US20030186391A1 - Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purpose - Google Patents

Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purpose Download PDF

Info

Publication number
US20030186391A1
US20030186391A1 US10/430,442 US43044203A US2003186391A1 US 20030186391 A1 US20030186391 A1 US 20030186391A1 US 43044203 A US43044203 A US 43044203A US 2003186391 A1 US2003186391 A1 US 2003186391A1
Authority
US
United States
Prior art keywords
seq
msrv
sequence
sequences
pcr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/430,442
Inventor
Herve Perron
Frederic Beseme
Frederic Bedin
Glaucia Paranhos-Baccala
Florence Komurian-Pradel
Colette Jolivet-Reynaud
Bernard Mandrand
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biomerieux SA
Original Assignee
Biomerieux SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biomerieux SA filed Critical Biomerieux SA
Priority to US10/430,442 priority Critical patent/US20030186391A1/en
Publication of US20030186391A1 publication Critical patent/US20030186391A1/en
Priority to US11/463,109 priority patent/US7932350B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/70Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving virus or bacteriophage
    • C12Q1/701Specific hybridization probes
    • C12Q1/702Specific hybridization probes for retroviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/10022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • MS Multiple sclerosis
  • CNS central nervous system
  • endogenous retroviruses are very important in the context of our discovery since, in the case of MSRV-1, it has been observed that endogenous retroviral sequences comprising sequences homologous to the MSRV-1 genome exist in normal human DNA.
  • endogenous retroviral elements ERP related to MSRV-1 by all or part of their genome explains the fact that the expression of the MSRV-1 retrovirus in human cells is able to interact with closely related endogenous sequences.
  • interactions consist mainly of (i) a trans-activation or coactivation of ERVs by the replicative retrovirus (ii) and “illegitimate” encapsidation of RNAs related to ERVS, or of ERVs—or even of cellular RNAs—simply possessing compatible encapsidation sequences, in the retroviral particles produced by the expression of the replicative strain, which are sometimes transmissible and sometimes with a pathogenicity of their own, and (iii) more or less substantial recombinations between the coencapsidated genomes, in particular in the phases of reverse transcription, which lead to the formation of hybrid genomes, which are sometimes transmissible and sometimes with a pathogenicity of their own.
  • the pathology caused by a retrovirus such as MSRV-1 may be a direct effect of its expression and of the proteins or peptides produced as a result thereof, but also an effect of the activation, the encapsidation or the recombination of related or heterologous genomes and of the proteins or peptides produced as a result thereof; thus, these genomes associated with the expression of and/or infection by MSRV-1 are an integral part of the potential pathogenicity of this virus, and hence constitute means of diagnostic detection and special therapeutic targets.
  • any agent associated with or cofactor of these interactions responsible for the pathogenesis in question such as MSRV-2 or the glyotoxic factor which are described in the patent application published under No. FR-2,716,198, may participate in the development of an overall and very effective strategy for the diagnosis, prognosis, therapeutic monitoring and/or integrated therapy of MS in particular, but also of any other disease associated with the same agents.
  • RA rheumatoid arthritis
  • the present invention relates, in the first place, to a viral material, in the isolated or purified state, which may be recognized or characterized in different ways:
  • its genome comprises a nucleotide sequence chosen from the group including the sequences SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:89, their complementary sequences and their equivalent sequences, in particular nucleotide sequences displaying, for any succession of 100 contiguous monomers, at least 50% and preferably at least 70% homology with the said sequences SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60 SEQ ID NO:61, SEQ ID NO:89, respectively, and their complementary sequences;
  • the region of its genome comprising the env and pol genes and a portion of the gag gene, excluding the subregion having a sequence identical or equivalent to SEQ ID NO:1, codes for any polypeptide displaying, for any contiguous succession of at least 30 amino acids, at least 50% and preferably at least 70% homology with a peptide sequence encoded by any nucleotide sequence chosen from the group including SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60 SEQ ID NO:61 SEQ ID NO:89 and their complementary sequences;
  • the pol gene comprises a nucleotide sequence partially or totally identical or equivalent to SEQ ID NO:57, excluding SEQ ID NO:1.
  • the gag gene comprises a nucleotide sequence partially or totally identical or equivalent to SEQ ID NO:88.
  • the viral material as defined above is associated with MS. And as defined by reference to the pol or gag gene of MSRV-1, and more especially to the sequences SEQ ID NOS 51, 56, 57, 59, 60, 61, 88 and 89, this viral material is associated with RA.
  • the present invention also relates to different nucleotide fragments each comprising a nucleotide sequence chosen from the group including:
  • genomic sequences partial or total, includes all sequences associated by coencapsidation or by coexpression, or recombined sequences.
  • such a fragment comprises:
  • nucleotide sequence identical to a partial or total genomic sequence of the pol gene of the MSRV-1 virus except for the total sequence of the nucleotide fragment defined by SEQ ID NO:1, or identical to any sequence equivalent to the said partial or total genomic sequence, in particular one which is homologous to the latter;
  • the invention relates to a nucleotide fragment comprising a coding nucleotide sequence which is partially or totally identical to a nucleotide sequence chosen from the group including:
  • the invention also relates to any nucleic acid probe for detection of a pathogenic and/or infective agent associated with MS, which is capable of hybridizing specifically with any fragment such as is defined above, belonging or lying within the genome of the said pathogenic agent. It relates, in addition, to any nucleic acid probe for detection of a pathogenic and/or infective agent associated with RA, which is capable of hybridizing specifically with any fragment as defined above by reference to the pol and gag genes, and especially with respect to the sequences SEQ ID NOS 40, 51, 56, 59, 60, 61, 62, 89 and SEQ ID NOS 39, 63 and 90.
  • the invention also relates to a primer for the amplification by polymerization of an RNA or a DNA of a viral material, comprising a nucleotide sequence identical or equivalent to at least one portion of the nucleotide sequence of any fragment such as is defined above, in particular a nucleotide sequence displaying, for any succession of 10 contiguous monomers, at least 70% homology with at least the said portion of the said fragment.
  • the nucleotide sequence of such a primer is identical to any one of the sequences chosen from the group including SEQ ID NO:47 to SEQ ID NO:50, SEQ ID NO:55 and SEQ ID NO:64 SEQ ID NO:86.
  • the invention also encompasses any RNA or DNA, and in particular replication vector, comprising a genomic fragment of the viral material such as is defined above, or a nucleotide fragment such as is defined above.
  • the invention also relates to the different peptides encoded by any open reading frame belonging to a nucleotide fragment such as is defined above, in particular any polypeptide, for example any oligopeptide forming or comprising an antigenic determinant recognized by sera of patients infected with the MSRV-1 virus and/or in whom the MSRV-1 virus has been reactivated.
  • this polypeptide is antigenic, and is encoded by the open reading frame beginning, in the 5′-3′ direction, at nucleotide 181 and ending at nucleotide 330 of SEQ ID NO:1.
  • the invention relates to an antigenic polypeptide recognized by the sera of patients infected with the MSRV-1 virus, and/or in whom the MSRV-1 virus has been reactivated, whose peptide sequence is partially or totally identical or is equivalent to the sequence defined by SEQ ID NO:39, SEQ ID NO:63 and SEQ ID NO:87; such a sequence is identical, for example, to any sequence chosen from the group including the sequences SEQ ID NO:41 to SEQ ID NO:44, SEQ ID NO:63 and SEQ ID NO:87.
  • the present invention also proposes mono- or polyclonal antibodies directed against the MSRV-1 virus, which are obtained by the immunological reaction of a human or animal body to an immunogenic agent consisting of an antigenic polypeptide such as is defined above.
  • the invention next relates to:
  • reagents for detection of the MSRV- virus, or of an exposure to the latter comprising, as reactive substance, a peptide, in particular an antigenic peptide, such as is defined above, or an anti-ligand, in particular an antibody to the said peptide;
  • compositions comprising one or more peptides, in particular antigenic peptides, such as are defined above, or one or more anti-ligands, in particular antibodies to the peptides, discussed above; such a composition is preferably, and by way of example, a vaccine composition.
  • the invention also relates to any diagnostic, prophylactic or therapeutic composition, in particular for inhibiting the expression of at least one pathogenic and/or infective agent associated with MS comprising a nucleotide fragment such as is defined above or a polynucleotide, in particular oligonucleotide, whose sequence is partially identical to that of the said fragment, except for that of the fragment having the nucleotide sequence SEQ ID NO:1.
  • any diagnostic, prophylactic or therapeutic composition in particular for inhibiting the expression of at least one pathogenic and/or infective agent associated with RA, comprising a nucleotide fragment such as is defined above by reference to the pol and gag genes, and especially with respect to the sequences SEQ ID NOS 40, 51, 56, 59, 60, 61, 62 and 89.
  • these same fragments or polynucleotides may participate in all suitable compositions for detecting, according to any suitable process or method, a pathological and/or infective agent associated with MS and with RA, respectively, in a biological sample.
  • a pathological and/or infective agent associated with MS and with RA respectively
  • an RNA and/or a DNA presumed to belong or originating from the said pathological and/or infective agent, and/or their complementary RNA and/or DNA is/are brought into contact with such a composition.
  • the present invention also relates to any process for detecting the presence or exposure to such a pathological and/or infective agent, in a biological sample, by bringing this sample into contact with a peptide, in particular an antigenic peptide such as is defined above, or an anti-ligand, in particular an antibody to this peptide, such as is defined above.
  • a peptide in particular an antigenic peptide such as is defined above, or an anti-ligand, in particular an antibody to this peptide, such as is defined above.
  • a device for detection of the MSRV-1 virus comprises a reagent such as is defined above, supported by a solid support which is immunologically compatible with the reagent, and a means for bringing the biological sample, for example a sample of blood or of cerebrospinal fluid, likely to contain anti-MSRV-1 antibodies, into contact with this reagent under conditions permitting a possible immunological reaction, the foregoing items being accompanied by means for detecting the immune complex formed with this reagent.
  • the invention also relates to the detection of anti-MSRV-1 antibodies in a biological sample, for example a sample of blood or of cerebrospinal fluid, according to which this sample is brought into contact with a reagent such as is defined above, consisting of an antibody, under conditions permitting their possible immunological reaction, and the presence of the immune complex thereby formed with the reagent is then detected.
  • a biological sample for example a sample of blood or of cerebrospinal fluid
  • strain or isolate is understood to mean any infective and/or pathogenic biological fraction containing, for example, viruses and/or bacteria and/or parasites, generating pathogenic and/or antigenic power, harboured by a culture or a living host; as an example, a viral strain according to the above definition can contain a coinfective agent, for example a pathogenic protist,
  • the term “MSRV” used in the present description denotes any pathogenic and/or infective agent associated with MS, in particular a viral species, the attenuated strains of the said viral species or the defective-interfering particles or particles containing coencapsidated genomes, or alternatively genomes recombined with a portion of the MSRV-1 genome, derived from this species.
  • Viruses, and especially viruses containing RNA are known to have a variability resulting, in particular, from relatively high rates of spontaneous mutation (7), which will be borne in mind below for defining the notion of equivalence,
  • human virus is understood to mean a virus capable of infecting, or of being harboured by human beings,
  • the variant of a virus or of a pathogenic and/or infective agent according to the invention comprises at least one antigen recognized by at least one antibody directed against at least one corresponding antigen of the said virus and/or said pathogenic and/or infective agent, and/or a genome any part of which is detected by at least one hybridization probe and/or at least one nucleotide amplification primer specific for the said virus and/or pathogenic and/or infective agent, such as, for example, for the MSRV-1 virus, the primers and probes having a nucleotide sequence chosen from SEQ ID No. 20 to SEQ ID No. 24, SEQ ID No. 26, SEQ ID No. 16 to SEQ ID No. 19, SEQ ID No.
  • SEQ ID No. 33 SEQ ID No. 45, SEQ ID No. 47, SEQ ID No. 48, SEQ ID No. 49, SEQ ID No. 50, SEQ ID No. 45 and their complementary sequences, under particular hybridization conditions well known to a person skilled in the art,
  • a nucleotide fragment or an oligonucleotide or polynucleotide is an arrangement of monomers, or a biopolymer, characterized by the informational sequence of the natural nucleic acids, which is capable of hybridizing with any other nucleotide fragment under predetermined conditions, it being possible for the arrangement to contain monomers of different chemical structures and to be obtained from a molecule of natural nucleic acid and/or by genetic recombination and/or by chemical synthesis;
  • a nucleotide fragment may be identical to a genomic fragment of the MSRV-1 virus discussed in the present invention, in particular a gene of this virus, for example pol or env in the case of the said virus,
  • a monomer can be a natural nucleotide of nucleic acid whose constituent elements are a sugar, a phosphate group and a nitrogenous base; in RNA the sugar is ribose, in DNA the sugar is 2-deoxyribose; depending on whether the nucleic acid is DNA or RNA, the nitrogenous base is chosen from adenine, guanine, uracil, cytosine and thymine; or the nucleotide can be modified in at least one of the three constituent elements; as an example, the modification can occur in the bases, generating modified bases such as inosine, 5-methyldeoxy- cytidine, deoxyuridine, 5-(dimethylamino)deoxyuridine, 2,6-diaminopurine, 5-bromodeoxyuridine and any other modified base promoting hybridization; in the sugar, the modification can consist of the replacement of at least one deoxyribose by a polyamide (8), and in the phosphate group,
  • “informational sequence” is understood to mean any ordered succession of monomers whose chemical nature and order in a reference direction constitute or otherwise an item of functional information of the same quality as that of the natural nucleic acids,
  • hybridization is understood to mean the process during which, under suitable working conditions, two nucleotide fragments having sufficiently complementary sequences pair to form a complex structure, in particular double or triple, preferably in the form of a helix,
  • a probe comprises a nucleotide fragment synthesized chemically or obtained by digestion or enzymatic cleavage of a longer nucleotide fragment, comprising at least six monomers, advantageously from 10 to 100 monomers and preferably 10 to 30 monomers, and possessing a specificity of hybridization under particular conditions; preferably, a probe possessing fewer than 10 monomers is not used alone, but is used in the presence of other probes of equally short size or otherwise; under certain special conditions, it may be useful to use probes of size greater than 100 monomers; a probe may be used, in particular, for diagnostic purposes, such molecules being, for example, capture and/or detection probes,
  • the capture probe may be immobilized on a solid support by any suitable means, that is to say directly or indirectly, for example by covalent bonding or passive adsorption,
  • the detection probe may be labelled by means of a label chosen, in particular, from radioactive isotopes, enzymes chosen, in particular, from peroxidase and alkaline phosphatase and those capable of hydrolysing a chromogenic, fluorogenic or luminescent substrate, chromophoric chemical compounds, chromogenic, fluorogenic or luminescent compounds, nucleotide base analogues and biotin,
  • a label chosen, in particular, from radioactive isotopes, enzymes chosen, in particular, from peroxidase and alkaline phosphatase and those capable of hydrolysing a chromogenic, fluorogenic or luminescent substrate, chromophoric chemical compounds, chromogenic, fluorogenic or luminescent compounds, nucleotide base analogues and biotin,
  • the probes used for diagnostic purposes of the invention may be employed in all known hybridization techniques, and in particular the techniques termed “DOT-BLOT” (9), “SOUTHERN BLOT” (10), “NORTHERN BLOT”, which is a technique identical to the “SOUTHERN BLOT” technique but which uses RNA as target, and the SANDWICH technique (11); advantageously, the SANDWICH technique is used in the present invention, comprising a specific capture probe and/or a specific detection probe, on the understanding that the capture probe and the detection probe must possess an at least partially different nucleotide sequence,
  • any probe according to the present invention can hybridize in vivo or in vitro with RNA and/or with DNA in order to block the phenomena of replication, in particular translation and/or transcription, and/or to degrade the said DNA and/or RNA,
  • a primer is a probe comprising at least six monomers, and advantageously from 10 to 30 monomers, possessing a specificity of hybridization under particular conditions for the initiation of an enzymatic polymerization, for example in an amplification technique such as PCR (polymerase chain reaction), in an elongation process such as sequencing, in a method of reverse transcription or the like,
  • nucleotide or peptide sequences are termed equivalent or derived with respect to one another, or with respect to a reference sequence, if functionally the corresponding biopolymers can perform substantially the same role, without being identical, as regards the application or use in question, or in the technique in which they participate; two sequences are, in particular, equivalent if they are obtained as a result of natural variability, in particular spontaneous mutation of the species from which they have been identified, or induced variability, as are two homologous sequences, homology being defined below,
  • variable is understood to mean any spontaneous or induced modification of a sequence, in particular by substitution and/or insertion and/or deletion of nucleotides and/or of nucleotide fragments, and/or extension and/or shortening of the sequence at one or both ends; an unnatural variability can result from the genetic engineering techniques used, for example the choice of synthesis primers, degenerate or otherwise, selected for amplifying a nucleic acid; this variability can manifest itself in modifications of any starting sequence, considered as reference, and capable of being expressed by a degree of homology relative to the said reference sequence,
  • homology characterizes the degree of identity of two nucleotide or peptide fragments compared; it is measured by the percentage identity which is determined, in particular, by direct comparison of nucleotide or peptide sequences, relative to reference nucleotide or peptide sequences,
  • this percentage identity has been specifically determined for the nucleotide fragments, clones in particular, dealt with in the present invention, which are homologous to the fragments identified, for the MSRV-1 Virus, by SEQ ID No. 1 to No.
  • any nucleotide fragment is termed equivalent or derived from a reference fragment if it possesses a nucleotide sequence equivalent to the sequence of the reference fragment; according to the above definition, the following in particular are equivalent to a reference nucleotide fragment:
  • polypeptide is understood to mean, in particular, any peptide of at least two amino acids, in particular an oligopeptide or protein, extracted, separated or substantially isolated or. synthesized through human intervention, in particular those obtained by chemical synthesis or by expression in a recombinant organism,
  • polypeptide partially encoded by a nucleotide fragment is understood to mean a polypeptide possessing at least three amino acids encoded by at least nine contiguous monomers lying within the said nucleotide fragment,
  • an amino acid is termed analogous to another amino acid when their respective physicochemical properties, such as polarity, hydrophobicity and/or basicity and/or acidity and/or neutrality are substantially the same; thus, a leucine is analogous to an isoleucine.
  • any polypeptide is termed equivalent or derived from a reference polypeptide if the polypeptides compared have substantially the same properties, and in particular the same antigenic, immunological, enzymological and/or molecular recognition properties; the following in particular are equivalent to a reference polypeptide:
  • any polypeptide into whose sequence a modification of the side chains of the amino acids has been introduced such as, for example, an acetylation of the amine functions, a carboxylation of the thiol functions, an esterification of the carboxyl functions,
  • any polypeptide in whose sequence one or more peptide bonds have been modified such as, for example, carba, retro, inverso, retro-inverso, reduced and methylenoxy bonds,
  • the percentage identity characterizing the homology of two peptide fragments compared is, according to the present invention, at least 50% and preferably at least 70%.
  • MSRV-1 the sequences relating to a virus possessing such reverse transcriptase activity
  • Detection of a substance or agent is understood below to mean both an identification and a quantification, or a separation or isolation, of the said substance or said agent.
  • FIG. 1 shows general consensus sequences of nucleic acids of the MSRV-1B clones amplified by the PCR technique in the “pol” region defined by Shih (12), from viral DNA originating from the LM7PC and PLI-2 lines, and identified under the references SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO: 5 and SEQ ID NO: 6, and the common consensus with amplification primers bearing the reference SEQ ID NO:7;
  • FIG. 2 gives the definition of a functional reading frame for each MSRV-1B/“PCR pol” type family, the said families A to D being defined, respectively, by the nucleotide sequences SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5 and SEQ ID NO:6 described in FIG. 1;
  • FIG. 3 gives an example of consensus of the MSRV-2B sequences, identified by SEQ ID NO:11;
  • FIG. 4 is a representation of the reverse transcriptase (RT) activity in dpm (disintegrations per minute) in the sucrose fractions taken from a purification gradient of the virions produced by the B lymphocytes in culture from a patient suffering from MS;
  • RT reverse transcriptase
  • FIG. 5 gives, under the same experimental conditions as in FIG. 4, the assay of the reverse transcriptase activity in the culture of a B lymphocyte line obtained from a control free from MS;
  • FIG. 6 shows the nucleotide sequence of the clone PSJ17 (SEQ ID NO:9)
  • FIG. 7 shows the nucleotide sequence SEQ ID NO:8 of the clone designated M003-P004;
  • FIG. 8 shows the nucleotide sequence SEQ ID NO:2 of the clone F11-1; the portion located between the two arrows in the region of the primer corresponds to a variability imposed by the choice of primer which was used for the cloning of F11-1; in this same figure, the translation into amino acids is shown;
  • FIG. 9 shows the nucleotide sequence SEQ ID NO:1, and a possible functional reading frame of SEQ ID NO:1 in terms of amino acids; on this sequence, the consensus sequences of the pol gene are underlined;
  • FIGS. 10 and 11 give the results of a PCR, in the form of a photograph under ultraviolet light of an ethidium bromide-impregnated agarose gel, of the amplification products obtained from the primers identified by SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18 and SEQ ID NO:19;
  • FIG. 12 gives a representation in matrix form of the homology between SEQ ID NO:1 of MSRV-1 and that of an endogenous retrovirus designated HSERV9; this homology of at least 65% is demonstrated by a continuous line, the absence of a line meaning a homology of less than 65%;
  • FIG. 13 shows the nucleotide sequence SEQ ID NO:46 of the clone FBd3;
  • FIG. 14 shows the sequence homology between the clone FBd3 and the HSERV-9 retrovirus
  • FIG. 15 shows the nucleotide sequence SEQ ID NO:51 of the clone t pol
  • FIGS. 16 and 17 show, respectively, the nucleotide sequences SEQ ID NO:52 and SEQ ID NO:53 of the clones JLBc1 and JLBc2, respectively;
  • FIG. 18 shows the sequence homology between the clone JLBc1 and the clone FBd3;
  • FIG. 19 the sequence homology between the clone JLBc2 and the clone FBd3;
  • FIG. 20 shows the sequence homology between the clones JLBc1 and JLBc2;
  • FIGS. 21 and 22 show the sequence homology between the HSERV-9 retrovirus and the clones JLBc1 and JLBc2, respectively;
  • FIG. 23 shows the nucleotide sequence SEQ ID NO:56 of the clone GM3;
  • FIG. 24 shows the sequence homology between the HSERV-9 retrovirus and the clone GM3;
  • FIG. 25 shows the localization of the different clones studied, relative to the genome of the known retrovirus ERV9
  • FIG. 26 shows the position of the clones F11-1, M003-P004, MSRV-1B and PSJ17 in the region hereinafter designated MSRV-1 pol*;
  • FIG. 27 split into three successive FIGS. 27 a, 27 b and 27 c , shows a possible reading frame covering the whole of the pol gene
  • FIG. 28 shows, according to SEQ ID NO:40, the nucleotide sequence coding for the peptide fragment POL2B, having the amino acid sequence identified by SEQ ID NO:39;
  • FIG. 29 shows the OD values (ELISA tests) at 492 nm obtained for 29 sera of MS patients and 32 sera of healthy controls tested with an anti-IgG antibody;
  • FIG. 30 shows the OD values (ELISA tests) at 492 nm obtained for 36 sera of MS patients and 42 sera of healthy controls tested with an anti-IgM antibody;
  • the bar at the far right-hand end represents a graphic scale standard unrelated to the serological test;
  • FIG. 34 shows the SEQ ID NO:41 and SEQ ID NO:42 of two polypeptides comprising immunodominant [lacuna], while SEQ ID NO:43 and 44 represent immunoreactive polypeptides specific to MS;
  • FIG. 35 shows the nucleotide sequence SEQ ID NO:59 of the clone LB19 and three potential reading frames of SEQ ID NO:59 in terms of amino acids;
  • FIG. 36 shows the nucleotide sequence SEQ ID NO:88 (GAG*) and a potential reading frame of SEQ ID NO:88 in terms of amino acids;
  • FIG. 37 shows the sequence homology between the clone FBd13 and the HSERV-9 retrovirus; according to this representation, the continuous line means a percentage homology greater than or equal to 70% and the absence of a line means a smaller percentage homology;
  • FIG. 38 shows the nucleotide sequence SEQ ID NO:61 of the clone FP6 and three potential reading frames of SEQ ID NO:61 in terms of amino acids;
  • FIG. 39 shows the nucleotide sequence SEQ ID NO:89 of the clone G+E+A and three potential reading frames of SEQ ID NO:89 in terms of amino acids;
  • FIG. 40 shows a reading frame found in the region E and coding for an MSRV-1 retroviral protease identified by SEQ ID NO:90;
  • FIG. 41 shows the response of each serum of patients suffering from MS, indicated by the symbol. (+), and of healthy patients, symbolised by ( ⁇ ), tested with an anti-IgG antibody, expressed as net optical density at 492 nm;
  • FIG. 42 shows the response of each serum of patients suffering from MS, indicated by the symbols (+) and (QS), and of healthy patients ( ⁇ ), tested with an anti-IgM antibody, expressed as net optical density at 492 nm.
  • a PCR technique derived from the technique published by Shih (12) was used. This technique enables all trace of contaminant DNA to be removed by treating all the components of the reaction medium with DNase. It concomitantly makes it possible, by the use of different but overlapping primers in two successive series of PCR amplification cycles, to increase the chances of amplifying a cDNA synthesized from an amount of RNA which is small at the outset and further reduced in the sample by the spurious action of the DNAse on the RNA.
  • the DNase is used under conditions of activity in excess which enable all trace of contaminant DNA to be removed before inactivation of this enzyme remaining in the sample by heating to 85° C. for 10 minutes.
  • sequences cloned and sequenced from these samples correspond, in particular, to two types of sequence: a first type of sequence, to be found in the majority of the clones (55% of the clones originating from the POL-2 isolates of the PLI-2 culture, and 67% of the clones originating from the MS7PG isolates of the LM7PC cultures), which corresponds to a family of “pol” sequences closely similar to, but different from, the endogenous human retrovirus designated ERV-9 or HSERV-9, and a second type of sequence which corresponds to sequences very strongly homologous to a sequence attributed to another infective and/or pathogenic agent designated MSRV-2.
  • the first type of sequence representing the majority of the clones, consists of sequences whose variability enables four subfamilies of sequences to be defined. These subfamilies are sufficiently similar to one another for it to be possible to consider them to be quasi-species originating from the same retrovirus, as is well known for the HIV-1 retrovirus (14), or to be the outcome of interference with several endogenous proviruses coregulated in the producing cells. These more or less defective endogenous elements are sensitive to the same regulatory signals possibly generated by a replicative provirus, since they belong to the same family of endogenous retroviruses (15).
  • This new family of endogenous retroviruses, or alternatively this new retroviral species from which the generation of quasi-species has been obtained in culture, and which contains a consensus of the sequences described below, is desig nated MSRV-1B.
  • FIG. 1 presents the general consensus sequences of the sequences of the different MSRV-1B clones sequenced in this experiment, these sequences being identified, respectively, by SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5 and SEQ ID NO:6. These sequences display a homology with respect to nucleic acids ranging from 70% to 88% with the HSERV9 sequence referenced X57147 and M37638 in the Genebankr data base.
  • Four “consensus” nucleic acid sequences representative of different quasi-species of a possibly exogenous retrovirus MSRV-1B, or of different subfamilies of an endogenous retrovirus MSRV-1B, have been defined. These representative consensus sequences are presented in FIG.
  • the second type of sequence representing the majority of the clones sequenced is represented by the sequence MSRV-2B presented in FIG. 3 and identified by SEQ ID NO:11. The differences observed in the sequences corresponding to the PCR primers are explained by the use of degenerate primers in mixture form used under different technical conditions.
  • the MSRV-2B sequence (SEQ ID NO:11) is sufficiently divergent from the retroviral sequences already described in the data banks for it to be suggested that the sequence region in question belongs to a new infective agent, designated MSRV-2.
  • This infective agent would, in principle, on the basis of the analysis of the first sequences obtained, be related to a retrovirus but, in view of the technique used for obtaining this sequence, it could also be a DNA virus whose genome codes for an enzyme which incidentally possesses reverse transcriptase activity, as is the case, for example, with the hepatitis B virus, HBV (12).
  • the random nature of the degenerate primers used for this PCR amplification technique may very well have permitted, as a result of unforeseen sequence homologies or of conserved sites in the gene for a related enzyme, the amplification of a nucleic acid originating from a prokaryotic or eukaryotic pathogenic and/or coinfective agent (protist).
  • protist a prokaryotic or eukaryotic pathogenic and/or coinfective agent
  • RNA nucleic acid material present in a purified fraction of virions at the peak of “LM7-like” reverse transcriptase activity on a sucrose gradient according to the technique described by H. Perron (13), and according to the protocols mentioned in Example 1, from a spontaneous lymphoblastoid line obtained by self-immortalization in culture of B lymphocytes from an MS patient who was seropositive for the Epstein-Barr virus (EBV), after setting up the blood lymphoid cells in culture in a suitable culture medium containing a suitable concentration of cyclosporin A.
  • EBV Epstein-Barr virus
  • FIG. 4 A representation of the reverse transcriptase activity in the sucrose fractions taken from a purification gradient of the virions produced by this line is presented in FIG. 4.
  • the culture supernatants of a B line obtained under the same conditions from a control free from MS were treated under the same conditions, and the assay of reverse transcriptase activity in the sucrose gradient fractions proved negative throughout (background), and is presented in FIG. 5.
  • Fraction 3 of the gradient corresponding to the MS B line and the same fraction without reverse transcriptase activity of the non-MS control gradient were analysed by the same RT-PCR technique as before, derived from Shih (12), followed by the same steps of cloning and sequencing as described in Example 1.
  • MSRV-1 and MSRV-2 type sequences are to be found only in the material associated with a peak of “LM7-like” reverse transcriptase activity originating from the MS B lymphoblastoid line. These sequences were not to be found with the material from the control (non-MS) B lymphoblastoid line in 26 recombinant clones taken at random. Only MoMuLV type contaminant sequences, originating from the commercial reverse transcriptase used for the CDNA synthesis step, and sequences without any particular retroviral analogy were to be found in this control, as a result of the “consensus” amplification of homologous polymerase sequences which is produced by this PCR technique. Furthermore, the absence of a concentrated target which competes for the amplification reaction in the control sample permits the amplification of dilute contaminants. The difference in results is manifestly highly significant (chi- squared, p ⁇ 0.001).
  • This approach is directed towards obtaining reverse-transcribed DNA sequences from the supposedly retroviral RNA in the isolate using the reverse transcriptase activity present in this same isolate.
  • This reverse transcriptase activity can theoretically function only in the presence of a retroviral RNA linked to a primer tRNA or hybridized with short strands of DNA already reverse-transcribed in the retroviral particles (16).
  • the obtaining of specific retroviral sequences in a material contaminated with cellular nucleic acids was optimized according to these authors by means of the specific enzymatic amplification of the portions of viral RNAs with a viral reverse transcriptase activity.
  • the molecular approach consisted in using a preparation of concentrated but unpurified virion obtained from the culture supernatants of the PLI-2 line, prepared according to the following method: the culture supernatants are collected twice weekly, precentrifuged at 10,000 rpm for 30 minutes to remove cell debris and then frozen at ⁇ 80° C. or used as they are for the following steps. The fresh or thawed supernatants are centrifuged on a cushion of 30% glycerol-PBS at 100,000 g (or 30,000 rpm in a type 45 T LKB-HITACHI rotor) for 2 h at 4° C.
  • the sedimented pellet After removal of the supernatant, the sedimented pellet is taken up in a small volume of PBS and constitutes the fraction of concentrated but unpurified virion.
  • This concentrated but unpurified viral sample was used to perform a so-called endogenous reverse transcription reaction, as described below.
  • a 5-fold concentrated buffer was prepared with the following components: 500 mM Tris-HCl pH 8.2; 75 mM NaCl; 25 mM MgC12; 75 mM DTT and 0.10% NP 40; 100 ⁇ l of 5 ⁇ buffer+25 ⁇ l of a 100 mM solution of dATP +25 ml of a 100 mM solution of dTTP+25 ml of a 100 ⁇ M solution of dGTP+25 ⁇ l of a 100 mM solution of dCTP +100 ml of sterile distilled water+200 ml of the virion suspension (RT activity of 5 million DPM) in PBS were mixed and incubated at 42° C. for 3 hours.
  • the reaction mixture is added directly to a buffered phenol/- chloroform/isoamyl alcohol mixture (Sigma ref. P 3803); the aqueous phase is collected and one volume of sterile distilled water is added to the organic phase to re-extract the residual nucleic acid material:
  • the collected aqueous phases are combined, and the nucleic acids contained are precipitated by adding 3M sodium acetate pH 5.2 to ⁇ fraction (1/10) ⁇ volume+2 volumes of ethanol+1 ⁇ l of glycogen (Boehringer-Mannheim ref. 901 393) and placing the sample at ⁇ 20° C. for 4 h or overnight at +4° C.
  • blunt-ended DNAs with unpaired adenines at the ends were generated: a “filling-in” reaction was first performed: 25 ⁇ l of the previously purified DNA solution were mixed with 2 ⁇ l of a 2.5 mM solution containing, in equimolar amounts, DATP+dGTP+dTTP+dCTP/1 ⁇ l of T4 DNA polymerase (Boehringer-Mannheim ref.
  • the DNA precipitated after centrifugation is resuspended in 10 ⁇ l of 10 mM Tris buffer pH 7.5. 5 ⁇ l of this suspension were then mixed with 20 ⁇ l of 5 ⁇ Taq buffer, 20 ⁇ l of 5 mM dATP, 1 ⁇ l (5U) of Taq DNA polymerase (AmplitaqTM) and 54 ⁇ l of sterile distilled water. This mixture is incubated for 2 h at 75° C. with a film of oil on the surface of the solution. The DNA suspended in the aqueous solution drawn off under the film of oil after incubation is precipitated as described above and resuspended in 2 ⁇ l of sterile distilled water.
  • the DNA obtained was inserted into a plasmid using the TA CloningTM kit.
  • the 2 ⁇ l of DNA solution were mixed with 5 ⁇ l of sterile distilled water, 1 ⁇ l of a 10-fold concentrated ligation buffer “ 10 ⁇ LIGATION BUFFER”, 2 ⁇ l of “pCRTM VECTOR” (25 ng/ml) and 1 ⁇ l of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology).
  • the white colonies of recombinant bacteria were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17).
  • the plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA cloning kit®.
  • oligonucleotides M001, M002-A, M003-BCD, P004 and P005, were defined in order to amplify the RNA originating from purified POL-2 virions. Control reactions were performed so as to check for the presence of contaminants (reaction with water).
  • the amplification consists of an RT-PCR step according to the protocol described in Example 2, followed by a “nested” PCR according to the PCR protocol described in the document EP-A-0,569,272.
  • the primers M001 and P004 or P005 are used.
  • the primers M002-A or M003-BCD and the primer P004 are used.
  • the primers are positioned as follows: M002-A M003-BCD M001 P004 P005 RNA POL-2 ⁇ -----------> ⁇ --------------------------> pol MSRV-1B PSJ17 primer GGTCITICCICAIGG (SEQ ID NO:20) M001: primer TTAGGGATAGCCCTCATCTCT (SEQ ID NO:21) M002-A: primer TCAGGGATAGCCCCCATCTAT (SEQ ID NO:22) M003-BCD: primer AACCCTTTGCCACTACATCAATTT (SEQ ID NO:23) P004: primer GCGTAAGGACTCCTAGAGCTATT (SEQ ID NO:24) P005:
  • the “nested” amplification product obtained, and designated M003-P004, is presented in FIG. 7, and corresponds to the sequence SEQ ID NO:8.
  • a PCR technique derived from the technique published by Frohman (19) was used.
  • the technique derived makes it possible, using a specific primer at the 3′ end of the genome to be amplified, to elongate the sequence towards the 5′ region of the genome to be analysed.
  • This technical variant is described in the documentation of the firm “Clontech Laboratories Inc.”, (Palo-Alto California, USA) supplied with its product “5′-AmpliFINDERTM RACE Kit”, which was used on a fraction of virion purified as described above.
  • the specific 3′ primers used in the kit protocol for the synthesis of the cDNA and the PCR amplification are, respectively, complementary to the following MSRV-1 sequences: cDNA: TCATCCATGTACCGAAGG (SEQ ID NO:25) amplification: ATGGGGTTCCCAAGTTCCCT (SEQ ID NO:26)
  • the 2 ⁇ l of DNA solution were mixed with 5 ⁇ l of sterile distilled water, 1 ⁇ l of a 10-fold concentrated ligation buffer “10′ LIGATION BUFFER”, 2 ⁇ l of “pCRTM VECTOR” (25 ng/ml) and 1 ⁇ l of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17).
  • the plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detecte under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®.
  • the reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer model 373 A” apparatus according to the manufacturer's instructions.
  • the sedimented pellet is taken up in a small volume of PBS and constitutes the fraction of concentrated but unpurified virions.
  • the concentrated virus is then applied to a sucrose gradient in sterile PBS buffer (15 to 50% weight/weight) and ultracentrifuged at 35,000 rpm (100,000 g) for 12 h at +4° C., in a swing-out rotor. 10 fractions are collected, and 20 ⁇ l are withdrawn from each fraction after homogenization to assay the reverse transcriptase activity therein according to the technique described by H. Perron (3).
  • the fractions containing the peak of “LM7-like” RT activity are then diluted in sterile PBS buffer and ultracentrifuged for one hour at 35,000 rpm (100,000 g) to sediment the viral particles.
  • the pellet of purified virion thereby obtained is then taken up in a small volume of a buffer which is appropriate for the extraction of RNA.
  • the cDNA synthesis reaction mentioned above is carried out on this RNA extracted from purified extracellular virion. PCR amplification according to the technique mentioned above enabled the clone F1-11 to be obtained, whose sequence, identified by SEQ ID NO:2, is presented in FIG. 8.
  • This clone makes it possible to define, with the different clones previously sequenced, a region of considerable length (1.2 kb) representative of the “pol” gene of the MSRV-1 retrovirus, as presented in FIG. 9.
  • This sequence designated SEQ ID NO:1
