functions as an U-II receptor (Ames, H., et al., Nature, 401:282-286 (1999)). Fish (goby) and human U-II reportedly bind to recombinant human GPR14 with high affinity, and the binding is functionally coupled to calcium mobilization. Human U-II is found within both vascular and cardiac tissue (including coronary atheroma) and effectively constricts isolated arteries from non-human primates (Ames, H., et al., supra). The potency of vasoconstriction of U-II is substantially greater than that of endothelin-1, making human U-II one of most potent mammalian vasoconstrictors currently known. In vivo, human U-II markedly increases total peripheral resistance in anaesthetized non-human primates, a response associated with profound cardiac contractile dysfunction (Ames, H., et al., supra). Since human U-II-like immunoreactivity is found within cardiac and vascular tissue (including coronary atheroma), U-II is believed to influence cardiovascular homeostasis and pathology (e.g., ischemic heart disease and congestive heart failure). Furthermore, the detection of U-II immunoreactivity within spinal cord and endocrine tissues suggests that U-II may have additional activities, including modulation of central nervous system and endocrine function in humans (Ames, H., et al., supra). Indeed, a number of maladies have been potentially linked to an excess or an under expression of U-II activity, including ischemic heart failure, hypotension, portal hypertension, angina pectoris, variceal bleeding, myocardial infarction, ulcers, and certain psychological and neurological disorders. Thus, there is a strong need for the development of potent compounds capable of modulating U-II activity, including U-II inhibitors or antagonists.
<SOH> SUMMARY OF THE INVENTION <EOH>The present invention features a novel class of cyclic polypeptides that have U-II antagonist activity. The polypeptides of the invention are octapeptides having the general formula: (R 1 ) a -AA 1 -cyclo[AA 2 -AA 3 -AA 4 -AA 5 -AA 6 -Cys]-AA 7 -R 2 (Formula I), wherein AA 1 is the L isomer of an aromatic amino acid; AA 2 is the L or D isomer of Cys; AA 3 is an L isomer of an aromatic amino acid; AA 4 is the L or D isomer of Trp; AA 5 is the L or D isomer of Lys, N-Me-Lys, or Orn; AA 6 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; AA 7 is the L or D isomer of Val, Thr, Leu, Ile, tert-Leu, Abu, Nle, or an aromatic amino acid; R 1 is H, a lower alkyl, lower alkanoyl, or a lower acyl; a is 1 or 2; and R 2 is OH, OR 3 , N(R 3 ) 2 , or NHR 3 , where R 3 is H, a lower alkyl, or arylalkyl; provided that the peptide is not Cpa-c[D-Cys-Pal-D-Trp-Lys-Val-Cys]-Cpa-NH 2 . In a preferred embodiment, AA 2 and AA 4 are D-Cys and L-Trp, respectively. In another preferred polypeptide, AA 1 is Cpa, AA 2 is D-Cys, AA 3 is Phe, AA 4 is Trp, AA 5 is Lys, AA 6 is Thr, and AA 7 is Val. In a particularly preferred embodiment, the polypeptide is an octapeptide having the formula Cpa-c[D-Cys-Phe-Trp-Lys-Thr-Cys]-Val-NH 2 . The invention also provides a Urotensin-II agonist polypeptide, and variants thereof, having the formula Asp-c[Cys-Phe-Trp-Lys-Tyr-Cys]-Val-OH. The polypeptides of the present invention are capable of altering U-II activity and can affect the binding of U-II to a receptor. Thus, these polypeptides may be administered to a subject as a means for preventing or treating medical or psychological conditions characterized by an excess or deficiency or under expression of Urotensin-It activity. Such conditions include, but are not limited to, ischaemic heart disease, congestive heart failure, portal hypertension, variceal bleeding, hypotension, angina pectoris, myocardial infarction, ulcers, anxiety, schizophrenia, manic depression, delirium, dementia, mental retardation, and dyskinesias. The present invention also provides pharmaceutical compositions that include a therapeutically effective amount of a polypeptide of Formula I in combination with a pharmaceutically acceptable carrier. Suitable carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The composition can be adapted for the mode of administration and can be in the form of a pill, tablet, capsule, spray, powder, or liquid. Other features and advantages of the invention will be apparent from the following detailed description thereof, and from the claims.
Method for verifying a perforation pattern serving as a security characteristic
A method of checking the authenticity of a perforation pattern which serves as a security feature and which when viewed in daylight back-lighting shows image information in the substrate of a document in particular in card form, wherein both the front side and also the rear side of the substrate is viewed in the region of the perforation pattern under incident light with light of a defined wavelength and in that case the image which is inherent to the perforation pattern shows on the front side while on the rear side thereof an area corresponding to the region of the perforation pattern shows up only in unspecified fashion.