  • SEQ ID NO:1 is reconstituted from different clones overlapping one another at their ends, correcting the artefacts associated with the primers and with the amplification or cloning techniques which would artificially interrupt the reading frame of the whole.
  • This sequence will be identified below under the designation “MSRV-1 pol* region”. Its degree of homology with the HSERV-9 sequence is shown in FIG. 12.
  • a PCR technique was used to detect the MSRV-1 and MSRV-2 genomes in plasmas obtained after taking blood samples from patients suffering from MS and from non-MS controls onto EDTA.
  • RNAs from plasma were extracted according to the technique described by P. Chomzynski (20), after adding one volume of buffer containing guanidinium thiocyanate to 1 ml of plasma stored frozen at ⁇ 80° C. after collection.
  • MSRV-2 the PCR was performed under the same conditions and with the following primers: 5′ primer, identified by 5′ GTAGTTCGATGTAGAAAGCG 3′; SEQ ID NO:14 3′ primer, identified by 5′ GCATCCGGCAACTGCACG 3′. SEQ ID NO:15
  • the primers used for this first step of 40 cycles with a hybridization temperature of 48° C. are the following:
  • 5′ primer identified by SEQ ID NO:27 5′ GCCGATATCACCCGCCATGG 3′, corresponding to a 5′ MSRV-2 PCR primer, for a first PCR on samples from patients,
  • 3′ primer identified by SEQ ID NO:28 5′ GCATCCGGCAACTGCACG 3′, corresponding to a 3′ MSRV-2 PCR primer, for a first PCR on samples from patients.
  • the primers used for this second step are the 10 following:
  • 5′ primer identified by SEQ ID NO:29 5′ CGCGATGCTGGTTGGAGAGC 3′, corresponding to a 5′ MSRV-2 PCR primer, for a nested PCR on samples from patients,
  • 3′ primer identified by SEQ ID NO:30 5′TCTCCACTCCGAATATTCCG 3′, corresponding to a 3′ MSRV-2 PCR primer, for a nested PCR on samples from patients.
  • MSRV-1 the amplification was performed in two steps. Furthermore, the nucleic acid sample is treated beforehand with DNase, and a control PCR without RT (AMV reverse transcriptase) is performed on the two amplification steps so as to verify that the RT-PCR amplification comes exclusively from the MSRV-1 RNA. In the event of a positive control without RT, the initial aliquot sample of RNA is again treated with DNase and amplified again.
  • AMV reverse transcriptase AMV reverse transcriptase
  • RNAse inhibitor Boehringer-Mannheim
  • RNAse-free DNAse Boehringer-Mannheim
  • pH 5 buffer containing 0.1 M/1 sodium acetate 35 and 5 mM/l MgSO 4 is added; the mixture is incubated for 15 min at 20° C. and brought to 95° C. for 1.5 min in a “thermocycler”.
  • the first MSRV-1 RT-PCR step is performed according to a variant of the RNA amplification method as described in Patent Application No. EP-A-0,569,272.
  • the CDNA synthesis step is performed at 42° C. for one hour; the PCR amplification takes place over 40 cycles, with a primer hybridization (“annealing”) temperature of 53° C.
  • the reaction volume is 100 ⁇ l.
  • the primers used for this first step are the following: 5′ primer, identified by 5′ AGGAGTAAGGAAACCCAACGGAC 3′; SEQ ID NO:16 3′ primer, identified by 5′ TAAGAGTTGCACAAGTGCG 3′. SEQ ID NO:17
  • the primers used for this second step are the following: 5′ primer, identified by 5′ TCAGGGATAGCCCCCATCTAT 3′; SEQ ID NO:18 3′ primer, identified by 5′ AACCCTTTGCCACTACATCAATTT 3′. SEQ ID NO:19
  • FIGS. 10 and 11 present the results of PCR in the form of photographs under ultraviolet light of ethidium bromide-impregnated agarose gels, in which an electrophoresis of the PCR amplification products applied separately to the different wells was performed.
  • FIG. 10 The top photograph (FIG. 10) shows the result of specific MSRV-2 amplification.
  • Well number 8 contains a mixture of DNA molecular weight markers, and wells 1 to 7 represent, in order, the products amplified from the total RNAs of plasmas originating from 4 healthy controls free from MS (wells 1 to 4) and from 3 patients suffering from MS at different stages of the disease (wells 5 to 7).
  • MSRV-2 nucleic acid material is detected in the plasma of one case of MS out of the 3 tested, and in none of the 4 control plasmas. Other results obtained on more extensive series confirm these results.
  • FIG. 11 The bottom photograph (FIG. 11) shows the result of specific amplification by MSRV-1 “nested” RT-PCR:
  • well No. 1 contains the PCR product produced with water alone, without the addition of AMV reverse transcriptase
  • well No. 2 contains the PCR product produced with water alone, with the addition of AMV reverse transcriptase
  • well number 3 contains a mixture of DNA molecular weight markers
  • wells 4 to 13 contain, in order, the products amplified from the total RNAs extracted from sucrose gradient fractions (collected in a downward direction), on which gradient a pellet of virion originating from a supernatant of a culture infected with MSRV-1 and MSRV-2 was centrifuged to equilibrium according to the protocol described by H. Perron (13); to well 14 nothing was applied; to wells 15 to 17, the amplified products of RNA extracted from plasmas originating from 3 different patients suffering from MS at different stages of the disease were applied.
  • the MSRV-1 retroviral genome is indeed to be found in the sucrose gradient fraction containing the peak of reverse transcriptase activity measured according to the technique described by H. Perron (3), with a very strong intensity (fraction 5 of the gradient, placed in well No. 8). A slight amplification has taken place in the first fraction (well No. 4), probably corresponding to RNA released by lysed particles which floated at the surface of the gradient; similarly, aggregated debris has sedimented in the last fraction (tube bottom), carrying with it a few copies of the MSRV-1 genome which have given rise to an amplification of low intensity.
  • MSRV-1 RNA turned up in one case, producing a very intense amplification (well No. 17).
  • the products of the nested PCR described above may be tested in two ELOSA systems enabling a consensus A and a consensus B+C+D of MSRV-1 to be detected separately, corresponding to the subfamilies described.in Example 1 and FIGS. 1 and 2.
  • the sequences closely resembling the consensus B+C+D are to be found essentially in the RNA samples originating from MSRV-1 virions purified from cultures or amplified in extracellular biological fluids of MS patients, whereas the sequences closely resembling the consensus A are essentially to be found in normal human cellular DNA.
  • the ELOSA/MSRV-1 system for the capture and specific hybridization of the PCR products of the subfamily A uses a capture oligonucleotide cpV1A with an amine bond at the 5′ end and a biotinylated detection oligonucleotide dpV1A having as their sequence, respectively:
  • dpV1A identified by SEQ ID NO:32;
  • 5′ CATCTITTTGGICAGGCAITAGC 3′ corresponding to the ELOSA capture oligonucleotide for the subfamily A of the products of MSRV-1 “nested” PCR performed with the primers identified by SEQ ID NO:16 and SEQ ID NO:17, optionally followed by amplification with the primers identified by SEQ ID NO:18 and SEQ ID NO:19 on samples from patients.
  • the ELOSA/MSRV-1 system for the capture and specific hybridization of the PCR products of the subfamily B+C+D uses the same biotinylated detection oligonucleotide dpV1A and a capture oligonucleotide cpV1B with an amine bond at the 5′ end having as its sequence:
  • MSRV-2 For MSRV-2, a similar ELOSA technique was evaluated on isolates originating from infected cell cultures, using the following PCR amplification primers,
  • the 2 ⁇ l of DNA solution were mixed with 5 ⁇ l of sterile distilled water, 1 ⁇ l of a 10-fold concentrated ligation buffer “10 ⁇ LIGATION BUFFER”, 2 ⁇ l of “pCRTM VECTOR” (25 ng/ml) and 1 ⁇ l of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17).
  • the plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®.
  • the reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “automatic sequencer, model 373 A [lacuna] apparatus according to the manufacturer's instructions.
  • the sedimented pellet constitutes the sample of concentrated but unpurified virions.
  • the pellet thereby obtained is then taken up in a small volume of an appropriate buffer for the extraction of RNA.
  • the cDNA synthesis reaction mentioned above is carried out on this RNA extracted from concentrated extracellular virion.
  • RT-PCR amplification according to the technique mentioned above enabled the clone FBd3 to be obtained, whose sequence, identified by SEQ ID NO:46, is presented in FIG. 13.
  • FIG. 14 the sequence homology between the clone FBd3 and the HSERV-9 retrovirus is shown on the matrix chart by a continuous line for any partial homology greater than or equal to 65%. It can be seen that there are homologies in the flanking regions of the clone (with the pol gene at the 5′ end and with the env gene and then the LTR at the 3′ end), but that the internal region is totally divergent and does not display any homology, even weak, with the “env” gene of HSERV9.
  • the clone FBd3 contains a longer “env” region than the one which is described for the defective endogenous HSERV-9; it may thus be seen that the internal divergent region constitutes an “insert” between the regions of partial homology with the HSERV-9 defective genes.
  • oligonucleotides F1, B4, F6 and B1 were defined for amplifying RNA originating from concentrated virions of the strains POL2 and MS7PG. Control reactions were performed so as to check for the presence of contaminants (reaction with water).
  • the amplification consists of a first step of RT-PCR according to the protocol described in Patent Application EP-A-0,569,272, followed by a second step of PCR performed on 10 ml of product of the first step with primers internal to the amplified first region (“nested” PCR). In the first RT-PCR cycle, the primers F1 and B4 are used. In the second PCR cycle, the primers F6 and the primer B1 are used.
  • the primers are positioned as follows: F1 F6 B1 B4 RNA MSRV-1 PSJ17 FBd3 -------> ⁇ ------------------/--- 5′pol MSRV-1 3′pol MSRV-1 / 5′env
  • Their composition is: primer TGATGTGAACGGCATACTCACTG (SEQ ID NO:47)
  • F1 primer CCCAGAGGTTAGGAACTCCCTTTC (SEQ ID N0 48)
  • B4 primer GCTAAAGGAGACTTGTGGTTGTCAG (SEQ ID N0 49)
  • F6 primer CAACATGGGCATTTCGGATTAG (SEQ ID N0 50)
  • a library of cDNA was produced according to the procedure described by the manufacturer of the “cDNA synthesis module, cDNA rapid adaptator ligation module, CDNA rapid cloning module and lambda gt10 in vitro packaging module” kits (Amersham, ref RPN1256Y/Z, RPN1712, RPN1713, RPN1717, N334Z), from the messenger RNA extracted from cells of a B lymphoblastoid line such as is described in Example 2, established from the lymphocytes of a patient suffering from MS and possessing reverse transcriptase activity which is detectable according to the technique described by Perron et al. (3).
  • Oligonucleotides were defined for amplifying the cDNA cloned into the nucleic acid library between the 3′ region of the clone PSJ17 (pol) and the 5′ (LTR) region of the clone FBd3. Control reactions were performed so as to check for the presence of contaminants (reaction with water). PCR reactions performed on the nucleic acids cloned into the library with different pairs of primers enabled a series [lacuna] clones linking pol sequences to the MSRV-1 type env or LTR sequences to be amplified.
  • JLB1, JLB2 and FBd3 comprises, with a few sequence and size variations of the “insert”, the additional sequence absent (“inserted”) in the HSERV-9 env sequence, as described in Example. 8.
  • the cloned “pol” region is very homologous to HSERV-9, does not possess a reading frame (bearing in mind the sequence errors induced by the techniques used, including even the automatic sequencer) and diverges from the MSRV-1 sequences obtained from virions.
  • these sequences were cloned from the RNA of cells expressing MSRV-1 particles, it is probable that they originate from endogenous retroviral elements related to the ERV9 family; this is all the more likely for the fact that the pol and env genes are present on the same RNA which is clearly not the MSRV-1 genomic RNA.
  • Some of these ERV9 elements possess functional LTRs which can be activated by replicative viruses coding for homologous or heterologous transactivators. Under these conditions, the relationship between MSRV-1 and HSERV-9 makes probable the transactivation of the defective (or otherwise) endogenous ERV9 elements by homologous, or even identical, MSRV-1 transactivating proteins.
  • Such a phenomenon may induce a viral interference between the expression of MSRV-1 and the related endogenous elements.
  • Such an interference generally leads to a so-called “defective-interfering” expression, some features of which were to be found in the MSRV-1-infected cultures studied.
  • a phenomenon does not lack generation of the expression of polypeptides, or even of endogenous retroviral proteins which are not necessarily tolerated by the immune system.
  • Such a scheme of aberrant expression of endogenous elements related to MSRV-1 and induced by the latter is liable to multiply the aberrant antigens, and hence to contribute to the induction of autoimmune processes such as are observed in MS.
  • the clones JLBc1 and JLBc2 differ from the ERV9 or HSERV9 sequence already described, in that they possess a longer env region comprising an additional region totally divergent from ERV9. Their kinship with the endogenous ERV9 family may hence be defined, but they clearly constitute novel elements never hitherto described. In effect, interrogation of the data banks of nucleic acid sequences available in version No. 15 (1995) of the “Entrez” software (NCBI, NIH, Bethesda, USA) did not enable a known homologous sequence in the env region of these clones to be identified.
  • MSRV-1 specific primers were defined.
  • the specific 3′ primers used in the kit protocol for the synthesis of the CDNA and the PCR amplification are, respectively, complementary to the following MSRV-1 sequences: cDNA: CCTGAGTTCTTGCACTAACCC (SEQ ID NO:54) amplification: GTCCGTTGGGTTTCCTTACTCCT (SEQ ID NO:55)
  • the 2 ⁇ l of DNA solution were mixed with 5 ⁇ l of sterile distilled water, 1 ⁇ l of a 10-fold concentrated ligation buffer “10 ⁇ LIGATION BUFFER”, 2 ⁇ l of “pCRTM VECTOR” (25 ng/ml) and 1 ⁇ l of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17).
  • the plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®.
  • the reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle seq uencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “automatic sequencer model 373 A [lacuna] apparatus according to the manufacturer's instructions.
  • the sedimented pellet constitutes the sample of concentrated but unpurified virions.
  • the pellet thereby obtained is then taken up in a small volume of an appropriate buffer for the extraction of RNA.
  • the CDNA synthesis reaction mentioned above is carried out on this RNA extracted from concentrated extracellular virion.
  • RT-PCR amplification according to the technique mentioned above enabled the clone GM3 to be obtained, whose sequence, identified by SEQ ID NO 56, is presented in FIG. 23.
  • sequence homology between the clone GMP3 and the HSERV-9 retrovirus is shown on the matrix chart by a continuous line, for any partial homology greater than or equal to 65%.
  • FIG. 25 shows the localization of the different clones studied above, relative to the known ERV9 genome.
  • the additional region is shown above the point of insertion according to a “V”, on the understanding that the inserted material displays a sequence and size vari-ability between the clones shown (JLBc1, JLBc2, FBd3).
  • FIG. 26 shows the position of different clones studied in the MSRV-1 pol* region.
  • the peptides were synthesized on a phenylacet-amidomethyl (PAM)/polystyrene/divinylbenzene resin (Applied Biosystems, Inc. Foster City, CA), using an “Applied Biosystems 430A” automatic synthesizer.
  • the amino acids are coupled in the form of hydroxybenzo-triazole (HOBT) esters.
  • HOBT hydroxybenzo-triazole
  • the chemical synthesis was performed using a double coupling protocol with N-methylpyrrolidone (NMP) as solvent.
  • NMP N-methylpyrrolidone
  • the peptides were cut from the resin, as well as the side-chain protective groups, simultaneously, using hydrofluoric acid (HF) in a suitable apparatus (type I cleavage apparatus, Peptide Institute, Osaka, Japan).
  • HF hydrofluoric acid
  • the peptides are purified by preparative high performance liquid chromatography on a VYDAC C18 type column (250 ⁇ 21 mm) (The Separation Group, Hesperia, CA, USA). Elution is carried out with an acetonitrile gradient at a flow rate of 22 ml/min. The fractions collected are monitored by an elution under isocratic conditions on a VYDAC® C18 analytical column (250 ⁇ 4.6 mm) at a flow rate of 1 ml/min. Fractions having the same retention time are pooled and lyophilized. The preponderant fraction is then analysed by analytical high performance liquid chromatography with the system described above. The peptide which is considered to be of acceptable purity manifests itself in a single peak representing not less than 95% of the chromatogram.
  • the purified peptides are then analysed with the object of monitoring their amino acid composition, using an Applied Biosystems 420H automatic amino acid analyser. Measurement of the (average) chemical molecular mass of the peptides is obtained using LSIMS mass spectrometry in the positive ion mode on a VG. ZAB.ZSEQ double focusing instrument connected to a DEC-VAX 2000 acquisition system (VG analytical Ltd, Manchester, England).
  • the lyophilized POL2B peptide was dissolved in sterile distilled water at a concentration of 1 mg/ml. This stock solution was aliquoted and kept at +4° C. for use over a fortnight, or frozen at ⁇ 20° C. for use within 2 months. An aliquot is diluted in PBS (phosphate buffered saline) solution so as to obtain a final peptide concentration of 1 microgram/ml. 100 microlitres of this dilution are placed in each well of microtitration plates (“high-binding” plastic, COSTAR ref: 3590). The plates are covered with a “plate-sealer” type adhesive and kept overnight at +4° C.
  • PBS phosphate buffered saline
  • test serum samples are diluted beforehand to ⁇ fraction (1/50) ⁇ in the solution B, and 100 microlitres of each dilute test serum are placed in the wells of each microtitration plate.
  • a negative control is placed in one well of each plate, in the form of 100 microlitres of buffer B.
  • the plates covered with an adhesive are then incubated for 1 to 3 hours at 37° C.
  • the plates are then washed three times with the solution A as described above.
  • a peroxidase-labelled goat antibody directed against human IgG Sigma Immunochemicals ref. A6029
  • IgM Cappel ref.
  • the peroxidase substrate is prepared according to the directions of the “Sigma fast OPD kit” (Sigma Immunochemicals, ref. P9187). 100 microlitres of substrate solution are placed in each well, and the plates are placed protected from light for 20 to 30 minutes at room temperature.
  • the plates are placed immediately in an ELISA plate spectrophotometric reader, and the optical density (OD) of each well is read at a wavelength of 492 nm.
  • OD optical density
  • 30 microlitres of 1N HCL are placed in each well to stop the reaction, and the plates are read in the spectrophotometer within 24 hours.
  • the serological samples are introduced in duplicate or in triplicate, and the optical density (OD) corresponding to the serum tested is calculated by taking the mean of the OD values obtained for the same sample at the same dilution.
  • the net OD of each serum corresponds to the mean OD of the serum minus the mean OD of the negative control (solution B: PBS, 0.05% Tween 20®, 10% goat serum).
  • FIG. 29 shows the results for each serum tested with an anti-IgG antibody.
  • Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested.
  • the ordinate axis gives the net OD at the top of the vertical bars.
  • the first 29 vertical bars lying to the left of the vertical broken line represent the sera of 29 cases of MS tested, and the 32 vertical bars lying to the right of the vertical broken line represent the sera of 32 healthy controls (blood donors).
  • the mean of the net OD values for the MS sera tested is 0.62.
  • the diagram enables 5 controls to be revealed whose net OD rises above the grouped values of the control population. These values may represent the presence of specific IgGs in symptomless seropositive patients. Two methods were hence evaluated in order to determine the statistical threshold of positivity of the test.
  • the mean of the net OD values for the controls is 0.36. Without the 5 controls whose net OD values are greater than or equal to 0.5, the mean of the “negative” controls is 0.33. The standard deviation of the negative controls is 0.10. A theoretical threshold of positivity may be calculated according to the formula:
  • threshold value (mean of the net OD values of the seronegative controls)+( 2 or 3 ⁇ standard deviation of the net OD values of the seronegative controls).
  • the test is specific for MS.
  • the test is specific for MS, since, as shown in Table 1, no control has a net OD above this threshold.
  • this result reflects the fact that the antibody titres in patients suffering from MS are, for the most part, higher than in vhealthy controls who have been in contact with MSRV-1. TABLE NO.
  • FIG. 30 shows the results for each serum tested with an anti-IgM antibody.
  • Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested.
  • the ordinate axis gives the net OD at the top of the vertical bars.
  • the first 36 vertical bars lying to the left of the vertical line cutting the abscissa axis represent the sera of 36 cases. of MS tested, and the vertical bars lying to the right of the vertical broken line represent the sera of 42 healthy controls (blood donors).
  • the horizontal line drawn in the middle of the diagram represents a theoretical threshold defining the boundary of the positive results (in which the top of the bar lies above) and the negative results (in which the top of the bar lies below).
  • the standard deviation of the negative controls is 0.05.
  • the threshold of theoretical positivity may be calculated according to the formula:
  • threshold value (mean of the net OD values of the seronegative controls)+(3 ⁇ standard deviation of the net OD values of the seronegative controls).
  • the negative results represent a non-specific “background” of the presence of antibodies directed specifically against an epitope of the peptide.
  • the IgM test is specific for MS, since no control has a net OD above the threshold. 7 of the 36 MS sera produce a positive IgM result; now, a study of the clinical data reveals that these positive sera were taken during a first attack of MS or an acute attack in untreated patients. It is known that IgMs directed against pathogenic agents are produced during primary infections or during reactivations following a latency phase of the said pathogenic agent.
  • the synthesis is carried out with esterified amino acids in which the ⁇ -amino group is protected with an FMOC group (Nova Biochem) and the side-chain groups with protective groups such as trityl, t-butyl ester or t-butyl ether.
  • the esterified amino acids are solubilized in N-methylpyrrolidone (NMP) at a concentration of 300 nM, and 0.9 ⁇ l are applied to spots of deposit of bromophenol blue. After incubation for 15 minutes, a further application of amino acids is carried out according to another 15-minute incubation. If the coupling between two amino acids has taken place correctly, a coloration modification (change from blue to yellow-green) is observed.
  • NMP N-methylpyrrolidone
  • an acetylation step is performed with acetic anhydride.
  • the terminal amino groups of the peptides in the process of synthesis are deprotected with 20% pyridine in DMF.
  • the spots of deposit are restained with a 1% solution of bromophenol blue in DMF, washed three times with methanol and dried. This set of operations constitutes one cycle of addition of an amino acid, and this cycle is repeated until the synthesis is complete.
  • the NH2-terminal group of the last amino acid is deprotected with 20% piperidine in DMF and acetylated with acetic anhydride.
  • the groups protecting the side chain are removed with a dichloromethane/trifluoroacetic acid/triisobutylsilane (5 ml/5 ml/250 ml) mixture.
  • the lmmunoreactivity of the peptides is then tested by ELISA.
  • a ⁇ -galactosidase-labelled anti-human immunoglobulin conjugate (marketed by Cambridge Research Biochemicals) is added at a dilution of ⁇ fraction (1/200) ⁇ , and the mixture is incubated for two hours at room temperature.
  • the immunoreactivity in the different spots is visualized by adding 5-bromo-4-chloro-3-indolyl ⁇ -D-galactopyranoside in potassium.
  • the intensity of coloration of the spots is estimated qualitatively with a relative value from 0 to 5 as shown in the attached FIGS. 31 to 33 .
  • cDNA AAGGGGCATG GACGAGGTGG TGGCTTATTT (SEQ ID NO:65) (antisense)
  • An anchoring PCR was carried out using the following 5′ and 3′ primers: AGATCTGCAG AATTCGATAT CACCCCCCCC CCCCCC (SEQ ID No. 91) (sense), and AAATGTCTGC GGCACCAATC TCCATGTT (SEQ ID No. 64) (antisense)
  • the 2 ⁇ l of DNA solution were mixed with 5 ⁇ l of sterile distilled water, 1 ⁇ l of 10-fold concentrated ligation buffer “10 ⁇ LIGATION BUFFER”, 2 ⁇ l of “pCRTM VECTOR” (25 ng/ml) and 1 ⁇ l of “T4 DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17).
  • the plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®.
  • the reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer, model 373 A” apparatus according to the manufacturer's instructions.
  • PCR amplification according to the technique mentioned above was used on a cDNA synthesized from the nucleic acids of fractions of infective particles purified on a sucrose gradient, according to the technique described by H. Perron (13), from culture supernatants of B lymphocytes of a patient suffering from MS, immortalized with Epstein-Barr virus (EBV) strain B95 and expressing retroviral particles associated with reverse transcriptase activity as described by Perron et al. (3) and in French Patent Applications MS 10, 11 and 12. the clone LB19, whose sequence, identified by SEQ ID NO:59, is presented in FIG. 35.
  • the clone makes it possible to define, with the clone GM3 previously sequenced and the clone G+E+A (see Example 15), a region of 690 base pairs representative of a significant portion of the gag gene of the MSRV-1 retrovirus, as presented in FIG. 36.
  • This sequence designated SEQ ID NO:88 is reconstituted from different clones overlapping at their ends. This sequence is identified under the name MSRV-1 “gag*” region.
  • FIG. 36 a potential reading frame with the translation into amino acids is presented below the nucleic acid sequence.
  • RNAs were extracted according to the method briefly described below.
  • a pool of culture supernatant of B lymphocytes of patients suffering from MS (650 ml) is centrifuged for 30 minutes at 10,000 g.
  • the viral pellet obtained is resuspended in 300 microlitres of PBS/10 mM MgCl2.
  • the material is treated with a DNAse (100 mg/ml)/RNAse (50 mg/ml) mixture for 30 minutes at 37° C. and then with proteinase K (50 mg/ml) for 30 minutes at 46° C.
  • nucleic acids are extracted with one volume of a phenol/0.1% SDS (V/V) mixture heated to 60° C., and then re-extracted with one volume of phenol/chloroform (1:1; V/V).
  • the pellet obtained after centrifugation is resuspended in 50 microlitres of sterile DEPC water.
  • RNA obtained is quantified by an OD reading at 260 nm.
  • the presence of MSRV-1 and the absence of DNA contaminant is monitored by a PCR and an MSRV-1-specific RTPCR associated with a specific ELOSA for the MSRV-1 genome.
  • RNA 5 mg are used to synthesize a cDNA primed with a poly(DT) oligonucleotide according to the instructions of the “cDNA Synthesis Module” kit (ref RPN 1256, Amersham) with a few modifications: The reverse transcription is performed at 45° C. instead of the recommended 42° C.
  • the synthesis product is purified by a double extraction and a double purification according to the manufacturer's instructions.
  • MSRV-1 The presence of MSRV-1 is verified by an MSRV-1 PCR associated with a specific ELOSA for the MSRV-1 genome.
  • LD-PCR reactions are conducted in a Perkin model 9600 PCR apparatus in thin-walled microtubes (Boehringer).
  • the amplification products are monitored by electrophoresis of 1 ⁇ 5th of the amplification volume (10 microlitres) in 1% agarose gel. For the pair of primers described above, a band of approximately 1.7 Kb is obtained.
  • PCR product was purified by passage through a preparative agarose gel and then through a Costar column (Spin; D. Dutcher) according to the supplier's instructions. 2 microlitres of the purified solution are joined up with 50 ng of vector PCRII according to the supplier's instructions (TA Cloning Kit; British Biotechnology)).
  • the recombinant vector obtained is isolated by transformation of competent DH5aF′ bacteria.
  • the molecular structure of the recombinant vector is confirmed by plasmid minipreparation and hydrolysis with the enzyme EcoR1.
  • FBd13 a positive clone for all these criteria, was selected.
  • a large-scale preparation of the recombinant plasmid was performed using the Midiprep Quiagen kit (ref 12243) according to the supplier's instructions.
  • Sequencing of the clone FBd13 is performed by means of the Perkin Prism Ready Amplitaq FS dye terminator kit (ref. 402119) according to the manufacturer's instructiions.
  • the sequence reactions are introduced into a Perkin type 377 or 373A automatic sequencer.
  • the sequencing strategy consists in gene walking carried out on both strands of the clone Fbd13.
  • sequence of the clone FBd13 is identified by SEQ ID NO 58.
  • FIG. 37 the sequence homology between the clone FBd13 and the HSERV-9 retrovirus is shown on the matrix chart by a continuous line for any partial homology greater than or equal to 70%. It can be seen that there are homologies in the flanking regions of the clone (with the pol gene at the 5′ end and with the env gene and then the LTR at the 3′ end) , but that the internal region is totally divergent and does not display any homology, even weak, with the env gene of HSERV-9.
  • the clone FBd13 contains a longer “env” region than the one which is described for the defective endogenous HSERV-9; it may thus be seen that the internal divergent region constitutes an “insert” between the regions of partial homology with the HSERV-9 defective genes.
  • ORF B13 This additional sequence determines a potential orf, designated ORF B13, which is represented by its amino acid sequence SEQ ID NO:87.
  • the protein does not have significant homology with already known sequences.
  • a 3′ RACE was performed on total RNA extracted from plasma of a patient suffering from MS.
  • a healthy control plasma treated under the same conditions was used as negative control.
  • the synthesis of cDNA was carried out with the following modified oligo(dT) primer: 5′ GACTCGCTGC AGATCGATTT TTTTTTTTTT TTTTTT 3′ (SEQ ID NO:68)
  • 5′ primer identified by SEQ ID NO:69 5′ GCCATCAAGC CACCCAAGAA CTCTTAACTT 3′;
  • a second, so-called “semi-nested” PCR was carried out with a 5′ primer located within the region already amplified. This second PCR was performed under the same experimental conditions as those used in the first PCR, using 10 ⁇ l of the amplification product originating from the first PCR.
  • 5′ primer identified by SEQ ID NO:70 5′ CCAATAGCCA GACCATTATA TACACTAATT 3′;
  • Primers SEQ ID NO:69 and SEQ ID NO:70 are specific for the pol* region: position No. 403 to No. 422 and No. 641 to No. 670, respectively.
  • the amplified DNA was inserted into a plasmid using the TA CloningTM kit.
  • the 2 ⁇ l of DNA solution were mixed with 5 ⁇ l of sterile distilled water, 1 ⁇ l of a 10-fold concentrated ligation buffer “10 ⁇ LIGATION BUFFER”, 2 ⁇ l of “pCRTM VECTOR” (25 ng/ml) and 1 ⁇ l of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning kit® (British Biotechnology).
  • the white columns of recombinant bacteria were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17).
  • the plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analyzed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide was selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA cloning kit®.
  • FP6 The clone obtained, designated FP6, enables a region of 467 bp which is 89% homologous to the pol* region of the MSRV-1 retrovirus and a region of 1167 bp which is 64% homologous to the pol region of ERV-9 (No. 1634 to 2856) to be defined.
  • the clone FP6 is represented in FIG. 38 by its nucleotide sequence identified by SEQ ID NO:61.
  • the three potential reading frames of this clone are indicated by their amino acid sequence under the nucleotide sequence.
  • Oligonucleotides specific for the MSRV-1 sequences already identified by the Applicant were defined in order to amplify the retroviral RNA originating from virions present in the plasma of patients suffering from MS. Control reactions were performed so as to monitor the presence of contaminants (reaction with water).
  • the amplification consists of a step of RT-PCR followed by a “nested” PCR. Pairs of primers were defined for amplifying three overlapping regions (designated G, E and A) on the regions defined by the sequences of the clones GM3 and pol* described above.
  • primer 1 SEQ ID NO:71 (sense)
  • primer 2 SEQ ID NO:72 (antisense)
  • primer 1 SEQ ID NO:73 (sense)
  • primer 4 SEQ ID NO:74 (antisense)
  • primer 5 SEQ ID NO:75 (sense)
  • primer 6 SEQ ID NO:76 (antisense)
  • primer 7 SEQ ID NO:77 (sense)
  • primer 8 SEQ ID NO:78 (antisense)
  • primer 9 SEQ ID NO:79 (sense)
  • primer 10 SEQ ID NO:80 (antisense)
  • primer 9 SEQ ID NO:81 (sense)
  • primer 11 SEQ ID NO:82 (antisense)
  • the clones G, E and A were assembled together by PCR with the primers 1 at the 5′ end of the fragment G and 11 at the 3′ end of the fragment A, the primers being described above.
  • An approximately 1580-bp fragment G+E+A was amplified and inserted into a plasmid using the TA Cloning (trademark) kit.
  • the sequence of the amplification product corresponding to G+E+A was determined and analysis of the G+E and E+A overlaps was carried out. The sequence is shown in FIG. 39, and corresponds to the sequence SEQ ID NO:89.
  • a reading frame coding for an MSRV-1 retroviral protease was found in the region E.
  • the amino acid sequence of the protease, identified by SEQ ID NO:90, is presented in FIG. 40.
  • a nested PCR was performed on the DNA extracted from a lymphoblastoid line (B lymphocytes immortalized with the EBV virus strain B95, as described above and as is well known to a person skilled in the art) expressing the MSRV-1 retrovirus and originating from peripheral blood lymphocytes of a patient suffering from MS.
  • a lymphoblastoid line B lymphocytes immortalized with the EBV virus strain B95, as described above and as is well known to a person skilled in the art
  • MSRV-1 retrovirus originating from peripheral blood lymphocytes of a patient suffering from MS.
  • primer CTCGATTTCT TGCTGGGCCT TA (SEQ ID NO:83) 4327: primer GTTGATTCCC TCCTCAAGCA (SEQ ID NO:84) 3512:
  • This step comprises 35 amplification cycles with the following conditions: 1 min at 94° C., 1 min at 54° C. and 4 min at 72° C.
  • primer 4294 CTCTACCAAT CAGCATGTGG (SEQ ID NO:85)
  • primer 3591 TGTTCCTCTT GGTCCCTAT (SEQ ID NO:86)
  • This step comprises 35 amplification cycles with the following conditions: 1 min at 94° C., 1 min at 54° C. and 4 min at 72° C.
  • the 2 ⁇ l of DNA solution were mixed with 5 ⁇ l of sterile distilled water, 1 ⁇ l of a 10-fold concentrated ligation buffer “10 ⁇ LIGATION BUFFER”, 2 ⁇ l of “pCRTM VECTOR” (25 ng/ml) and 1 ⁇ l of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17).
  • the plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analyzed on agarose gel.
  • the plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization witn a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®.
  • the reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle. sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer, model 373 A” apparatus according to the manufacturer's instructions.
  • LTRGAG12 a clone designated LTRGAG12 could be obtained, and is represented by its internal sequence identified by SEQ ID NO:60.
  • This clone is probably representative of endogenous elements close to ERV-9, present in human DNA, in particular in the DNA of patients suffering from MS, and capable of interfering with the expression of the MSRV-1 retrovirus, hence capable of having a role in the pathogenesis associated with the MSRV-1 retrovirus and capable of serving as marker for a specific expression in the pathology in question.
  • the peptides were synthesized on a phenylacetamidomethyl (PAM)/polystyrene/divinylbenzene resin (Applied Biosystems, Inc. Foster City, CA), using an “Applied Biosystems 430A” automatic synthesizer.
  • the amino acids are coupled in the form of hydroxybenzotriazole (HOBT) esters.
  • the amino acids used are obtained from Novabiochem (Läuflerlfingen, Switzerland) or Bachem (Bubendorf, Switzerland).
  • the peptides are purified by preparative high performance liquid chromatography on a VYDAC C18 type column (250 ⁇ 21 mm) (The Separation Group, Hesperia, CA, USA). Elution is carried out with an acetonitrile gradient at a flow rate of 22 ml/min. The fractions collected are monitored by an elution under isocratic conditions on a VYDAC® C18 analytical column (250 ⁇ 4.6 mm) at a flow rate of 1 ml/min. Fractions having the same retention time are pooled and lyophilized. The preponderant fraction is then analysed by analytical high performance liquid chromatography with the system described above. The peptide which is considered to be of acceptable purity manifests itself in a single peak representing not less than 95% of the chromatogram.
  • the purified peptides are then analysed with the object of monitoring their amino acid composition, using an Applied Biosystems 420H automatic amino acid analyser. Measurement of the (average) chemical molecular mass of the peptides is obtained using LSIMS mass spectrometry in the positive ion mode on a VG. ZAB.ZSEQ double focusing instrument connected to a DEC-VAX 2000 acquisition system (VG analytical Ltd, Manchester, England).
  • the lyophilized S24Q peptide was dissolved in 10 % acetic acid at a concentration of 1 mg/ml. This stock solution was aliquoted and kept at +4° C. for use over a fortnight, or frozen at ⁇ 20° C. for use within 2 months. An aliquot is diluted in PBS (phosphate buffered saline) solution so as to obtain a final peptide concentration of 5 micrograms/ml. 100 microlitres of this dilution are placed in each well of Nunc Maxisorb (trade name) microtitration plates. The plates are covered with a “plate-sealer” type adhesive and kept for 2 hours at +37° C.
  • PBS phosphate buffered saline
  • test serum samples are diluted beforehand to ⁇ fraction (1/100) ⁇ in the solution B, and 100 microlitres of each dilute test serum are placed in the wells of each microtitration plate.
  • a negative control is placed in one well of each plate, in the form of 100 microlitres of buffer B.
  • the plates covered with an adhesive are then incubated for 1 hour 30 min at 37° C.
  • the plates are then washed three times with the solution A as described above.
  • a peroxidase-labelled goat antibody directed against human IgG (marketed by Jackson Immuno Research Inc.) is diluted in the solution B (dilution ⁇ fraction (1/10,000) ⁇ ).
  • the peroxidase substrate is prepared according to the directions of the BioMérieux kits. 100 microlitres of substrate solution are placed in each well, and the plates are placed protected from light for 20 to 30 minutes at room temperature.
  • the serological samples are introduced in duplicate or in triplicate, and the optical density (OD) corresponding to the serum tested is calculated by taking the mean of the OD values obtained for the same sample at the same dilution.
  • the net OD of each serum corresponds to the mean OD of the serum minus the mean OD of the negative control (solution B: PBS, 0.05% Tween 20®, 10% goat serum).
  • FIG. 41 shows the results for each serum tested with an anti-IgG antibody.
  • Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested.
  • the ordinate axis gives the net OD at the top of the vertical bars.
  • the first 15 vertical bars lying to the left of the vertical broken line represent the sera of 15 healthy controls (blood donors), and the 15 vertical bars lying to the right of the vertical broken line represent the sera of 15 cases of MS tested.
  • the diagram enables 2 controls to be revealed whose OD rises above the grouped values of the control population. These values may represent the presence of specific IgGs in symptomless seropositive patients. Two methods were hence evaluated in order to determine the statistical threshold of positivity of the test.
  • the mean of the net OD values for the controls is 0.129 and the standard deviation is 0.06. Without the 2 controls whose OD values are greater than 0.2, the mean of the “negative” controls is 0.107 and the standard deviation is 0.03.
  • a theoretical threshold of positivity may be calculated according to the formula:
  • threshold value (mean of the net OD values of the negative controls)+(2 or 3 ⁇ standard deviation of the net OD values of the negative controls).
  • the test is specific for MS.
  • the test is specific for MS, since, as shown in Table 1, no control has a net OD above this threshold.
  • this result reflects the fact that the antibody titres in patients suffering from MS are, for the most part, higher than in healthy controls who have been in contact with MSRV-1.
  • MSRV-1 retrovirus constitutes all or part of this “pathogenic agent” at the source of MS, and it is hence normal for controls taken from a healthy population to possess IgG type antibodies against components of the MSRV-1 retrovirus.
  • FIG. 42 shows the results for each serum tested with an anti-IgM antibody.
  • Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested.
  • the ordinate axis gives the net OD at the top of the vertical bars.
  • the first 15 vertical bars lying to the left of the vertical line cutting the abscissa axis represent the sera of 15 healthy controls (blood donors), and the vertical bars lying to the right of the vertical broken line represent the sera of 15 cases of MS tested.
  • the standard deviation of the negative controls is 0.6.
  • the threshold of theoretical positivity may be calculated according to the formula:
  • threshold value (mean of the OD values of the negative controls)+(3 ⁇ standard deviation of the OD values of the negative controls).
  • the negative results represent a non-specific “background” of the presence of antibodies directed specifically against an epitope of the peptide.
  • the IgM test is specific for MS, since no control has a net OD above the threshold. 6 of the 15 MS sera produce a positive IgM result

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Rheumatology (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Neurology (AREA)
  • Pain & Pain Management (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Communicable Diseases (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)

Abstract

Viral material, in the isolated or purified state, in which the genome comprises a nucleotide sequence chosen from the group including sequences SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:56, their complementary sequences and their equivalent sequences, in particular nucleotide sequences displaying, for any succession of 100 contiguous monomers, at least 50% and preferably at least 70% homology with the said sequences SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53 and SEQ ID NO:56, respectively, and their complementary sequences.

Description

  • Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS) the cause of which remains as yet unknown. [0001]
  • Many studies have supported the hypothesis of a viral aetiology of the disease, but none of the known viruses tested has proved to be the causal agent sought: a review of the viruses sought for several years in MS has been compiled by E. Norrby (1) and R. T. Johnson (2). [0002]
  • Recently, a retrovirus different from the known human retroviruses has been isolated in patients suffering from MS (3, 4, and 5). The authors were also able to show that this retrovirus could be transmitted in vitro, that patients suffering from MS produced antibodies capable of recognizing proteins associated with the infection of leptomeningeal cells by this retrovirus, and that the expression of the latter could be strongly stimulated by the immediate-early genes of some herpes-viruses (6). [0003]
  • All these results point to the role in MS of at least one unknown retrovirus or of a virus having reverse transcriptase activity which is detectable according to the method published by H. Perron (3) and qualified as “LM7-like RT” activity. The content of the publication identified by (3) is incorporated in the present description by reference. [0004]
  • Recently, the Applicant's studies have enabled two continuous cell lines infected with natural isolates originating from two different patients suffering from MS to be obtained by a culture method as described in the document WO-A-93/20188, the content of which is incorporated in the present description by reference. These two lines, derived from human choroid plexus cells, designated LM7PC and PLI-2, were deposited with the ECACC on Jul. 22, 1992 and Jan. 8, 1993, respectively, under numbers 92072201 and 93010817, in accordance with the provisions of the Budapest Treaty. Moreover, the viral isolates possessing LM7-like RT activity were also deposited with the ECACC under the overall designation of “strains”. The “strain” or isolate harboured by the PLI-2 line, designated POL-2, was deposited with the ECACC on Jul. 22, 1992 under No. V92072202. The “strain” or isolate harboured by the LM7PC line, designated MS7PG, was deposited with the ECACC on Jan. 8, 1993 under No. V93010816. [0005]
  • Starting from the cultures and isolates mentioned above, characterized by biological and morphological criteria, the next step was to endeavour to characterize the nucleic acid material associated with the viral particles produced in these cultures. [0006]
  • The portions of the genome which have already been characterized have been used to develop tests for molecular detection of the viral genome and immunoserological tests, using the amino acid sequences encoded by the nucleotide sequences of the viral genome, in order to detect the immune response directed against epitopes associated with the infection and/or viral expression. [0007]
  • These tools have already enabled an association to be confirmed between MS and the expression of the sequences identified in the patents cited later. However, the viral system discovered by the Applicant is related to a complex retroviral system. In effect, the sequences to be found encapsidated in the extracellular viral particles produced by the different cultures of cells of patients suffering from MS show clearly that there is coencapsidation of retroviral genomes which are related but different from the “wild-type” retroviral genome which produces the infective viral particles. This phenomenon has been observed between replicative retroviruses and endogenous retroviruses belonging to the same family, or even heterologous retroviruses. The notion of endogenous retroviruses is very important in the context of our discovery since, in the case of MSRV-1, it has been observed that endogenous retroviral sequences comprising sequences homologous to the MSRV-1 genome exist in normal human DNA. The existence of endogenous retroviral elements (ERV) related to MSRV-1 by all or part of their genome explains the fact that the expression of the MSRV-1 retrovirus in human cells is able to interact with closely related endogenous sequences. These interactions are to be found in the case of pathogenic and/or infectious endogenous retroviruses (for example some ecotropic strains of the murine leukaemia virus), and in the case of exogenous retroviruses whose nucleotide sequence may be found partially or wholly, in the form of ERVS, in the host animal's genome (e.g. mouse exogenous mammary tumor virus transmitted via the milk). These interactions consist mainly of (i) a trans-activation or coactivation of ERVs by the replicative retrovirus (ii) and “illegitimate” encapsidation of RNAs related to ERVS, or of ERVs—or even of cellular RNAs—simply possessing compatible encapsidation sequences, in the retroviral particles produced by the expression of the replicative strain, which are sometimes transmissible and sometimes with a pathogenicity of their own, and (iii) more or less substantial recombinations between the coencapsidated genomes, in particular in the phases of reverse transcription, which lead to the formation of hybrid genomes, which are sometimes transmissible and sometimes with a pathogenicity of their own. [0008]
  • Thus, (i) different sequences related to MSRV-1 have been found in the purified viral particles; (ii) molecular analysis of the different regions of the MSRV-1 retroviral genome should be carried out by systematically analyzing the coencapsidated, interfering and/or recombined sequences which are generated by the infection and/or expression of MSRV-1; furthermore, some clones may have defective sequence portions produced by the retroviral replication and template errors and/or errors of transcription of the reverse transcriptase; (iii) the families of sequences related to the same retroviral genomic region provide the means for an overall diagnostic detection which may be optimized by the identification of invariable regions among the clones expressed, and by the identification of reading frames responsible for the production. of antigenic and/or pathogenic polypeptides which may be produced only by a portion, or even by just one, of the clones expressed, and, under these conditions, the systematic analysis of the clones expressed in the region of a given gene enables the frequency of variation and/or of recombination of the MSRV-1 genome in this region to be evaluated and the optimal sequences for the applications, in particular diagnostic applications, to be defined; (iv) the pathology caused by a retrovirus such as MSRV-1 may be a direct effect of its expression and of the proteins or peptides produced as a result thereof, but also an effect of the activation, the encapsidation or the recombination of related or heterologous genomes and of the proteins or peptides produced as a result thereof; thus, these genomes associated with the expression of and/or infection by MSRV-1 are an integral part of the potential pathogenicity of this virus, and hence constitute means of diagnostic detection and special therapeutic targets. Similarly, any agent associated with or cofactor of these interactions responsible for the pathogenesis in question, such as MSRV-2 or the glyotoxic factor which are described in the patent application published under No. FR-2,716,198, may participate in the development of an overall and very effective strategy for the diagnosis, prognosis, therapeutic monitoring and/or integrated therapy of MS in particular, but also of any other disease associated with the same agents. [0009]
  • In this context, a parallel discovery has been made in another autoimmune disease, rheumatoid arthritis (RA), which has been described in the French Patent Application filed under No. 95/02960. This discovery shows that, by applying methodological approaches similar to the ones which were used in the Applicant's work on MS, it was possible to identify a retrovirus expressed in RA which shares the sequences described for MSRV-1 in MS, and also the coexistence of an associated MSRV-2 sequence also described in MS. As regards MSRV-1, the sequences detected in common in MS and RA relate to the pol and gag genes. In the current state of knowledge, it is possible to associate the gag and pol sequences described with the MSRV-1 strains expressed in these two diseases. [0010]
  • The present patent application relates to various results which are additional to those already protected by the following French Patent Applications:[0011]
  • No. 92/04322 of 03.04.1992, published under No. 2,689,519; [0012]
  • No. 92/13447 of 03.11.1992, published under No. 2,689,521; [0013]
  • No. 92/13443 of 03.11.1992, published under No. 2,689,520; [0014]
  • No. 94/01529 of 04.02.1994, published under No. 2,715,936; [0015]
  • No. 94/01531 of 04.02.1994, published under No. 2,715,939; [0016]
  • No. 94/01530 of 04.02.1994, published under No. 2,715,936; [0017]
  • No. 94/01532 of 04.02.1994, published under No. 2,715,937; [0018]
  • No. 94/14322 of 24.11.1994, published under No. 2,727,428; [0019]
  • and No. 94/15810 of 23.12.1994; published under No. 2,728,585. [0020]
  • The present invention relates, in the first place, to a viral material, in the isolated or purified state, which may be recognized or characterized in different ways: [0021]
  • its genome comprises a nucleotide sequence chosen from the group including the sequences SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:89, their complementary sequences and their equivalent sequences, in particular nucleotide sequences displaying, for any succession of 100 contiguous monomers, at least 50% and preferably at least 70% homology with the said sequences SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60 SEQ ID NO:61, SEQ ID NO:89, respectively, and their complementary sequences; [0022]
  • the region of its genome comprising the env and pol genes and a portion of the gag gene, excluding the subregion having a sequence identical or equivalent to SEQ ID NO:1, codes for any polypeptide displaying, for any contiguous succession of at least 30 amino acids, at least 50% and preferably at least 70% homology with a peptide sequence encoded by any nucleotide sequence chosen from the group including SEQ ID NO:46, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO:53, SEQ ID NO:56, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60 SEQ ID NO:61 SEQ ID NO:89 and their complementary sequences; [0023]
  • the pol gene comprises a nucleotide sequence partially or totally identical or equivalent to SEQ ID NO:57, excluding SEQ ID NO:1. [0024]
  • the gag gene comprises a nucleotide sequence partially or totally identical or equivalent to SEQ ID NO:88. [0025]
  • As indicated above, according to the present invention, the viral material as defined above is associated with MS. And as defined by reference to the pol or gag gene of MSRV-1, and more especially to the sequences [0026] SEQ ID NOS 51, 56, 57, 59, 60, 61, 88 and 89, this viral material is associated with RA.
  • The present invention also relates to different nucleotide fragments each comprising a nucleotide sequence chosen from the group including: [0027]
  • (a) all the genomic sequences, partial and total, of the pol gene of the MSRV-1 virus, except for the total sequence of the nucleotide fragment defined by SEQ ID NO:1; [0028]
  • (b) all the genomic sequences, partial and total, of the env gene of MSRV-1; [0029]
  • (c) all the partial genomic sequences of the gag gene of MSRV-1; [0030]
  • (d) all the genomic sequences overlapping the pol gene and the env gene of the MSRV-1 virus, and overlapping the pol gene and the gag gene; [0031]
  • (e) all the sequences, partial and total, of a clone chosen from the group including the clones FBd3 (SEQ ID NO:46), t pol (SEQ ID NO:51), JLBc1 (SEQ ID NO:52), JLBc2 (SEQ ID NO:53) and GM3 (SEQ ID NO:56), FBd13 (SEQ ID NO:58), LB19 (SEQ ID NO:59), LTRGAG12 (SEQ ID NO:60), FP6 (SEQ ID NO:61), G+E+A (SEQ ID [0032]
  • NO:89), excluding any nucleotide sequence identical to or lying within the sequence defined by SEQ ID NO:1; [0033]
  • (f) sequences complementary to the said genomic sequences; [0034]
  • (g) sequences equivalent to the said sequences (a) to (e), in particular nucleotide sequences displaying, for any succession of 100 contiguous monomers, at least 50% and preferably at least 70% homology with the said sequences (a) to (d). provided that this nucleotide fragment does not comprise or consist of the sequence ERV-9 as described in LA MANTIA et al. (18). [0035]
  • The term genomic sequences, partial or total, includes all sequences associated by coencapsidation or by coexpression, or recombined sequences. [0036]
  • Preferably, such a fragment comprises: [0037]
  • either a nucleotide sequence identical to a partial or total genomic sequence of the pol gene of the MSRV-1 virus, except for the total sequence of the nucleotide fragment defined by SEQ ID NO:1, or identical to any sequence equivalent to the said partial or total genomic sequence, in particular one which is homologous to the latter; [0038]
  • or a nucleotide sequence identical to a partial or total genomic sequence of the env gene of the MSRV-1 virus, or identical to any sequence complementary to the said nucleotide sequence, or identical to any sequence equivalent to the said nucleotide sequence, in particular one which is homologous to the latter. [0039]
  • In particular, the invention relates to a nucleotide fragment comprising a coding nucleotide sequence which is partially or totally identical to a nucleotide sequence chosen from the group including: [0040]
  • the nucleotide sequence defined by SEQ ID NO:40, SEQ ID NO:62 or SEQ ID NO:89; [0041]
  • sequences complementary to SEQ ID NO:40, SEQ ID NO:62 or SEQ ID NO:89; [0042]
  • sequences equivalent, and in particular homologous to SEQ ID NO:40, SEQ ID NO:62 or SEQ ID NO:89; [0043]
  • sequences coding for all or part of the peptide sequence defined by SEQ ID NO:39, SEQ ID NO:63 or SEQ ID NO:90; [0044]
  • sequences coding for all or part of a peptide sequence equivalent, in particular homologous to SEQ ID NO:39, SEQ ID NO:63 or SEQ ID NO:90, which is capable of being recognized by sera of patients infected with the MSRV-1 virus, or in whom the MSRV-1 virus has been reactivated. [0045]
  • The invention also relates to any nucleic acid probe for detection of a pathogenic and/or infective agent associated with MS, which is capable of hybridizing specifically with any fragment such as is defined above, belonging or lying within the genome of the said pathogenic agent. It relates, in addition, to any nucleic acid probe for detection of a pathogenic and/or infective agent associated with RA, which is capable of hybridizing specifically with any fragment as defined above by reference to the pol and gag genes, and especially with respect to the sequences [0046] SEQ ID NOS 40, 51, 56, 59, 60, 61, 62, 89 and SEQ ID NOS 39, 63 and 90.
  • The invention also relates to a primer for the amplification by polymerization of an RNA or a DNA of a viral material, comprising a nucleotide sequence identical or equivalent to at least one portion of the nucleotide sequence of any fragment such as is defined above, in particular a nucleotide sequence displaying, for any succession of 10 contiguous monomers, at least 70% homology with at least the said portion of the said fragment. Preferably, the nucleotide sequence of such a primer is identical to any one of the sequences chosen from the group including SEQ ID NO:47 to SEQ ID NO:50, SEQ ID NO:55 and SEQ ID NO:64 SEQ ID NO:86. [0047]
  • Generally speaking the invention also encompasses any RNA or DNA, and in particular replication vector, comprising a genomic fragment of the viral material such as is defined above, or a nucleotide fragment such as is defined above. [0048]
  • The invention also relates to the different peptides encoded by any open reading frame belonging to a nucleotide fragment such as is defined above, in particular any polypeptide, for example any oligopeptide forming or comprising an antigenic determinant recognized by sera of patients infected with the MSRV-1 virus and/or in whom the MSRV-1 virus has been reactivated. Preferably, this polypeptide is antigenic, and is encoded by the open reading frame beginning, in the 5′-3′ direction, at nucleotide 181 and ending at [0049] nucleotide 330 of SEQ ID NO:1.
  • In particular, the invention relates to an antigenic polypeptide recognized by the sera of patients infected with the MSRV-1 virus, and/or in whom the MSRV-1 virus has been reactivated, whose peptide sequence is partially or totally identical or is equivalent to the sequence defined by SEQ ID NO:39, SEQ ID NO:63 and SEQ ID NO:87; such a sequence is identical, for example, to any sequence chosen from the group including the sequences SEQ ID NO:41 to SEQ ID NO:44, SEQ ID NO:63 and SEQ ID NO:87. [0050]
  • The present invention also proposes mono- or polyclonal antibodies directed against the MSRV-1 virus, which are obtained by the immunological reaction of a human or animal body to an immunogenic agent consisting of an antigenic polypeptide such as is defined above. [0051]
  • The invention next relates to: [0052]
  • reagents for detection of the MSRV- virus, or of an exposure to the latter, comprising, as reactive substance, a peptide, in particular an antigenic peptide, such as is defined above, or an anti-ligand, in particular an antibody to the said peptide; [0053]
  • all diagnostic, prophylactic or therapeutic compositions comprising one or more peptides, in particular antigenic peptides, such as are defined above, or one or more anti-ligands, in particular antibodies to the peptides, discussed above; such a composition is preferably, and by way of example, a vaccine composition. [0054]
  • The invention also relates to any diagnostic, prophylactic or therapeutic composition, in particular for inhibiting the expression of at least one pathogenic and/or infective agent associated with MS comprising a nucleotide fragment such as is defined above or a polynucleotide, in particular oligonucleotide, whose sequence is partially identical to that of the said fragment, except for that of the fragment having the nucleotide sequence SEQ ID NO:1. Likewise, it relates to any diagnostic, prophylactic or therapeutic composition, in particular for inhibiting the expression of at least one pathogenic and/or infective agent associated with RA, comprising a nucleotide fragment such as is defined above by reference to the pol and gag genes, and especially with respect to the sequences [0055] SEQ ID NOS 40, 51, 56, 59, 60, 61, 62 and 89.
  • According to the invention, these same fragments or polynucleotides, in particular oligonucleotides, may participate in all suitable compositions for detecting, according to any suitable process or method, a pathological and/or infective agent associated with MS and with RA, respectively, in a biological sample. In such a process, an RNA and/or a DNA presumed to belong or originating from the said pathological and/or infective agent, and/or their complementary RNA and/or DNA, is/are brought into contact with such a composition. [0056]
  • The present invention also relates to any process for detecting the presence or exposure to such a pathological and/or infective agent, in a biological sample, by bringing this sample into contact with a peptide, in particular an antigenic peptide such as is defined above, or an anti-ligand, in particular an antibody to this peptide, such as is defined above. [0057]
  • In practice, and for example, a device for detection of the MSRV-1 virus comprises a reagent such as is defined above, supported by a solid support which is immunologically compatible with the reagent, and a means for bringing the biological sample, for example a sample of blood or of cerebrospinal fluid, likely to contain anti-MSRV-1 antibodies, into contact with this reagent under conditions permitting a possible immunological reaction, the foregoing items being accompanied by means for detecting the immune complex formed with this reagent. [0058]
  • Lastly, the invention also relates to the detection of anti-MSRV-1 antibodies in a biological sample, for example a sample of blood or of cerebrospinal fluid, according to which this sample is brought into contact with a reagent such as is defined above, consisting of an antibody, under conditions permitting their possible immunological reaction, and the presence of the immune complex thereby formed with the reagent is then detected. [0059]
  • Before describing the invention in detail, different terms used in the description and the claims are now defined: [0060]
  • strain or isolate is understood to mean any infective and/or pathogenic biological fraction containing, for example, viruses and/or bacteria and/or parasites, generating pathogenic and/or antigenic power, harboured by a culture or a living host; as an example, a viral strain according to the above definition can contain a coinfective agent, for example a pathogenic protist, [0061]
  • the term “MSRV” used in the present description denotes any pathogenic and/or infective agent associated with MS, in particular a viral species, the attenuated strains of the said viral species or the defective-interfering particles or particles containing coencapsidated genomes, or alternatively genomes recombined with a portion of the MSRV-1 genome, derived from this species. Viruses, and especially viruses containing RNA, are known to have a variability resulting, in particular, from relatively high rates of spontaneous mutation (7), which will be borne in mind below for defining the notion of equivalence, [0062]
  • human virus is understood to mean a virus capable of infecting, or of being harboured by human beings, [0063]
  • in view of all the natural or induced variations and/or recombination which may be encountered when implementing the present invention, the subjects of the latter, defined above and in the claims, have been expressed including the equivalents or derivatives of the different biological materials defined below, in particular of the homologous nucleotide or peptide sequences, [0064]
  • the variant of a virus or of a pathogenic and/or infective agent according to the invention comprises at least one antigen recognized by at least one antibody directed against at least one corresponding antigen of the said virus and/or said pathogenic and/or infective agent, and/or a genome any part of which is detected by at least one hybridization probe and/or at least one nucleotide amplification primer specific for the said virus and/or pathogenic and/or infective agent, such as, for example, for the MSRV-1 virus, the primers and probes having a nucleotide sequence chosen from SEQ ID No. 20 to SEQ ID No. 24, SEQ ID No. 26, SEQ ID No. 16 to SEQ ID No. 19, SEQ ID No. 31 to SEQ ID No. 33, SEQ ID No. 45, SEQ ID No. 47, SEQ ID No. 48, SEQ ID No. 49, SEQ ID No. 50, SEQ ID No. 45 and their complementary sequences, under particular hybridization conditions well known to a person skilled in the art, [0065]
  • according to the invention, a nucleotide fragment or an oligonucleotide or polynucleotide is an arrangement of monomers, or a biopolymer, characterized by the informational sequence of the natural nucleic acids, which is capable of hybridizing with any other nucleotide fragment under predetermined conditions, it being possible for the arrangement to contain monomers of different chemical structures and to be obtained from a molecule of natural nucleic acid and/or by genetic recombination and/or by chemical synthesis; a nucleotide fragment may be identical to a genomic fragment of the MSRV-1 virus discussed in the present invention, in particular a gene of this virus, for example pol or env in the case of the said virus, [0066]
  • thus, a monomer can be a natural nucleotide of nucleic acid whose constituent elements are a sugar, a phosphate group and a nitrogenous base; in RNA the sugar is ribose, in DNA the sugar is 2-deoxyribose; depending on whether the nucleic acid is DNA or RNA, the nitrogenous base is chosen from adenine, guanine, uracil, cytosine and thymine; or the nucleotide can be modified in at least one of the three constituent elements; as an example, the modification can occur in the bases, generating modified bases such as inosine, 5-methyldeoxy- cytidine, deoxyuridine, 5-(dimethylamino)deoxyuridine, 2,6-diaminopurine, 5-bromodeoxyuridine and any other modified base promoting hybridization; in the sugar, the modification can consist of the replacement of at least one deoxyribose by a polyamide (8), and in the phosphate group, the modification can consist of its replacement by esters chosen, in particular, from diphosphate, alkyl- and arylphosphonate and phosphorothioate esters, [0067]
  • “informational sequence” is understood to mean any ordered succession of monomers whose chemical nature and order in a reference direction constitute or otherwise an item of functional information of the same quality as that of the natural nucleic acids, [0068]
  • hybridization is understood to mean the process during which, under suitable working conditions, two nucleotide fragments having sufficiently complementary sequences pair to form a complex structure, in particular double or triple, preferably in the form of a helix, [0069]
  • a probe comprises a nucleotide fragment synthesized chemically or obtained by digestion or enzymatic cleavage of a longer nucleotide fragment, comprising at least six monomers, advantageously from 10 to 100 monomers and preferably 10 to 30 monomers, and possessing a specificity of hybridization under particular conditions; preferably, a probe possessing fewer than 10 monomers is not used alone, but is used in the presence of other probes of equally short size or otherwise; under certain special conditions, it may be useful to use probes of size greater than 100 monomers; a probe may be used, in particular, for diagnostic purposes, such molecules being, for example, capture and/or detection probes, [0070]
  • the capture probe may be immobilized on a solid support by any suitable means, that is to say directly or indirectly, for example by covalent bonding or passive adsorption, [0071]
  • the detection probe may be labelled by means of a label chosen, in particular, from radioactive isotopes, enzymes chosen, in particular, from peroxidase and alkaline phosphatase and those capable of hydrolysing a chromogenic, fluorogenic or luminescent substrate, chromophoric chemical compounds, chromogenic, fluorogenic or luminescent compounds, nucleotide base analogues and biotin, [0072]
  • the probes used for diagnostic purposes of the invention may be employed in all known hybridization techniques, and in particular the techniques termed “DOT-BLOT” (9), “SOUTHERN BLOT” (10), “NORTHERN BLOT”, which is a technique identical to the “SOUTHERN BLOT” technique but which uses RNA as target, and the SANDWICH technique (11); advantageously, the SANDWICH technique is used in the present invention, comprising a specific capture probe and/or a specific detection probe, on the understanding that the capture probe and the detection probe must possess an at least partially different nucleotide sequence, [0073]
  • any probe according to the present invention can hybridize in vivo or in vitro with RNA and/or with DNA in order to block the phenomena of replication, in particular translation and/or transcription, and/or to degrade the said DNA and/or RNA, [0074]
  • a primer is a probe comprising at least six monomers, and advantageously from 10 to 30 monomers, possessing a specificity of hybridization under particular conditions for the initiation of an enzymatic polymerization, for example in an amplification technique such as PCR (polymerase chain reaction), in an elongation process such as sequencing, in a method of reverse transcription or the like, [0075]
  • two nucleotide or peptide sequences are termed equivalent or derived with respect to one another, or with respect to a reference sequence, if functionally the corresponding biopolymers can perform substantially the same role, without being identical, as regards the application or use in question, or in the technique in which they participate; two sequences are, in particular, equivalent if they are obtained as a result of natural variability, in particular spontaneous mutation of the species from which they have been identified, or induced variability, as are two homologous sequences, homology being defined below, [0076]
  • “variability” is understood to mean any spontaneous or induced modification of a sequence, in particular by substitution and/or insertion and/or deletion of nucleotides and/or of nucleotide fragments, and/or extension and/or shortening of the sequence at one or both ends; an unnatural variability can result from the genetic engineering techniques used, for example the choice of synthesis primers, degenerate or otherwise, selected for amplifying a nucleic acid; this variability can manifest itself in modifications of any starting sequence, considered as reference, and capable of being expressed by a degree of homology relative to the said reference sequence, [0077]
  • homology characterizes the degree of identity of two nucleotide or peptide fragments compared; it is measured by the percentage identity which is determined, in particular, by direct comparison of nucleotide or peptide sequences, relative to reference nucleotide or peptide sequences, [0078]
  • this percentage identity has been specifically determined for the nucleotide fragments, clones in particular, dealt with in the present invention, which are homologous to the fragments identified, for the MSRV-1 Virus, by SEQ ID No. 1 to No. 9, SEQ ID NO:46, SEQ ID NO:51 to SEQ ID NO:53, SEQ ID NO:40, SEQ ID NO:56 and SEQ ID NO:57, as well as for the probes and primers homologous to the probes and primers identified by SEQ ID NO:20 to SEQ ID NO:24, SEQ ID NO:26, SEQ ID NO:16 to SEQ ID NO:19, SEQ ID NO:31 to SEQ ID NO:33, SEQ ID NO:45, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:50, SEQ ID NO:55, SEQ ID NO:40, SEQ ID NO:56 and SEQ ID NO:57; as an example, the smallest percentage identity observed between the different general consensus sequences of nucleic acids obtained from fragments of MSRV-1 viral RNA, originating from the LM7PC and PLI-2 lines according to a protocol detailed later, is 67% in the region described in FIG. 1, [0079]
  • any nucleotide fragment is termed equivalent or derived from a reference fragment if it possesses a nucleotide sequence equivalent to the sequence of the reference fragment; according to the above definition, the following in particular are equivalent to a reference nucleotide fragment: [0080]
  • a) any fragment capable of hybridizing at least partially with the complement of the reference fragment, [0081]
  • b) any fragment whose alignment with the reference fragment results in the demonstration of a larger number of identical contiguous bases than with any other fragment originating from another taxonomic group, [0082]
  • c) any fragment resulting, or capable of resulting, from the natural variability of the species from which it is obtained, [0083]
  • d) any fragment capable of resulting from the genetic engineering techniques applied to the reference fragment, [0084]
  • e) any fragment containing at least eight contiguous nucleotides encoding a peptide which is homologous or identical to the peptide encoded by the reference fragment, [0085]
  • f) any fragment which is different from the reference fragment by insertion, deletion or substitution of at least one monomer, or extension or shortening at one or both of its ends; for example, any fragment corresponding to the reference fragment flanked at one or both of its ends by a nucleotide sequence not coding for a polypeptide, [0086]
  • polypeptide is understood to mean, in particular, any peptide of at least two amino acids, in particular an oligopeptide or protein, extracted, separated or substantially isolated or. synthesized through human intervention, in particular those obtained by chemical synthesis or by expression in a recombinant organism, [0087]
  • polypeptide partially encoded by a nucleotide fragment is understood to mean a polypeptide possessing at least three amino acids encoded by at least nine contiguous monomers lying within the said nucleotide fragment, [0088]
  • an amino acid is termed analogous to another amino acid when their respective physicochemical properties, such as polarity, hydrophobicity and/or basicity and/or acidity and/or neutrality are substantially the same; thus, a leucine is analogous to an isoleucine. [0089]
  • any polypeptide is termed equivalent or derived from a reference polypeptide if the polypeptides compared have substantially the same properties, and in particular the same antigenic, immunological, enzymological and/or molecular recognition properties; the following in particular are equivalent to a reference polypeptide: [0090]
  • a) any polypeptide possessing a sequence in which at least one amino acid has been replaced by an analogous amino acid, [0091]
  • b) any polypeptide having an equivalent peptide sequence, obtained by natural or induced variation of the said reference polypeptide and/or of the nucleotide fragment coding for the said polypeptide, [0092]
  • c) a mimotope of the said reference polypeptide, [0093]
  • d) any polypeptide in whose sequence one or more amino acids of the L series are replaced by an amino acid of the D series, and vice versa, [0094]
  • e) any polypeptide into whose sequence a modification of the side chains of the amino acids has been introduced, such as, for example, an acetylation of the amine functions, a carboxylation of the thiol functions, an esterification of the carboxyl functions, [0095]
  • f) any polypeptide in whose sequence one or more peptide bonds have been modified, such as, for example, carba, retro, inverso, retro-inverso, reduced and methylenoxy bonds, [0096]
  • (g) any polypeptide at least one antigen of which is recognized by an antibody directed against a reference polypeptide, [0097]
  • the percentage identity characterizing the homology of two peptide fragments compared is, according to the present invention, at least 50% and preferably at least 70%. [0098]
  • In view of the fact that a virus possessing reverse transcriptase enzymatic activity may be genetically characterized equally well in RNA and in DNA form, both the viral DNA and RNA will be referred to for characterizing the sequences relating to a virus possessing such reverse transcriptase activity, termed MSRV-1 according to the present description. [0099]
  • The expressions of order used in the present description and the claims, such as “first nucleotide sequence”, are not adopted so as to express a particular order, but so as to define the invention more clearly. [0100]
  • Detection of a substance or agent is understood below to mean both an identification and a quantification, or a separation or isolation, of the said substance or said agent.[0101]
  • A better understanding of the invention will be gained on reading the detailed description which follows, prepared with reference to the attached figures, in which: [0102]
  • FIG. 1 shows general consensus sequences of nucleic acids of the MSRV-1B clones amplified by the PCR technique in the “pol” region defined by Shih (12), from viral DNA originating from the LM7PC and PLI-2 lines, and identified under the references SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO: 5 and SEQ ID NO: 6, and the common consensus with amplification primers bearing the reference SEQ ID NO:7; [0103]
  • FIG. 2 gives the definition of a functional reading frame for each MSRV-1B/“PCR pol” type family, the said families A to D being defined, respectively, by the nucleotide sequences SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5 and SEQ ID NO:6 described in FIG. 1; [0104]
  • FIG. 3 gives an example of consensus of the MSRV-2B sequences, identified by SEQ ID NO:11; [0105]
  • FIG. 4 is a representation of the reverse transcriptase (RT) activity in dpm (disintegrations per minute) in the sucrose fractions taken from a purification gradient of the virions produced by the B lymphocytes in culture from a patient suffering from MS; [0106]
  • FIG. 5 gives, under the same experimental conditions as in FIG. 4, the assay of the reverse transcriptase activity in the culture of a B lymphocyte line obtained from a control free from MS; [0107]
  • FIG. 6 shows the nucleotide sequence of the clone PSJ17 (SEQ ID NO:9); [0108]
  • FIG. 7 shows the nucleotide sequence SEQ ID NO:8 of the clone designated M003-P004; [0109]
  • FIG. 8 shows the nucleotide sequence SEQ ID NO:2 of the clone F11-1; the portion located between the two arrows in the region of the primer corresponds to a variability imposed by the choice of primer which was used for the cloning of F11-1; in this same figure, the translation into amino acids is shown; [0110]
  • FIG. 9 shows the nucleotide sequence SEQ ID NO:1, and a possible functional reading frame of SEQ ID NO:1 in terms of amino acids; on this sequence, the consensus sequences of the pol gene are underlined; [0111]
  • FIGS. 10 and 11 give the results of a PCR, in the form of a photograph under ultraviolet light of an ethidium bromide-impregnated agarose gel, of the amplification products obtained from the primers identified by SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18 and SEQ ID NO:19; [0112]
  • FIG. 12 gives a representation in matrix form of the homology between SEQ ID NO:1 of MSRV-1 and that of an endogenous retrovirus designated HSERV9; this homology of at least 65% is demonstrated by a continuous line, the absence of a line meaning a homology of less than 65%; [0113]
  • FIG. 13 shows the nucleotide sequence SEQ ID NO:46 of the clone FBd3; [0114]
  • FIG. 14 shows the sequence homology between the clone FBd3 and the HSERV-9 retrovirus; [0115]
  • FIG. 15 shows the nucleotide sequence SEQ ID NO:51 of the clone t pol; [0116]
  • FIGS. 16 and 17 show, respectively, the nucleotide sequences SEQ ID NO:52 and SEQ ID NO:53 of the clones JLBc1 and JLBc2, respectively; [0117]
  • FIG. 18 shows the sequence homology between the clone JLBc1 and the clone FBd3; [0118]
  • and FIG. 19 the sequence homology between the clone JLBc2 and the clone FBd3; [0119]
  • FIG. 20 shows the sequence homology between the clones JLBc1 and JLBc2; [0120]
  • FIGS. 21 and 22 show the sequence homology between the HSERV-9 retrovirus and the clones JLBc1 and JLBc2, respectively; [0121]
  • FIG. 23 shows the nucleotide sequence SEQ ID NO:56 of the clone GM3; [0122]
  • FIG. 24 shows the sequence homology between the HSERV-9 retrovirus and the clone GM3; [0123]
  • FIG. 25 shows the localization of the different clones studied, relative to the genome of the known retrovirus ERV9; [0124]
  • FIG. 26 shows the position of the clones F11-1, M003-P004, MSRV-1B and PSJ17 in the region hereinafter designated MSRV-1 pol*; [0125]
  • FIG. 27, split into three successive FIGS. 27[0126] a, 27 b and 27 c, shows a possible reading frame covering the whole of the pol gene;
  • FIG. 28 shows, according to SEQ ID NO:40, the nucleotide sequence coding for the peptide fragment POL2B, having the amino acid sequence identified by SEQ ID NO:39; [0127]
  • FIG. 29 shows the OD values (ELISA tests) at 492 nm obtained for 29 sera of MS patients and 32 sera of healthy controls tested with an anti-IgG antibody; [0128]
  • FIG. 30 shows the OD values (ELISA tests) at 492 nm obtained for 36 sera of MS patients and 42 sera of healthy controls tested with an anti-IgM antibody; [0129]
  • FIGS. [0130] 31 to 33 show the results obtained (relative intensity of the spots) for 43 overlapping octapeptides covering the amino acid sequence 61-110, according to the Spotscan technique, respectively with a pool of MS sera, with a pool of control sera and with the pool of MS sera after deduction of a background corresponding to the maximum signal detected on at least one octapeptide with the control serum (intensity =1), on the understanding that these sera were diluted to {fraction (1/50)}. The bar at the far right-hand end represents a graphic scale standard unrelated to the serological test;
  • FIG. 34 shows the SEQ ID NO:41 and SEQ ID NO:42 of two polypeptides comprising immunodominant [lacuna], while SEQ ID NO:43 and 44 represent immunoreactive polypeptides specific to MS; [0131]
  • FIG. 35 shows the nucleotide sequence SEQ ID NO:59 of the clone LB19 and three potential reading frames of SEQ ID NO:59 in terms of amino acids; [0132]
  • FIG. 36 shows the nucleotide sequence SEQ ID NO:88 (GAG*) and a potential reading frame of SEQ ID NO:88 in terms of amino acids; [0133]
  • FIG. 37 shows the sequence homology between the clone FBd13 and the HSERV-9 retrovirus; according to this representation, the continuous line means a percentage homology greater than or equal to 70% and the absence of a line means a smaller percentage homology; [0134]
  • FIG. 38 shows the nucleotide sequence SEQ ID NO:61 of the clone FP6 and three potential reading frames of SEQ ID NO:61 in terms of amino acids; [0135]
  • FIG. 39 shows the nucleotide sequence SEQ ID NO:89 of the clone G+E+A and three potential reading frames of SEQ ID NO:89 in terms of amino acids; [0136]
  • FIG. 40, shows a reading frame found in the region E and coding for an MSRV-1 retroviral protease identified by SEQ ID NO:90; [0137]
  • FIG. 41 shows the response of each serum of patients suffering from MS, indicated by the symbol. (+), and of healthy patients, symbolised by (−), tested with an anti-IgG antibody, expressed as net optical density at 492 nm; [0138]
  • FIG. 42 shows the response of each serum of patients suffering from MS, indicated by the symbols (+) and (QS), and of healthy patients (−), tested with an anti-IgM antibody, expressed as net optical density at 492 nm. [0139]
  • EXAMPLE 1
  • Obtaining Clones Designated MSRV-1B and MSRV-2B, Defining, Respectively, a Retrovirus MSRV-1 and a Coinfective Agent MSRV2, by “Nested” PCR Amplification of the Conserved POL Regions of Retroviruses on Virion Preparations Originating from the LM7PC and PLI-2 Lines [0140]
  • A PCR technique derived from the technique published by Shih (12) was used. This technique enables all trace of contaminant DNA to be removed by treating all the components of the reaction medium with DNase. It concomitantly makes it possible, by the use of different but overlapping primers in two successive series of PCR amplification cycles, to increase the chances of amplifying a cDNA synthesized from an amount of RNA which is small at the outset and further reduced in the sample by the spurious action of the DNAse on the RNA. In effect, the DNase is used under conditions of activity in excess which enable all trace of contaminant DNA to be removed before inactivation of this enzyme remaining in the sample by heating to 85° C. for 10 minutes. This variant of the PCR technique described by Shih (12) was used on a cDNA synthesized from the nucleic acids of fractions of infective particles purified on a sucrose gradient according to the technique described by H. Perron (13) from the “POL-2” isolate (ECACC No. V92072202) produced by the PLI-2 line (ECACC No. 92072201) on the one hand, and from the MS7PG isolate (ECACC No. V93010816) produced by the LM7PC line (ECACC No. 93010817) on the other hand. These cultures were obtained according to the methods which formed the subject of the patent applications published under Nos WO 93/20188 and WO 93/20189. [0141]
  • After cloning the products amplified by this technique with the TA Cloning Kit® and analysis of the sequence using an Applied Biosystems model 373A Automatic Sequencer, the sequences were analysed using tne Geneworks® software on the latest available version of the Genebank® data bank. [0142]
  • The sequences cloned and sequenced from these samples correspond, in particular, to two types of sequence: a first type of sequence, to be found in the majority of the clones (55% of the clones originating from the POL-2 isolates of the PLI-2 culture, and 67% of the clones originating from the MS7PG isolates of the LM7PC cultures), which corresponds to a family of “pol” sequences closely similar to, but different from, the endogenous human retrovirus designated ERV-9 or HSERV-9, and a second type of sequence which corresponds to sequences very strongly homologous to a sequence attributed to another infective and/or pathogenic agent designated MSRV-2. [0143]
  • The first type of sequence, representing the majority of the clones, consists of sequences whose variability enables four subfamilies of sequences to be defined. These subfamilies are sufficiently similar to one another for it to be possible to consider them to be quasi-species originating from the same retrovirus, as is well known for the HIV-1 retrovirus (14), or to be the outcome of interference with several endogenous proviruses coregulated in the producing cells. These more or less defective endogenous elements are sensitive to the same regulatory signals possibly generated by a replicative provirus, since they belong to the same family of endogenous retroviruses (15). This new family of endogenous retroviruses, or alternatively this new retroviral species from which the generation of quasi-species has been obtained in culture, and which contains a consensus of the sequences described below, is desig nated MSRV-1B. [0144]
  • FIG. 1 presents the general consensus sequences of the sequences of the different MSRV-1B clones sequenced in this experiment, these sequences being identified, respectively, by SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5 and SEQ ID NO:6. These sequences display a homology with respect to nucleic acids ranging from 70% to 88% with the HSERV9 sequence referenced X57147 and M37638 in the Genebankr data base. Four “consensus” nucleic acid sequences representative of different quasi-species of a possibly exogenous retrovirus MSRV-1B, or of different subfamilies of an endogenous retrovirus MSRV-1B, have been defined. These representative consensus sequences are presented in FIG. 2, with the translation into amino acids. A functional reading frame exists for each subfamily of these MSRV-1B sequences, and it can be seen that the functional open reading frame corresponds in each instance to the amino acid sequence appearing on the second line under the nucleic acid sequence. The general consensus of the MSRV-1B sequence, identified by SEQ ID NO:7 and obtained by this PCR technique in the “pol” region, is presented in FIG. 1. [0145]
  • The second type of sequence representing the majority of the clones sequenced is represented by the sequence MSRV-2B presented in FIG. 3 and identified by SEQ ID NO:11. The differences observed in the sequences corresponding to the PCR primers are explained by the use of degenerate primers in mixture form used under different technical conditions. [0146]
  • The MSRV-2B sequence (SEQ ID NO:11) is sufficiently divergent from the retroviral sequences already described in the data banks for it to be suggested that the sequence region in question belongs to a new infective agent, designated MSRV-2. This infective agent would, in principle, on the basis of the analysis of the first sequences obtained, be related to a retrovirus but, in view of the technique used for obtaining this sequence, it could also be a DNA virus whose genome codes for an enzyme which incidentally possesses reverse transcriptase activity, as is the case, for example, with the hepatitis B virus, HBV (12). Furthermore, the random nature of the degenerate primers used for this PCR amplification technique may very well have permitted, as a result of unforeseen sequence homologies or of conserved sites in the gene for a related enzyme, the amplification of a nucleic acid originating from a prokaryotic or eukaryotic pathogenic and/or coinfective agent (protist). [0147]
  • EXAMPLE 2
  • Obtaining Clones Designated MSRV-1B and MSRV-2B, Defining a Family MSRV-1 and MSRV2, by “Nested” PCR Amplification of the Conserved POL Regions of Retroviruses on Preparations of B Lymphocytes from a New Case of MS [0148]
  • The same PCR technique, modified according to the technique of Shih (12), was used to amplify and sequence the RNA nucleic acid material present in a purified fraction of virions at the peak of “LM7-like” reverse transcriptase activity on a sucrose gradient according to the technique described by H. Perron (13), and according to the protocols mentioned in Example 1, from a spontaneous lymphoblastoid line obtained by self-immortalization in culture of B lymphocytes from an MS patient who was seropositive for the Epstein-Barr virus (EBV), after setting up the blood lymphoid cells in culture in a suitable culture medium containing a suitable concentration of cyclosporin A. A representation of the reverse transcriptase activity in the sucrose fractions taken from a purification gradient of the virions produced by this line is presented in FIG. 4. Similarly, the culture supernatants of a B line obtained under the same conditions from a control free from MS were treated under the same conditions, and the assay of reverse transcriptase activity in the sucrose gradient fractions proved negative throughout (background), and is presented in FIG. 5. [0149] Fraction 3 of the gradient corresponding to the MS B line and the same fraction without reverse transcriptase activity of the non-MS control gradient were analysed by the same RT-PCR technique as before, derived from Shih (12), followed by the same steps of cloning and sequencing as described in Example 1.
  • It is particularly noteworthy that the MSRV-1 and MSRV-2 type sequences are to be found only in the material associated with a peak of “LM7-like” reverse transcriptase activity originating from the MS B lymphoblastoid line. These sequences were not to be found with the material from the control (non-MS) B lymphoblastoid line in 26 recombinant clones taken at random. Only MoMuLV type contaminant sequences, originating from the commercial reverse transcriptase used for the CDNA synthesis step, and sequences without any particular retroviral analogy were to be found in this control, as a result of the “consensus” amplification of homologous polymerase sequences which is produced by this PCR technique. Furthermore, the absence of a concentrated target which competes for the amplification reaction in the control sample permits the amplification of dilute contaminants. The difference in results is manifestly highly significant (chi- squared, p<0.001). [0150]
  • EXAMPLE 3
  • Obtaining a Clone PSJ17, Defining a Retrovirus MSRV-1, by Reaction of Endogenous Reverse Transcriptase with a Virion Preparation Originating from the PLI-2 Line. [0151]
  • This approach is directed towards obtaining reverse-transcribed DNA sequences from the supposedly retroviral RNA in the isolate using the reverse transcriptase activity present in this same isolate. This reverse transcriptase activity can theoretically function only in the presence of a retroviral RNA linked to a primer tRNA or hybridized with short strands of DNA already reverse-transcribed in the retroviral particles (16). Thus, the obtaining of specific retroviral sequences in a material contaminated with cellular nucleic acids was optimized according to these authors by means of the specific enzymatic amplification of the portions of viral RNAs with a viral reverse transcriptase activity. To this end, the authors determined the particular physicochemical conditions under which this enzymatic activity of reverse transcription on RNAs contained in virions could be effective in vitro. These conditions correspond to the technical description of the protocols presented below (endogenous RT reaction, purification, cloning and sequencing). [0152]
  • The molecular approach consisted in using a preparation of concentrated but unpurified virion obtained from the culture supernatants of the PLI-2 line, prepared according to the following method: the culture supernatants are collected twice weekly, precentrifuged at 10,000 rpm for 30 minutes to remove cell debris and then frozen at −80° C. or used as they are for the following steps. The fresh or thawed supernatants are centrifuged on a cushion of 30% glycerol-PBS at 100,000 g (or 30,000 rpm in a type 45 T LKB-HITACHI rotor) for 2 h at 4° C. After removal of the supernatant, the sedimented pellet is taken up in a small volume of PBS and constitutes the fraction of concentrated but unpurified virion. This concentrated but unpurified viral sample was used to perform a so-called endogenous reverse transcription reaction, as described below. [0153]
  • A volume of 200 μl of virion purified according to the protocol described above, and containing a reverse transcriptase activity of approximately 1-5 million dpm, is thawed at 37° C. until a liquid phase appears, and then placed on ice. A 5-fold concentrated buffer was prepared with the following components: 500 mM Tris-HCl pH 8.2; 75 mM NaCl; 25 mM MgC12; 75 mM DTT and 0.10[0154] % NP 40; 100 μl of 5× buffer+25 μl of a 100 mM solution of dATP +25 ml of a 100 mM solution of dTTP+25 ml of a 100 μM solution of dGTP+25 μl of a 100 mM solution of dCTP +100 ml of sterile distilled water+200 ml of the virion suspension (RT activity of 5 million DPM) in PBS were mixed and incubated at 42° C. for 3 hours. After this incubation, the reaction mixture is added directly to a buffered phenol/- chloroform/isoamyl alcohol mixture (Sigma ref. P 3803); the aqueous phase is collected and one volume of sterile distilled water is added to the organic phase to re-extract the residual nucleic acid material: The collected aqueous phases are combined, and the nucleic acids contained are precipitated by adding 3M sodium acetate pH 5.2 to {fraction (1/10)}volume+2 volumes of ethanol+1 μl of glycogen (Boehringer-Mannheim ref. 901 393) and placing the sample at −20° C. for 4 h or overnight at +4° C. The precipitate obtained after centrifugation is then washed with 70% ethanol and resuspended in 60 ml of distilled water. The products of this reaction were then purified, cloned and sequenced according to the protocol which will now be described: blunt-ended DNAs with unpaired adenines at the ends were generated: a “filling-in” reaction was first performed: 25 μl of the previously purified DNA solution were mixed with 2 μl of a 2.5 mM solution containing, in equimolar amounts, DATP+dGTP+dTTP+dCTP/1 μl of T4 DNA polymerase (Boehringer-Mannheim ref. 1004 786) / 5 μl of 10× “incubation buffer for restriction enzyme” (Boehringer-Mannheim ref. 1417 975) / 1 μl of a 1% bovine serum albumin solution / 16 μl of sterile distilled water. This mixture was incubated for 20 minutes at 11° C. 50 μl of TE buffer and 1 μl of glycogen (Boehringer-Mannheim ref. 901 393) were added thereto before extraction of the nucleic acids with phenol/chloroform/isoamyl alcohol (Sigma ref. P 3803) and precipitation with sodium acetate as described above. The DNA precipitated after centrifugation is resuspended in 10 μl of 10 mM Tris buffer pH 7.5. 5 μl of this suspension were then mixed with 20 μl of 5× Taq buffer, 20 μl of 5 mM dATP, 1 μl (5U) of Taq DNA polymerase (Amplitaq™) and 54 μl of sterile distilled water. This mixture is incubated for 2 h at 75° C. with a film of oil on the surface of the solution. The DNA suspended in the aqueous solution drawn off under the film of oil after incubation is precipitated as described above and resuspended in 2 μl of sterile distilled water. The DNA obtained was inserted into a plasmid using the TA Cloning™ kit. The 2 μl of DNA solution were mixed with 5 μl of sterile distilled water, 1 μl of a 10-fold concentrated ligation buffer “10× LIGATION BUFFER”, 2 μl of “pCR™ VECTOR” (25 ng/ml) and 1 μl of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17). The plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA cloning kit®. The reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer, model 373 A” apparatus according to the manufacturer's instructions.
  • Discriminating analysis on the computerized data banks of the sequences cloned from the DNA fragments present in the reaction mixture enabled a retroviral type sequence to be revealed. The corresponding clone PSJ17 was completely sequenced, and the sequence obtained, presented in FIG. 6 and identified by SEQ ID No. 9, was analysed using the “Geneworks®” software on the updated “Genebank®” data banks. An identical sequence already described could not be found by analysis of the data banks. Only a partial homology with some known retroviral elements was to be found. The most useful relative homology relates to an endogenous retrovirus designated ERV-9, or HSERV-9, according to the references (18). [0155]
  • EXAMPLE 4
  • PCR Amplification of the Nucleic Acid Sequence Contained Between the 5′ Region Defined by the Clone *POL NSRV-1B′ and the 3′ REGION DEFINED BY THE CLONE PSJ17. [0156]
  • Five oligonucleotides, M001, M002-A, M003-BCD, P004 and P005, were defined in order to amplify the RNA originating from purified POL-2 virions. Control reactions were performed so as to check for the presence of contaminants (reaction with water). The amplification consists of an RT-PCR step according to the protocol described in Example 2, followed by a “nested” PCR according to the PCR protocol described in the document EP-A-0,569,272. In the first RT-PCR cycle, the primers M001 and P004 or P005 are used. In the second PCR cycle, the primers M002-A or M003-BCD and the primer P004 are used. The primers are positioned as follows: [0157]
                 M002-A
                 M003-BCD
           M001               P004  P005
                                        