1. A method of checking a perforation pattern which serves as a security feature and which when viewed in daylight back-lighting shows image information in the substrate of a document in particular in card form, characterized in that both the front side and also the rear side of the substrate is viewed in the region of the perforation pattern under incident light with light of a defined wavelength and in that case the image which is inherent to the perforation pattern shows on the front side while on the rear side thereof an area corresponding to the region of the perforation pattern shows up only in unspecified fashion. 2. A method as set forth in one of the preceding claims characterized in that viewing in daylight back-lighting is effected both from the front side and also from the rear side of the substrate and the image information must be visible in both cases. 3. A method as set forth in one of the preceding claims characterized in that viewing is effected both in daylight back-lighting and/or under incident light with light of a defined wavelength with the naked eye. 4. A method as set forth in one of the preceding claims characterized in that the wavelength of the incident light is between 10 nm and 500 nm, in particular between 100 nm and 400 nm and is in particular wide-band UV-light. 5. A method as set forth in one of the preceding claims characterized in that on the rear side the mouth openings of the perforation pattern are all of equal size and in particular are arranged in regular relationship with each other, but on the front side the mouth openings are of different sizes. 6. A method as set forth in one of the preceding claims characterized in that the diameters of the mouth openings on the front side are between 20 μm and 140 μm. 7. A method as set forth in one of the preceding claims characterized in that the perforations were burnt in by means of laser light. 8. A method as set forth in one of the preceding claims characterized in that the substrate is a plastic material, in particular a thermosetting plastic material, in particular polycarbonate. 9. A method as set forth in one of the preceding claims characterized in that the thickness of the substrate is between 0.1 and 1.0 millimeter. 10. A method as set forth in one of the preceding claims characterized in that the wavelength of the laser light used for burning in the perforations is between 6 μm and 12 μm. 11. A method as set forth in one of the preceding claims characterized in that the diameters of the mouth openings on the front side are between 100 times and 500 times the wavelength of the incident light used. 12. A method as set forth in one of the preceding claims characterized in that the thickness of the substrate is at least 300 thousand times the wavelength of the incident light used. 13. A method as set forth in one of the preceding claims characterized in that the wavelength of the laser light used for burning in the perforations is between 10 times and 70 times, in particular between 20 times and 40 times, the wavelength of the incident light used.
<SOH> II. TECHNICAL BACKGROUND <EOH>It is known from WO 98/19869 to incorporate a security feature in the form of a perforation pattern into documents such as banknotes, passes, or plastic cards, for example payment cards. In that case a plurality of perforations are applied extensively in side-by-side relationship. In that respect the attempt is made to produce an extensive two-dimensional image by control of the diameters of the perforations, that is to say the mouth openings of the perforations on one of the outward sides, or by way of the spacings between the mouth openings, that is to say the distribution of the mouth openings on the surface. In accordance with WO 00/43216 that is possible even when the arrangement involves blind holes which do not at all reach the viewing side. In practice that method is implemented by a procedure whereby perforations are burnt in by means of laser light in mutually juxtaposed relationship from the rear side of the document, that is to say the substrate, in regular or irregular raster configurations. Accordingly the mouth openings of the perforations on that rear side are all of the same size, but not the mouth openings of the corresponding perforations on the front side. Accordingly, by virtue of the variation in the size of the mouth openings on the front side, when viewing the front side in back-lighting, for example in daylight, it is possible to see an image with gray scales. This image may involve digits, letters, symbols or representations for example of a face. The differing diameters of the mouth openings on the front side are implemented by the duration for which the substrate is subjected to the action of laser light being varied in dependence on the desired diameter of the mouth opening. Although a laser beam is very well focused, ultimately, as it increasingly burns into a substrate from the rear side, it produces a hole which is conical at least in the deepest region, due to the Gaussian distribution of the energy within the cross-section of the laser beam and at least as long as the duration of action of the laser beam is not still substantially prolonged after piercing the substrate. That conicity can be increased or attenuated by suitable optical systems arranged upstream of the point of impingement, for shaping the laser beam. For, after piercing the substrate, the burnt passage is expanded to the full thickness of the laser beam and it is only after this has been achieved that the mouth opening on the exit side is approximately of the same size as the mouth opening on the entry side. As this is precisely not wanted for influencing the size of the mouth openings on the exit side, that is to say the front side of the document, the through openings through the substrate are slightly conical when considered in longitudinal section. In addition, due to the generation of heat by the laser light, the edges of the mouth openings, in particular on the exit side of the laser light, are rounded, at least insofar as the materials involved are relatively soft such as plastic materials. By virtue of the conical shape of the holes which constitute the perforation pattern, the intended image, in contrast to the assertion in the above-mentioned patent application, can be recognized when considered in the back-lighting mode, irrespective of the side from which the substrate is viewed. This is due to the fact that, even when viewing from the rear side, at which in fact the mouth openings are arranged regularly or irregularly in mutually juxtaposed relationship and are of the same diameter, the amount of the light passing thereto is nonetheless determined by the size of the mouth openings on the opposite side, the front side. Accordingly therefore the intended image effect can be recognized when viewing from both sides. Such a security feature however is relatively easy to circumvent: thus for example a corresponding perforation pattern could be produced by means of a kind of sewing machine, for example by using two needles of differing thicknesses, or also by means of one and the same needle which is of a conical configuration at least in the front region, and simultaneous control of the depth of engagement of the needle.