                                             RNA
    POL-2
    <-------------->       <----------------------->
         pol MSRV-1B            PSJ17
    primer GGTCITICCICAIGG (SEQ ID NO:20)
    M001:
    primer TTAGGGATAGCCCTCATCTCT (SEQ ID NO:21)
    M002-A:
    primer TCAGGGATAGCCCCCATCTAT (SEQ ID NO:22)
    M003-BCD:
    primer AACCCTTTGCCACTACATCAATTT (SEQ ID NO:23)
    P004:
    primer GCGTAAGGACTCCTAGAGCTATT (SEQ ID NO:24)
    P005:
  • The “nested” amplification product obtained, and designated M003-P004, is presented in FIG. 7, and corresponds to the sequence SEQ ID NO:8. [0158]
  • EXAMPLE 5
  • Amplification and Cloning of a Portion of the MSRV-1 Retroviral Genome using a Sequence Already Identified, in a Sample of Virus Purified at the Peak of Reverse Transcriptinve Activity [0159]
  • A PCR technique derived from the technique published by Frohman (19) was used. The technique derived makes it possible, using a specific primer at the 3′ end of the genome to be amplified, to elongate the sequence towards the 5′ region of the genome to be analysed. This technical variant is described in the documentation of the firm “Clontech Laboratories Inc.”, (Palo-Alto California, USA) supplied with its product “5′-AmpliFINDER™ RACE Kit”, which was used on a fraction of virion purified as described above. [0160]
  • The specific 3′ primers used in the kit protocol for the synthesis of the cDNA and the PCR amplification are, respectively, complementary to the following MSRV-1 sequences: [0161]
    cDNA: TCATCCATGTACCGAAGG (SEQ ID NO:25)
    amplification: ATGGGGTTCCCAAGTTCCCT (SEQ ID NO:26)
  • The products originating from the PCR were purified after purification on agarose gel according to conventional methods (17), and then resuspended in 1.0 ml of distilled water. Since one of the properties of Taq polymerase consists in adding an adenine at the 3′ end of each of the two DNA strands, the DNA obtained was inserted directly into a plasmid using the TA Cloning™ kit (British Biotechnology). The 2 μl of DNA solution were mixed with 5 μl of sterile distilled water, 1 μl of a 10-fold concentrated ligation buffer “10′ LIGATION BUFFER”, 2 μl of “pCR™ VECTOR” (25 ng/ml) and 1 μl of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17). The plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detecte under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®. The reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer model 373 A” apparatus according to the manufacturer's instructions. [0162]
  • This technique was applied first to two fractions of virion purified as described below on sucrose from the “POL-2” isolate produced by the PLI-2 line on the one hand, and from the MS7PG isolate produced by the LM7PC line on the other hand. The culture supernatants are collected twice weekly, precentrifuged at 10,000 rpm for 30 minutes to remove cell debris and then frozen at −80° C. or used as they are for the following steps. The fresh or thawed supernatants are centrifuged on a cushion of 30% glycerol-PBS at 100,000 g (or 30,000 rpm in a type 45 T LKB-HITACHI rotor) for 2 h at 4° C. After removal of the supernatant, the sedimented pellet is taken up in a small volume of PBS and constitutes the fraction of concentrated but unpurified virions. The concentrated virus is then applied to a sucrose gradient in sterile PBS buffer (15 to 50% weight/weight) and ultracentrifuged at 35,000 rpm (100,000 g) for 12 h at +4° C., in a swing-out rotor. 10 fractions are collected, and 20 μl are withdrawn from each fraction after homogenization to assay the reverse transcriptase activity therein according to the technique described by H. Perron (3). The fractions containing the peak of “LM7-like” RT activity are then diluted in sterile PBS buffer and ultracentrifuged for one hour at 35,000 rpm (100,000 g) to sediment the viral particles. The pellet of purified virion thereby obtained is then taken up in a small volume of a buffer which is appropriate for the extraction of RNA. The cDNA synthesis reaction mentioned above is carried out on this RNA extracted from purified extracellular virion. PCR amplification according to the technique mentioned above enabled the clone F1-11 to be obtained, whose sequence, identified by SEQ ID NO:2, is presented in FIG. 8. [0163]
  • This clone makes it possible to define, with the different clones previously sequenced, a region of considerable length (1.2 kb) representative of the “pol” gene of the MSRV-1 retrovirus, as presented in FIG. 9. This sequence, designated SEQ ID NO:1, is reconstituted from different clones overlapping one another at their ends, correcting the artefacts associated with the primers and with the amplification or cloning techniques which would artificially interrupt the reading frame of the whole. This sequence will be identified below under the designation “MSRV-1 pol* region”. Its degree of homology with the HSERV-9 sequence is shown in FIG. 12. [0164]
  • In FIG. 9, the potential reading frame with its translation into amino acids is presented below the nucleic acid sequence. [0165]
  • EXAMPLE 6
  • Detection of Specific MSRV-1 and MSRV-2 Sequences in Different Samlpes of Plasma Originating from Patients Suffering from MS or from Controls. [0166]
  • A PCR technique was used to detect the MSRV-1 and MSRV-2 genomes in plasmas obtained after taking blood samples from patients suffering from MS and from non-MS controls onto EDTA. [0167]
  • Extraction of the RNAs from plasma was performed according to the technique described by P. Chomzynski (20), after adding one volume of buffer containing guanidinium thiocyanate to 1 ml of plasma stored frozen at −80° C. after collection. [0168]
  • For MSRV-2, the PCR was performed under the same conditions and with the following primers: [0169]
    5′ primer, identified by
    5′ GTAGTTCGATGTAGAAAGCG 3′; SEQ ID NO:14
    3′ primer, identified by
    5′ GCATCCGGCAACTGCACG 3′. SEQ ID NO:15
  • However, similar results were also obtained with the following PCR primers in two successive amplifica tions by “nested” PCR on samples of nucleic acids not treated with DNase. [0170]
  • The primers used for this first step of 40 cycles with a hybridization temperature of 48° C. are the following: [0171]
  • 5′ primer, identified by SEQ ID NO:27 5′ [0172] GCCGATATCACCCGCCATGG 3′, corresponding to a 5′ MSRV-2 PCR primer, for a first PCR on samples from patients,
  • 3′ primer, identified by SEQ ID NO:28 5′ [0173] GCATCCGGCAACTGCACG 3′, corresponding to a 3′ MSRV-2 PCR primer, for a first PCR on samples from patients.
  • After this step, 10 μl of the amplification product are taken and used to carry out a second, so-called “nested” PCR amplification with primers located within the region already amplified. This second step takes place over 35 cycles, with a primer hybridization (“annealing”) temperature of 50° C. The reaction volume is 100 μl. [0174]
  • The primers used for this second step are the 10 following: [0175]
  • 5′ primer, identified by SEQ ID NO:29 5′ [0176] CGCGATGCTGGTTGGAGAGC 3′, corresponding to a 5′ MSRV-2 PCR primer, for a nested PCR on samples from patients,
  • 3′ primer, identified by SEQ ID NO:30 5′[0177] TCTCCACTCCGAATATTCCG 3′, corresponding to a 3′ MSRV-2 PCR primer, for a nested PCR on samples from patients.
  • For MSRV-1, the amplification was performed in two steps. Furthermore, the nucleic acid sample is treated beforehand with DNase, and a control PCR without RT (AMV reverse transcriptase) is performed on the two amplification steps so as to verify that the RT-PCR amplification comes exclusively from the MSRV-1 RNA. In the event of a positive control without RT, the initial aliquot sample of RNA is again treated with DNase and amplified again. [0178]
  • The protocol for treatment with DNase lacking RNAse activity is as follows: the extracted RNA is aliquoted in the presence of “RNAse inhibitor” (Boehringer-Mannheim) in water treated with DEPC at a final concentration of 1 μg in 10 μl; to these 10 μl, 1 μl of “RNAse-free DNAse” (Boehringer-Mannheim) and 1.2 μl of [0179] pH 5 buffer containing 0.1 M/1 sodium acetate 35 and 5 mM/l MgSO4 is added; the mixture is incubated for 15 min at 20° C. and brought to 95° C. for 1.5 min in a “thermocycler”.
  • The first MSRV-1 RT-PCR step is performed according to a variant of the RNA amplification method as described in Patent Application No. EP-A-0,569,272. In particular, the CDNA synthesis step is performed at 42° C. for one hour; the PCR amplification takes place over 40 cycles, with a primer hybridization (“annealing”) temperature of 53° C. The reaction volume is 100 μl. [0180]
  • The primers used for this first step are the following: [0181]
    5′ primer, identified by
    5′ AGGAGTAAGGAAACCCAACGGAC 3′; SEQ ID NO:16
    3′ primer, identified by
    5′ TAAGAGTTGCACAAGTGCG 3′. SEQ ID NO:17
  • After this step, 10 μl of the amplification product are taken and used to carry out a second, so-called “nested” PCR amplification with primers located within the region already amplified. This second step takes place over 35 cycles, with a primer hybridization (“annealing”) temperature of 53° C. The reaction volume is 100 μl. [0182]
  • The primers used for this second step are the following: [0183]
    5′ primer, identified by
    5′ TCAGGGATAGCCCCCATCTAT 3′; SEQ ID NO:18
    3′ primer, identified by
    5′ AACCCTTTGCCACTACATCAATTT 3′. SEQ ID NO:19
  • FIGS. 10 and 11 present the results of PCR in the form of photographs under ultraviolet light of ethidium bromide-impregnated agarose gels, in which an electrophoresis of the PCR amplification products applied separately to the different wells was performed. [0184]
  • The top photograph (FIG. 10) shows the result of specific MSRV-2 amplification. [0185]
  • [0186] Well number 8 contains a mixture of DNA molecular weight markers, and wells 1 to 7 represent, in order, the products amplified from the total RNAs of plasmas originating from 4 healthy controls free from MS (wells 1 to 4) and from 3 patients suffering from MS at different stages of the disease (wells 5 to 7).
  • In this series, MSRV-2 nucleic acid material is detected in the plasma of one case of MS out of the 3 tested, and in none of the 4 control plasmas. Other results obtained on more extensive series confirm these results. [0187]
  • The bottom photograph (FIG. 11) shows the result of specific amplification by MSRV-1 “nested” RT-PCR: [0188]
  • well No. 1 contains the PCR product produced with water alone, without the addition of AMV reverse transcriptase; well No. 2 contains the PCR product produced with water alone, with the addition of AMV reverse transcriptase; [0189] well number 3 contains a mixture of DNA molecular weight markers; wells 4 to 13 contain, in order, the products amplified from the total RNAs extracted from sucrose gradient fractions (collected in a downward direction), on which gradient a pellet of virion originating from a supernatant of a culture infected with MSRV-1 and MSRV-2 was centrifuged to equilibrium according to the protocol described by H. Perron (13); to well 14 nothing was applied; to wells 15 to 17, the amplified products of RNA extracted from plasmas originating from 3 different patients suffering from MS at different stages of the disease were applied.
  • The MSRV-1 retroviral genome is indeed to be found in the sucrose gradient fraction containing the peak of reverse transcriptase activity measured according to the technique described by H. Perron (3), with a very strong intensity ([0190] fraction 5 of the gradient, placed in well No. 8). A slight amplification has taken place in the first fraction (well No. 4), probably corresponding to RNA released by lysed particles which floated at the surface of the gradient; similarly, aggregated debris has sedimented in the last fraction (tube bottom), carrying with it a few copies of the MSRV-1 genome which have given rise to an amplification of low intensity.
  • Of the 3 MS plasmas tested in this series, MSRV-1 RNA turned up in one case, producing a very intense amplification (well No. 17). [0191]
  • In this series, the MSRV-1 retroviral RNA genome, probably corresponding to particles of extracellular virus present in the plasma in extremely small numbers, was detected by “nested” RT-PCR in one case of MS out of the 3 tested. Other results obtained on more extensive series confirm these results. [0192]
  • Furthermore, the specificity of the sequences amplified by these PCR techniques may be verified and evaluated by the “ELOSA” technique as described by F. Mallet (21) and in the document FR- A-2,663,040. [0193]
  • For MSRV-1, the products of the nested PCR described above may be tested in two ELOSA systems enabling a consensus A and a consensus B+C+D of MSRV-1 to be detected separately, corresponding to the subfamilies described.in Example 1 and FIGS. 1 and 2. In effect, the sequences closely resembling the consensus B+C+D are to be found essentially in the RNA samples originating from MSRV-1 virions purified from cultures or amplified in extracellular biological fluids of MS patients, whereas the sequences closely resembling the consensus A are essentially to be found in normal human cellular DNA. [0194]
  • The ELOSA/MSRV-1 system for the capture and specific hybridization of the PCR products of the subfamily A uses a capture oligonucleotide cpV1A with an amine bond at the 5′ end and a biotinylated detection oligonucleotide dpV1A having as their sequence, respectively: [0195]
  • cpV1A identified by SEQ ID NO:31 [0196]
  • [0197] 5GATCTAGGCCACTTCTCAGGTCCAGS 3′, corresponding to the ELOSA capture oligonucleotide for the products of MSRV-1 nested PCR performed with the primers identified by SEQ ID NO:16 and SEQ ID NO:17, optionally followed by amplification with the primers identified by SEQ ID NO18 and SEQ ID NO:19 on samples from patients;
  • dpV1A identified by SEQ ID NO:32; [0198]
  • 5′ [0199] CATCTITTTGGICAGGCAITAGC 3′, corresponding to the ELOSA capture oligonucleotide for the subfamily A of the products of MSRV-1 “nested” PCR performed with the primers identified by SEQ ID NO:16 and SEQ ID NO:17, optionally followed by amplification with the primers identified by SEQ ID NO:18 and SEQ ID NO:19 on samples from patients.
  • The ELOSA/MSRV-1 system for the capture and specific hybridization of the PCR products of the subfamily B+C+D uses the same biotinylated detection oligonucleotide dpV1A and a capture oligonucleotide cpV1B with an amine bond at the 5′ end having as its sequence: [0200]
  • dpV1B identified by SEQ ID NO:33 [0201]
  • [0202] 5° CTTGAGCCAGTTCTCATACCTGGA 3′, corresponding to the ELOSA capture oligonucleotide for the subfamily B+C+D of the products of MSRV-1 “nested” PCR performed with the primers identified by SEQ ID NO:16 and SEQ ID NO:17, optionally. followed by amplification with the primers identified by SEQ ID NO:18 and SEQ ID NO:19 on samples from patients.
  • This ELOSA detection system enabled it to be verified that none of the PCR products thus amplified from DNase-treated plasmas of MS patients contained a sequence of the subfamily A, and that all were positive with the consensus of the subfamilies B, C and D. [0203]
  • For MSRV-2, a similar ELOSA technique was evaluated on isolates originating from infected cell cultures, using the following PCR amplification primers, [0204]
  • 5′ primer, identified by SEQ ID NO:34 [0205]
  • 5′ [0206] AGTGYTRCCMCARGGCGCTGAA 3′, corresponding to a 5′ MSRV-2 PCR primer, for PCR on samples from cultures,
  • 3′ primer, identified by SEQ ID NO:35 [0207]
  • 5′ [0208] GMGGCCAGCAGSAKGTCATCCA 3′, corresponding to a 3′ MSRV-2 PCR primer, for PCR on samples from cultures,
  • and the capture oligonucleotides with an amine bond at the 5′ end cpV2.and the biotinylated detection oligonucleotide dpV2 having as their respective sequences: [0209]
  • cpV2 identified by SEQ ID NO:36 [0210]
  • 5 GGATGCCGCCTATAGCCTCTAC 3′, corresponding to an ELOSA capture oligonucleotide for the products of MSRV-2 PCR performed with the primers SEQ ID NO:34 and SEQ ID NO:35, or optionally with the degenerate primers defined by Shih (12). [0211]
  • dpV2 identified by SEQ ID NO:37 [0212]
  • 5′ [0213] AAGCCTATCGCGTGCAGTTGCC 3′, corresponding to an ELOSA detection oligonucleotide for the products of MSRV-2 PCR performed with the primers SEQ ID NO:34 and SEQ ID NO:35, or optionally with the degenerate primers defined by Shih (12)
  • This PCR amplification system with a pair of primers different from those which were described previously for amplification on the samples from patients made it possible to confirm the infection with MSRV-2 of in vitro cultures and of samples of nucleic acids used for the molecular biology studies. [0214]
  • All things considered, the first results of PCR detection of the genome of pathogenic and/or infective agents show that it is possible that free “virus” may circulate in the blood stream of patients in an acute, virulent phase, outside the nervous system. This is compatible with the almost invariable presence of “gaps” in the blood-brain barrier of patients in an active phase of MS. [0215]
  • EXAMPLE 7
  • Obtaining Sequences of the “env” Gene of the MSRV-1 Retroviral Genome [0216]
  • As has already been described in Example 5, a PCR technique derived from the technique published by Frohman (19) was used. The technique derived makes it possible, using a specific primer at the 3′ end of the genome to be amplified, to elongate the sequence towards the 5′ region of the genome to be analysed. This technical variant is described in the documentation of “Clontech Laboratories Inc., (Palo-Alto Calif., USA) supplied with its product “5′-AmpliFINDER™ RACE Kit”, which was used on a fraction of virion purified as described above. [0217]
  • In order to carry out an amplification of the 3′ region of the MSRV-1 retroviral genome encompassing the region of the “env” gene, a study was carried out to determine a consensus sequence in the LTR regions of the same type as those of the defective endogenous retrovirus HSERV-9 (18, 24), with which the MSRV-1 retrovirus displays partial homologies. [0218]
  • The same specific 3′ primer was used in the kit protocol for the synthesis of the CDNA and the PCR amplification; its sequence is as follows: [0219]
  • GTGCTGATTGGTGTATTTACAATCC (SEQ ID NO 45) [0220]
  • Synthesis of the complementary DNA (CDNA) and unidirectional PCR amplification with the above primer were carried out in one step according to the method described in Patent EP-A-0,569,272. [0221]
  • The products originating from the PCR were extracted after purification of agarose gel according to conventional methods (17), and then resuspended in 10 ml of distilled water. Since one of the properties of Taq polymerase consists in adding an adenine at the 3′ end of each of the two DNA strands, the DNA obtained was inserted directly into a plasmid using the TA Cloning™ kit (British Biotechnology). The 2 μl of DNA solution were mixed with 5 μl of sterile distilled water, 1 μl of a 10-fold concentrated ligation buffer “10× LIGATION BUFFER”, 2 μl of “pCR™ VECTOR” (25 ng/ml) and 1 μl of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17). The plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®. The reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “automatic sequencer, model 373 A [lacuna] apparatus according to the manufacturer's instructions. [0222]
  • This technical approach was applied to a sample of virion concentrated as described below from a mixture of culture supernatants produced by B lymphoblastoid lines such as are described in Example 2, established from lymphocytes of patients suffering from MS and possessing reverse transcriptase activity which is detectable according to the technique described by Perron et al. (3): the culture supernatants are collected twice weekly, precentrifuged at 10,000 rpm for 30 minutes to remove cell debris and then frozen at −80° C. or used as they are for the following steps. The fresh or thawed supernatants are centrifuged on a cushion of 30% glycerol-PBS at 100,000 g for 2 h at 4° C. After removal of the supernatant, the sedimented pellet constitutes the sample of concentrated but unpurified virions. The pellet thereby obtained is then taken up in a small volume of an appropriate buffer for the extraction of RNA. The cDNA synthesis reaction mentioned above is carried out on this RNA extracted from concentrated extracellular virion. [0223]
  • RT-PCR amplification according to the technique mentioned above enabled the clone FBd3 to be obtained, whose sequence, identified by SEQ ID NO:46, is presented in FIG. 13. [0224]
  • In FIG. 14, the sequence homology between the clone FBd3 and the HSERV-9 retrovirus is shown on the matrix chart by a continuous line for any partial homology greater than or equal to 65%. It can be seen that there are homologies in the flanking regions of the clone (with the pol gene at the 5′ end and with the env gene and then the LTR at the 3′ end), but that the internal region is totally divergent and does not display any homology, even weak, with the “env” gene of HSERV9. Furthermore, it is apparent that the clone FBd3 contains a longer “env” region than the one which is described for the defective endogenous HSERV-9; it may thus be seen that the internal divergent region constitutes an “insert” between the regions of partial homology with the HSERV-9 defective genes. [0225]
  • EXAMPLE 8
  • Amplification, Cloning and Sequencing of the Region of the MSRV-1 Retroviral Genome Located Between the Clones PSJ17 and FBd3 [0226]
  • Four oligonucleotides, F1, B4, F6 and B1, were defined for amplifying RNA originating from concentrated virions of the strains POL2 and MS7PG. Control reactions were performed so as to check for the presence of contaminants (reaction with water). The amplification consists of a first step of RT-PCR according to the protocol described in Patent Application EP-A-0,569,272, followed by a second step of PCR performed on 10 ml of product of the first step with primers internal to the amplified first region (“nested” PCR). In the first RT-PCR cycle, the primers F1 and B4 are used. In the second PCR cycle, the primers F6 and the primer B1 are used. The primers are positioned as follows: [0227]
         F1   F6                      B1   B4
                                             