Reengineering event-driven field processes with a self-managed team approach
The invention provides a globally scalable, business-to-business (B2B) Mobile Process Service (MPS) that creates value for users and furthermore, through a franchise arrangement, allows value to be captured especially by telecom service providers. The MPS is characterized by conducting mass-customized, event-driven processes with teams dynamically maintained by requisition of these resources from virtual competence centers using an information system that notifies team members of changes of state that require activity. The information required, derived from both the field and from back-office, to execute that activity is made available at local, single-user server, distributed-databases maintained by replication over packet-switched radio. This required information is identified from business process object collaboration models in which the parameters (rows/columns) are specified in the modelled messages between the business objects. Main and supporting process frameworks are specified.
1. Resolution of the Productivity Paradox of Information Technology by a mobile process service (abbreviated to MPS) that increases the effectiveness and efficiency of any (main or supporting) event-driven, business-to-business (abbreviated to B2B) mobile, processes and is realized by combining three components: object-engineered processes (abbreviated to P), competence-centric organizations (abbreviated to O), and an information handling software that includes distributed relational databases maintained over radio (abbreviated to S); furthermore this claim is made only when all three components (P, O and S) are invoked and offered as a MPS, rather than as individual components, since it is the synergy between multi-disciplines (P, O and S) that is claimed as innovative and not obvious. 2. Mass customization of the mobile process service of claim-1 by rapid application development (RAD) wherein the object-oriented model of the intended business process and organization shall semi-automatically create system models (including data models) and these system models shall be one input for semi-automatic creation of the system executable programs (optionally including replication rules and triggers) to be employed as a component of the ultimate mobile process service (MPS) the intention being that such automation makes mass customization (for each individual end-user company) of processes economically feasible. 3. An ‘ABC’ refinement of the mobile process service of any preceding claim in which the main processes are enhanced by invoking three engineered supporting processes: Assessing (abbreviated to A), Building team (abbreviated to B), under the conditions that a) participant competence centers are within a single organization, or if b) several companies cooperate by allowing one or more companies to requisition resources from another's virtual, or more formal, competence centers (for example when a company requisitions resources from an up- or down-stream channel member), or if c) partner companies agree that resources in one company will conduct processes traditionally performed by the other (for example, if a service organization spots sales leads for the selling company), or if d) partner companies (for example one being a vendor of capital equipment and the other a vendor of related supplies) share the activities and information related to the customer, or if e) two or more vendor companies co-fund and collaboratively conduct the marketing/sales of a solution that utilizes the products/services from such participating vendors. Coordinating contact occasions (abbreviated to C). furthermore this claim is made only when all three components (A, B and C) are invoked since it is the synergy between these three processes that is claimed as innovative and not obvious. 4. An interface database that enables a refinement of the mobile process service (MPS) of any preceding claim, inserted as an information link between the resource team-members as engaging in preceding claims, and the so-called ‘back-office’ (or other) information storage sites the interface database being a replicated subset of the primary database, the latter maintained at the data center, wherein the interface database contains only those data elements flowing between the teams in the field and the back-office (or other) information storage sites, there being an option for the interface database to reside either at the data center of the mobile process service provider or at the premises of the mobile process user (MPU). 5. Worldwide deployment of the mobile process service of preceding claims, economically scalable, and achieved by information communication technology (ICT) wherein the data flow is orchestrated by data service (DS) from a single data center, and delivery executed by network service (NS) through, usually national, carriers to the teams in the local, usually national, field sites. 6. A selling process that is dependent on any preceding claim and has the following sequence of sub-processes: understanding customer fulfillment (abbreviated to UCF), in which Need and Solution are explicitly represented as business objects and Fulfillment (satisfaction) represents the relationship, with associated ‘states’ between Need and Solution reflecting the customer satisfaction level, inspiring customer (abbreviated to IC), and pursuing prospect (abbreviated to PP) 7. A mobile process service that globally deploys the SELLING process (which is a main, event-driven, B2B process) and is dependent on any preceding claim; the claimed delivering process service supports sub-processes that include, but are not limited to: understanding customer fulfillment (abbreviated to UCF), inspiring customer (abbreviated to IC), and pursuing prospect (abbreviated to PP) 8. A mobile process service that globally deploys the DELIVERING process (which is a main, event-driven, B2B process) and is dependent on any preceding claim; the claimed delivering process service supports sub-processes that include, but are not limited to: recognising (abbreviated to RE), planning (abbreviated to PL), committing and dispatching (abbreviated to CD), delivering (abbreviated to DE), documenting (abbreviated to DO), financial transactions (FT). 9. The organization and mechanisms to grow a market for mobile process service as articulated in the preceding claims, then explicit capture of a fair share of the value created during execution of any of the preceding claims within the telecom industry, by establishing telecom infrastructure providers as Franchisers that will: appoint expert resources by certification to design, construct and deploy the mobile process service for the business-to-business (B2B) mobile process users (MPUs); appoint ‘Inspiration Franchisees’ that will build the market for these mobile process services; appoint ‘System Service’ franchisees that will be responsible for constructing and continuously updating the system consisting of processes, organization and software; appoint ‘ICT Service’ franchisees that will be responsible for operating the system that distributes the data by radio and/or wire-line communication as a mobile process service (MPS), all this being explicitly such that the Franchiser may, at its discretion, appoint companies (such as network operators) as ICT Service franchisees that are customers, or potential customers, for products and services (such as base-stations) offered by the Franchiser and thus capture value indirectly. 10. A service specific basis for billing the user for mobile process service support to each instance of the process articulated in any of the preceding claims which may include, but are not limited to, a specific charge for supporting each selling instance, or each delivering instance, or each assessing instance, or each building team instance, or each coordinating contact occasion instance. This is in contrast to billing for products (like computer hardware), consulting services (utilized to design the system), horizontal applications (such as E-mail) and low-level service (such as time on the network or packets of data transported). 11. An inter-franchisee pricing mechanism, dependent on any of the preceding claims, mandated by the Franchiser such that all members of the channel, including certified expert resources, Inspiration Franchisees, System Service Franchisees and ICT Franchisees will be remunerated partly at the time when services are delivered to the next downstream level in the channel (for example when the system simulator is delivered to ICT provider) and partly at the time when the mobile process service is delivered to the mobile process user.
<SOH> E. BACKGROUND OF THE INVENTION <EOH>E.1 Field of Invention The invention concerns the reengineering of event-driven, field business processes (for example sales, or delivery of articles and/or services at customer sites), enabled by teams assembled within a competence-centric organization that is, in turn, enabled by an information system exploiting data distribution over radio. Of particular interest is the deployment as a Mobile Process Service—with emphasis on ‘process’ rather than ‘product’ or ‘application’. Deployment can be worldwide due to advances in ICT (information communication technology). E.2 Background Art and Assessment Thereof E.2.1 Theoretical Background E.2.1.1 Relationship Between Effort and Result The generic S-curve nature of the relationship between effort and result is well established. In the ‘selling process’ case, for example, the aspiration is to optimize the combination of sales revenue and M&S (marketing and selling) costs so as to maximize profit. It's easy to show arithmetically how modest increases in sales revenues and/or slight reductions in M&S costs have high impact on net profit. See Drawing 38 : Creating Value for End-Users Choosing the best point (TODAY) on the current (red) curve is done somewhat intuitively today. It's natural to increase resources (like adding another salesman) if expected additional gross profit generated exceeds the extra selling cost. Increasing M&S costs has little or no effect on Sales revenue once on the plateau. See Drawing 1 : Current Selling Process In the ‘service process’ case companies aspire to providing the highest affordable level of service and try to improve service level both in response time and in delivered quality. Any attempt to increase TODAY's level of service increases cost exponentially unless the service process is re-engineered. See Drawing 2 : Current Service Process Due to economies of scale there is a range in which sales-revenues are highly responsive to applied M&S resources (costs) and in which levels of service can be economically improved (red curve). However, in both the selling and service cases both curves hit an asymptote—sales revenue does not respond to further M&S costs and attempts to deliver higher service causes exponential increases in cost of providing that service. These ‘plateaus’ are due to diminishing returns. Most companies are at a point nearing the plateau—otherwise they would simply move along the curve until they do reach the plateau (red star on diagrams)—in other words they would just do ‘more of the same’ as long as return is greater than cost. See Drawing 3 : Selling—Shift to a Reengineered Process See Drawing 4 : Service—Shift to a Reengineered Process Modest gains might be made by automation of the existing processes. But the ‘automated process’ (orange) curve is identical in shape to today's more manual process—just displaced. Savings from pure automation are often not worth the cost of automating. Significant sales-revenue increases and level-of-service improvements, respectively, require reengineering the processes. Then new curves of a different shape emerge (green) in which increasing sales-revenue and level-of-service occur in an economical manner. E.2.1.2 Processes Conducted by Teams Sales and service processes are event-driven because of the unpredictable behavior of buying organizations and competitors (for sales) and machines (for delivery of service). Therefore it seems reasonable to consider possible ways of responding quickly to these events with appropriate competence. Thus, it is axiomatic (self-evident) that: IF event-driven processes, such as selling and service, are conducted by dynamically formed and informed, self-managed, customer focused teams, consisting of expert team members as appropriate to the progression of the process THEN sales revenues achieved (at a particular M&S cost level) and the cost-of-providing-service (at a particular service-level) can be dramatically improved. For event-driven field processes particularly, there is still a need to find more effective and efficient ways to achieve 1:1 (one