                                                RNA
    MSRV-1
    PSJ17                           FBd3
    ---------->           <------------------/---
    5′pol MSRV-1          3′pol MSRV-1      /
    5′env
    Their composition is:
    primer TGATGTGAACGGCATACTCACTG (SEQ ID NO:47)
    F1:
    primer CCCAGAGGTTAGGAACTCCCTTTC (SEQ ID N0 48)
    B4:
    primer GCTAAAGGAGACTTGTGGTTGTCAG (SEQ ID N0 49)
    F6:
    primer CAACATGGGCATTTCGGATTAG (SEQ ID N0 50)
    B1:
  • The product of “nested” amplification obtained and designated “t pol” is presented in FIG. 15, and corresponds to the sequence SEQ ID NO:51. [0228]
  • EXAMPLE 9
  • Obtaining New Sequences, Expressed as RNA in Cells in Culture Producing MSRV-1, and Comprising an “env” Region of the NSRV-1 Retroviral Genome [0229]
  • A library of cDNA was produced according to the procedure described by the manufacturer of the “cDNA synthesis module, cDNA rapid adaptator ligation module, CDNA rapid cloning module and lambda gt10 in vitro packaging module” kits (Amersham, ref RPN1256Y/Z, RPN1712, RPN1713, RPN1717, N334Z), from the messenger RNA extracted from cells of a B lymphoblastoid line such as is described in Example 2, established from the lymphocytes of a patient suffering from MS and possessing reverse transcriptase activity which is detectable according to the technique described by Perron et al. (3). [0230]
  • Oligonucleotides were defined for amplifying the cDNA cloned into the nucleic acid library between the 3′ region of the clone PSJ17 (pol) and the 5′ (LTR) region of the clone FBd3. Control reactions were performed so as to check for the presence of contaminants (reaction with water). PCR reactions performed on the nucleic acids cloned into the library with different pairs of primers enabled a series [lacuna] clones linking pol sequences to the MSRV-1 type env or LTR sequences to be amplified. [0231]
  • Two clones are representative of the sequences obtained in the cellular cDNA library: [0232]
  • the clone JLBc1, whose sequence SEQ ID NO:52 is presented in FIG. 16; [0233]
  • the clone JLBc2, whose sequence SEQ ID NO:53 is presented in FIG. 17. [0234]
  • The sequences of the clones JLBc1 and JLBc2 are homologous to that of the clone FBd3, as is apparent in FIGS. 18 and 19. The homology between the clone JLBc1 and the clone JLBc2 is shown in FIG. 20. [0235]
  • The homologies between the clones JLBc1 and JLBc2 on the one hand and the HSERV9 sequence on the other hand are presented, respectively, in FIGS. 21 and 22. [0236]
  • It will be noted that the region of homology between JLB1, JLB2 and FBd3 comprises, with a few sequence and size variations of the “insert”, the additional sequence absent (“inserted”) in the HSERV-9 env sequence, as described in Example. 8. [0237]
  • It will also be noted that the cloned “pol” region is very homologous to HSERV-9, does not possess a reading frame (bearing in mind the sequence errors induced by the techniques used, including even the automatic sequencer) and diverges from the MSRV-1 sequences obtained from virions. In view of the fact that these sequences were cloned from the RNA of cells expressing MSRV-1 particles, it is probable that they originate from endogenous retroviral elements related to the ERV9 family; this is all the more likely for the fact that the pol and env genes are present on the same RNA which is clearly not the MSRV-1 genomic RNA. Some of these ERV9 elements possess functional LTRs which can be activated by replicative viruses coding for homologous or heterologous transactivators. Under these conditions, the relationship between MSRV-1 and HSERV-9 makes probable the transactivation of the defective (or otherwise) endogenous ERV9 elements by homologous, or even identical, MSRV-1 transactivating proteins. [0238]
  • Such a phenomenon may induce a viral interference between the expression of MSRV-1 and the related endogenous elements. Such an interference generally leads to a so-called “defective-interfering” expression, some features of which were to be found in the MSRV-1-infected cultures studied. Furthermore, such a phenomenon does not lack generation of the expression of polypeptides, or even of endogenous retroviral proteins which are not necessarily tolerated by the immune system. Such a scheme of aberrant expression of endogenous elements related to MSRV-1 and induced by the latter is liable to multiply the aberrant antigens, and hence to contribute to the induction of autoimmune processes such as are observed in MS. [0239]
  • It is, however, essential to note that the clones JLBc1 and JLBc2 differ from the ERV9 or HSERV9 sequence already described, in that they possess a longer env region comprising an additional region totally divergent from ERV9. Their kinship with the endogenous ERV9 family may hence be defined, but they clearly constitute novel elements never hitherto described. In effect, interrogation of the data banks of nucleic acid sequences available in version No. 15 (1995) of the “Entrez” software (NCBI, NIH, Bethesda, USA) did not enable a known homologous sequence in the env region of these clones to be identified. [0240]
  • EXAMPLE 10
  • OBTAINING SEQUENCES LOCATED IN [0241] TEE 5′ pol and 3′ gag REGION OF THE NSRV-1 RETROVIRAL GENOME As has already been described in Example 5, a PCR technique derived from the technique published by Frohman (19) was used. The technique derived makes it possible, using a specific primer at the 3′ end of the genome to be amplified, to elongate the sequence towards the 5′ region of the genome to be analysed. This technical variant is described in the documentation of the firm “Clontech Laboratories Inc., (Palo-Alto California, USA) supplied with its product “5′ -AmpliFINDER™ RACE Kit”, which was used on a fraction of virion purified as described above.
  • In order to carry out an amplification of the 5′ region of the MSRV-1 retroviral genome starting from the pol sequence already sequenced (clone F11-1) and extending towards the gag gene, MSRV-1 specific primers were defined. [0242]
  • The specific 3′ primers used in the kit protocol for the synthesis of the CDNA and the PCR amplification are, respectively, complementary to the following MSRV-1 sequences: [0243]
    cDNA:
    CCTGAGTTCTTGCACTAACCC (SEQ ID NO:54)
    amplification:
    GTCCGTTGGGTTTCCTTACTCCT (SEQ ID NO:55)
  • The products originating from the PCR were extracted after purification on agarose gel according to conventional methods (17), and then resuspended in 10 ml of distilled water. Since one of the properties of Taq polymerase consists in adding an adenine at the 3′ end of each ot the two DNA strands, the DNA obtained was inserted directly into a plasmid using the TA Cloning™ kit (British Biotechnology). The 2 μl of DNA solution were mixed with 5 μl of sterile distilled water, 1 μl of a 10-fold concentrated ligation buffer “10× LIGATION BUFFER”, 2 μl of “pCR™ VECTOR” (25 ng/ml) and 1 μl of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17). The plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®. The reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle seq uencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “automatic sequencer model 373 A [lacuna] apparatus according to the manufacturer's instructions. [0244]
  • This technical approach was applied to a sample of virion concentrated as described below from a mixture of culture supernatants produced by B lymphoblastoid lines such as are described in Example 2, established from lymphocytes of patients suffering from MS and possessing reverse transcriptase activity- which is detectable according to the technique described by Perron et al. (3): the culture supernatants are collected twice weekly, precentrifuged at 10,000 rpm for 30 minutes to remove cell debris and then frozen at −80° C. or used as they are for the following steps. The fresh or thawed supernatants are centrifuged on a cushion of 30% glycerol-PBS at 100,000 g for 2 h at 4° C. After removal of the supernatant, the sedimented pellet constitutes the sample of concentrated but unpurified virions. The pellet thereby obtained is then taken up in a small volume of an appropriate buffer for the extraction of RNA. The CDNA synthesis reaction mentioned above is carried out on this RNA extracted from concentrated extracellular virion. [0245]
  • RT-PCR amplification according to the technique mentioned above enabled the clone GM3 to be obtained, whose sequence, identified by [0246] SEQ ID NO 56, is presented in FIG. 23.
  • In FIG. 24, the sequence homology between the clone GMP3 and the HSERV-9 retrovirus is shown on the matrix chart by a continuous line, for any partial homology greater than or equal to 65%. [0247]
  • In summary, FIG. 25 shows the localization of the different clones studied above, relative to the known ERV9 genome. In FIG. 25, since the MSRV-1 env region is longer than the reference ERV9 env gene, the additional region is shown above the point of insertion according to a “V”, on the understanding that the inserted material displays a sequence and size vari-ability between the clones shown (JLBc1, JLBc2, FBd3). And FIG. 26 shows the position of different clones studied in the MSRV-1 pol* region. [0248]
  • By means of the clone GM3 described above, a possible reading frame could be defined, covering the whole of the pol gene, referenced according to SEQ ID NO:57, shown in the successive FIGS. 27[0249] a to 27 c.
  • EXAMPLE 11
  • Detection of Anti-MSRV-1 Specific Antibodies in Human Serum. [0250]
  • Identification of the sequence of the pol gene of the MSRV-1 retrovirus and of an open reading frame of this gene enabled the amino acid sequence SEQ ID NO:39 of a region of the said gene, referenced SEQ ID NO:40, to be determined (see FIG. 28). [0251]
  • Different synthetic peptides corresponding to fragments of the protein sequence of MSRV-1 reverse transcriptase encoded by the pol gene were tested for their antigenic specificity with respect to sera of patients suffering from MS and of healthy controls. [0252]
  • The peptides were synthesized chemically by solid-phase synthesis according to the Merrifield technique (Barany G, and Merrifielsd R. B, 1980, In the Peptides, 2, 1-284, Gross E and Meienhofer J, Eds., Academic Press, New York). The practical details are those described below. [0253]
  • a) Peptide Synthesis
  • The peptides were synthesized on a phenylacet-amidomethyl (PAM)/polystyrene/divinylbenzene resin (Applied Biosystems, Inc. Foster City, CA), using an “Applied Biosystems 430A” automatic synthesizer. The amino acids are coupled in the form of hydroxybenzo-triazole (HOBT) esters. The amino acids used are obtained from Novabiochem (Lauflerlfingen, Switzerland) or Bachem (Bubendorf, Switzerland). [0254]
  • The chemical synthesis was performed using a double coupling protocol with N-methylpyrrolidone (NMP) as solvent. The peptides were cut from the resin, as well as the side-chain protective groups, simultaneously, using hydrofluoric acid (HF) in a suitable apparatus (type I cleavage apparatus, Peptide Institute, Osaka, Japan). [0255]
  • For 1 g of peptidyl resin, 10 ml of HF, 1 ml of anisole and 1 ml of dimethyl sulphide 5DMS are used. The mixture is stirred for 45 minutes at −2° C. The HF is then evaporated off under vacuum. After intensive washes with ether, the peptide is eluted from the resin with 10% acetic acid and then lyophilized. [0256]
  • The peptides are purified by preparative high performance liquid chromatography on a VYDAC C18 type column (250×21 mm) (The Separation Group, Hesperia, CA, USA). Elution is carried out with an acetonitrile gradient at a flow rate of 22 ml/min. The fractions collected are monitored by an elution under isocratic conditions on a VYDAC® C18 analytical column (250×4.6 mm) at a flow rate of 1 ml/min. Fractions having the same retention time are pooled and lyophilized. The preponderant fraction is then analysed by analytical high performance liquid chromatography with the system described above. The peptide which is considered to be of acceptable purity manifests itself in a single peak representing not less than 95% of the chromatogram. [0257]
  • The purified peptides are then analysed with the object of monitoring their amino acid composition, using an Applied Biosystems 420H automatic amino acid analyser. Measurement of the (average) chemical molecular mass of the peptides is obtained using LSIMS mass spectrometry in the positive ion mode on a VG. ZAB.ZSEQ double focusing instrument connected to a DEC-[0258] VAX 2000 acquisition system (VG analytical Ltd, Manchester, England).
  • The reactivity of the different peptides was tested against sera of patients suffering from MS and against sera of healthy controls. This enabled a peptide designated POL2B to be selected, whose sequence is shown in FIG. 28 in the identifier SEQ ID NO:39, below, encoded by the pol gene of MSRV-1 (nucleotides 181 to 330). [0259]
  • b) Antigenic Properties
  • The antigenic properties of the POL2B peptide were demonstrated according to the ELISA protocol described below. [0260]
  • The lyophilized POL2B peptide was dissolved in sterile distilled water at a concentration of 1 mg/ml. This stock solution was aliquoted and kept at +4° C. for use over a fortnight, or frozen at −20° C. for use within 2 months. An aliquot is diluted in PBS (phosphate buffered saline) solution so as to obtain a final peptide concentration of 1 microgram/ml. 100 microlitres of this dilution are placed in each well of microtitration plates (“high-binding” plastic, COSTAR ref: 3590). The plates are covered with a “plate-sealer” type adhesive and kept overnight at +4° C. for the phase of adsorption of the peptide to the plastic. The adhesive is removed and the plates are washed three times with a volume of 300 micro-litres of a solution A (1 ×PBS, 0.05[0261] % Tween 20®), then inverted over an absorbent tissue. The plates thus drained are filled with 200 microlitres per well of a solution B (solution A+10% of goat serum), then covered with an adhesive and incubated for 45 minutes to 1 hour at 37° C. The plates are then washed three times with the solution A as described above.
  • The test serum samples are diluted beforehand to {fraction (1/50)} in the solution B, and 100 microlitres of each dilute test serum are placed in the wells of each microtitration plate. A negative control is placed in one well of each plate, in the form of 100 microlitres of buffer B. The plates covered with an adhesive are then incubated for 1 to 3 hours at 37° C. The plates are then washed three times with the solution A as described above. In parallel, a peroxidase-labelled goat antibody directed against human IgG (Sigma Immunochemicals ref. A6029) or IgM (Cappel ref. 55228) is diluted in the solution B (dilution {fraction (1/5000)}for the anti-IgG and {fraction (1/1000)}for the anti-IgM). 100 microlitres of the appropriate dilution of the labelled antibody are then placed in each well of the microtitration plates, and the plates covered with an adhesive are incubated for 1 to 2 hours at 37° C. A further washing of the plates is then performed as described above. In parallel, the peroxidase substrate is prepared according to the directions of the “Sigma fast OPD kit” (Sigma Immunochemicals, ref. P9187). 100 microlitres of substrate solution are placed in each well, and the plates are placed protected from light for 20 to 30 minutes at room temperature. [0262]
  • When the colour reaction has stabilized, the plates are placed immediately in an ELISA plate spectrophotometric reader, and the optical density (OD) of each well is read at a wavelength of 492 nm. Alternatively, 30 microlitres of 1N HCL are placed in each well to stop the reaction, and the plates are read in the spectrophotometer within 24 hours. [0263]
  • The serological samples are introduced in duplicate or in triplicate, and the optical density (OD) corresponding to the serum tested is calculated by taking the mean of the OD values obtained for the same sample at the same dilution. [0264]
  • The net OD of each serum corresponds to the mean OD of the serum minus the mean OD of the negative control (solution B: PBS, 0.05[0265] % Tween 20®, 10% goat serum).
  • c) Detection of Anti-MSRV-1 IgG Antibodies By ELISA:
  • The technique described above was used with the POLB2 peptide to test for the presence of anti-MSRV-1 specific IgG antibodies in the serum of 29 patients for whom a definite or probable diagnosis of MS was established according to the criteria of Poser (23), and of 32 healthy controls (blood donors). [0266]
  • FIG. 29 shows the results for each serum tested with an anti-IgG antibody. Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested. The ordinate axis gives the net OD at the top of the vertical bars. The first 29 vertical bars lying to the left of the vertical broken line represent the sera of 29 cases of MS tested, and the 32 vertical bars lying to the right of the vertical broken line represent the sera of 32 healthy controls (blood donors). [0267]
  • The mean of the net OD values for the MS sera tested is 0.62. The diagram enables 5 controls to be revealed whose net OD rises above the grouped values of the control population. These values may represent the presence of specific IgGs in symptomless seropositive patients. Two methods were hence evaluated in order to determine the statistical threshold of positivity of the test. [0268]
  • The mean of the net OD values for the controls, including the controls with high net OD values, is 0.36. Without the 5 controls whose net OD values are greater than or equal to 0.5, the mean of the “negative” controls is 0.33. The standard deviation of the negative controls is 0.10. A theoretical threshold of positivity may be calculated according to the formula: [0269]
  • threshold value (mean of the net OD values of the seronegative controls)+( 2 or 3×standard deviation of the net OD values of the seronegative controls). [0270]
  • In the first case, there are considered to be symptomless seropositives, and the threshold value is equal to 0.33+(2×0.10)=0.53. The negative results represent a non-specific “background” of the presence of antibodies directed specifically against an epitope of the peptide. [0271]
  • In the second case, if the set of controls consisting of blood donors in apparent good health is taken as a reference basis, without excluding the sera which are, on the face of it, seropositive, the standard deviation of the “non-MS controls” is 0.116. The threshold value then becomes 0.36+(2×0.116)=0.59. [0272]
  • According to this analysis, the test is specific for MS. In this respect, it is seen that the test is specific for MS, since, as shown in Table 1, no control has a net OD above this threshold. In fact, this result reflects the fact that the antibody titres in patients suffering from MS are, for the most part, higher than in vhealthy controls who have been in contact with MSRV-1. [0273]
    TABLE NO. 1
    MS CONTROLS
    0.681 0.3515
    1.0425 0.56
    0.5675 0.3565
    0.63 0.449
    0.588 0.2825
    0.645 0.55
    0.6635 0.52
    0.576 0.2535
    0.7765 0.55
    0.5745 0.51
    0.513 0.426
    0.4325 0.451
    0.7255 0.227
    0.859 0.3905
    0.6435 0.265
    0.5795 0.4295
    0.8655 0.291
    0.671 0.347
    0.596 0.4495
    0.662 0.3725
    0.602 0.181
    0.525 0.2725
    0.53 0.426
    0.565 0.1915
    0.517 0.222
    0.607 0.395
    0.3705 0.34
    0.397 0.307
    0.4395 0.219
    0.491
    0.2265
    0.2605
    MEAN 0.62 0.33
    STD DEV 0.14 0.10
    THRESHOLD VALUE 0.53
  • In accordance with the first method of calculation, and as shown in FIG. 29 and in the corresponding Table 1, 26 of the 29 MS sera give a positive result (net OD greater than or equal to 0.50), indicating the presence of IgGs specifically directed against the POL2B peptide, hence against a portion of the reverse transcriptase enzyme of the MSRV-1 retrovirus encoded by its pol gene, and consequently against the MSRV-1 retrovirus. Thus, approximately 90% of the MS patients tested have reacted against an epitope carried by the POL2B peptide and possess circulating IgGs directed against the latter. [0274]
  • Five out of 32 blood donors in apparent good health show a positive result. Thus, it is apparent that approximately 15% of the symptomless population may have been in contact with an epitope carried by the POL2B peptide under conditions which have led to an active immunization which manifests itself in the persistence of specific serum IgGs. These conditions are compatible with an immunization against the MSRV-1 retrovirus reverse transcriptase during an infection with (and/or reactivation of) the MSRV-1 retrovirus. The absence of apparent neurological pathology recalling MS in these seropositive controls may indicate that they are healthy carriers and have eliminated an infectious virus after immunizing themselves, or that they constitute an at-risk population of chronic carriers. In effect, epidemiological data showing that a pathogenic agent present in the environment of regions of high prevalence of MS may be the cause of this disease imply that a fraction of the population free from MS has necessarily been in contact with such a pathogenic agent. It has been shown that the MSRV-1 retrovirus constitutes all or part of this “pathogenic agent” at the source of MS, and it is hence normal for controls taken from a healthy population to possess IgG type antibodies against components of the MSRV-1 retrovirus. Thus, the difference in seroprevalence between the MS and control populations is extremely significant: “chi-squared”; test, p<0.001. These results hence point to an aetiopathogenic role of MSRV-1 in MS. [0275]
  • d) Detection of Anti-MSRV-1 IgM Antibodies By ELISA
  • The ELISA technique with the POL2B peptide was used to test for the presence of anti-MSRV-1 IgM specific antibodies in the serum of 36 patients for whom a definite or probable diagnosis of MS was established according to the criteria of Poser (23), and of 42 healthy controls (blood donors). [0276]
  • FIG. 30 shows the results for each serum tested with an anti-IgM antibody. Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested. The ordinate axis gives the net OD at the top of the vertical bars. The first 36 vertical bars lying to the left of the vertical line cutting the abscissa axis represent the sera of 36 cases. of MS tested, and the vertical bars lying to the right of the vertical broken line represent the sera of 42 healthy controls (blood donors). The horizontal line drawn in the middle of the diagram represents a theoretical threshold defining the boundary of the positive results (in which the top of the bar lies above) and the negative results (in which the top of the bar lies below). [0277]
  • The mean of the net OD values for the MS cases tested is 0.19. [0278]
  • The mean of the net OD values for the controls is 0.09. [0279]
  • The standard deviation of the negative controls is 0.05. [0280]
  • In view of the small difference between the mean and the standard deviation of the controls, the threshold of theoretical positivity may be calculated according to the formula: [0281]
  • threshold value=(mean of the net OD values of the seronegative controls)+(3×standard deviation of the net OD values of the seronegative controls). [0282]
  • The threshold value is hence equal to 0.09+(3×0.05)=0.26; or, in practice, 0.25. [0283]
  • The negative results represent a non-specific “background” of the presence of antibodies directed specifically against an epitope of the peptide. [0284]
  • According to this analysis, and as shown in FIG. 30 and in the corresponding Table 2, the IgM test is specific for MS, since no control has a net OD above the threshold. 7 of the 36 MS sera produce a positive IgM result; now, a study of the clinical data reveals that these positive sera were taken during a first attack of MS or an acute attack in untreated patients. It is known that IgMs directed against pathogenic agents are produced during primary infections or during reactivations following a latency phase of the said pathogenic agent. [0285]
  • The difference in seroprevalence between the MS and control populations is extremely significant: “chi-squared” test, p <0.001. [0286]
  • These results point to an aetiopathogenic role of MSRV-1 in MS. [0287]
  • The detection of IgM and IgG antibodies against the POL2B peptide enables the course of an MSRV-1 infection and/or of the viral reactivation of MSRV-1 to be evaluated. [0288]
    TABLE NO. 2
    MS CONTROLS
    0.064 0.243
    0.087 0.11
    0.044 0.098
    0.115 0.028
    0.089 0.094
    0.025 0.038
    0.097 0.176
    0.108 0.146
    0.018 0.049
    0.234 0.161
    0.274 0.113
    0.225 0.079
    0.314 0.093
    0.522 0.127
    0.306 0.02
    0.143 0.052
    0.375 0.062
    0.142 0.074
    0.157 0.043
    0.168 0.046
    1.051 0.041
    0.104 0.13
    0.187 0.153
    0.044 0.107
    0.053 0.178
    0.153 0.114
    0.07 0.078
    0.033 0.118
    0.104 0.177
    0.187 0.026
    0.044 0.024
    0.053 0.046
    0.153 0.116
    0.07 0.04
    0.033 0.028
    0.973 0.073
    0.008
    0.074
    0.141
    0.219
    0.047
    0.017
    MEAN 0.19 0.09
    STD. DEV. 0.23 0.05
    THRESHOLD VALUE 0.26
  • e) Search for Immunodominant Epitopes in the POL2B Peptide
  • In order to reduce the non-specific background and to optimize the detection of the responses of the anti-MSRV-1 antibodies, the synthesis of octapeptides, advancing in successive one amino acid steps, covering the whole of the sequence determined by POL2B, was carried out according to the protocol described below. [0289]
  • The chemical synthesis of overlapping octapeptides covering the amino acid sequence 61-110 shown in the identifier SEQ ID NO:39 was carried out on an activated cellulose membrane according to the technique of BERG et al. (1989.-J. Ann. Chem. Soc., 111, 8024-8026) marketed by Cambridge Research Biochemicals under the trade name Spotscan. This technique permits the simultaneous synthesis of a large number of peptides and their analysis. [0290]
  • The synthesis is carried out with esterified amino acids in which the α-amino group is protected with an FMOC group (Nova Biochem) and the side-chain groups with protective groups such as trityl, t-butyl ester or t-butyl ether. The esterified amino acids are solubilized in N-methylpyrrolidone (NMP) at a concentration of 300 nM, and 0.9 μl are applied to spots of deposit of bromophenol blue. After incubation for 15 minutes, a further application of amino acids is carried out according to another 15-minute incubation. If the coupling between two amino acids has taken place correctly, a coloration modification (change from blue to yellow-green) is observed. After three washes in DMF, an acetylation step is performed with acetic anhydride. Next, the terminal amino groups of the peptides in the process of synthesis are deprotected with 20% pyridine in DMF. The spots of deposit are restained with a 1% solution of bromophenol blue in DMF, washed three times with methanol and dried. This set of operations constitutes one cycle of addition of an amino acid, and this cycle is repeated until the synthesis is complete. When all the amino acids have been added, the NH2-terminal group of the last amino acid is deprotected with 20% piperidine in DMF and acetylated with acetic anhydride. The groups protecting the side chain are removed with a dichloromethane/trifluoroacetic acid/triisobutylsilane (5 ml/5 ml/250 ml) mixture. The lmmunoreactivity of the peptides is then tested by ELISA. [0291]
  • After synthesis of the different octapeptides in duplicate on two different membranes, the latter are rinsed with methanol and washed in TBS (0.1M Tris pH 7.2), then incubated overnight at room temperature in a saturation buffer. After several washes in TBS-T (0.1M Tris pH 7.2 - 0.05% Tween 20), one membrane is incubated with a {fraction (1/50)} dilution of a reference serum originating from a patient suffering from MS, and the other membrane with a {fraction (1/50)} dilution of a pool of sera of healthy controls. The membranes are incubated for 4 hours at room temperature. After washes with TBS-T, a β-galactosidase-labelled anti-human immunoglobulin conjugate (marketed by Cambridge Research Biochemicals) is added at a dilution of {fraction (1/200)}, and the mixture is incubated for two hours at room temperature. After washes of the membranes with 0.05% TBS-T and PBS, the immunoreactivity in the different spots is visualized by adding 5-bromo-4-chloro-3-indolyl β-D-galactopyranoside in potassium. The intensity of coloration of the spots is estimated qualitatively with a relative value from 0 to 5 as shown in the attached FIGS. [0292] 31 to 33.
  • In this way, it is possible to determine two immunodominant regions at each end of the POL2B peptide, corresponding, respectively, to the amino acid sequences 65-75 (SEQ ID NO:41) and 92-109 (SEQ ID NO:42), according to FIG. 34, and lying, respectively, between the octapeptides Phe-Cys-Ile-Pro-Val-Arg-Pro-Asp (FCIPVRPD) and Arg-Pro-Asp-Ser-Gln-Phe-Leu-Phe (RPDSQFLF), and Thr-Val-Leu-Pro-Gln-Gly-Phe-Arg (TVLPQGFR) and Leu-Phe-Gly-Gln-Ala-Leu-Ala-Gln (LFGQALAQ), and a region which is less reactive but apparently more specific, since it does not produce any background with the control serum, represented by the octapeptides Leu-Phe-Ala-Phe-Glu-Asp-Pro-Leu (LFAFEDPL) (SEQ ID NO:43) and Phe-Ala-Phe-Glu-Asp-Pro-Leu-Asn (FAFEDPLN) (SEQ ID NO:44). [0293]
  • These regions make it possible to define new peptides which are more specific and more immunoreactive according to the usual techniques. [0294]
  • It is thus possible, as a result of the discoveries made and the methods developed by the inventors, to carry out a diagnosis of MSRV-1 infection and/or reactivation and to evaluate a therapy in MS on the basis of its efficacy in “negativing” the detection of these agents in the patients' biological fluids. Furthermore, early detection in individuals not yet displaying neurological signs of MS could make it possible to institute a treatment which would be all the more effective with respect to the subsequent clinical course for the fact that it would precede the lesion stage which corresponds to the onset of neurological disorders. Now, at the present time, a diagnosis of MS cannot be established before a symptomatology of neurological lesions has set in, and hence no treatment is instituted before the emergence of a clinical picture suggestive of lesions of the central nervous system which are already significant. The diagnosis of an MSRV-1 and/or MSRV-2 infection and/or reactivation in man is hence of decisive importance, and the present invention provides the means of doing this. [0295]
  • It is thus possible, apart from carrying out a diagnosis of MSRV-1 infection and/or reactivation, to evaluate a therapy in MS on the basis of its efficacy in “negativing” the detection of these agents in the patients' biological fluids. [0296]
  • EXAMPLE 12
  • Obtaining a Clone LB19 Containing a Portion of the Gag Gene of the MSRV-1 Retrovirus [0297]
  • A PCR technique derived from the technique published by Gonzalez-Quintial R et al. (19) and PLAZA et al. (25) was used. From the total RNAs extracted from a fraction of virion purified as described above, the cDNA was synthesized using a specific primer (SEQ ID No.64) at the 3′ end of the genome to be amplified, using EXPAND™ REVERSE TRANSCRIPTASE (BOEHRINGER MANNHEIM). [0298]
  • cDNA: AAGGGGCATG GACGAGGTGG TGGCTTATTT (SEQ ID NO:65) (antisense) [0299]
  • After purification, a poly(G) tail was added at the 5′ end of the CDNA using the “Terminal transferases kit” marketed by the company Boehringer Mannheim, according to the manufacturer's protocol. [0300]
  • An anchoring PCR was carried out using the following 5′ and 3′ primers: AGATCTGCAG AATTCGATAT CACCCCCCCC CCCCCC (SEQ ID No. 91) (sense), and AAATGTCTGC GGCACCAATC TCCATGTT (SEQ ID No. 64) (antisense) [0301]
  • Next, a semi-nested anchoring PCR was carried out with the following 5′ and 3′ primers: AGATCTGCAG AATTCGATAT CA (SEQ ID No.92) (sense), and AAATGTCTGC GGCACCAATC TCCATGTT (SEQ ID No.64) (antisense) [0302]
  • The products originating from the PCR were purified after purification on agarose gel according to conventional methods (17), and then resuspended in 10 microlitres of distilled water. Since one of the properties of Taq polymerase consists in adding an adenine at the 3′ end of each of the two DNA strands, the DNA obtained was inserted directly into a plasmid using the TA CloningTm kit (British Biotechnology). The 2 μl of DNA solution were mixed with 5 μl of sterile distilled water, 1 μl of 10-fold concentrated ligation buffer “10×LIGATION BUFFER”, 2 μl of “pCR™ VECTOR” (25 ng/ml) and 1 μl of “T4 DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17). The plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analysed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®. The reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer, model 373 A” apparatus according to the manufacturer's instructions. [0303]
  • PCR amplification according to the technique mentioned above was used on a cDNA synthesized from the nucleic acids of fractions of infective particles purified on a sucrose gradient, according to the technique described by H. Perron (13), from culture supernatants of B lymphocytes of a patient suffering from MS, immortalized with Epstein-Barr virus (EBV) strain B95 and expressing retroviral particles associated with reverse transcriptase activity as described by Perron et al. (3) and in French [0304] Patent Applications MS 10, 11 and 12. the clone LB19, whose sequence, identified by SEQ ID NO:59, is presented in FIG. 35.
  • The clone makes it possible to define, with the clone GM3 previously sequenced and the clone G+E+A (see Example 15), a region of 690 base pairs representative of a significant portion of the gag gene of the MSRV-1 retrovirus, as presented in FIG. 36. This sequence designated SEQ ID NO:88 is reconstituted from different clones overlapping at their ends. This sequence is identified under the name MSRV-1 “gag*” region. In FIG. 36, a potential reading frame with the translation into amino acids is presented below the nucleic acid sequence. [0305]
  • EXAMPLE 13
  • Obtaining a Clone FBd13 Containing a pol Gene Region Related to the MSRV-1 Retrovirus and an Apparently Incomplete ENV Region Containing a Potential Reading Frame (ORF) for a Glycoprotein. [0306]
  • Extraction of viral RNAs
  • The RNAs were extracted according to the method briefly described below. [0307]
  • A pool of culture supernatant of B lymphocytes of patients suffering from MS (650 ml) is centrifuged for 30 minutes at 10,000 g. The viral pellet obtained is resuspended in 300 microlitres of PBS/10 mM MgCl2. The material is treated with a DNAse (100 mg/ml)/RNAse (50 mg/ml) mixture for 30 minutes at 37° C. and then with proteinase K (50 mg/ml) for 30 minutes at 46° C. [0308]
  • The nucleic acids are extracted with one volume of a phenol/0.1% SDS (V/V) mixture heated to 60° C., and then re-extracted with one volume of phenol/chloroform (1:1; V/V). [0309]
  • Precipitation of the material is performed with 2.5 V of ethanol in the presence of 0.1 V of sodium acetate pH =5.2. The pellet obtained after centrifugation is resuspended in 50 microlitres of sterile DEPC water. [0310]
  • The sample is treated again with 50 mg/ml of “RNAse free” DNAse for 30 minutes at room temperature, extracted with one volume of phenol/chloroform and precipitated in the presence of sodium acetate and ethanol. [0311]
  • The RNA obtained is quantified by an OD reading at 260 nm. The presence of MSRV-1 and the absence of DNA contaminant is monitored by a PCR and an MSRV-1-specific RTPCR associated with a specific ELOSA for the MSRV-1 genome. [0312]
  • Synthesis of cDNA
  • 5 mg of RNA are used to synthesize a cDNA primed with a poly(DT) oligonucleotide according to the instructions of the “cDNA Synthesis Module” kit (ref RPN 1256, Amersham) with a few modifications: The reverse transcription is performed at 45° C. instead of the recommended 42° C. [0313]
  • The synthesis product is purified by a double extraction and a double purification according to the manufacturer's instructions. [0314]
  • The presence of MSRV-1 is verified by an MSRV-1 PCR associated with a specific ELOSA for the MSRV-1 genome. [0315]
  • “Long Distance PCR”: (LD-PCR)
  • 500 ng of cDNA are used for the LD-PCR step (Expand Long Template System; Boehringer (ref.1681 842)). [0316]
  • Several pairs of oligonucleotides were used. Among these, the pair defined by the following primers: 5′ primer: GGAGAAGAGC AGCATAAGTG G (SEQ ID No. 66) 3′ primer: GTGCTGATTG GTGTATTTAC AATCC (SEQ ID No. 67). [0317]
  • The amplification conditions are as follows: [0318]
  • 94° C. 10 seconds [0319]
  • 56° C. 30 seconds [0320]
  • 68° C. 5 minutes; [0321]
  • 10 cycles, then 20 cycles with an increment of 20 seconds in each cycle on the elongation time. At the end of this first amplification, 2 microlitres of the amplification product are subjected to a second amplification under the same conditions as before. [0322]
  • The LD-PCR reactions are conducted in a Perkin model 9600 PCR apparatus in thin-walled microtubes (Boehringer). [0323]
  • The amplification products are monitored by electrophoresis of ⅕th of the amplification volume (10 microlitres) in 1% agarose gel. For the pair of primers described above, a band of approximately 1.7 Kb is obtained. [0324]
  • Cloning of the Amplified Fragment:
  • The PCR product was purified by passage through a preparative agarose gel and then through a Costar column (Spin; D. Dutcher) according to the supplier's instructions. 2 microlitres of the purified solution are joined up with 50 ng of vector PCRII according to the supplier's instructions (TA Cloning Kit; British Biotechnology)). [0325]
  • The recombinant vector obtained is isolated by transformation of competent DH5aF′ bacteria. The bacteria are selected using their resistance to ampicillin and the loss of metabolism for Xgal (=white colonies). The molecular structure of the recombinant vector is confirmed by plasmid minipreparation and hydrolysis with the enzyme EcoR1. [0326]
  • FBd13, a positive clone for all these criteria, was selected. A large-scale preparation of the recombinant plasmid was performed using the Midiprep Quiagen kit (ref 12243) according to the supplier's instructions. [0327]
  • Sequencing of the clone FBd13 is performed by means of the Perkin Prism Ready Amplitaq FS dye terminator kit (ref. 402119) according to the manufacturer's instructiions. The sequence reactions are introduced into a Perkin type 377 or 373A automatic sequencer. The sequencing strategy consists in gene walking carried out on both strands of the clone Fbd13. [0328]
  • The sequence of the clone FBd13 is identified by SEQ ID NO 58. [0329]
  • In FIG. 37, the sequence homology between the clone FBd13 and the HSERV-9 retrovirus is shown on the matrix chart by a continuous line for any partial homology greater than or equal to 70%. It can be seen that there are homologies in the flanking regions of the clone (with the pol gene at the 5′ end and with the env gene and then the LTR at the 3′ end) , but that the internal region is totally divergent and does not display any homology, even weak, with the env gene of HSERV-9. Furthermore, it is apparent that the clone FBd13 contains a longer “env” region than the one which is described for the defective endogenous HSERV-9; it may thus be seen that the internal divergent region constitutes an “insert” between the regions of partial homology with the HSERV-9 defective genes. [0330]
  • This additional sequence determines a potential orf, designated ORF B13, which is represented by its amino acid sequence SEQ ID NO:87. [0331]
  • The molecular structure of the clone FBd13 was analyzed using the GeneWork software and Genebank and SwissProt data banks. [0332]
  • 5 glycosylation sites were found. [0333]
  • The protein does not have significant homology with already known sequences. [0334]
  • It is probable that this clone originates from a recombination of an endogenous retroviral element (ERV), linked to the replication of MSRV-1. [0335]
  • Such a phenomenon does not lack generation of the expression of polypeptides, or even of endogenous retroviral proteins which are not necessarily tolerated by the immune system. Such a scheme of aberrant expression of endogenous elements related to MSRV-1 and/or induced by the latter is liable to multiply the aberrant antigens, and hence tends to contribute to the induction of autoimmune processes such as are observed in MS. It clearly constitutes a novel element never hitherto described. In effect, interrogation of the data banks of nucleic acid sequences available in version No. 19 (1996) of the “Entrez” software (NCBI, NIH, Bethesda, USA) did not enable a known homologous sequence comprising the whole of the env region of this clone to be identified. [0336]
  • EXAMPLE 14
  • Obtaining a Clone FP6 Containing a Portion of the pol Gene, with a Region Coding for the Reverse Transcriptase Enzyme Homologous to the Clone POL* MSRV-1, and a 3′ pol Region Divergent=from the Equivalent Sequences Described in the Clones POL*, tpol, FBd3, JLBc1 and JLBc2. [0337]
  • A 3′ RACE was performed on total RNA extracted from plasma of a patient suffering from MS. A healthy control plasma treated under the same conditions was used as negative control. The synthesis of cDNA was carried out with the following modified oligo(dT) primer: 5′ GACTCGCTGC [0338] AGATCGATTT TTTTTTTTTT TTTT 3′ (SEQ ID NO:68)
  • and Boehringer “Expand RT” reverse transcriptase according to the conditions recommended by the company. A PCR was performed with the enzyme Klentaq (Clontech) under the following conditions: 94° C. 5 min then 93° C. 1 min, 58° C. 1 min, 68° C. 3 min for 40 cycles and 68° C. for 8 min, and with a final reaction volume of 50 μl. [0339]
  • Primers used for the PCR: [0340]
  • 5′ primer, identified by SEQ ID NO:69 5′ [0341] GCCATCAAGC CACCCAAGAA CTCTTAACTT 3′;
  • 3′ primer, identified by SEQ ID NO:68 (=the same as for the cDNA) [0342]
  • A second, so-called “semi-nested” PCR was carried out with a 5′ primer located within the region already amplified. This second PCR was performed under the same experimental conditions as those used in the first PCR, using 10 μl of the amplification product originating from the first PCR. [0343]
  • Primers used for the semi-nested PCR: [0344]
  • 5′ primer, identified by SEQ ID NO:70 5′ [0345] CCAATAGCCA GACCATTATA TACACTAATT 3′;
  • 3′ primer, identified by SEQ ID NO:68 (=the same as for the cDNa) Primers SEQ ID NO:69 and SEQ ID NO:70 are specific for the pol* region: position No. 403 to No. 422 and No. 641 to No. 670, respectively. [0346]
  • An amplification product was thus obtained from the extracellular RNA extracted from the plasma of a patient suffering from MS. The corresponding fragment was not observed for the plasma of the healthy control. This amplification product was cloned in the following manner. [0347]
  • The amplified DNA was inserted into a plasmid using the TA Cloning™ kit. The 2 μl of DNA solution were mixed with 5 μl of sterile distilled water, 1 μl of a 10-fold concentrated ligation buffer “10× LIGATION BUFFER”, 2 μl of “pCR™ VECTOR” (25 ng/ml) and 1 μl of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning kit® (British Biotechnology). At the end of the procedure, the white columns of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17). The plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analyzed on agarose gel. Plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide was selected for sequencing of the insert, after hybridization with a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA cloning kit®. The reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer, model 373 A” apparatus according to the manufacturer's instructions. [0348]
  • The clone obtained, designated FP6, enables a region of 467 bp which is 89% homologous to the pol* region of the MSRV-1 retrovirus and a region of 1167 bp which is 64% homologous to the pol region of ERV-9 (No. 1634 to 2856) to be defined. [0349]
  • The clone FP6 is represented in FIG. 38 by its nucleotide sequence identified by SEQ ID NO:61. The three potential reading frames of this clone are indicated by their amino acid sequence under the nucleotide sequence. [0350]
  • EXAMPLE 15
  • Obtaining a Region Designated G+E+A Containing an ORF for a Retroviral Protease, by PCR Amplification of the Nucleic Acid Sequance Contained Between the 5′ Region Defined by the Clone “GM3” and the 3′ Region Defined by the Clone POL*, from the RNA Extracted from a Pool of Plasmas of Patients Suffering from MS. [0351]
  • Oligonucleotides specific for the MSRV-1 sequences already identified by the Applicant were defined in order to amplify the retroviral RNA originating from virions present in the plasma of patients suffering from MS. Control reactions were performed so as to monitor the presence of contaminants (reaction with water). The amplification consists of a step of RT-PCR followed by a “nested” PCR. Pairs of primers were defined for amplifying three overlapping regions (designated G, E and A) on the regions defined by the sequences of the clones GM3 and pol* described above. [0352]
  • Semi-Nested RT-PCR for Amplification of the Region G:
  • in the first RT-PCR cycle, the following primers are used: [0353]
  • primer 1: SEQ ID NO:71 (sense) [0354]
  • primer 2: SEQ ID NO:72 (antisense) [0355]
  • in the second.PCR cycle, the following primers are used: [0356]
  • primer 1: SEQ ID NO:73 (sense) [0357]
  • primer 4: SEQ ID NO:74 (antisense) [0358]
  • Nested RT-PCR for Amplification of the Region E:
  • in the first RT-PCR cycle, the following primers are used: [0359]
  • primer 5: SEQ ID NO:75 (sense) [0360]
  • primer 6: SEQ ID NO:76 (antisense) [0361]
  • in the second PCR cycle, the following primers are used: [0362]
  • primer 7: SEQ ID NO:77 (sense) [0363]
  • primer 8: SEQ ID NO:78 (antisense) [0364]
  • Semi-Nested RT-PCR for Amplification of the Region A:
  • in the first RT-PCR cycle, the following primers are used: [0365]
  • primer 9: SEQ ID NO:79 (sense) [0366]
  • primer 10: SEQ ID NO:80 (antisense) [0367]
  • in the second PCR cycle, the following primers are used: [0368]
  • primer 9: SEQ ID NO:81 (sense) [0369]
  • primer 11: SEQ ID NO:82 (antisense) [0370]
  • The primers and the regions G, E and A which they define are positioned as follows: [0371]
                                                           