person to one person), 1:N (one person to a team), N: 1 (a team to one person) and N:N (team to a team) interactions. Such team: team interactions are desirable within and between the inbound (such as buyer) and outbound (such as seller) process participants. Analogous to a football game, both teams must keep their eye-on-the-ball, being prepared at all times that the ball be passed to them to apply their particular skill or to respond to action by the other team. E.2.2 The State-of-the-Art E.2.2.1 Background Art—the Productivity Paradox of Information Technology. Modern communication technology has resulted in exponential growth of information distribution from the ‘hubs’ of centralized organizations but, on average, the information distributed has low added value beyond quick dispatching. Centralization is also contrary to the way people like to work. The high cost, low speed and the incomplete radio network coverage (except in the cities) are facts that will not go away soon. Productivity corresponding to exponentially increasing information communication traffic is thus not being realized. This is part of the long-standing ‘Productivity Paradox of Information Technology’. E. 2.2.2 Background Art—, Object-Oriented Business Process Engineering Background art includes the availability (for at least five years) of tools to model business processes in an object-oriented manner (such as Rational Rose from the Rational Software company). Business objects, including objects that represent worker role in processes, are identified by a so-called ‘Use-case’ approach. These objects are later mirrored electronically in a supporting information system. A software tool capable of converting an object-oriented business-engineering model to a system model was released to the market only in 1998. Therefore we believe that there are few published cases in which systems have been developed as a waterfall from business model to system model to working system and there are certainly no cases of systems created in such a manner that are an integral part of offerings of entire mobile process service (MPS) delivered over ICT. E. 2.2.3 Background Art—Competence-Centric Organization The literature includes rather vague references to Competence Centers (Beyond Reengineering—Champy). However, there are very few concrete adoptions—for reasons that include the unwillingness to give up ‘turf’ (hierarchical organizations) and the many other human factors such as recruitment, education, motivation and compensation of teams. See Drawing 5 : the Process-Managed Company The need for such organizations applies to, for example, consultative selling processes where there is discussion of need/solution between seller/buyer. In the delivery processes, especially delivery of engineering service that achieve the repair/maintenance of increasingly sophisticated equipment, such organizations are also needed. Hierarchical, departmental organization is still the norm in most companies. This results in allocation of tasks to resources from smaller, and more generally (rather than specially) qualified units. Furthermore, a person's competence (education/training/experience) is still applied repetitively to few (usually one) processes (such as selling) rather than being spread over several processes. In contrast, a competence-centric organization has qualified (educated, trained and experienced) personnel in (virtual) competence centers. An engineer (for example) may be assigned to activities within development, production or marketing/sales processes when/where required as the process progresses. Today such organizations rarely exist because the department (marketing, sales, development, production) ‘owns’ the resource. See Drawing 36 : Matching Competence and Task E.2.2.4 Background Art—Access to Data Stored at a Central Site Wire-line can collect and deliver data as far as the radio base-station. From there it must travel by radio to mobile people. Today communication by radio is still slow, expensive (typically up to 25 cents/minute during business hours). Good radio coverage is limited to cities—and there circuits are becoming overloaded. Wide coverage (especially internationally), convenient, fast, low cost direct radio access to data stored at a central site is still several years away. The development of good coverage has been handicapped by the recent auction of operating licenses by governments at high prices making subsequent investment in more base-stations (needed to give better coverage) a severe financial strain for operators. E. 2.2.5 Background Art—Data Replication Data can be replicated from Data Base Management Systems (DBMS) based on rules that identify which rows/columns of data tables shall be exchanged between stations (for example using Adaptive Server DBMS from Sybase). However, the rules for replication are still primitive—based on intuitive understanding of data requirement. This means that data delivered over radio is far from optimized and has low average value-added. See Drawing 6 : Replication—Publish and Subscribe E.2.2.6 Background Art—Ultra-Lite DBMS Recently DBMSs with minute ‘finger-print’ have appeared (such as from Sybase). These can carry a database on palmtop (Psion) and mobile phone. New operating systems (Windows CE from Microsoft and EPOC from Symbian) capable of working with these DBMSs are available. Present applications are limited to storing small personal databases and two-way paging and certainly do not include the orchestration of self-managed teams. E.2.2.7 Background Art—DBMS Triggers Some DBMSs (data base management systems) have the capability of creating notification messages when the ‘state’ of an object changes. This is achieved by using ‘triggers’. Such capability is hardly exploited today because the critical state-transitions have not been defined in the processes therefore what is notified to whom is ad hoc. E.2.2.8 Background Art—Simple Notification The most advanced combination of appropriate notification to the appropriate person in effect today is a short message sent to a mobile telephone to advise the recipient that there is a waiting E-Mail—the title of the E-Mail may also be displayed. But the E-Mail content itself is generally very unstructured, it is sent manually on a 1:1 or 1:N basis, is expensive to retrieve over radio and the E-Mail becomes yet another fragment in the unorganized in-box of the recipient.
<SOH> F. BRIEF SUMMARY OF THE INVENTION <EOH>
Security feature
The invention concerns a process for producing a security feature and a print medium which is equipped in that respect, which affords the possibility of not having to implement any change and in particular mechanical change in the surface of the card body. In that respect the substrate includes at least one change-over substance which by irradiation with light of a given wavelength experiences an irreversible change in colour from a starting colour to a final colour, wherein the substrate in the initial condition is so irradiated with a controlled light beam of that wavelength, in particular a laser light beam that, due to the change in colour caused thereby in the change-over substance, an image which can be recognised with the naked eye is produced within the volume of the substrate.
1. A process for producing a security feature, in particular on print media, in particular passes and identity cards, plastic payment cards, credit cards, memory cards etc, wherein the substrate (1, 1a, 1b) includes at least one change-over substance which by virtue of irradiation with light of a given wavelength (λ, λ1, λ2) experiences an irreversible change in color from a starting color to a final color, characterized in that the substrate when in the initial condition is so irradiated by a controlled light beam of that wavelength (λ, λ1, λ2), in particular a laser beam, that due to the change in color caused thereby in the change-over substance an image which can be recognized especially with the naked eye is produced on the substrate (1). 2. A process according to claim 1 characterized in that an image is produced within the volume of the substrate (1). 3. A process according to claim 1 characterized in that the change in color is a change from opaque to transparent. 4. A process according to one of the preceding claims characterized in that the change-over substance is present in the substrate in such a level of concentration that the change in color of the change-over substance appears to the viewer as a corresponding change in color of the substrate, in particular the substrate in the initial condition becomes thereby opaque and in the final condition becomes thereby transparent in the irradiated region. 5. A process according to claim 1 characterized in that the substrate is a plastic material, in particular ABS, PVC, PTE. 6. A process according to claim 1 characterized in that the change-over substance is distributed, in particular uniformly distributed, in the form of microcapsules or granules (2) or powder in the substrate. 7. A process according to claim 1 characterized in that the change-over substance is arranged only in a layer of the substrate, as viewed in cross-section through the substrate, in the interior of the substrate. 8. A process according to claim 1 characterized in that the print medium comprises a single substrate layer (1). 9. A process according to claim 1 characterized in that the print medium has a single substrate layer (1) having the change-over substance and is covered on at least one side by a cover layer (1′) of transparent material which does not react to irradiation with light of the wavelength (λ, λ1, λ2), and in particular the change-over substance is arranged on one of the outward sides of the substrate (1). 10. A process according to claim 1 characterized in that the print medium has a single substrate layer (1) having the change-over substance and said substrate (1) is coated on the rear side remote from the irradiation side with an opaque cover layer (1″). 11. A process according to claim 1 characterized in that the print medium comprises at least two interconnected substrate layers (1a, 1b) having different change-over substances which react to different wavelengths (λ, λ1, λ2) and which are so irradiated simultaneously or successively with light beams of different wavelengths (λ, λ1, λ2) that considered in plan view in terms of surface at most partially overlapping images are produced by the color changes in the two substrate layers (1a, 1b). 12. A process according to claim 11 characterized in that the starting colors of the substrate layers (1a, 1b) are different and the final colours colors are in particular the same and in particular are transparent.
<SOH> II. BACKGROUND OF THE INVENTION <EOH>In order to improve the anti-forgery security features of passes, identity cards, plastic payment cards or credit cards used for payments, various security features will be formed on appropriate print mediums. For example, one possibility consists in having the plastic payment card made with a multilayer part whose medium layer is opaque, and is used for printing the specific data, whereupon at least the front print side, mostly the rear side too, is coated in a transparent cover layer. On the upper side of the opaque medium layer there is printed an image, a symbol or something alike, and in the area of the above arranged transparent cover layer there is printed a surface three-dimensional structure having such a configuration that—by deflection and the lens effect of the elements that structure is consisting of—on viewing it from two different direction, usually forming a right angle between them, the different images and symbols respectively of that colours configuration become visible. Since on the one hand, from a technical point of view, such security features may be forged only by using an expensive equipment, and on the other hand as far as the type of the created image and the symbol respectively are concerned, and in regard of the serial number, the date of fabrication, a.s.o these may be modified, that provides first of all a relatively enhanced protection against short forgeries. However, the drawback of this security feature consists in having at least some parts of the arrangement on the card body surface which may be smeared, deteriorated or otherwise brought into a non-operating situation.