    cDNA
         1     G     4 2
                 5 7           E            8 6
                                             3    A   11 10
    <-------------------------><-------------------------->
                   GM3                      POL*
  • The sequence of the region defined by the different clones G, E and A was determined after cloning and sequencing of the “nested” amplification products. [0372]
  • The clones G, E and A were assembled together by PCR with the [0373] primers 1 at the 5′ end of the fragment G and 11 at the 3′ end of the fragment A, the primers being described above. An approximately 1580-bp fragment G+E+A was amplified and inserted into a plasmid using the TA Cloning (trademark) kit. The sequence of the amplification product corresponding to G+E+A was determined and analysis of the G+E and E+A overlaps was carried out. The sequence is shown in FIG. 39, and corresponds to the sequence SEQ ID NO:89.
  • A reading frame coding for an MSRV-1 retroviral protease was found in the region E. The amino acid sequence of the protease, identified by SEQ ID NO:90, is presented in FIG. 40. [0374]
  • EXAMPLE 16
  • Obtaining a Clone LTRGAG12, Related to an Endogenous Retroviral Element (ERV) Close to MSRV-1, in the DNA of an MS Lymphoblastoid Line Producing Virions and Expressing the MSRV-1 Retrovirus. [0375]
  • A nested PCR was performed on the DNA extracted from a lymphoblastoid line (B lymphocytes immortalized with the EBV virus strain B95, as described above and as is well known to a person skilled in the art) expressing the MSRV-1 retrovirus and originating from peripheral blood lymphocytes of a patient suffering from MS. [0376]
  • In the first PCR step, the following primers are used: [0377]
    primer CTCGATTTCT TGCTGGGCCT TA (SEQ ID NO:83)
    4327:
    primer GTTGATTCCC TCCTCAAGCA (SEQ ID NO:84)
    3512:
  • This step comprises 35 amplification cycles with the following conditions: 1 min at 94° C., 1 min at 54° C. and 4 min at 72° C. [0378]
  • In the second PCR step, the following primers are used: [0379]
    primer 4294: CTCTACCAAT CAGCATGTGG (SEQ ID NO:85)
    primer 3591: TGTTCCTCTT GGTCCCTAT (SEQ ID NO:86)
  • This step comprises 35 amplification cycles with the following conditions: 1 min at 94° C., 1 min at 54° C. and 4 min at 72° C. [0380]
  • The products originating from the PCR were purified after purification on agarose gel according to conventional methods (17), and then resuspended in 10 ml of distilled water. Since one of the properties of Taq polymerase consists in adding an adenine at the 3′ end of each of the two DNA strands, the DNA obtained was inserted directly into a plasmid using the TA Cloning™ kit (British Biotechnology). The 2 μl of DNA solution were mixed with 5 μl of sterile distilled water, 1 μl of a 10-fold concentrated ligation buffer “10× LIGATION BUFFER”, 2 μl of “pCR™ VECTOR” (25 ng/ml) and 1 μl of “TA DNA LIGASE”. This mixture was incubated overnight at 12° C. The following steps were carried out according to the instructions of the TA Cloning® kit (British Biotechnology). At the end of the procedure, the white colonies of recombinant bacteria (white) were picked out in order to be cultured and to permit extraction of the plasmids incorporated according to the so-called “miniprep” procedure (17). The plasmid preparation from each recombinant colony was cut with a suitable restriction enzyme and analyzed on agarose gel. The plasmids possessing an insert detected under UV light after staining the gel with ethidium bromide were selected for sequencing of the insert, after hybridization witn a primer complementary to the Sp6 promoter present on the cloning plasmid of the TA Cloning Kit®. The reaction prior to sequencing was then performed according to the method recommended for the use of the sequencing kit “Prism ready reaction kit dye deoxyterminator cycle. sequencing kit” (Applied Biosystems, ref. 401384), and automatic sequencing was carried out with an Applied Biosystems “Automatic Sequencer, model 373 A” apparatus according to the manufacturer's instructions. [0381]
  • Thus, a clone designated LTRGAG12 could be obtained, and is represented by its internal sequence identified by SEQ ID NO:60. [0382]
  • This clone is probably representative of endogenous elements close to ERV-9, present in human DNA, in particular in the DNA of patients suffering from MS, and capable of interfering with the expression of the MSRV-1 retrovirus, hence capable of having a role in the pathogenesis associated with the MSRV-1 retrovirus and capable of serving as marker for a specific expression in the pathology in question. [0383]
  • EXAMPLE 17
  • Detection of Anti-MSRV-1 Specific Antibodies in Human Serum. [0384]
  • Identification of the sequence of the pol gene of the MSRV-1 retrovirus and of an open reading frame of this gene enabled the amino acid sequence SEQ ID NO:63 of a region of the said gene, referenced SEQ ID NO:62, to be determined. [0385]
  • Different synthetic peptides corresponding to fragments of the protein sequence of MSRV-1 reverse transcriptase encoded by the pol gene were tested for their antigenic specificity with respect to sera of patients suffering from MS and of healthy controls. [0386]
  • The peptides were synthesized chemically by solid-phase synthesis according to the Merrifield technique (22). The practical details are those described below. [0387]
  • a) Peptide Synthesis:
  • The peptides were synthesized on a phenylacetamidomethyl (PAM)/polystyrene/divinylbenzene resin (Applied Biosystems, Inc. Foster City, CA), using an “Applied Biosystems 430A” automatic synthesizer. The amino acids are coupled in the form of hydroxybenzotriazole (HOBT) esters. The amino acids used are obtained from Novabiochem (Läuflerlfingen, Switzerland) or Bachem (Bubendorf, Switzerland). [0388]
  • The chemical synthesis was performed using a double coupling protocol with N-methylpyrrolidone (NMP) as solvent. The peptides were cut from the resin, as well as the side-chain protective groups, simultaneously, using hydrofluoric acid (HF) in a suitable apparatus (type I cleavage apparatus, Peptide Instiute, Osaka, Japan). [0389]
  • For,1 g of peptidyl resin, 10 ml of HF, 1 ml of anisole and 1 ml of dimethyl sulphide SDMS are used. The mixture is stirred for 45 minutes at −2° C. The HP is then evaporated off under vacuum. After intensive washes with ether, the peptide is eluted from the resin with 10% acetic acid and then lyophilized. [0390]
  • The peptides are purified by preparative high performance liquid chromatography on a VYDAC C18 type column (250×21 mm) (The Separation Group, Hesperia, CA, USA). Elution is carried out with an acetonitrile gradient at a flow rate of 22 ml/min. The fractions collected are monitored by an elution under isocratic conditions on a VYDAC® C18 analytical column (250×4.6 mm) at a flow rate of 1 ml/min. Fractions having the same retention time are pooled and lyophilized. The preponderant fraction is then analysed by analytical high performance liquid chromatography with the system described above. The peptide which is considered to be of acceptable purity manifests itself in a single peak representing not less than 95% of the chromatogram. [0391]
  • The purified peptides are then analysed with the object of monitoring their amino acid composition, using an Applied Biosystems 420H automatic amino acid analyser. Measurement of the (average) chemical molecular mass of the peptides is obtained using LSIMS mass spectrometry in the positive ion mode on a VG. ZAB.ZSEQ double focusing instrument connected to a DEC-[0392] VAX 2000 acquisition system (VG analytical Ltd, Manchester, England).
  • The reactivity of the different peptides was tested against sera of patients suffering from MS and against sera of healthy controls. This enabled a peptide designated S24Q to be selected, whose sequence is identified by SEQ ID NO:63, encoded by a nucleotide sequence of the pol gene of MSRV-1 (SEQ ID NO:62). [0393]
  • b) Antigenic Properties:
  • The antigenic properties of the S24Q peptide were demonstrated according to the ELISA protocol described below. [0394]
  • The lyophilized S24Q peptide was dissolved in 10 % acetic acid at a concentration of 1 mg/ml. This stock solution was aliquoted and kept at +4° C. for use over a fortnight, or frozen at −20° C. for use within 2 months. An aliquot is diluted in PBS (phosphate buffered saline) solution so as to obtain a final peptide concentration of 5 micrograms/ml. 100 microlitres of this dilution are placed in each well of Nunc Maxisorb (trade name) microtitration plates. The plates are covered with a “plate-sealer” type adhesive and kept for 2 hours at +37° C. for the phase of adsorption of the peptide to the plastic. The adhesive is removed and the plates are washed three times with a volume of 300 microlitres of a solution A (1 ×PBS, 0.05[0395] % Tween 20®), then inverted over an absorbent tissue. The plates thus drained are filled with 250 microlitres per well of a solution B (solution A+10% of goat serum), then covered with an adhesive and incubated for 1 hour at 37° C. The plates are then washed three times with the solution A as described above.
  • The test serum samples are diluted beforehand to {fraction (1/100)} in the solution B, and 100 microlitres of each dilute test serum are placed in the wells of each microtitration plate. A negative control is placed in one well of each plate, in the form of 100 microlitres of buffer B. The plates covered with an adhesive are then incubated for 1 [0396] hour 30 min at 37° C. The plates are then washed three times with the solution A as described above. For the IgG response, a peroxidase-labelled goat antibody directed against human IgG (marketed by Jackson Immuno Research Inc.) is diluted in the solution B (dilution {fraction (1/10,000)}). 100 microlitres of the appropriate dilution of the labelled antibody are then placed in each well of the microtitration plates, and the plates covered with an adhesive are incubated for 1 hour at 37° C. A further washing of the plates is then performed as described above. In parallel, the peroxidase substrate is prepared according to the directions of the BioMérieux kits. 100 microlitres of substrate solution are placed in each well, and the plates are placed protected from light for 20 to 30 minutes at room temperature.
  • When the colour reaction has stabilized, 50 microlitres of Color 2 (BioMérieux trade name) are placed in each well in order to stop the reaction. The plates are placed immediately in an ELISA plate spectrophotometric reader, and the optical density (OD) of each well is read at a wavelength of 492 nm. [0397]
  • The serological samples are introduced in duplicate or in triplicate, and the optical density (OD) corresponding to the serum tested is calculated by taking the mean of the OD values obtained for the same sample at the same dilution. [0398]
  • The net OD of each serum corresponds to the mean OD of the serum minus the mean OD of the negative control (solution B: PBS, 0.05[0399] % Tween 20®, 10% goat serum).
  • c) Detection of Anti-MSRV-1 IgG Antibodies (S240) by ELISA
  • The technique described above was used with the S24Q peptide to test for the presence of anti-MSRV-1 specific IgG antibodies in the serum of 15 patients for whom a definite diagnosis of MS was established according to the criteria of Poser (23), and of 15 healthy controls (blood donors). [0400]
  • FIG. 41 shows the results for each serum tested with an anti-IgG antibody. Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested. The ordinate axis gives the net OD at the top of the vertical bars. The first 15 vertical bars lying to the left of the vertical broken line represent the sera of 15 healthy controls (blood donors), and the 15 vertical bars lying to the right of the vertical broken line represent the sera of 15 cases of MS tested. The diagram enables 2 controls to be revealed whose OD rises above the grouped values of the control population. These values may represent the presence of specific IgGs in symptomless seropositive patients. Two methods were hence evaluated in order to determine the statistical threshold of positivity of the test. [0401]
  • The mean of the net OD values for the controls, including the controls with high net OD values, is 0.129 and the standard deviation is 0.06. Without the 2 controls whose OD values are greater than 0.2, the mean of the “negative” controls is 0.107 and the standard deviation is 0.03. A theoretical threshold of positivity may be calculated according to the formula: [0402]
  • threshold value (mean of the net OD values of the negative controls)+(2 or 3×standard deviation of the net OD values of the negative controls). [0403]
  • In the first case, there are considered to be symptomless seropositives, and the threshold value is equal to 0.11+(3×0.03)=0.20. The negative results represent a non-specific “background” of the presence of antibodies directed specifically against an epitope of the peptide. [0404]
  • In the second case, if the set of controls consisting of blood donors in apparent good health is taken as a reference basis, without excluding the sera which are, on the face of it, seropositive, the standard deviation of the “non-MS controls” is 0.116. The threshold value then becomes 0.13 +(3×0.06)=0.31. [0405]
  • According to this latter analysis, the test is specific for MS. In this respect, it is seen that the test is specific for MS, since, as shown in Table 1, no control has a net OD above this threshold. In fact, this result reflects the fact that the antibody titres in patients suffering from MS are, for the most part, higher than in healthy controls who have been in contact with MSRV-1. [0406]
  • In accordance with the first method of calculation, and as shown in FIG. 41 and in Table 3, 6 of the 15 MS sera give a positive result (OD greater than or equal to 0.2), indicating the presence of IgGs specifically directed against the S24Q peptide, hence against a portion of the reverse transcriptase enzyme of the MSRV-1 retrovirus encoded by its pol gene, and consequently against the MSRV-1 retrovirus. [0407]
  • Thus, approximately 40% of the MS patients tested have reacted against an epitope carried by the S24Q peptide and possess circulating IgGs directed against the latter. [0408]
  • Two out of 15 blood donors in apparent good health show a positive result. Thus, it is apparent that approximately 13% of the symptomless population may have been in contact with an epitope carried by the S24Q peptide under conditions which have led to an active immunization which manifests itself in the persistence of specific serum IgGs. These conditions are compatible with an immunization against the MSRV-1 retrovirus reverse transcriptase during an infection with (and/or reactivation of) the MSRV-1 retrovirus. The absence of apparent neurological pathology recalling MS in these seropositive controls may indicate that they are healthy carriers and have eliminated an infectious virus after immunizing themselves, or that they constitute an at-risk population of chronic carriers. In effect, epidemiological data showing that a pathogenic agent present in the environment of regions of high prevalence of MS may be the cause of this disease imply that a fraction of the population free from MS has necessarily been in contact with such a pathogenic agent. It has been shown that the MSRV-1 retrovirus constitutes all or part of this “pathogenic agent” at the source of MS, and it is hence normal for controls taken from a healthy population to possess IgG type antibodies against components of the MSRV-1 retrovirus. [0409]
  • Lastly, the detection of anti-S24Q antibodies in only one out of two MS cases tested here may reflect the fact that this peptide does not represent an immunodominant MSRV-1 epitope, that inter-individual strain variations may induce an immunization against a divergent peptide motif in the same region, or that the course of the disease and the treatments followed may modulate over time the antibody response against the S24Q peptide. [0410]
    TABLE NO. 3
    CONTROLS
    MS
    0.101
    0.136
    0.058
    0.391
    0.126
    0.37
    0.131
    0.119
    0.105
    0.267
    0.294
    0.141
    0.116
    0.102
    0.088
    0.18
    0.105
    0.411
    0.172
    0.164
    0.137
    0.049
    0.223
    0 644
    0.08
    0.268
    0.073
    0.065
    0.132
    0.074
    Mean 0.129
    Standard Deviation 0.06
    Threshold 0.31
  • d) Detection of Anti-MSRV-1 IgM Antibodies by ELISA
  • The ELISA technique with the S24Q peptide was used to test for the presence of anti-MSRV-1 IgM specific antibodies in the same sera as above. [0411]
  • FIG. 42 shows the results for each serum tested with an anti-IgM antibody. Each vertical bar represents the net optical density (OD at 492 nm) of a serum tested. The ordinate axis gives the net OD at the top of the vertical bars. The first 15 vertical bars lying to the left of the vertical line cutting the abscissa axis represent the sera of 15 healthy controls (blood donors), and the vertical bars lying to the right of the vertical broken line represent the sera of 15 cases of MS tested. [0412]
  • The mean of the OD values for the MS cases tested is 1.6. [0413]
  • The mean of the net OD values for the controls is 0.7. [0414]
  • The standard deviation of the negative controls is 0.6. [0415]
  • The threshold of theoretical positivity may be calculated according to the formula: [0416]
  • threshold value=(mean of the OD values of the negative controls)+(3×standard deviation of the OD values of the negative controls). [0417]
  • The threshold value is hence equal to 0.7+(3×0.6)=2.5; [0418]
  • The negative results represent a non-specific “background” of the presence of antibodies directed specifically against an epitope of the peptide. [0419]
  • According to this analysis, and as shown in FIG. 42 and in the corresponding Table 4, the IgM test is specific for MS, since no control has a net OD above the threshold. 6 of the 15 MS sera produce a positive IgM result [0420]
  • The difference in seroprevalence between the MS and control populations is extremely significant: “chi-squared” test, p <0.002. [0421]
  • These results point to an aetiopathogenic role of MSRV-1 in MS. [0422]
  • Thus, the detection of IgM and IgG antibodies against the S24Q peptide makes it possible to evaluate, alone or in combination with other MSRV-1 peptides, the course of an MSRV-1 infection and/or of the viral reactivation of MSRV-1. [0423]
    TABLE NO. 4
    CONTROLS
    MS
    1,449
    0,974
    0,371
    6,117
    0,448
    2,883
    0,456
    1,945
    0,885
    1,787
    2,235
    0,273
    0,301
    1,766
    0,138
    0,668
    0,16
    2,603
    1,073
    0,802
    1,366
    0,245
    0,283
    0,147
    0,262
    2,441
    0,585
    0,287
    0,356
    0,589
    Mean 0,7
    Standard Deviation 0,6
    Threshold 2,5
  • It is possible, as a result of the new discoveries made and the new methods developed by the inventors, to permit the improved implementation of diagnostic tests for MSRV-1 infection and/or reactivation and to evaluate a therapy in MS and/or RA on the basis of its efficacy in “negativing” the detection of these agents in the patient's biological fluids. Furthermore, early detection in individuals not yet displaying neurological signs of MS or rheumatological signs of RA could make it possible to institute a treatment which would be all the more effective with respect to the subsequent clinical course for the fact that it would precede the lesion stage which corresponds to the onset of the clinical disorders. Now, at the present time, a diagnosis of MS or RA cannot be established before a symptomatology of lesions has set in, and hence no treatment is instituted before the emergence of a clinical picture suggestive of lesions which are already significant. The diagnosis of an MSRV-1 and/or MSRV-2 infection and/or reactivation in man is hence of decisive importance, and the present invention provides the means of doing this. [0424]
  • It is thus possible, apart from carrying out a diagnosis of MSRV-1 infection and/or reactivation, to evaluate a therapy in MS on the basis of its efficacy in “negativing” the detection of these agents in the patients' biological fluids. [0425]
  • BIBLIOGRAPHY
  • (1) Norrby E., Prog. Med. Virol., 1978; 24, 1-39. [0426]
  • (2) Johnson R. T., “Handbook of clinical neurology, 47 Demyelinating diseases”, Vinken P. and Bruyn G. W., eds. Amsterdam, Elsevier Science Publishing, 1985, 319-336. [0427]
  • (3) Perron R. et al., Res. Virol. 1989, 140, 551-561. [0428]
  • (4) Perron H. et al., “Current concepts in multiple sclerosis” Wiethölter et al., eds. Amsterdam, Elsevier, 1991, 111-116. [0429]
  • (5) Perron H. et al., The Lancet 1991, 337, 862-863. [0430]
  • (6) Perron H. et al., J. Gen. Virol. 1993, 74, 65-72. [0431]
  • (7) Fields and Knipe, Fondamental Virology 1986, Rev Press N.Y. [0432]
  • (8) Nielsen P. E. et al., Science 1991; 254, 1497-1500. [0433]
  • (9) Maniatis et al., Molecular Cloning, Cold Spring Harbour, 1982. [0434]
  • (10) Southern. E. M., J. Mol. Biol. 1975, 98, 503. [0435]
  • (11) Dunn A. R. and Hassel J. A., [0436] Cell 1977, 12, 23,
  • (12) Shih et al., J. Virol. 1989, 63, 64-75. [0437]
  • (13) Perron H. et al., Res. Vir. 1992, 143, 337-350. [0438]
  • (14) Meyerhans et al., Cell 1989, 58, 901-910. [0439]
  • (15) Linial M. L. and Miller A. D., “Current topics in microbiology and immunobiology. Retroviruses, strategies of replication” vol. 157, 125-152; Swanstrom R. and Vogt P. K., editors, Springer-Verla-g, Heidelberg 1990. [0440]
  • (16) Lori F. et al., J. Virol. 1992, 66, 5067-5074. [0441]
  • (17) Sambrook J., Fritsch E. F. and Maniatis T., Molecular cloning, a laboratory manual. Cold Spring Harbour Laboratory Press, 1989. [0442]
  • (18) La Mantia et al., Nucleic Acids Research 1991, 19, 1513-1520. [0443]
  • (19) Gonzales-Quintial R, Baccala R, Pope R M and Theofilopoulos N, J. Clin. Invest, Vol. 97, [0444] Number 5, ppl335-1343, 1996.
  • (20) Chomzynski P. and N. Sacchi, Analytical Biochemistry 1987, 162, 156-159. [0445]
  • (21) F. Mallet et al., Journal of Clinical Microbiology 1993; 31, 1444-1449. [0446]
  • (22) G. Barany and R. B. Merrifielsd, 1980, In the Peptides, 2, 1-284, Gross E and Meienhofer J, Eds., Academic Press, New York. [0447]
  • (23) Poser et al., Gbers G. C. eds. The diagnosis of multiple sclerosis Thieme Stratton Inc, New York 1984: 225-229. [0448]
  • (24) La Mantia et al., [0449] Nucleic Acid Research 1989, 17, 5913-22.
  • (25) PLAZA, A; KONO, D. H.; THEOFILOPOULOS, A. N. NEWHUMAN [0450]
  • Vβ 12DD GENES AND POLYMORPHIC VARIANTS. J. Tm; [0451]
  • 147(12): 4360-4365, 1991. [0452]
  • 1 92 1158 base pairs nucleotide single linear cDNA 1 CCCTTTGCCA CTACATCAAT TTTAGGAGTA AGGAAACCCA ACGGACAGTG GAGGTTAGTG 60 CAAGAACTCA GGATTATCAA TGAGGCTGTT GTTCCTCTAT ACCCAGCTGT ACCTAACCCT 120 TATACAGTGC TTTCCCAAAT ACCAGAGGAA GCAGAGTGGT TTACAGTCCT GGACCTTAAG 180 GATGCCTTTT TCTGCATCCC TGTACGTCCT GACTCTCAAT TCTTGTTTGC CTTTGAAGAT 240 CCTTTGAACC CAACGTCTCA ACTCACCTGG ACTGTTTTAC CCCAAGGGTT CAGGGATAGC 300 CCCCATCTAT TTGGCCAGGC ATTAGCCCAA GACTTGAGTC AATTCTCATA CCTGGACACT 360 CTTGTCCTTC AGTACATGGA TGATTTACTT TTAGTCGCCC GTTCAGAAAC CTTGTGCCAT 420 CAAGCCACCC AAGAACTCTT AACTTTCCTC ACTACCTGTG GCTACAAGGT TTCCAAACCA 480 AAGGCTCGGC TCTGCTCACA GGAGATTAGA TACTNAGGGC TAAAATTATC CAAAGGCACC 540 AGGGCCCTCA GTGAGGAACG TATCCAGCCT ATACTGGCTT ATCCTCATCC CAAAACCCTA 600 AAGCAACTAA GAGGGTTCCT TGGCATAACA GGTTTCTGCC GAAAACAGAT TCCCAGGTAC 660 ASCCCAATAG CCAGACCATT ATATACACTA ATTANGGAAA CTCAGAAAGC CAATACCTAT 720 TTAGTAAGAT GGACACCTAC AGAAGTGGCT TTCCAGGCCC TAAAGAAGGC CCTAACCCAA 780 GCCCCAGTGT TCAGCTTGCC AACAGGGCAA GATTTTTCTT TATATGCCAC AGAAAAAACA 840 GGAATAGCTC TAGGAGTCCT TACGCAGGTC TCAGGGATGA GCTTGCAACC CGTGGTATAC 900 CTGAGTAAGG AAATTGATGT AGTGGCAAAG GGTTGGCCTC ATNGTTTATG GGTAATGGNG 960 GCAGTAGCAG TCTNAGTATC TGAAGCAGTT AAAATAATAC AGGGAAGAGA TCTTNCTGTG 1020 TGGACATCTC ATGATGTGAA CGGCATACTC ACTGCTAAAG GAGACTTGTG GTTGTCAGAC 1080 AACCATTTAC TTAANTATCA GGCTCTATTA CTTGAAGAGC CAGTGCTGNG ACTGCGCACT 1140 TGTGCAACTC TTAAACCC 1158 297 base pairs nucleotide single linear cDNA 2 CCCTTTGCCA CTACATCAAT TTTAGGAGTA AGGAAACCCA ACGGACAGTG GAGGTTAGTG 60 CAAGAACTCA GGATTATCAA TGAGGCTGTT GTTCCTCTAT ACCCAGCTGT ACCTAACCCT 120 TATACAGTGC TTTCCCAAAT ACCAGAGGAA GCAGAGTGGT TTACAGTCCT GGACCTTAAG 180 GATGCCTTTT TCTGCATCCC TGTACGTCCT GACTCTCAAT TCTTGTTTGC CTTTGAAGAT 240 CCTTTGAACC CAACGTCTCA ACTCACCTGG ACTGTTTTAC CCCAAGGGTT CAAGGGA 297 85 base pairs nucleotide single linear cDNA 3 GTTTAGGGAT ANCCCTCATC TCTTTGGTCA GGTACTGGCC CAAGATCTAG GCCACTTCTC 60 AGGTCCAGSN ACTCTGTYCC TTCAG 85 86 base pairs nucleotide single linear cDNA 4 GTTCAGGGAT AGCCCCCATC TATTTGGCCA GGCACTAGCT CAATACTTGA GCCAGTTCTC 60 ATACCTGGAC AYTCTYGTCC TTCGGT 86 85 base pairs nucleotide single linear cDNA 5 GTTCARRGAT AGCCCCCATC TATTTGGCCW RGYATTAGCC CAAGACTTGA GYCAATTCTC 60 ATACCTGGAC ACTCTTGTCC TTYRG 85 85 base pairs nucleotide single linear cDNA 6 GTTCAGGGAT AGCTCCCATC TATTTGGCCT GGCATTAACC CGAGACTTAA GCCAGTTCTY 60 ATACGTGGAC ACTCTTGTCC TTTGG 85 111 base pairs nucleotide single linear cDNA 7 GTGTTGCCAC AGGGGTTTAR RGATANCYCY CATCTMTTTG GYCWRGYAYT RRCYCRAKAY 60 YTRRGYCAVT TCTYAKRYSY RGSNAYTCTB KYCCTTYRGT ACATGGATGA C 111 645 base pairs nucleotide single linear cDNA 8 TCAGGGATAG CCCCCATCTA TTTGGCCAGG CATTAGCCCA AGACTTGAGT CAATTCTCAT 60 ACCTGGACAC TCTTGTCCTT CAGTACATGG ATGATTTACT TTTAGTCGCC CGTTCAGAAA 120 CCTTGTGCCA TCAAGCCACC CAAGAACTCT TAACTTTCCT CACTACCTGT GGCTACAAGG 180 TTTCCAAACC AAAGGCTCGG CTCTGCTCAC AGGAGATTAG ATACTNAGGG CTAAAATTAT 240 CCAAAGGCAC CAGGGCCCTC AGTGAGGAAC GTATCCAGCC TATACTGGCT TATCCTCATC 300 CCAAAACCCT AAAGCAACTA AGAGGGTTCC TTGGCATAAC AGGTTTCTGC CGAAAACAGA 360 TTCCCAGGTA CASCCCAATA GCCAGACCAT TATATACACT AATTANGGAA ACTCAGAAAG 420 CCAATACCTA TTTAGTAAGA TGGACACCTA CAGAAGTGGC TTTCCAGGCC CTAAAGAAGG 480 CCCTAACCCA AGCCCCAGTG TTCAGCTTGC CAACAGGGCA AGATTTTTCT TTATATGCCA 540 CAGAAAAAAC AGGAATAGCT CTAGGAGTCC TTACGCAGGT CTCAGGGATG AGCTTGCAAC 600 CCGTGGTATA CCTGAGTAAG GAAATTGATG TAGTGGCAAA GGGTT 645 741 base pairs nucleotide single linear cDNA 9 CAAGCCACCC AAGAACTCTT AAATTTCCTC ACTACCTGTG GCTACAAGGT TTCCAAACCA 60 AAGGCTCAGC TCTGCTCACA GGAGATTAGA TACTTAGGGT TAAAATTATC CAAAGGCACC 120 AGGGGCCTCA GTGAGGAACG TATCCAGCCT ATACTGGGTT ATCCTCATCC CAAAACCCTA 180 AAGCAACTAA GAGGGTTCCT TAGCATGATC AGGTTTCTGC CGAAAACAAG ATTCCCAGGT 240 ACAACCAAAA TAGCCAGACC ATTATATACA CTAATTAAGG AAACTCAGAA AGCCAATACC 300 TATTTAGTAA GATGGACACC TAAACAGAAG GCTTTCCAGG CCCTAAAGAA GGCCCTAACC 360 CAAGCCCCAG TGTTCAGCTT GCCAACAGGG CAAGATTTTT CTTTATATGG CACAGAAAAA 420 ACAGGAATCG CTCTAGGAGT CCTTACACAG GTCCGAGGGA TGAGCTTGCA ACCCGTGGCA 480 TACCTGAATA AGGAAATTGA TGTAGTGGCA AAGGGTTGGC CTCATNGTTT ATGGGTAATG 540 GNGGCAGTAG CAGTCTNAGT ATCTGAAGCA GTTAAAATAA TACAGGGAAG AGATCTTNCT 600 GTGTGGACAT CTCATGATGT GAACGGCATA CTCACTGCTA AAGGAGACTT GTGGTTGTCA 660 GACAACCATT TACTTAANTA TCAGGCTCTA TTACTTGAAG AGCCAGTGCT GNGACTGCGC 720 ACTTGTGCAA CTCTTAAACC C 741 93 base pairs nucleotide single linear cDNA 10 TGGAAAGTGT TGCCACAGGG CGCTGAAGCC TATCGCGTGC AGTTGCCGGA TGCCGCCTAT 60 AGCCTCTACA TGGATGACAT CCTGCTGGCC TCC 93 96 base pairs nucleotide single linear cDNA 11 TTGGATCCAG TGYTGCCACA GGGCGCTGAA GCCTATCGCG TGCAGTTGCC GGATGCCGCC 60 TATAGCCTCT ACGTGGATGA CCTSCTGAAG CTTGAG 96 748 base pairs nucleotide single linear cDNA 12 TGCAAGCTTC ACCGCTTGCT GGATGTAGGC CTCAGTACCG GNGTGCCCCG CGCGCTGTAG 60 TTCGATGTAG AAAGCGCCCG GAAACACGCG GGACCAATGC GTCGCCAGCT TGCGCGCCAG 120 CGCCTCGTTG CCATTGGCCA GCGCCACGCC GATATCACCC GCCATGGCGC CGGAGAGCGC 180 CAGCAGACCG GCGGCCAGCG GCGCATTCTC AACGCCGGGC TCGTCGAACC ATTCGGGGGC 240 GATTTCCGCA CGACCGCGAT GCTGGTTGGA GAGCCAGGCC CTGGCCAGCA ACTGGCACAG 300 GTTCAGGTAA CCCTGCTTGT CCCGCACCAA CAGCAGCAGG CGGGTCGGCT TGTCGCGCTC 360 GTCGTGATTG GTGATCCACA CGTCAGCCCC GACGATGGGC TTCACGCCCT TGCCACGCGC 420 TTCCTTGTAG ANGCGCACCA GCCCGAAGGC ATTGGCGAGA TCGGTCAGCG CCAAGGCGCC 480 CATGCCATCT TTGGCGGCAG CCTTGACGGC ATCGTCGAGA CGGACATTGC CATCGACGAC 540 GGAATATTCG GAGTGGAGAC GGAGGTGGAC GAAGCGCGGC GAATTCATCC GCGTATTGTA 600 ACGGGTGACA CCTTCCGCAA AGCATTCCGG ACGTGCCCGA TTGACCCGGA GCAACCCCGC 660 ACGGCTGCGC GGGCAGTTAT AATTTCGGCT TACGAATCAA CGGGTTACCC CAGGGCGCTG 720 AAGCCTATCG CGTGCAGTTG CCGGATGC 748 18 base pairs nucleotide single linear cDNA 13 GCATCCGGCA ACTGCACG 18 20 base pairs nucleotide single linear cDNA 14 GTAGTTCGAT GTAGAAAGCG 20 18 base pairs nucleotide single linear cDNA 15 GCATCCGGCA ACTGCACG 18 23 base pairs nucleotide single linear cDNA 16 AGGAGTAAGG AAACCCAACG GAC 23 19 base pairs nucleotide single linear cDNA 17 TAAGAGTTGC ACAAGTGCG 19 21 base pairs nucleotide single linear cDNA 18 TCAGGGATAG CCCCCATCTA T 21 24 base pairs nucleotide single linear cDNA 19 AACCCTTTGC CACTACATCA ATTT 24 15 base pairs nucleotide single linear cDNA 5, 7, 10, 13 N represents inosine (i) 20 GGTCNTNCCN CANGG 15 21 base pairs nucleotide single linear cDNA 21 TTAGGGATAG CCCTCATCTC T 21 21 base pairs nucleotide single linear cDNA 22 TCAGGGATAG CCCCCATCTA T 21 24 base pairs nucleotide single linear cDNA 23 AACCCTTTGC CACTACATCA ATTT 24 23 base pairs nucleotide single linear cDNA 24 GCGTAAGGAC TCCTAGAGCT ATT 23 18 base pairs nucleotide single linear cDNA 25 TCATCCATGT ACCGAAGG 18 20 base pairs nucleotide single linear cDNA 26 ATGGGGTTCC CAAGTTCCCT 20 20 base pairs nucleotide single linear cDNA 27 GCCGATATCA CCCGCCATGG 20 18 base pairs nucleotide single linear cDNA 28 GCATCCGGCA ACTGCACG 18 20 base pairs nucleotide single linear cDNA 29 CGCGATGCTG GTTGGAGAGC 20 20 base pairs nucleotide single linear cDNA 30 TCTCCACTCC GAATATTCCG 20 26 base pairs nucleotide single linear cDNA 31 GATCTAGGCC ACTTCTCAGG TCCAGS 26 23 base pairs nucleotide single linear cDNA 6, 12, 19 N represents inosine (i) 32 CATCTNTTTG GNCAGGCANT AGC 23 24 base pairs nucleotide single linear cDNA 33 