<SOH> III. SUMMARY OF THE INVENTION <EOH>a) Technical Object Therefore, the object of this invention is to provide another security feature and a process for producing it, and in particular a security feature which is not implying the necessity of operating modification, in particular mechanical ones, on the surface of the card's body. b) Attainment of the Object This object is attained by features of claim 1 . Advantageous embodiments are set forth in the append ant claims. In this case the basic idea consists in arranging at least a substance in a substrate of the plastic card body or in a layer of that card body which may be made of plastic, paper or a composite material, substance which experiences an irreversible colour change upon being irradiated with a light having a given wavelength, for example a laser beam. Such a changeover substance may be arranged in a substrate layer uniformly distributed in the form of microcapsules or granules or powder. Thereby there is possible to produce in the card a visible colour change for the viewer by irradiating it with a laser beam of a given wavelength the changeover substance reacts to. And as long as this substance is directly arranged in the substrate beneath the visible surface for the viewer, and there is a sufficient amount of it, for the viewer there is not visible the changing of the component colour particles but the colour effect of the substrate itself wherein the area irradiated with light of a given wavelength experiences a changing for the viewer. Thereby there is possible to produce images of a different colour inside a previously monochromatic substrate by irradiating it with light, in particular by means of a focused beam or by preventing it from being irradiated by means of an appropriately shaped shutter. Such an optical effect on a print medium may be indeed theoretically provided by printing on the medium surface too, but however there is another optical effect since on a closer examination there may be noticed that in case of employing a process according to the present invention the optical effect emerges inside the substrate and not on its surface. Even this optical effect may disappear if the change in colour experienced by the changeover substance which will thereby be produced in the entire substrate, is a change in colour from opaque to transparent. Thereby a previously entirely coloured card body may become completely transparent upon being irradiated with a given wavelength if the distribution and the density of the changeover substance are sufficiently high and homogeneous. Thereby the viewer may undertake the examinations relying on the fact that inside the card body there should be a pre-arranged image or a visible symbol which may be best noticed if is viewed in front of a source of light. In spite of this effect, the rear side of this substrate as seen in regard of the viewer may be coated in an opaque layer, preferably having another colour than the substrate in its previous state, thereby providing a different colouring in the irradiated area, which however becomes visible only in the rear side of the card body, therefore inside it. Likewise, such a substrate may be coated on the front and/or the rear side with a transparent, durable cover layer. The advantage of all these features consists in that, on the one hand the surface is always smooth and therefore it should not be mechanically printed or otherwise mechanically influenced, making thus the smearing almost impossible, and on the other hand the employing of this process allows the making of the transparent or otherwise coloured areas, therefore images and symbols, both in different shapes and locations on the initial card. Further on, the image effects thus created, in particular in the shape of transparent areas, may be anytime later improved too by irradiating them with a light of a given wavelength. Thereby the passports may be properly endowed with supplementary security features or the existing security feature may be improved by means of adding supplementary images a.s.o. Another advantage consists in that the print medium has several overlapping substrate layers, whereupon there is in every substrate layer another changeover substance which therefore reacts to another wavelength. Thereby, upon being simultaneously irradiated with different wavelength or with time delayed lights having both the given wavelengths, there may be created separate images in each of the overlapping layers, in particular transparent areas. When there are created partially overlapping transparent areas and the initial colour of those two substrate layers is different, there appears a special image effect because, the card body is transparent in the overlapping area of those two substrate layers, which do not overlap as seen from the front and rear side and provide thus another colour in every case and—according to the volume of the irradiated area—another image contour too. Likewise, there is possible to arrange more different changeover substances which react to different wavelengths in one and the same substrate layer.
Non-photochromic, colored, borosilicate inorganic glasses which absorb ultraviolet, and preparations
The object of the present invention is non-photochromic, colored, borosilicate inorganic glasses, which absorb ultraviolet and which contain effective amounts of silver, of copper, of halogen(s), of reducing agent(s) and of colorant(s). The present invention also relates to the preparation of said glasses. Within the context of said preparation, borosilicate glasses, which absorb ultraviolet and which are of various colors, can be obtained from a single crude borosilicate glass.
1. A non-photochromic, colored, borosilicate inorganic glass, which absorbs ultraviolet and which contains effective amounts of silver, of copper, of halogen(s), of reducing agent(s) and of colorant(s). 2. The glass according to claim 1, characterised in that it contains, per 100 parts by weight of its borosilicate base: from 0.0020 to 1.5 parts by weight of Ag; from 0.15 to 1.5 parts by weight of Cu; and an effective amount of at least one halogen selected from Cl, Br, I and F, which is conjugated to: an effective amount of at least one reducing agent, said effective amounts of halogen(s) and of reducing agent(s) generating sufficient copper halide(s) for the absorption of the ultraviolet; as well as an effective amount of at least one colorant. 3. The glass according to claim 1, characterised in that said reducing agent(s) is(are) selected from SnO, SnO2, Sb2O3 and As2O3. 4. The glass according to claim 1, characterised in that said effective amount of reducing agent(s) is between 0.1 and 5 parts by weight, per 100 parts by weight of its borosilicate base. 5. The glass according to claim 1, characterised in that said effective amount of halogen(s) is between 0.25 and 10 parts by weight, per 100 parts by weight of its borosilicate base; each halogen being advantageously incorporated in an amount less than or equal to 3 parts by weight. 6. The glass according to claim 1, characterised in that said colorant(s) is(are) selected from Fe2O3, NiO, CoO, V2O5, MnO, SeO2, Cr2O3 and Nd2O3. 7. The glass according to claim 1, characterised in that it contains, per 100 parts by weight of its borosilicate base, up to 7 parts by weight of Fe2O3, and/or up to 2 parts by weight of NiO, and/or up to 3 parts by weight of CoO, and/or up to 10 parts by weight of V2O5, and/or up to 2 parts by weight of SeO2, and/or up to 2 parts by weight of Cr2O3, and/or up to 4 parts by weight of Nd2O3. 8. The glass according to claim 1, characterised in that it contains an effective amount of NiO and/or of CoO. 9. The glass according to claim 1, characterised in that it contains, per 100 parts by weight of its borosilicate base from 0.0050 to 1 part by weight of Ag; from 0.2 to 1 part by weight of Cu; from 0 to 2 parts by weight of Cl; from 0 to 2 parts by weight of Br, with at least 0.25 parts by weight of Br+Cl; from 0 to 2 parts by weight of I; from 0 to 2 parts by weight of F; from 0.2 to 3 parts by weight of SnO; from 0 to 3 parts by weight of SnO2; from 0 to 3 parts by weight of Sb2O3; from 0 to 3 parts by weight of As2O3; from 0 to 3 parts by weight of Fe2O3; from 0.0100 to 1 part by weight of NiO; from 0.0050 to 1 part by weight of CoO; from 0 to 5 parts by weight of V2O5; from 0 to 0.5 part by weight of MnO; from 0 to 0.5 part by weight of SeO2; from 0 to 0.5 part by weight of Cr2O3; and from 0 to 0.5 part by weight of Nd2O3. 10. The glass according to claim 1, characterised in that its borosilicate base has the following composition by weight: SiO2 25-60% B2O3 10-35% Al2O3 3-17% ZrO2 0-13% Li2O 0-15% Na2O 0-15% K2O 0-15% with Li2O + Na2O + K2O > 2% MgO 0-10% CaO 0-15% SrO 0-15% BaO 0-15% with MgO + CaO + SrO + BaO > 1% ZnO 0-15% PbO 0-8% TiO2 0-8% Nb2O5 0-8% La2O3 0-8% Y2O3 0-8%; advantageously the following composition by weight: SiO2 30-55% B2O3 15-28% Al2O3 6-12% ZrO2 3-8% Li2O 1.5-3% Na2O 2-5% K2O 2.5-8% MgO 0-3% CaO 0-5% SrO 0-5% BaO 3-8% ZnO 0-11% PbO 0-5% TiO2 0-5% Nb2O5 0-5% La2O3 0-5% Y2O3 0-5%. 11. Sunglasses, which are corrective or non-corrective, made from a glass according to claim 1. 12. A method of preparing a glass according to claim 1, characterised in that it comprises: preparing a crude borosilicate glass which contains the suitable amounts of silver, of copper, of halogen(s), of reducing agent(s) and of colorant(s); and heat-treating said crude glass in order to generate within it the crystal phase of the halide(s) which is responsible for the absorption of the ultraviolet. 13. A method of preparing a glass according to claim to 1, characterised in that it comprises: preparing a crude borosilicate glass which contains the suitable amounts of silver, of copper, of halogen(s), of reducing agent(s) and of colorant(s); optionally heat-treating said crude glass in order to generate within it the crystal phase of the halide(s) which is responsible for the absorption of the ultraviolet; polishing the surface of said borosilicate glass which is heat-treated or non-heat-treated; and heat-treating said polished borosilicate glass under a reducing atmosphere; said heat-treatment being carried out under conditions of duration and temperature which are suitable for obtaining the coloration sought after; said heat-treatment also ensuring the generation of the crystal phase of the halide(s) which is responsible for the absorption of the ultraviolet if this crystal phase has not been generated beforehand. 14. The method according to claim 12, characterised in that said heat-treatment of the crude glass is carried out, for 10 minutes to 2 hours, at a temperature between 450 and 650° C. 15. The method according to claim 12, characterised in that said heat-treatment under reducing atmosphere is carried out, for 2 minutes to 12 hours, at a temperature between 250 and 650° C. 16. A method for preparing the glasses according to claim 1, which are of various colors, characterised in that it comprises heat-treating, under a reducing atmosphere, under various conditions of duration or(and) of temperature, a polished crude borosilicate glass which has a defined composition containing suitable amounts of silver, of copper, of halogen(s), of reducing agent(s) and of colorant(s).
Preparation of 4-methyl-2, 3, 5, 6-tetrafluorobenzyl alcohol
A process for the preparation of 1,4-bis(hydroxymethyl)-2,3,5,6-tetrafluorobenzene comprising fluorinating 2,3,5,6-tetrachloroterephthalonitrile to obtain 2,3,5,6-tetrafluoroterephthalonitrile, hydrogenating 2,3,5,6-tetrafluoroterephthalonitrile to give 1,4-bis(aminomethyl)-2,3,5,6-tetrafluorobenzene and converting 1,4-bis(aminomethyl)-2,3,5,6-tetrafluorobenzene to 1,4-bis(hydroxymethyl)-2,3,5,6-tetrafluorobenzene.