CTTGAGCCAG TTCTCATACC TGGA 24 22 base pairs nucleotide single linear cDNA 34 AGTGYTRCCM CARGGCGCTG AA 22 22 base pairs nucleotide single linear cDNA 35 GMGGCCAGCA GSAKGTCATC CA 22 22 base pairs nucleotide single linear cDNA 36 GGATGCCGCC TATAGCCTCT AC 22 22 base pairs nucleotide single linear cDNA 37 AAGCCTATCG CGTGCAGTTG CC 22 40 base pairs nucleotide single linear cDNA 38 TAAAGATCTA GAATTCGGCT ATAGGCGGCA TCCGGCAAGT 40 50 amino acids amino acid linear peptide 39 Asp Ala Phe Phe Cys Ile Pro Val Arg Pro Asp Ser Gln Phe Leu Phe 1 5 10 15 Ala Phe Glu Asp Pro Leu Asn Pro Thr Ser Gln Leu Thr Trp Thr Val 20 25 30 Leu Pro Gln Gly Phe Arg Asp Ser Pro His Leu Phe Gly Gln Ala Leu 35 40 45 Ala Gln 50 299 base pairs nucleic acid single linear cDNA 40 GATGCCTTTT TCTGCATCCC TGTACGTCCT GACTCTCAAT TCTTGTTTGC CTTTGAAGAT 60 CCTTTGAACC CAACGTCTCA ACTCACCTGG ACTGTTTTAC CCCAAGGGTT CAGGGATAGC 120 CCCATCTATT TGGCCAGGCA TTAGCCCAAG ATGCCTTTTG CATCCCTGTA CGTGACTCTC 180 AATTCTTGTT TGCCTTTGCC TTTGAAGATG CTTTGAACCC AACGTCTCAA CTCACCTGGA 240 CTGTTTTACG CCAAGGGTTC AGGGATAGCC CCCATCTATT TGGCCAGGCA TTAGCCCAA 299 11 amino acids amino acid linear peptide 41 Cys Ile Pro Val Arg Pro Asp Ser Gln Phe Leu 1 5 10 17 amino acids amino acid linear peptide 42 Val Leu Pro Gln Gly Phe Arg Asp Ser Pro His Leu Phe Gly Glu Ala 1 5 10 15 Leu 17 8 amino acid amino acid linear peptide 43 Leu Phe Ala Phe Glu Asp Pro Leu 1 5 8 8 amino acids amino acid linear peptide 44 Phe Ala Phe Glu Asp Pro Leu Asn 1 5 8 25 base pairs nucleic acid single linear cDNA 45 GTGCTGATTG GTGTATTTAC AATCC 25 1859 base pairs nucleic acid single linear cDNA 46 GTGCTGATTG GTGTATTTAC AATCCTTTAT CTAATCCGAA ATGCCCATGT TGCAATATGG 60 AAAGAAAGGG AGTTCCTAAC CTCTGGGGGA ACCCCCATTA AATACCACAA GTAAATCATG 120 GAGTTATTGC ACACAGTGCA AAAACTCAAG GAGGTGGAAG TCTTACACTG CCAAAGCCAT 180 CAGAAAAGGG AAGAGGGGAG AAGAGCAGCA TAAGTGGCTA CAGAGGCAAG GAAAGACTAG 240 CAGAAAGGAA AGAGAGAAAG AGACAGAAAG TCAGAGAGAG AGAGAGGAAG AGACAGAGCA 300 CAAAGAGGGA GTCAGAGAGA GAGAGAGACA GAGAGTCAGA GAGAAGGAAA GAGAGAGAGG 360 AAGAGACAAA GAATGAATCA AACAGAGAGA CAGAAAGTCA GAGAGAGAGA GAGAGAGGAA 420 GAGACAGAGA AAAAGAGGGA GTCAGAAAAA GAGAGACCAA AGAAGAAGTC CAAAGAGAAA 480 GAAAGAGAGA TGGAAGTAGT AAAGGAAAAA CAGTGTACCC TATTCCTTTA AAAGCCGGGG 540 TAAATTTAAA ACCTATAATT GATAACTGAA GGTCTTCTCT GTAACCCTGT AACACTCCAA 600 TACCACCTTG TTGTCAAGTG TAAACAAGGG CGTAGCCCAA AAGCACTGAG GCCACTAACA 660 ACCCATAGCC TTCCTATCAA AATTCCTTAA CCCAGCAGGT TTCCTAACAG GGGATCTAAA 720 TCTTAATTAA TTACCATACA ATGGTCCAAC CAGACTTAGG AGGAATTCCC TTCAGGACGG 780 GAAGATAGAT GCTTCCTCCC AGGCGATTAA GGGAGAAAGA CACAATGGGT ATTCAGTAAG 840 TGCCAAGGGG AACACTTGTA GAAGCAAAGT TAGGAAAATT GCCAAATAAT TGGTTTGCTC 900 AAGAGTTGTT TGCACTCAGC CAAACCTTGA AGTACTTGCA GAATCAGAAA GGAGCCATCT 960 ATACCAATTC TAAGTTAATA TGGACTGAAG GAGGTTTTAT TAATACCAAA GAGAAATTAA 1020 AATCCCAAAC TTATAAGGTT TTCAACCAAA GTAAAGTTTG CTAAAAGTTA ACAGCGTAAC 1080 ATGTATTATC CTACTACCAC ACACTCTCAA AGGATTTCTC AGACAGTTTG CAAGAAATAA 1140 TGATATCTAT CCTTACTCTA CAATCCCAAA TAGACTCTTT GGCAGCAGTG ACTCTCCAAA 1200 ACCGTCAAGG CCTAGACCTC CTCACTGCTG AGAAAGGAGG ACTCTGCACC TTCTTAAGGG 1260 AAGAGTGTTG TCTTTACACT AACCAGTCAG GGATAGTATG AGATGCTGCC CGGCATTTAC 1320 AGAAAAAGGC TTCTGAAATC AGACAACGCC TTTCAAATTC CTATACCAAC CTCTGGAGTT 1380 GGGCAACATG GTTTCTTCCC TTTCTATGTC CCATGGCTGC CATCTTGCTA TTACTCGCCT 1440 TTGGGCCCTG TATTTTTAAC CTCCTTGTCA AATTTGTTTC TTCTAGGATC GAGGCCATCA 1500 AGCTACAGAT GGTCTTACAA ATGGAACCCC AAATGAGCTC AACTATCAAC TTCTACTGAG 1560 GACCCCTAGA CCAACCCCCT GGCCCTTTCA CTGGCCTAAA GAGTTCCCCT CTGGAGGACA 1620 CTACCACTGC AGGGCCCCAT CTTTGCCCCT ATCCAGAAGG AAGTAGCTAG AGCAGTCATT 1680 GCCCAATTCC CAAGAGCAGC TGGGGTGTCC CGTTTAGAGT GGGGATTGAG AGGTGAAGCC 1740 AGCTGGACTT CTGGGTCGGG TGGGGACTTG GAGAACTTTT GTGTCTAGCT AAAGGATTGT 1800 AAATGCAACA ATCAGTGCTC TGTGTCTAGC TAAAGGATTG TAAATACACC AATCAGCAC 1859 23 base pairs nucleic acid single linear cDNA 47 TGATGTGAAC GGCATACTCA CTG 23 24 base pairs nucleic acid single linear cDNA 48 CCCAGAGGTT AGGAACTCCC TTTC 24 25 base pairs nucleic acid single linear cDNA 49 GCTAAAGGAG ACTTGTGGTT GTCAG 25 22 base pairs nucleotide single linear cDNA 50 CAACATGGGC ATTTCGGATT AG 22 400 base pairs nucleotide single linear cDNA 51 GGCTGCTAAA GGAGACTTGT GGTTGTCAGA CAATCGCCTA CTTAGGTACC AGGCCTTATT 60 ACTTGAGGGA CTGGTGCTTC AGATGCGCAC TTGTGCAGCT CTTAACCCAA ACTTATGCTG 120 CCCAGAAGGA TCTTTTAGAG GTCCCCTTAG CCAACCCTGA CCTCAACCTA TATATATACT 180 GATGGAAGTT CGTTTGTAGA AAAGGGATTA CAAAGGGNAG GATATNCCAT AGGTTAGTGA 240 TAAAGCAGTA CTTGAAAGTA AGCCTCTTCC CCCCAGGGAC CAGCGCCCCC GTTAGCAGAA 300 CTAGTGGCAC TGACCCCGAG CCTTAGAACT TGGAAAGGGA GGAGGATAAA TGTGTATACA 360 GATAGCAAGT ATGCTTATCT AATCCGAAAT GCCCATGTTG 400 2389 base pairs nucleotide single linear cDNA 52 TCAGGGATAG CCCCCATCTA TTTGGTCAGG CACTGGCCCA AGATCTAGGG ACATGCCACT 60 TTTAAGAGCC ATTTCTCAAG TCCAGGTACT CTGGTCCTTC GGTATGTGGA TGATTTACTT 120 TTGGCTACCA GTTCAGTAGC CTCATGCCAG CAGGCTACTC TAGATCTCTT GAACTTTCTA 180 GCTAATCAAG GGTACAAGGC ATCTAGGTTG AAGGCCCAGC TTTGCCTACA GCAGGTCAAA 240 TATCTAGGCC TAATCTTAGC CAGAGGGACC AGGGCACTCA GCAAGGAACA AATACAGCCT 300 ATACTGGCTT ATCCTCACCC TAAGACATTA AAACAGTTGC GGGGGTTCCT TGGAATCACT 360 GGCTTTTTGG TGACTATGGA TTCCCAGATA CAGCAAGATT GGCAGGCCCC TCTATACTGT 420 AATCAAGGAG ACTCACGAGG GCAAGTACTC ATCTAGTAGA ATGGGAACTA GGGACAGAAA 480 CAGCCTTCAA AACCTTAAAG CAGGCCCTAG TACAATCTCC AGCTTTAAGC CTTCCCACAG 540 GACAAAACTT CTCTTTATAC ATCACAGAGA GGGCAGAGAT AGCTCTTGGT GTCCTTATTC 600 AGACTCATGG GACTACCCCA CAACCAGTGG CACACCTAAG TAAGGAAATT GATGTAGTAG 660 CAAAAGGCTG GCCTCACTGT TTATGGGTAG CTGTGGTGGT GGCTGTCTTA GTGTCAGAAG 720 CTATCAAAAT AATACAAGGA AAGGATCTCA CTGTCTGGAC TACTCATGAT GTAATGGCAT 780 ACTAGGTGCC AAAAGAAGTT TATGGGTATC AGACAACCAC CTGCTTAGAT ACCAGGGACT 840 ACTCCTGGAG GATTGGGCTT CAAGTGCGTT TTTTGTGGCC TCAACCCTGC CACTTTTCCT 900 CCAGAGGATG GAGAGCCGCT TGAGCATGCT TGCCAACAGG TTGTAGGCCA GAATTATTCC 960 ACCCGAGATG ATCTCTTAGA GTACCCTTAG CTAATCCTGA CCTTAACCTA TATACCAATG 1020 GAAGTTCATT TGTGGAAAAC GGGATATGAA GGGCAGGTTA TGTCATAGTT AGTGATGTAA 1080 TCATACTTGC AAGTAAGCCT CTTACCCCAG GGGCCAGCAC TCAGTTAGCA GAACTAGTCA 1140 CACTTACCTT AACCTTAGAA CTGGGAAAGG GAAAAAGAAT AAATATGTAT ACAGATAGTA 1200 AGTATGCTTA TCTAATCCTA CATGCCCATG CTGCAATATG GAAGGAAAGG GAGTTCCTAA 1260 CCCCTGGGGG AACCCCCATT AAATACCACA AGGYAAATCA TGGAGTTATT GCACGCAGTG 1320 CAAAAACTCA AGGAGGTGGC AGTCTTACAC TGCCGAAGCY ATCAAAAAGG GGAAGGAGAG 1380 GGGAGAACAG CAGCATAAGT GGTTGGCAGA GGCAGTGAAA GACCAGCAGA GAGAAGGAGA 1440 GAGACAACGT CAACGACAGA AGGAAAGAAG AGGAGGAGAC AGAGAGGAAG AGACAGAGAG 1500 ACAGTTAGTC CAAGAGAGAG ACAGAGAGAG GAAGAGACAG ACAGAAAGTC CAAGAGAGAA 1560 GGAAAGAGAG GAAGAGACCA AGGAGTCCNA GAGAGAGAAA GAGATAGAAG TAGTAAAGAA 1620 AAAACATTGT ACCCTATTCC TTTAAAAGCC GGGGTATATT TAAAACCTAT AATTGATAAT 1680 TGAGTTCTTG CACCCTCCTC CAGGGGATYG CTGGGAGGAA ACCCTCAACC GATATGTGAA 1740 AATTGTGGGT CGTCCCTATG TCTCAATTAC CAGCCAATAC CCCCTTGTTT TTAGTGTGAA 1800 CGAGGGTGTA GAGCGCAGAC AGGGAGACCT CTGACAATCC ATACCCTTCC TATCCAAAAT 1860 CCTTAACCCA GCAGGTTTTC TAAAAGGGGA TCTAAATCTT AATTAATTAC CATACAAAGG 1920 TCAAACCAGA TCTAGGAGGA ACTTCCTTCA GGACAGGATG ATAGATGGTT CCTCCCAGGC 1980 GATTAAAGAA AATAAAAAGA CACATGGGCA GCCAGTAAGT GATAAGGGAA CACTAGTAGA 2040 AGCAGTTAGG AGAAGTTGCC TAATAATTGG TCTACTCCAA ATGTGTGAGT TGTTCGCACT 2100 CAGCCCAAAT CTTAAAGTAC TTACAGAATT AGGGAGGAGC CATTTACACC AATTCTAAGT 2160 TAATATGGAC TGGATGAGGT TTTATTAATA GCGAAGGAGA ATTAAATCCT AAACTNACAA 2220 GGTTTTCAAC TAAAGTAAAT TTTACTAAAA GCTAACAGTG TAACATGCAT TATCCTACTA 2280 CAACACACTC TCANAGGATT CCTCAGACAG TTTACAAGAA ATAACAAAAT CTATCTGGTA 2340 AGGATAGTAA CTACAATCCC AAATACATTC TTTGGCAGCA GTGACTCTC 2389 2448 base pairs nucleotide single linear cDNA 53 TCAGGGATAG CCCCCATCTA TTTGATCAGG CACTAGCCCA AGATCTAGGC CACTTCTGAA 60 GTCCAGGCAT TCTAGTCCTT CAGTATGTGG ATGATTTACT TTTGGCTACC AGTTTGGAAG 120 CCTCATGCCA GCAGGCTACT TGAGATCTCT TGAACTTTCT AGCTAATCAA GGGTGTATGG 180 CATCTAAATT GAAAGTCCAG CTCTGCCTAC AACAAGTCAA ATATCTAGGC CTAATCTTAG 240 ATAGAAGAAC CAGGGCCCTC AGCAAGGAAT GAATAAAGCC TATGCTGGCT TATCGGCACC 300 CTAAGACATT AAAACAATTG TGGGGGTTCC TTGGAATCAC TGGCTTTTGC CGACTATGGA 360 TCCCTGGATA GAGTGAGATA GCCAGGCCCC CTCTATTACT CTTATCAAGG AGACCCAGAG 420 GGCAAATACT TATCTAGTAT TATGGGNACC AGAGGCAGAA AAAGCCTTCC AAACCTTAAA 480 GGAGACCCTA GTACAAGCTC CAGCTTTAAG CCTTCCCACA GGACAAANCT TCTCTTTATA 540 TGTCACAGAG AGAGCAGGAA TAGCTCCTGG AGTCCTTACT CAGACTTTTG GACGACCCCA 600 CGGCCAGTGG CRTACCTAAG TAAGGAAATT GATGTAGTAG CAAAAGGCTG GCCTCACTGT 660 TTATGGGTAG TTGCGGCTGT GGCAGTCTTA CTGTCAAAGG CTATCAAAAT AATACAAGGA 720 AAGGATTTCA CTATCTGGAC TACTCATGAG GAAAATGGCA TATTAGGTGC CAAAGGAAGT 780 TTTTGGCTAT CAGACAACCA CCTGCTCAGA TTCCAGGCAC TACTGATTGA GAGACCAGTG 840 CTTTAAATAT GTATGTGTGT GTGTGGCCCT CAACCCTGCC ACTGTTCTCC CAGAAGATGG 900 AGAACCAATG AAGCATTACT GTCAACAAAT TAGAGTCCAG AGTTATGCTG CCTGAGAGGA 960 TCTCTTAGAA GTCCCCTTAG CTAATCCTGA CCTTAACCTA TATGCTGATG GAAGTTCACT 1020 TGTGGAGAAT GGGATACGAA AAGCACATTA TGCCATAGTT AGTGAGGTAA CAGTACTTGA 1080 AAGTAAGCCT ATTCCCCCAT GGACCAGAGC CCAGTTAGCA GAACTAGTGG CACTTACCCA 1140 AGCCTTAGAA CTAGGAAAGG GAAAAATAAT AAATGTGTAT ACAGATAGCA AGTATGCTTA 1200 TCTAATCCTA CATGCCCATG CTGCAGTATG GAAAGAAAGG GAGTTCCTAA CCTCTGGGGG 1260 AACCCCCATT AAATACCACA AGGCAAATCA TGGAGTTATT GCATGTAGTG CAAAACCTCA 1320 AGTAGGTGGC AGTTTTACAC TGCCTGAAGC TATGGGGAAG GAGAGAGGAG AACAGCAGCA 1380 TAAGTGGCTA GCAGAGGCAG CGAAAGACTA GCAGAGAGGA GAGGTAGGGG AAAGACAGAA 1440 AGTCAAAGAA AAGAAGTCAA AGACAGACAG AGAAAGAGAC AGAGGGAGCC AGAGAGAAAG 1500 AAAAGAGAGA ACGAAAGAGA CAGAATGTCA AAGAACAGAA GAGAGAGGCA GCGCCAGAAG 1560 AGTTAAGAAA GTGAGAAAGA GAGATGGAAA TAGTAAAGAA AAAACAGTGT ACCCTATTCC 1620 TTTAAAAGCC AGGGTAAATT TAAAACGTAT AATTTTATAA TTGGAAGGTC TTCTCCATAA 1680 CCCTATAACA TTAAAATACC ACCTTGTTGT CAGTGTAAAC AAGAGCATAG CCCAAAAGCA 1740 CTGAGGCCAC TGACAACCCA TAGCCTTCCT ATCAAAAATC CTTAACTCTG CAGGTTTCCT 1800 AACAGGGGAT CTAAATCTCA ACTAATCACC ATACAATGGT CCGACCAGAC CTAGGAGCGA 1860 CTCCCCTCAG GACAGAAGGA TGGATGGTTC CTCCCAGGCC ATTAAGGGAA AGAGACACAA 1920 TGGGTATTCA GTAAGTGATA AGGGAACTCT TGTAGAAGCA GTTAGGAAGA TTGCCTAATA 1980 TTTGGTCTGC TCAAATGTGC CAGCTGTTTG CACTCAGCTA AACCTTAAAT TACTTACAGA 2040 ATTAGGAAGG AGCCATCTAT ACCAATTCTG AGTTAATATG AGCTGAACAA GTTCTTATTA 2100 ATAGCAAAGA ATCATTGAAA TCTCAAACTT GCAAAGTTTT CAACAAAAGT AAAGTTTGCT 2160 GAAAGTTAGC AGTGTAACAT GTATTATCCT AACTTCTAAT CTTGTGGAAA TCAGACCCTA 2220 TCAGTGCCCC TCAAAGCTGA AGTCCATCAG CATATGGCCA TACAACTAAT ACCCCTATTT 2280 ATAGGGTTAG GAATGGCCAC TGCTACAGGA ATGGGAGTAA CAGGTTTATC TACTTCATTA 2340 TCCTATTACC ACACACTCTT AAAGGATTTC TCAGACAGTT TACAAGAAAT AACAAAATCT 2400 ATCCTTACTC TNTARTCCCA AATAGRTTCT TTGGCAGCAG TGACTCTC 2448 21 base pairs nucleotide single linear cDNA 54 CCTGAGTTCT TGCACTAACC C 21 23 base pairs nucleotide single linear cDNA 55 GTCCGTTGGG TTTCCTTACT CCT 23 1196 base pairs nucleotide single linear cDNA 56 TTCCTGAGTT CTTGCACTAA CCTCAAATGA GAGAAGTGCC GCCATAACTG CAACCCAAGA 60 GTTTGGCGAT CCCTGGTATC TCAGTCAGGT CAATGACAGG ATGACAACAG AGGAAAGATA 120 ATGATTCCCC ACAGGCCAGC AGGCAGTTCC CAGTGTAGAC CCTCATTAGG ACACAGAATC 180 AGAACATGGA GATTGGTGCC GCAGACATTT GCTAACTTGC GTGCTAGAAG GACTAAGGAA 240 AACTAGGAAG ATATGAATTA TTCAATGATG TCCACTATAA CACAGGGGAA AGGAAGAAAA 300 TCCTACTGCC TTTCTGGAGA GACTAAGGGA GGCATTGAGG AAGCATACCA GGCAAGTGGA 360 CATTGGAGGC TCTGGAAAAG GGAAAAGTTG GGAAAAGTAT ATGTCTAATA GGGCTTGCTT 420 CCAGTGTGGT CTACAAGGAC ACTTTAAAAA AGATTGTCCA ATAGAAATAA GCCACCACCT 480 CGTCCATGCC CCTTATGTCA AGGGAATCAC TGGAAGGCCC ACTGCCCCAG GGGATGAAGG 540 TCCTCTGAGT CAGAAGCCAC TAACCAGATG ATCCAGCAGC AGGACTGAGG GTGCCCGGGG 600 CAAGCGCCAG CCCATGCCAT CACCCTCACA GAGCCCCAGG TATGCTTGAC CATTGAGGGT 660 CAGAAGGGTA CTGTCTCCTG GACACTGGCG GGCCTTCTCA GTCTTACTTT CCTGTCCTGG 720 ACAACTGTCC TCCAGATCTG TCACTGTCCG AGGGGTCCTA GGACAGCCAG TCACTAGATA 780 CTTCTCCCAG CCACTAAGTT GTGACTGGGG AACTTTACTC TTCCACATGC TTTTCTAATT 840 ATGCCTGAAA GCCCCACTCT CTTGTTAGGG GAGAGACATT CTAGCAAAAG CAGGGGCCAT 900 TATACATGTG AATATAGGAG AAGGAACAAC TGTTTGTTGT CCCCTGCTTG AGGAAGGAAT 960 TAATCCTGAA GTCCGGGCAA CAGAAGGACA ATATGGACAA GCAAAGAATG CCCGTCCTGT 1020 TCAAGTTAAA CTAAAGGATT CCACCTCCTT TCCCTACCAA AGGCAGTACC CCCTCAGACC 1080 CGAGACCCAA CAAGAACTCC AAAAGATTGT AAAGGACCTA AAAGCCCAAG GCCTAGTAAA 1140 ACCAAGCAAT AGCCCTTGCA AGACTCCAAT TTTAGGAGTA AGGAAACCCA ACGGAC 1196 2391 base pairs nucleotide single linear cDNA 57 ATGATCCAGC AGCAGGACNG AGGGTGCCCG GGGCAAGCGC CAGCCCATGC CATCACCCTC 60 ACAGAGCCCC AGGTATGCTT GACCATTGAG GGTCAGAAGG GTNACTGTCT CCTGGACACT 120 GGCGGNGCCT TCTCAGTCTT ACTTTCCTGT CCTGGACAAC TGTCCTCCAG ATCTGTCACT 180 GTCCGAGGGG TCCTAGGACA GCCAGTCACT AGATACTTCT CCCAGCCACT AAGTTGTGAC 240 TGGGGAACTT TACTCTTCCC ACATGCTTTT CTAATTATGC CTGAAAGCCC CACTCTCTTG 300 TTGGGGAGAG ACATTCTAGC AAAAGCAGGG GCCATTATAC ATGTGAATAT AGGAGAAGGA 360 ACAACTGTTT GTTGTCCCCT GCTTGAGGAA GGAATTAATC CTGAAGTCCG GGCAACAGAA 420 GGACAATATG GACAAGCAAA GAATGCCCGT CCTGTTCAAG TTAAACTAAA GGATTCCACC 480 TCCTTTCCCT ACCAAAGGCA GTACCCCCTC AGACCCGAGA CCCAACAAGA ACTCCAAAAG 540 ATTGTAAAGG ACCTAAAAGC CCAAGGCCTA GTAAAACCAA GCAATAGCCC TTGCAAGACT 600 CCAATTTTAG GAGTAAGGAA ACCCAACGGA CAGTGGAGGT TAGTGCAAGA ACTCAGGATT 660 ATCAATGAGG CTGTTGTTCC TCTATACCCA GCTGTACCTA ACCCTTATAC AGTGCTTTCC 720 CAAATACCAG AGGAAGCAGA GTGGTTTACA GTCCTGGACC TTAAGGATGC CTTTTTCTGC 780 ATCCCTGTAC GTCCTGACTC TCAATTCTTG TTTGCCTTTG AAGATCCTTT GAACCCAACG 840 TCTCAACTCA CCTGGACTGT TTTACCCCAA GGGTTCAGGG ATAGCCCCCA TCTATTTGGC 900 CAGGCATTAG CCCAAGACTT GAGTCAATTC TCATACCTGG ACACTCTTGT CCTTCAGTAC 960 ATGGATGATT TACTTTTAGT CGCCCGTTCA GAAACCTTGT GCCATCAAGC CACCCAAGAA 1020 CTCTTAACTT TCCTCACTAC CTGTGGCTAC AAGGTTTCCA AACCAAAGGC TCGGCTCTGC 1080 TCACAGGAGA TTAGATACTN AGGGCTAAAA TTATCCAAAG GCACCAGGGC CCTCAGTGAG 1140 GAACGTATCC AGCCTATACT GGCTTATCCT CATCCCAAAA CCCTAAAGCA ACTAAGAGGG 1200 TTCCTTGGCA TAACAGGTTT CTGCCGAAAA CAGATTCCCA GGTACASCCC AATAGCCAGA 1260 CCATTATATA CACTAATTAN GGAAACTCAG AAAGCCAATA CCTATTTAGT AAGATGGACA 1320 CCTACAGAAG TGGCTTTCCA GGCCCTAAAG AAGGCCCTAA CCCAAGCCCC AGTGTTCAGC 1380 TTGCCAACAG GGCAAGATTT TTCTTTATAT GCCACAGAAA AAACAGGAAT AGCTCTAGGA 1440 GTCCTTACGC AGGTCTCAGG GATGAGCTTG CAACCCGTGG TATACCTGAG TAAGGAAATT 1500 GATGTAGTGG CAAAGGGTTG GCCTCATNGT TTATGGGTAA TGGNGGCAGT AGCAGTCTNA 1560 GTATCTGAAG CAGTTAAAAT AATACAGGGA AGAGATCTTN CTGTGTGGAC ATCTCATGAT 1620 GTGAACGGCA TACTCACTGC TAAAGGAGAC TTGTGGTTGT CAGACAACCA TTTACTTAAN 1680 TATCAGGCTC TATTACTTGA AGAGCCAGTG CTGNGACTGC GCACTTGTGC AACTCTTAAA 1740 CCCAAACTTA TGCTGCCCAG AAGGATCTTT NTAGAGGTCC CCTTAGCCAA CCCTGACCTC 1800 AACTATATAT ATACTGATGG AAGTTCGTTT GTAGAAAAGG GATTACAAAG GGNAGGATAT 1860 NCCATAGGTG TTAGTGATAA AGCAGTACTT GAAAGTAAGC CTCTTCCCCC CCAGGGACCA 1920 GCGCCCCCGT TAGCAGAACT AGTGGCACTG ACCCCGCGAG CCTTAGAACT TTGGAAAGGG 1980 AGGAGGATAA ATGTGTATAC AGATAGCAAG TATGCTTATC TAATCCGAAA TGCCCATGTT 2040 GTTTATCTAA TCCGAAATGC CCATGTTGCA ATATGGAAAG AAAGGGAGTT CCTAACCTCT 2100 GGGGGAACCC CCATTAAATA CCACAAGTTA ATCATGGAGT TATTGCACAC AGTGCAAAAA 2160 CTCAAGGAGG TGGAAGTCTT ACACTGCCAA AGCCATCAGA AAAGGGAAAG GGGAGAAGAG 2220 CAGCATAAGT GGCTACAGAG GCAAGGAAAG ACTAGCAGAA AGGAAAGAGA GAAAGAGACA 2280 GAAAGTCAGA GAGAGAGAGA GGAAGAGACA GAGCACAAAG AGGGAGTCAG AGAGAGAGAG 2340 AGACAGAGAG TCAGAGAGAA GGAAAGAGAG AGAGGAAGAG ACAAAGAATG A 2391 1722 base pairs nucleotide single linear cDNA 58 TGGAGAATAG CAGCATAAGT TGGCTGGCAG AAGTAGGGAA AGACAGCAAG AAGTAAAGAA 60 AAAAARGAGA AAGTCAGAGA AAGAAAAAAA GAGAGGAAGA AACAAAGAAG AACTTGAAGA 120 GAGAAAGAAG TAGTAAAGAA AAAACAGTAT ACCCTATTCC TTTAAAAGCC AGGGTAAATT 180 TCTGTCTACC TAGCCAAGGC ATATTCTTCT TATGTGGAAC ATCAACCTAT ATCTGCCTCC 240 CCACTAACTG GACAGGCACC TGAACCTTAG TCTTTCTAAG TCCCAACATT AACATTGCCC 300 CAGGAAATCA GACCCTATTG GTACCTGTCA AAGCTAAAGT CCCGTCAGTG CAGAGCCATA 360 CAACTAATAT CCCTATTTAT AGGGTTAGGA ATGGCTACTG CTACAGGAAC TGGAATAGCC 420 GGTTTATCTA CTTCATTATC CTACTACCAT ACACTCTCAA AGAATTTCTC AGACAGTTTG 480 CAAGAAATAA TGAAATCTAT TCTTACTTTA CAATCCCAAT TAGACTCTTT GGCAGCAATG 540 ACTCTCCAAA ACCGCCGAGG CCCACACCTC CTCACTGCTG AGAAAGGAGG ACTCTGCACC 600 TTCTTAGGGG AAGAGTGTTG TTTTTACACT AACCAGTCAG GGATAGTACG AGATGCCACC 660 TGGCATTTAC AGGAAAGGGC TTCTGATATC AGACAATGCC TTTCAAACTC TTATACCAAC 720 CTCTGGAGTT GGGCAACATG GCTTCTTCCA TTTCTAGGTC CCATGGCAGC CATCTTGCTG 780 TTACTCACCT TTGGGCCCTG TATTTTTAAG CTTCTTGTCA AATTTGTTTC CTCTAGGATC 840 GAAGCCATCA AGCTACAGAT GGTCTTACAA ATGGAACCCC AAATGAGTTC AACTAACAAC 900 TTCTACCAAG GACCCCTGGA ACGATCCACT GGCACTTCCA CTAGCCTAGA GATTCCCCTC 960 TGGAAGACAC TACAACTGCA GGGCCCCTTC TTTGCCCCTA TCCAGCAGGA AGTAGCTAGA 1020 GCGGTCATCG GCCAAATTCC CAACAGCAGT TGGGGTGTCC TGTTTAGAGG GGGGATTGAA 1080 GAGGTGACAG CCTGCTGGCA GCCTCACAGC CCTCGTTGGY TCTCAGTGCC TCCTCAGCCT 1140 TGGTGCCCAC TCTGGCCGTG CTTGAGGAGC CCTTCAGCCT GCCACTGCAC TGTGGGAGCC 1200 TCTTTCTGGG CTGGACAAGG CCGGAGCCAG CTCCCTCAGC TTGCAGGGAG GTATGGAGGG 1260 AGAGATGCAG GCGGGAACCA GGGCTGCGCA TGGCGCTTGC GGGCCAGCAT GAGTTCCAGG 1320 TGGGCGTGGG CTCGGCGGGC CCCACACTCG GGCAGTGAGG GGCTTAGCAC CTGGGCCAGA 1380 CAGATGCTGT GCTCAACTTC TTCGCTGGGC CTTAGCTGCC TTCCCCGTGG GGCAGGGCTY 1440 CGGGAACMTG CAGCCTGCCC ATGCTTGAGC CCCCCACCCC GCCGTGGGTT CYTGCACAGC 1500 CCAAGCTTCC CGGACAAGCA CCACCCCTTA TCCACGGTGC CCAGTCCCAT CAACCACCCA 1560 AGGGTTGAGG AGTGCGGGCA CACAGCGCGG GATTGGCAGG CAGTTCCACT TGCGGCCTTG 1620 GTGCGGGATC CACTGCGTGA AGCCAGCTGG GCTCCTGAGT CTGGTGGGGA CTTGGAGAAT 1680 CTTTATGTCT AGCTAAGGGA TTGTAAATAC ACCAATCAGC AC 1722 495 base pairs nucleotide single linear cDNA 59 CTTCCCCAAC TAATAAGGAC CCCCCTTTCA ACCCAAACAG TCCAAAAGGA CATAGACAAA 60 GGAGTAAACA ATGAACCAAA GAGTGCCAAT ATTCCCTGGT TATGCACCCT CCAAGCGGTG 120 GGAGAAGAAT TCGGCCCAGC CAGAGTGCAT GTACCTTTTT CTCTCTCACA CTTGAAGCAA 180 ATTAAAATAG ACNTAGGTNA ATTNTCAGAT AGCCCTGATG GYTATATTGA TGTTTTACAA 240 GGATTAGGAC AATCCTTTGA TCTGACATGG AGAGATATAA TATTACTGCT AAATCAGACG 300 CTAACCTCAA ATGAGAGAAG TGCTGCCATA ACTGGAGCCC GAGAGTTTGG CAATCTCTGG 360 TATCTCAGTC AGGTCAATGA TAGGATGACA ACGGAGGAAA GAGAACGATT CCCCACAGGG 420 CAGCAGGCAG TTCCCAGTGT AGCTCCTCAT TGGGACACAG AATCAGAACA TGGAGATTGG 480 TGCCGCAGAC ATTTA 495 2503 base pairs nucleotide single linear cDNA 60 CCAAGAACCC ACCAATTCCG GANCACATTT TGGCGACCAC GAAGGGACTT TCGCATATCG 60 CCAAGCGGTG AGACAATAGC CGAGCGGTGA GACCTTTCCC AATCGCCAAG CAGTGAGTAC 120 CATCAGACCC CTTTCACTTG CTATTCTGTC CTATCTTTCT TTAGAATTCG GGGGCTAAAT 180 ACCGGGCATC TGTCAGCCAT TTAAAAGTGA CTAGCGGGCC GCCGGACTAA AGACACGGGT 240 GTCAAGCTTT CTGGGAAAGG GCTCTCTAAC AACCCCCAAC TCTTTGGAGT TGGGACCGTT 300 GGTTTGCCTA GAACCAGCTT CCGCTTTTCC TGTACTTCTG GGCTGAGCCG TGGGTTGACA 360 GTGAAGGAAA GCCATGCATC TCCGGGGTCT CGMCAACATG TTGGTTGACC CTGCGGCCAT 420 GAGTGGAACT CTCAAAAGCA TGTCGCCCAA GCGACACTCG CCTATCTATC CTATCTATCC 480 TGACCCTTGC CCTCTGGGTC CTAATGCCTG CCAGACAAAC TTCCTCTCGC CTCTCTTCTC 540 TGAAGCTAGA ACCGCTTCTA AAAATTGCTA CCTGGTCTCT GGTGCTTTTC CTARTTTCTC 600 CTATAAAGAA TGAWTTCTAG TATTAAACTC CAGGACTCTG TTACCTTCTT TAGGCACCCG 660 GGCTCACCAA TCAGAAAGAC ACAGTTTTTG CCCAAGGCCC CATCGTAGTG GGGACTACCT 720 GGAATTTTAG GATCCCTCCT CAGACTAACA GGCCTAACAA AAGTTATTCC TGAAGCTAGG 780 ATATGGGGAG CCTCAGAAAT TGTATCCCTC CTATTCATAT AAGTGAGAAC AAAAGGTGTC 840 ACTCTTCCAA CCCTGAAGAT CCCCTCCCTC CCTCAGGGTA TGGCCCTCCA TTTCATTTTT 900 GTGGCATAAC ATCTTTATAG GATGGGGTAA AGTCCCAATA CTAACAGGAG AATGCTTAGG 960 ACTCTAACAG GTTTTTGAGA ATGCGTCAGT AAGGGCCACT AAATCTGATT TTTCTCAGTC 1020 GGTCCTCCTT GTGGTCTAGG AGGACAGGCA AGGTTGTGCA GGTTTTCGAG AATGCGTCAG 1080 TAAGGACCAC TAAATCCGAC CTTCCTCGGT CCTCCATGTG GTCTGGGAGG AAAACTAGTG 1140 TTTCTGCTGC TGCGTCGGTG AGCGCAACTA TTCAAGTCAG CAGGGTCCAG GGACCGTTGC 1200 AGGTTCTTGG GCAGGGGTTG TTTCTGCTGC TGCATTGGTG AATGCAACTA TTCTGATCAG 1260 CAGGGTCCCA GGACCATTGC AGGTCCTTGG GCAGGGAGAG AAACAAAACA AACCAAAACT 1320 GTGGGCGGTT TTGTCTTTCA TATGGGAAAC ACTCAGGCAT CAACAGGTTC ACCCTTGAAA 1380 TGCATCCTAA GCCATTGGGA CCAATTTGAC CCACAAACCC TGAAAAAGAG GAGGCTCATT 1440 TTTTCCTGCA CTACGGCTTG GCCCCAATAT TCTCTTTYTG ATGGGGAAAA ATGGCCACCT 1500 GAGGGAAGCA CAAATTACAA TAYTATCCTA CAGCYTGATC TTTTCTGTAA GAGGGAAGGC 1560 AAATGGAGTG AATACCTTAT GTCCAAGCTT TCTTTTCATT GAGGGAGAAT ACACAACTAT 1620 GCAAAGCTTG CAATTTACAT CCCACAGGAG GACCCTTCAG CTTACCCCCA TATCCTAGCC 1680 TCCCTATAGC TTCCCTTCCT ATTGATGATA CTCCTCCTCT AATCTCCCCT GCCCAGAAGG 1740 AAATAAGCAA AGAAATCTCC AAAGGTCCAC AAAAACCCCC GGGCTATCGG TTATGTCCCT 1800 TCAAGYTGTA GGGGGAGGGG AATTTGGCCC AACCCGGGTG CATGTCCCTT CTCCCTCTCT 1860 GATTTAAAGC AGATCAAGGC AGACCTGGGG AAGTTTTCAG ATGATCCTGA TAGGTACATA 1920 GATGTCCTAC AGGGTCTAGG GCAAACCTTT GACCTCACTT GGAGAGACGT CATGCTACTG 1980 TTAGATCAAA CCCTGGCCTT TAATGAAAAG AATGCGGCTT TAGCTGCAGC CTGAGAGTTT 2040 GGAGATACCT GGTATCCTAG TCAAGTAAAT GAAAGAATGA CAGCCGAAGA AAGGGACAAC 2100 TTCCTTACTG GTCAGCAACC CATCCCCAGT ATGGATCCCC ACTGGGACTT TGACTCAGAT 2160 CATGGGGACT GGAGTCGTAA ACATCTGTTG ATCTGTGTTC TGGAAGGACT AAGGAGAATT 2220 GGGAAAAAGC CCATGAATTA TTCAATGATA TCCACCATAA CCCAGGGAAA GGAAGAAAAT 2280 CCTTCTGCCT TCCTCGAGCG GCTACAAGAG GCCTTAAGAA AATATACTCC CCTGTCACCC 2340 GAATCACTCG AGGGTCAATT GATTCTAAAA GATAAGTTTA TTACCCAATC AGCCACAGAT 2400 ATCAGGAGAA AGCTCCAAAA GCAAGCCCTG AGCCTGAACA AAATCTAGAG ACATTATTAA 2460 ACCTGGCAAC CTTGGTGTTC TATAATAGGG ACCAAGAGGA ACA 2503 1167 base pairs nucleotide single linear cDNA 61 AAGGAAACTC AGAAAGCCAA TACCCATTTA GTAAGATGGA CACCAGAAGC AGAAGCAGCT 60 TTCCAGGCCC TAAAGAAATC CCTAACCCAA GCCCCAGTGT TAAGCTTGCC AACGGGGCAA 120 GACTTTTCTT TATATGTCAC AGAAAAACAG GAATAGCTCT AGGAGTCCTT ACACAGGTCC 180 AAGGGACAAG CTTGCAACCT GTGGCATACC TGAGTAAGGA AACTGATGTA NTGGCAAAGG 240 GTTGGCCTCA TTGTTTACAG GTAGGGCAGC AGTAGCAGTC TTAGTTTCTG AAACAGTTAA 300 AATAATACAG GGAAGAGATC TTACTGTGTG GACATCTCAT GATGTGAACG GCATACTCAC 360 TGCTAAAGAG GACTTGTGGC TGTCAGACAA CCATTTACTT AAATAGCAGG TTCTATTACT 420 TGAAGTGCCA GTGCTGCGAC TGCACATTTG TGCAACTCTT AACCCAGCCA CATTTCTTCC 480 AGACAATGAA GAAAAGATAG AACATAACTG TCAACAAGTA ATTGCTCAAA CCTATGCTGC 540 TCGAGGGGAC CTTCTAGAGG TTCCCTTGAC TGATCCCGAC CTCAACTTGT ATACTGATGG 600 AAGTTCCTTG GCAGAAAAAG GACTTTGAAA AGCGGGGTAT GCAGTGATCA GTGATAATGG 660 AATACTTGAA AGTAATCGCC TCACTCCAGG AACTAGTGCT CACCTGGCAG AACTAATAGC 720 CCTCACTTGG GCACTAGAAT TAGGAGAAGG AAAAAGGGTA AATATATATT CAGACTCTAA 780 GTATGCTTAC CTAGTCCTCC ATGCCCATGC AGCAATATGG AGAGAGAGGG AATTCCTAAC 840 TTCTGAGGGA ACACCTATCA ACCATCAGGG AAGCCATTAG GAGATTATTA TTGGCTGTAC 900 AGAAACCTAA AGAGGTGGCA GTCTTACACT GCCAGGGTCA TCAGGAAGAA GAGGAAAGGG 960 AAATAGAAGG CAATCGCCAA GCGGATATTG AAGCAAAAAA AGCCGCAAGG CAGGACTCTC 1020 CATTAGAAAT GCTTATAGAA GGACCCCTAG TATGGGGTAA TCCCCTCTGG GAAACCAAGC 1080 CCCAGTACTC AGCAGGAAAA ATAGAATAGG AAACCTCACA AGGACATACT TTCCTCCCCT 1140 CCAGATGGCT AGCCACTGAG GAAGGAA 1167 78 base pairs nucleotide single linear cDNA 62 TCCAAAGGCA CCAGGGCCCT CAGTGAGGAA CGTATCCAGC CTATACTGGC TTATCCTCAT 60 CCCAAAACCC TAAAGCAA 78 26 amino acids amino acid linear peptide 63 Ser Lys Gly Thr Arg Ala Leu Ser Glu Glu Arg Ile Gln Pro Ile Leu 1 5 10 15 Ala Tyr Pro His Pro Lys Thr Leu Lys Gln 20 25 28 base pairs nucleotide single linear cDNA 64 AAATGTCTGC GGCACCAATC TCCATGTT 28 30 base pairs nucleotide single linear cDNA 65 AAGGGGCATG GACGAGGTGG TGGCTTATTT 30 21 base pairs nucleotide single linear cDNA 66 GGAGAAGAGC AGCATAAGTG G 21 25 base pairs nucleotide single linear cDNA 67 GTGCTGATTG GTGTATTTAC AATCC 25 34 base pairs nucleotide single linear cDNA 68 GACTCGCTGC AGATCGATTT TTTTTTTTTT TTTT 34 30 base pairs nucleotide single linear cDNA 69 GCCATCAAGC CACCCAAGAA CTCTTAACTT 30 30 base pairs nucleotide single linear cDNA 70 CCAATAGCCA GACCATTATA TACACTAATT 30 23 base pairs nucleotide single linear cDNA 71 GCCATAACTG CAACCCAAGA GTT 23 23 base pairs nucleotide single linear cDNA 72 GGACGAGGTG GTGGCTTATT TCT 23 25 base pairs nucleotide single linear cDNA 73 AACTTGCGTG CTAGAAGGAC TAAGG 25 24 base pairs nucleotide single linear cDNA 74 AACTTTTCCC TTTTCCAGAT CCTC 24 22 base pairs nucleotide single linear cDNA 75 GCATACCAGG CAAGTGGACA TT 22 25 base pairs nucleotide single linear cDNA 76 CTGTCCGTTG GGTTTCCTTA CTCCT 25 24 base pairs nucleotide single linear cDNA 77 GAGGCTCTGG AAAAGGGAAA AGTT 24 25 base pairs nucleotide single linear cDNA 78 CTGTCCGTTG GGTTTCCTTA CTCCT 25 25 base pairs nucleotide single linear cDNA 79 AGGAGTAAGG AAACCCAACG GACAG 25 25 base pairs nucleotide single linear cDNA 80 TGTATATAAT GGTCTGGCTA TTGGG 25 25 base pairs nucleotide single linear cDNA 81 AGGAGTAAGG AAACCCAACG GACAG 25 26 base pairs nucleotide single linear cDNA 82 TTCGGCAGAA ACCTGTTATG CCAAGG 26 22 base pairs nucleotide single linear cDNA 83 CTCGATTTCT TGCTGGGCCT TA 22 20 base pairs nucleotide single linear cDNA 84 GTTGATTCCC TCCTCAAGCA 20 20 base pairs nucleotide single linear cDNA 85 CTCTACCAAT CAGCATGTGG 20 19 base pairs nucleotide single linear cDNA 86 TGTTCCTCTT GGTCCCTAT 19 433 amino acids amino acid linear peptide 87 Met Ala Thr Ala Thr Gly Thr Gly Ile Ala Gly Leu Ser Thr Ser Leu 1 5 10 15 Ser Tyr Tyr His Thr Leu Ser Lys Asn Phe Ser Asp Ser Leu Gln Glu 20 25 30 Ile Met Lys Ser Ile Leu Thr Leu Gln Ser Gln Leu Asp Ser Leu Ala 35 40 45 Ala Met Thr Leu Gln Asn Arg Arg Gly Pro His Leu Leu Thr Ala Glu 50 55 60 Lys Gly Gly Leu Cys Thr Phe Leu Gly Glu Glu Cys Cys Phe Tyr Thr 65 70 75 80 Asn Gln Ser Gly Ile Val Arg Asp Ala Thr Trp His Leu Gln Glu Arg 85 90 95 Ala Ser Asp Ile Arg Gln Cys Leu Ser Asn Ser Tyr Thr Asn Leu Trp 100 105 110 Ser Trp Ala Thr Trp Leu Leu Pro Phe Leu Gly Pro Met Ala Ala Ile 115 120 125 Leu Leu Leu Leu Thr Phe Gly Pro Cys Ile Phe Lys Leu Leu Val Lys 130 135 140 Phe Val Ser Ser Arg Ile Glu Ala Ile Lys Leu Gln Met Val Leu Gln 145 150 155 160 Met Glu Pro Gln Met Ser Ser Thr Asn Asn Phe Tyr Gln Gly Pro Leu 165 170 175 Glu Arg Ser Thr Gly Thr Ser Thr Ser Leu Glu Ile Pro Leu Trp Lys 180 185 190 Thr Leu Gln Leu Gln Gly Pro Phe Phe Ala Pro Ile Gln Gln Glu Val 195 200 205 Ala Arg Ala Val Ile Gly Gln Ile Pro Asn Ser Ser Trp Gly Val Leu 210 215 220 Phe Arg Gly Gly Ile Glu Glu Val Thr Ala Cys Trp Gln Pro His Ser 225 230 235 240 Pro Arg Trp Xaa Ser Val Pro Pro Gln Pro Trp Cys Pro Leu Trp Pro 245 250 255 Cys Leu Arg Ser Pro Ser Ala Cys His Cys Thr Val Gly Ala Ser Phe 260 265 270 Trp Ala Gly Gln Gly Arg Ser Gln Leu Pro Gln Leu Ala Gly Arg Tyr 275 280 285 Gly Gly Arg Asp Ala Gly Gly Asn Gln Gly Cys Ala Trp Arg Leu Arg 290 295 300 Ala Ser Met Ser Ser Arg Trp Ala Trp Ala Arg Arg Ala Pro His Ser 305 310 315 320 Gly Ser Glu Gly Leu Ser Thr Trp Ala Arg Gln Met Leu Cys Ser Thr 325 330 335 Ser Ser Leu Gly Leu Ser Cys Leu Pro Arg Gly Ala Gly Leu Arg Glu 340 345 350 Xaa Ala Ala Cys Pro Cys Leu Ser Pro Pro Pro Arg Arg Gly Phe Leu 355 360 365 His Ser Pro Ser Phe Pro Asp Lys His His Pro Leu Ser Thr Val Pro 370 375 380 Ser Pro Ile Asn His Pro Arg Val Glu Glu Cys Gly His Thr Ala Arg 385 390 395 400 Asp Trp Gln Ala Val Pro Leu Ala Ala Leu Val Arg Asp Pro Leu Arg 405 410 415 Glu Ala Ser Trp Ala Pro Glu Ser Gly Gly Asp Leu Glu Asn Leu Tyr 420 425 430 Val 433 693 base pairs nucleotide single linear cDNA 88 CTTCCCCAAC TAATAAGGAC CCCCCTTTCA ACCCAAACAG TCCAAAAGGA CATAGACAAA 60 GGAGTAAACA ATGAACCAAA GAGTGCCAAT ATTCCCTGGT TATGCACCCT CCAAGCGGTG 120 GGAGAAGAAT TCGGCCCAGC CAGAGTGCAT GTACCTTTTT CTCTCTCACA CTTGAAGCAA 180 ATTAAAATAG ACNTAGGTNA ATTNTCAGAT AGCCCTGATG GYTATATTGA TGTTTTACAA 240 GGATTAGGAC AATCCTTTGA TCTGACATGG AGAGATATAA TATTACTGCT AAATCAGACG 300 CTAACCTCAA ATGAGAGAAG TGCTGCCATA ACTGGAGCCC GAGAGTTTGG CAATCTCTGG 360 TATCTCAGTC AGGTCAATGA TAGGATGACA ACGGAGGAAA GAGAACGATT CCCCACAGGG 420 CAGCAGGCAG TTCCCAGTGT AGCTCCTCAT TGGGACACAG AATCAGAACA TGGAGATTGG 480 TGCCGCAGAC ATTTACTAAC TTGCGTGCTA GAAGGACTAA GGAAAACTAG GAAGACTATG 540 AATTATTCAA TGATGTCCAC TATAACACAG GGGAAAGGAA GAAAATCCTA CTGCCTTTCT 600 GGAGAGACTA AGGGAGGCAT TGAGGAAGCA TACCAGGCAA GTGGACATTG GAGGCTCTGG 660 AAAAGGGAAA AGTTGGGCAA ATTGAATGCC TAA 693 1577 base pairs nucleotide single linear cDNA 89 AACTTGCGTG CTAGAAGGAC TAAGGAAAAC TAGGAAGACT ATGAATTATT CAATGATGTC 60 CACTATAACA CAGGGGAAAG GAAGAAAATC CTACTGCCTT TCTGGAGAGA CTAAGGGAGG 120 CATTGAGGAA GCATACCAGG CAAGTGGACA TTGGAGGCTC TGGAAAAGGG AAAAGTTGGG 180 CAAATTGAAT GCCTAATAGG GCTTGCTTCC AGTGCAGTCT ACAAGGACGC TTTAGAAAAG 240 ATTGTCCAAG TAGAAATAAG CCGCCCCTCG TCCATGCCCC TTATGTCAAG GGAATCACTG 300 GAAGGCCTAC TGCCCCAGGG GACGAAGGTC CTCTGAGTCA GAAGCCACTA ACCTGATGAT 360 CCAGCAGCAG GACTGAGGGT GCCCGGGGCA AGTGCCAGCC CATGCCATCA CCCTCAGAGC 420 CCCGGGTATG TTTGACCATT GAGAGCCAGG AAGTTAACTG TCTCCTGGAC ACTGGCGCAG 480 CCTTCTCAGT CTTACTTTCC TGTCCCAGAC AATTGTCCTC CAGATCTGTC ACTATCCGAG 540 GGGTCCTAAG ACAGCCAGTC ACTACATACT TCTCTCAGCC ACTAAGTTGT GACTGGGGAA 600 CTTTACTCTT TTCACATGCT TTTCTAATTA TGCCTGAAAG CCCCACTCCC TTGTTAGGGA 660 GAGACATTTT AGCAAAAGCA GGGGCCATTA TACACCTGAA CATAGGAAAA GGAATACCCA 720 TTTGCTGTCC CCTGCTTGAG GAAGGAATTA ATCCTGAAGT CTGGGCAATA GAAGGACAAT 780 ATGGACAAGC AAAGAATGCC CGTCCTGTTC AAGTTAAACT AAAGGATTCT GCCTCCTTTC 840 CCTACCAAAG GAAGTACCCT CTTAGACCCG AGGCCCTACA AGGACTCAAA AGATTGTTAA 900 GGACCTAAAA GCCCAAGGCC TAGTAAAACC ATGCAGTAGC CCCTGCAATA CTCCAATTTT 960 AGGAGTAAGG AAACCCAACG GACAGTGGAG GTTAGTGCAA GATCTCAGGA TTATTAATGA 1020 GGCTGTTTTT CCTCTATACC CAGCTGTATC TAGCCCTTAT ACTCTGCTTT CCCTAATACC 1080 AGAGGAAGCA GAGTAGTTTA CAGTCCTGGA CCTTAAGGAT GCCTCTTTCT GCATCCCTGT 1140 ACATCCTGAT TCTCAATTCT TGTTTGTCTT TGAAGATCCT TTGAACCCAA TGTCTCAATT 1200 CACCTGGACT GTTTTACCCC AGGGGTTCCG GGATAGCCCC CATCTATTTG GCCAGGCATT 1260 AGCCCAAGAC TTGAGCCAAT TCTCATACCT GGACATCTTG TCCTTCGGTA TGGGATGATT 1320 TAATTTTAGC CACCCGTTCA GAAACCTTGT GCCATCAAGC CACCCAAGCG TTCTTAAATT 1380 TCCTCACTCC GTGTGGCTAC AAGGTTTCCA AACCAAAGGC TCAGCTCTGC TCACAGCAGG 1440 TTAAATACTT AGGGTTAAAA TTATCCAAAG GCACCAGGGC CCTCTGTGAG GAATGTATCC 1500 AACCTGTACT GGCTTATCTT CATCCCAAAA CCCTAAAGCA ACTAAGAAGG TCCTTGGCAT 1560 AACAGGTTTC TGCCGAA 1577 182 amino acids amino acid linear peptide 90 Ser Ser Ser Arg Thr Glu Gly Ala Arg Gly Lys Cys Gln Pro Met Pro 1 5 10 15 Ser Pro Ser Glu Pro Arg Val Cys Leu Thr Ile Glu Ser Gln Glu Val 20 25 30 Asn Cys Leu Leu Asp Thr Gly Ala Ala Phe Ser Val Leu Leu Ser Cys 35 40 45 Pro Arg Gln Leu Ser Ser Arg Ser Val Thr Ile Arg Gly Val Leu Arg 50 55 60 Gln Pro Val Thr Thr Tyr Phe Ser Gln Pro Leu Ser Cys Asp Trp Gly 65 70 75 80 Thr Leu Leu Phe Ser His Ala Phe Leu Ile Met Pro Glu Ser Pro Thr 85 90 95 Pro Leu Leu Gly Arg Asp Ile Leu Ala Lys Ala Gly Ala Ile Ile His 100 105 110 Leu Asn Ile Gly Lys Gly Ile Pro Ile Cys Cys Pro Leu Leu Glu Glu 115 120 125 Gly Ile Asn Pro Glu Val Trp Ala Ile Glu Gly Gln Tyr Gly Gln Ala 130 135 140 Lys Asn Ala Arg Pro Val Gln Val Lys Leu Lys Asp Ser Ala Ser Phe 145 150 155 160 Pro Tyr Gln Arg Lys Tyr Pro Leu Arg Pro Glu Ala Leu Gln Gly Leu 165 170 175 Lys Arg Leu Leu Arg Thr 180 36 base pairs nucleotide single linear cDNA 91 AGATCTGCAG AATTCGATAT CACCCCCCCC CCCCCC 36 22 base pairs nucleotide single linear cDNA 92 AGATCTGCAG AATTCGATAT CA 22

Claims (8)

What is claimed is:
1. A process for detecting a pathological and/or infective agent associated with multiple sclerosis and/or rheumatoid arthritis, in a biological sample, comprising bringing a nucleic acid into contact with the biological sample, and detecting hybridization, said nucleic acid comprising a nucleotide sequence having a succession of at least 100 contiguous monomers of a nucleotide sequence selected from the group consisting of said sequences SEQ ID NO: 46, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 56, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 89 and their complementary sequences.
2. A process for detecting a pathological and/or infective agent associated with multiple sclerosis and/or rheumatoid arthritis, in a biological sample, comprising bringing a nucleic acid into contact with the biological sample, and detecting hybridization, said nucleic acid consisting of 100 or more contiguous monomers of a nucleotide sequence selected from the group consisting of SEQ ID NO: 46, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 56, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 89 and their complementary sequences.
3. A process for detecting a pathological and/or infective agent associated with multiple sclerosis and/or rheumatoid arthritis, in a biological sample, comprising bringing a nucleic acid into contact with the biological sample, and detecting hybridization, said nucleic acid comprising 100 or more contiguous monomers of a nucleotide sequence selected from the group consisting of SEQ ID NO: 46, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 56, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 89 and their complementary sequences.
4. A probe for specifically hybridizing with a nucleic acid of a virus associated with multiple sclerosis, said probe comprising at least ten contiguous monomers of the nucleotide sequence of a nucleic acid, said nucleic acid comprising a nucleotide sequence having a succession of at least 100 contiguous monomers of a nucleotide sequence selected from the group consisting of SEQ ID NO: 46, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 56, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 89 and their complementary sequences.
5. A probe as defined in claim 4, having 10 to 100 contiguous monomers of said nucleotide sequence of the nucleic acid.
6. A specific primer for amplification by polymerization of a nucleic acid of a virus associated with multiple sclerosis, said primer comprising at least ten contiguous monomers of the nucleotide sequence of a nucleic acid, said nucleic acid comprising a nucleotide sequence having a succession of at least 100 contiguous monomers of a nucleotide sequence selected from the group consisting of SEQ ID NO: 46, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 56, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 89 and their complementary sequences.
7. A primer as defined in claim 6, having 10 to 100 contiguous monomers of said nucleotide sequence of the nucleic acid.
8. A process for detecting a pathological and/or infective agent associated with multiple sclerosis, in a biological sample, comprising bringing a probe according to claim 4 into contact with the biological sample, and detecting hybridization.
US10/430,442 1995-08-03 2003-05-07 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purpose Abandoned US20030186391A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/430,442 US20030186391A1 (en) 1995-08-03 2003-05-07 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purpose
US11/463,109 US7932350B2 (en) 1995-08-03 2006-08-08 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
FR9509643A FR2737500B1 (en) 1995-08-03 1995-08-03 VIRAL MATERIAL AND NUCLEOTIDE FRAGMENTS ASSOCIATED WITH MULTIPLE SCLEROSIS, FOR DIAGNOSIS, PROPHYLACTICS AND THERAPEUTICS
FR9509643 1995-08-03
US08/691,563 US6001987A (en) 1995-08-03 1996-08-02 Isolated nucleotide sequences associated with Multiple sclerosis
US09/374,766 US6579526B1 (en) 1995-08-03 1999-08-16 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes
US10/430,442 US20030186391A1 (en) 1995-08-03 2003-05-07 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purpose

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/374,766 Division US6579526B1 (en) 1995-08-03 1999-08-16 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/463,109 Division US7932350B2 (en) 1995-08-03 2006-08-08 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes

Publications (1)

Publication Number Publication Date
US20030186391A1 true US20030186391A1 (en) 2003-10-02

Family

ID=9481811

Family Applications (4)

Application Number Title Priority Date Filing Date
US08/691,563 Expired - Lifetime US6001987A (en) 1995-08-03 1996-08-02 Isolated nucleotide sequences associated with Multiple sclerosis
US09/374,766 Expired - Lifetime US6579526B1 (en) 1995-08-03 1999-08-16 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes
US10/430,442 Abandoned US20030186391A1 (en) 1995-08-03 2003-05-07 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purpose
US11/463,109 Expired - Fee Related US7932350B2 (en) 1995-08-03 2006-08-08 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/691,563 Expired - Lifetime US6001987A (en) 1995-08-03 1996-08-02 Isolated nucleotide sequences associated with Multiple sclerosis
US09/374,766 Expired - Lifetime US6579526B1 (en) 1995-08-03 1999-08-16 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/463,109 Expired - Fee Related US7932350B2 (en) 1995-08-03 2006-08-08 Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes

Country Status (19)

Country Link
US (4) US6001987A (en)
EP (2) EP1916304B1 (en)
JP (2) JP4444372B2 (en)
AT (2) ATE374252T1 (en)
AU (1) AU730080B2 (en)
BG (1) BG62977B1 (en)
BR (1) BR9606566A (en)
CA (1) CA2201282C (en)
CZ (1) CZ135797A3 (en)
DE (1) DE69637264T2 (en)
ES (2) ES2379011T3 (en)
FR (1) FR2737500B1 (en)
HU (1) HUP9900425A3 (en)
IS (1) IS4455A (en)
NO (1) NO971493L (en)
NZ (1) NZ316080A (en)
PL (1) PL319512A1 (en)
SK (1) SK56797A3 (en)
WO (1) WO1997006260A1 (en)

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2737500B1 (en) * 1995-08-03 1997-08-29 Bio Merieux VIRAL MATERIAL AND NUCLEOTIDE FRAGMENTS ASSOCIATED WITH MULTIPLE SCLEROSIS, FOR DIAGNOSIS, PROPHYLACTICS AND THERAPEUTICS
US7241876B2 (en) 1996-01-11 2007-07-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US7785842B2 (en) * 1996-03-26 2010-08-31 Oncomedx, Inc. Comparative analysis of extracellular RNA species
US8043835B1 (en) 1996-03-26 2011-10-25 Oncomedx, Inc. Methods for detecting and monitoring cancer using extracellular RNA
PT938320E (en) * 1996-03-26 2010-09-22 Michael S Kopreski Method enabling use of extracellular rna extracted from plasma or serum to detect, monitor or evaluate cancer
US6759217B2 (en) * 1996-03-26 2004-07-06 Oncomedx, Inc. Method enabling use of extracellular RNA extracted from plasma or serum to detect, monitor or evaluate cancer
WO1998014617A1 (en) * 1996-10-04 1998-04-09 Chronix Biomedical Diagnostic detection of nucleic acids
CA2272845C (en) * 1996-11-26 2010-01-12 Bio Merieux Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes
US8440396B2 (en) * 1997-03-14 2013-05-14 Oncomedx, Inc. Method enabling use of extracellular RNA extracted from plasma or serum to detect, monitor or evaluate cancer
US20070009934A1 (en) * 1997-03-14 2007-01-11 Kopreski Michael S Method enabling use of extracellular RNA extracted from plasma or serum to detect, monitor or evaluate cancer
FR2762601B3 (en) * 1997-04-29 1999-06-04 Bio Merieux POLYPEPTIDE CAPABLE OF RESPONDING TO ANTIBODIES OF PATIENTS WITH MULTIPLE SCLEROSIS AND USES
FR2762600B1 (en) * 1997-04-29 1999-11-05 Bio Merieux POLYPEPTIDE CAPABLE OF REACTING WITH ANTIBODIES OF PATIENTS WITH MULTIPLE SCLEROSIS AND USES
DE69836484T2 (en) * 1997-07-07 2007-09-13 Bio Merieux Endogenous retroviral genomic cliniin element associated with autoimmune disease and / or perturbation, use as a marker
US20040176314A1 (en) * 1997-07-07 2004-09-09 Bio Merieux Endogenetic retroviral sequences, associated with autoimmune diseases or with pregnancy disorders
FR2765588A1 (en) 1997-07-07 1999-01-08 Bio Merieux RETROVIRAL NUCLEIC MATERIAL AND NUCLEOTIDE FRAGMENTS ESPECIALLY ASSOCIATED WITH MULTIPLE SCLEROSIS AND / OR RHUMATOID POLYARTHRITIS, FOR DIAGNOSIS, PROPHYLACTICS AND THERAPEUTICS
JP2002511277A (en) * 1998-04-08 2002-04-16 エムエス・リサーチ・アクティーゼルスカブ Diagnosis of multiple sclerosis and other demyelinating diseases
FR2780069B1 (en) * 1998-06-23 2002-06-28 Inst Nat Sante Rech Med FAMILY OF NUCLEIC SEQUENCES AND DEDUCTED PROTEIN SEQUENCES WITH HUMAN ENDOGENOUS RETROVIRAL PATTERNS AND THEIR APPLICATIONS
US20090233276A1 (en) * 1998-09-22 2009-09-17 Oncomedx, Inc. Method Enabling the Use of Extracellular Ribonucleic Acid (RNA) Extracted from Plasma or Serum to Detect, Monitor or Evaluate Cancer or Premalignant Conditions
US8163524B2 (en) * 1998-09-22 2012-04-24 Oncomedx, Inc. Comparative analysis of extracellular RNA species
US20060204989A1 (en) * 1998-09-22 2006-09-14 Kopreski Michael S Comparative analysis of extracellular RNA species
FR2788784A1 (en) * 1999-01-21 2000-07-28 Bio Merieux New nucleic acid from human endogenous retrovirus, useful e.g. for diagnosis of autoimmune disease and complications of pregnancy, contains at least part of the gag gene
EP1029917A1 (en) 1999-02-15 2000-08-23 Bio Merieux The LTR region of MSRV-1 and the proteins it encodes, and probes and methods for detecting MSRV-1 retrovirus
EP1204754A2 (en) * 1999-08-20 2002-05-15 Curagen Corporation Polynucleotides expressed in activated t-lymphocytes and proteins encoded thereby
DE10012875B4 (en) * 2000-03-16 2004-04-01 Infineon Technologies Ag pager
IL151868A0 (en) * 2000-03-21 2003-04-10 Ms Res As An infective endogenous retrovirus and its association with demyelinating diseases and other diseases
DE60239470D1 (en) * 2001-07-25 2011-04-28 Oncomedx Inc METHOD FOR EVALUATING PATHOLOGICAL ILLICIT STATES USING EXTRACELLULAR RNA
US20100159464A1 (en) * 2001-11-05 2010-06-24 Oncomedx, Inc. Method for Detection of DNA Methyltransferase RNA in Plasma and Serum
US20030104454A1 (en) * 2001-11-05 2003-06-05 Kopreski Michael S. Method for detection of DNA methyltransferase RNA in plasma and serum
US20090136942A1 (en) * 2007-09-18 2009-05-28 Oncomedx, Inc. Analysis of Extracellular RNA
WO2010030931A1 (en) * 2008-09-11 2010-03-18 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Oligonucleotides targeting human endogenous retrovirus 9 (erv-9) long terminal repeat (ltr) and methods of use

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4311686A (en) * 1979-04-30 1982-01-19 The Immunology Development Corporation Methodology for the immunodiagnosis of multiple sclerosis and/or malignant diseases from blood sample analysis
US4346074A (en) * 1978-08-24 1982-08-24 National Research Development Corp. Pasteurellosis vaccines
US4388298A (en) * 1982-07-14 1983-06-14 The United States Of America As Represented By The Secretary Of Agriculture Propagation of hemorrhagic enteritis virus in a turkey cell line and vaccine produced
US4396600A (en) * 1980-12-18 1983-08-02 Gus Gallucci Adult schistosome worm-derived antigenic substance and method of obtaining same
US4520113A (en) * 1984-04-23 1985-05-28 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Serological detection of antibodies to HTLV-III in sera of patients with AIDS and pre-AIDS conditions
US4708818A (en) * 1983-09-15 1987-11-24 Institut Pasteur Human immunodeficiency viruses associated with Acquired Immune Deficiency Syndrome (AIDS), a diagnostic method for AIDS and pre-AIDS, and a kit therefor
US4900553A (en) * 1987-09-11 1990-02-13 Case Western Reserve University Method of reducing glial scar formation and promoting axon and blood vessel growth and/or regeneration through the use of activated immature astrocytes
US5158976A (en) * 1990-10-29 1992-10-27 The Children's Medical Center Corporation Controlling glutamine/glutamate related neuronal injury
US5219837A (en) * 1990-06-21 1993-06-15 Trustees Of The University Of Pennsylvania Method of stimulating myelination of cells
US5225352A (en) * 1989-09-06 1993-07-06 Centre National De La Recherche Scientifique Agents for the diagnosis of demyelinating neuropathies, in particular multiple sclerosis
US5494807A (en) * 1991-03-07 1996-02-27 Virogenetics Corporation NYVAC vaccinia virus recombinants comprising heterologous inserts
US5580766A (en) * 1994-01-14 1996-12-03 Alexion Pharmaceuticals, Inc. Retroviral vector particles for transducing non-proliferating cells
US5585262A (en) * 1992-04-03 1996-12-17 Bio Merieux Process for the production of a viable cell culture infected by a multiple sclerosis-associated virus
US5650318A (en) * 1992-04-03 1997-07-22 Bio Merieux Process and culture medium for the production of cells infected by a multiple sclerosis-associated virus
US5691147A (en) * 1994-06-02 1997-11-25 Mitotix, Inc. CDK4 binding assay
US5710037A (en) * 1994-06-10 1998-01-20 The United States Of America As Represented By The Department Of Health And Human Services Retroviral vector particles
US5800980A (en) * 1899-02-04 1998-09-01 Bio Merieux Detection of MSRV1 virus and MSRV2 pathogen and/or infective agent associated with multiple sclerosis, by nucleic acid hybridization
US6001987A (en) * 1995-08-03 1999-12-14 Bio Merieux Isolated nucleotide sequences associated with Multiple sclerosis

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4647773A (en) * 1984-04-23 1987-03-03 The United States Of America As Represented By The Department Of Health And Human Services Method of continuous production of retroviruses (HTLV-III) from patients with AIDS and pre-AIDS
NZ218050A (en) * 1985-11-13 1989-05-29 Wistar Inst Test for the presence of htlv-1v
EP0326395A2 (en) * 1988-01-29 1989-08-02 City Of Hope Method of detecting and identifying certain viral sequences
DK173091D0 (en) * 1991-10-11 1991-10-11 Schleroseforeningen The Danish BIOLOGICAL MATERIALS
EP0642585B1 (en) * 1992-05-18 2005-12-28 Genentech, Inc. Activation of oligomerizing receptors by using fused receptor ligands
GB9310657D0 (en) * 1993-05-24 1993-07-07 Univ London Retrovirus
FR2731356B1 (en) * 1995-03-09 1997-04-30 Bio Merieux MSRV-1 VIRUS AND PATHOGENIC AND / OR INFECTING MSRV-2 AGENT ASSOCIATED WITH RHUMATOID POLYARTHRITIS

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5800980A (en) * 1899-02-04 1998-09-01 Bio Merieux Detection of MSRV1 virus and MSRV2 pathogen and/or infective agent associated with multiple sclerosis, by nucleic acid hybridization
US4346074A (en) * 1978-08-24 1982-08-24 National Research Development Corp. Pasteurellosis vaccines
US4311686A (en) * 1979-04-30 1982-01-19 The Immunology Development Corporation Methodology for the immunodiagnosis of multiple sclerosis and/or malignant diseases from blood sample analysis
US4396600A (en) * 1980-12-18 1983-08-02 Gus Gallucci Adult schistosome worm-derived antigenic substance and method of obtaining same
US4396600B1 (en) * 1980-12-18 1986-02-11
US4388298A (en) * 1982-07-14 1983-06-14 The United States Of America As Represented By The Secretary Of Agriculture Propagation of hemorrhagic enteritis virus in a turkey cell line and vaccine produced
US4708818A (en) * 1983-09-15 1987-11-24 Institut Pasteur Human immunodeficiency viruses associated with Acquired Immune Deficiency Syndrome (AIDS), a diagnostic method for AIDS and pre-AIDS, and a kit therefor
US4520113A (en) * 1984-04-23 1985-05-28 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Serological detection of antibodies to HTLV-III in sera of patients with AIDS and pre-AIDS conditions
US4900553A (en) * 1987-09-11 1990-02-13 Case Western Reserve University Method of reducing glial scar formation and promoting axon and blood vessel growth and/or regeneration through the use of activated immature astrocytes
US5225352A (en) * 1989-09-06 1993-07-06 Centre National De La Recherche Scientifique Agents for the diagnosis of demyelinating neuropathies, in particular multiple sclerosis
US5219837A (en) * 1990-06-21 1993-06-15 Trustees Of The University Of Pennsylvania Method of stimulating myelination of cells
US5158976A (en) * 1990-10-29 1992-10-27 The Children's Medical Center Corporation Controlling glutamine/glutamate related neuronal injury
US5494807A (en) * 1991-03-07 1996-02-27 Virogenetics Corporation NYVAC vaccinia virus recombinants comprising heterologous inserts
US5585262A (en) * 1992-04-03 1996-12-17 Bio Merieux Process for the production of a viable cell culture infected by a multiple sclerosis-associated virus
US5650318A (en) * 1992-04-03 1997-07-22 Bio Merieux Process and culture medium for the production of cells infected by a multiple sclerosis-associated virus
US5580766A (en) * 1994-01-14 1996-12-03 Alexion Pharmaceuticals, Inc. Retroviral vector particles for transducing non-proliferating cells
US5871745A (en) * 1994-02-04 1999-02-16 Bio Merieux Multiple sclerosis related virus
US5871996A (en) * 1994-02-04 1999-02-16 Bio Merieux Retrovirus agents MSRV1 and MSRV2 associated with multiple sclerosis
US5962217A (en) * 1994-02-04 1999-10-05 Bio Merieux MSRV1 virus and MSRV2 pathogen and/or infective agent associated with multiple sclerosis, and biopolymer constituents thereof
US5691147A (en) * 1994-06-02 1997-11-25 Mitotix, Inc. CDK4 binding assay
US5710037A (en) * 1994-06-10 1998-01-20 The United States Of America As Represented By The Department Of Health And Human Services Retroviral vector particles
US6001987A (en) * 1995-08-03 1999-12-14 Bio Merieux Isolated nucleotide sequences associated with Multiple sclerosis

Also Published As

Publication number Publication date
WO1997006260A1 (en) 1997-02-20
BG62977B1 (en) 2000-12-29
US6001987A (en) 1999-12-14
EP0789077B1 (en) 2007-09-26
HUP9900425A2 (en) 1999-05-28
FR2737500A1 (en) 1997-02-07
CA2201282C (en) 2013-04-02
HUP9900425A3 (en) 1999-11-29
US20070117189A1 (en) 2007-05-24
NZ316080A (en) 1999-04-29
IS4455A (en) 1997-03-26
AU730080B2 (en) 2001-02-22
US7932350B2 (en) 2011-04-26
ATE374252T1 (en) 2007-10-15
DE69637264D1 (en) 2007-11-08
JP4444372B2 (en) 2010-03-31
SK56797A3 (en) 1998-09-09
ES2379011T3 (en) 2012-04-19
NO971493D0 (en) 1997-04-02
NO971493L (en) 1997-06-03
JP5143814B2 (en) 2013-02-13
DE69637264T2 (en) 2008-06-26
PL319512A1 (en) 1997-08-18
CA2201282A1 (en) 1997-02-20
BG101355A (en) 1997-12-30
EP1916304B1 (en) 2012-01-18
ATE541933T1 (en) 2012-02-15
US6579526B1 (en) 2003-06-17
AU6823296A (en) 1997-03-05
FR2737500B1 (en) 1997-08-29
CZ135797A3 (en) 1998-06-17
EP1916304A2 (en) 2008-04-30
ES2293645T3 (en) 2008-03-16
EP0789077A1 (en) 1997-08-13
JPH11502416A (en) 1999-03-02
BR9606566A (en) 1997-12-30
JP2010047604A (en) 2010-03-04
EP1916304A3 (en) 2008-05-07

Similar Documents

Publication Publication Date Title
US6579526B1 (en) Viral material and nucleotide fragments associated with multiple sclerosis, for diagnostic, prophylactic and therapeutic purposes
US6582703B2 (en) Isolated nucleotide sequences associated with multiple sclerosis or rheumatoid arthritis and a process of detecting
US6342383B1 (en) Cell lines and viral isolates associated with multiple sclerosis
AU625174B2 (en) Hiv-2 virus variants
US6548635B1 (en) Retrovirus from the HIV type O and its use (MVP-2901/94)
US20040176314A1 (en) Endogenetic retroviral sequences, associated with autoimmune diseases or with pregnancy disorders
US20040043381A1 (en) The LTR region of MSRV-1 and the proteins it encodes and probes and methods for detecting MSRV-1 retrovirus

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION