WO1997033610A1 - DNA ENCODING 28 kDa GLUTATHIONE S-TRANSFERASE OF SCHISTOSOMA MANSONI AND USES THEREOF - Google Patents
DNA ENCODING 28 kDa GLUTATHIONE S-TRANSFERASE OF SCHISTOSOMA MANSONI AND USES THEREOF Download PDFInfo
- Publication number
- WO1997033610A1 WO1997033610A1 PCT/US1997/003977 US9703977W WO9733610A1 WO 1997033610 A1 WO1997033610 A1 WO 1997033610A1 US 9703977 W US9703977 W US 9703977W WO 9733610 A1 WO9733610 A1 WO 9733610A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- plasmid
- dna
- mice
- cells
- protein
- Prior art date
Links
- 241000242680 Schistosoma mansoni Species 0.000 title claims abstract description 43
- 102000005720 Glutathione transferase Human genes 0.000 title claims abstract description 40
- 108010070675 Glutathione transferase Proteins 0.000 title claims abstract description 40
- 238000000034 method Methods 0.000 claims abstract description 35
- 208000015181 infectious disease Diseases 0.000 claims abstract description 25
- 229960005486 vaccine Drugs 0.000 claims abstract description 17
- 108091028043 Nucleic acid sequence Proteins 0.000 claims abstract description 8
- 230000002458 infectious effect Effects 0.000 claims abstract description 7
- 239000013612 plasmid Substances 0.000 claims description 82
- 108020004414 DNA Proteins 0.000 claims description 73
- 108090000623 proteins and genes Proteins 0.000 claims description 62
- 102000004169 proteins and genes Human genes 0.000 claims description 35
- 241001465754 Metazoa Species 0.000 claims description 20
- 210000003205 muscle Anatomy 0.000 claims description 16
- 230000024932 T cell mediated immunity Effects 0.000 claims description 14
- 238000011282 treatment Methods 0.000 claims description 13
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 9
- 239000000203 mixture Substances 0.000 claims description 8
- 229920001184 polypeptide Polymers 0.000 claims description 5
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 5
- 208000030852 Parasitic disease Diseases 0.000 abstract description 2
- 241000699670 Mus sp. Species 0.000 description 85
- 210000004027 cell Anatomy 0.000 description 60
- 235000018102 proteins Nutrition 0.000 description 33
- 230000004044 response Effects 0.000 description 26
- 239000011324 bead Substances 0.000 description 25
- 235000013601 eggs Nutrition 0.000 description 25
- 238000002347 injection Methods 0.000 description 24
- 239000007924 injection Substances 0.000 description 24
- 108091061960 Naked DNA Proteins 0.000 description 23
- 238000002255 vaccination Methods 0.000 description 23
- 210000004698 lymphocyte Anatomy 0.000 description 19
- 210000001744 T-lymphocyte Anatomy 0.000 description 18
- 230000000694 effects Effects 0.000 description 18
- 230000014509 gene expression Effects 0.000 description 18
- 239000000427 antigen Substances 0.000 description 15
- 108091007433 antigens Proteins 0.000 description 15
- 102000036639 antigens Human genes 0.000 description 15
- 241000699666 Mus <mouse, genus> Species 0.000 description 14
- 239000012634 fragment Substances 0.000 description 14
- 238000012360 testing method Methods 0.000 description 14
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 13
- 210000004185 liver Anatomy 0.000 description 13
- 239000000047 product Substances 0.000 description 13
- 108010005774 beta-Galactosidase Proteins 0.000 description 12
- 238000002965 ELISA Methods 0.000 description 11
- 238000011161 development Methods 0.000 description 11
- 244000045947 parasite Species 0.000 description 11
- 241000701022 Cytomegalovirus Species 0.000 description 10
- 238000011238 DNA vaccination Methods 0.000 description 10
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 10
- 238000002474 experimental method Methods 0.000 description 10
- 239000000499 gel Substances 0.000 description 10
- 230000033687 granuloma formation Effects 0.000 description 10
- 239000002953 phosphate buffered saline Substances 0.000 description 10
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 9
- 206010018691 Granuloma Diseases 0.000 description 9
- 241000282414 Homo sapiens Species 0.000 description 9
- 101001068429 Schistosoma mansoni Glutathione S-transferase class-mu 26 kDa isozyme Proteins 0.000 description 9
- 238000003556 assay Methods 0.000 description 9
- 238000001516 cell proliferation assay Methods 0.000 description 9
- 239000002299 complementary DNA Substances 0.000 description 9
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 9
- 239000011780 sodium chloride Substances 0.000 description 9
- 238000004519 manufacturing process Methods 0.000 description 8
- 201000004409 schistosomiasis Diseases 0.000 description 8
- 210000002966 serum Anatomy 0.000 description 8
- 101710149506 28 kDa protein Proteins 0.000 description 7
- 241000894006 Bacteria Species 0.000 description 7
- 101710137943 Complement control protein C3 Proteins 0.000 description 7
- 241000588724 Escherichia coli Species 0.000 description 7
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 7
- 101710152003 Suppressor of silencing P0 Proteins 0.000 description 7
- 108700019146 Transgenes Proteins 0.000 description 7
- 239000002671 adjuvant Substances 0.000 description 7
- 229940037003 alum Drugs 0.000 description 7
- 238000006243 chemical reaction Methods 0.000 description 7
- 201000010099 disease Diseases 0.000 description 7
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 7
- 230000028996 humoral immune response Effects 0.000 description 7
- 230000028993 immune response Effects 0.000 description 7
- 210000004072 lung Anatomy 0.000 description 7
- 230000001404 mediated effect Effects 0.000 description 7
- 210000002027 skeletal muscle Anatomy 0.000 description 7
- 210000003491 skin Anatomy 0.000 description 7
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 7
- 239000000243 solution Substances 0.000 description 7
- 229920000936 Agarose Polymers 0.000 description 6
- VYZAHLCBVHPDDF-UHFFFAOYSA-N Dinitrochlorobenzene Chemical compound [O-][N+](=O)C1=CC=C(Cl)C([N+]([O-])=O)=C1 VYZAHLCBVHPDDF-UHFFFAOYSA-N 0.000 description 6
- 102000004190 Enzymes Human genes 0.000 description 6
- 108090000790 Enzymes Proteins 0.000 description 6
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 6
- 239000012980 RPMI-1640 medium Substances 0.000 description 6
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 6
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 6
- 241000242678 Schistosoma Species 0.000 description 6
- WQZGKKKJIJFFOK-FPRJBGLDSA-N beta-D-galactose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@H]1O WQZGKKKJIJFFOK-FPRJBGLDSA-N 0.000 description 6
- 230000009089 cytolysis Effects 0.000 description 6
- 229960003180 glutathione Drugs 0.000 description 6
- 230000035755 proliferation Effects 0.000 description 6
- 210000001519 tissue Anatomy 0.000 description 6
- 238000001890 transfection Methods 0.000 description 6
- 238000012546 transfer Methods 0.000 description 6
- 241000283690 Bos taurus Species 0.000 description 5
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 5
- 241000754688 Cercaria Species 0.000 description 5
- 108010074328 Interferon-gamma Proteins 0.000 description 5
- 102000000743 Interleukin-5 Human genes 0.000 description 5
- 229920001213 Polysorbate 20 Polymers 0.000 description 5
- 241000700159 Rattus Species 0.000 description 5
- 241000700605 Viruses Species 0.000 description 5
- 238000004458 analytical method Methods 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 235000011089 carbon dioxide Nutrition 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- 239000013604 expression vector Substances 0.000 description 5
- 239000008188 pellet Substances 0.000 description 5
- 108091033319 polynucleotide Proteins 0.000 description 5
- 102000040430 polynucleotide Human genes 0.000 description 5
- 239000002157 polynucleotide Substances 0.000 description 5
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 5
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 5
- 108091008146 restriction endonucleases Proteins 0.000 description 5
- 239000002002 slurry Substances 0.000 description 5
- 210000000952 spleen Anatomy 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- 239000006228 supernatant Substances 0.000 description 5
- 239000000725 suspension Substances 0.000 description 5
- 238000001262 western blot Methods 0.000 description 5
- 102100026189 Beta-galactosidase Human genes 0.000 description 4
- 108010062580 Concanavalin A Proteins 0.000 description 4
- 108010041986 DNA Vaccines Proteins 0.000 description 4
- 241000287828 Gallus gallus Species 0.000 description 4
- 241000238631 Hexapoda Species 0.000 description 4
- 108010002616 Interleukin-5 Proteins 0.000 description 4
- 108060001084 Luciferase Proteins 0.000 description 4
- 239000005089 Luciferase Substances 0.000 description 4
- 241000283973 Oryctolagus cuniculus Species 0.000 description 4
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 4
- 230000005540 biological transmission Effects 0.000 description 4
- AIYUHDOJVYHVIT-UHFFFAOYSA-M caesium chloride Chemical compound [Cl-].[Cs+] AIYUHDOJVYHVIT-UHFFFAOYSA-M 0.000 description 4
- 239000013592 cell lysate Substances 0.000 description 4
- 235000013330 chicken meat Nutrition 0.000 description 4
- 230000001419 dependent effect Effects 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 239000000835 fiber Substances 0.000 description 4
- 102000054766 genetic haplotypes Human genes 0.000 description 4
- 238000010255 intramuscular injection Methods 0.000 description 4
- 239000007927 intramuscular injection Substances 0.000 description 4
- 238000002955 isolation Methods 0.000 description 4
- 210000002540 macrophage Anatomy 0.000 description 4
- 229940022007 naked DNA vaccine Drugs 0.000 description 4
- JYVLIDXNZAXMDK-UHFFFAOYSA-N pentan-2-ol Chemical compound CCCC(C)O JYVLIDXNZAXMDK-UHFFFAOYSA-N 0.000 description 4
- 238000000746 purification Methods 0.000 description 4
- 210000003314 quadriceps muscle Anatomy 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 102000007469 Actins Human genes 0.000 description 3
- 108010085238 Actins Proteins 0.000 description 3
- 238000011725 BALB/c mouse Methods 0.000 description 3
- 241000283707 Capra Species 0.000 description 3
- 102100038784 Carbohydrate sulfotransferase 4 Human genes 0.000 description 3
- 101710158423 Carbohydrate sulfotransferase 4 Proteins 0.000 description 3
- 108010054576 Deoxyribonuclease EcoRI Proteins 0.000 description 3
- 238000012286 ELISA Assay Methods 0.000 description 3
- 241000237858 Gastropoda Species 0.000 description 3
- 108010024636 Glutathione Proteins 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 206010061218 Inflammation Diseases 0.000 description 3
- 102100037850 Interferon gamma Human genes 0.000 description 3
- 239000000020 Nitrocellulose Substances 0.000 description 3
- 241001442514 Schistosomatidae Species 0.000 description 3
- 229930006000 Sucrose Natural products 0.000 description 3
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 3
- 206010044269 Toxocariasis Diseases 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 230000007969 cellular immunity Effects 0.000 description 3
- 230000003111 delayed effect Effects 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 239000003814 drug Substances 0.000 description 3
- 210000003979 eosinophil Anatomy 0.000 description 3
- 239000013613 expression plasmid Substances 0.000 description 3
- 239000000284 extract Substances 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 230000036039 immunity Effects 0.000 description 3
- 230000003053 immunization Effects 0.000 description 3
- 238000002649 immunization Methods 0.000 description 3
- 238000003018 immunoassay Methods 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 230000004054 inflammatory process Effects 0.000 description 3
- 230000001418 larval effect Effects 0.000 description 3
- 230000004807 localization Effects 0.000 description 3
- 239000006166 lysate Substances 0.000 description 3
- 210000004962 mammalian cell Anatomy 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 239000003226 mitogen Substances 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 229920001220 nitrocellulos Polymers 0.000 description 3
- 230000001681 protective effect Effects 0.000 description 3
- 238000011084 recovery Methods 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 230000000717 retained effect Effects 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 210000004989 spleen cell Anatomy 0.000 description 3
- 230000003393 splenic effect Effects 0.000 description 3
- 239000005720 sucrose Substances 0.000 description 3
- 241000701447 unidentified baculovirus Species 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 2
- OPIFSICVWOWJMJ-AEOCFKNESA-N 5-bromo-4-chloro-3-indolyl beta-D-galactoside Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1OC1=CNC2=CC=C(Br)C(Cl)=C12 OPIFSICVWOWJMJ-AEOCFKNESA-N 0.000 description 2
- 206010014950 Eosinophilia Diseases 0.000 description 2
- 208000000624 Esophageal and Gastric Varices Diseases 0.000 description 2
- 101150066516 GST gene Proteins 0.000 description 2
- 102100030943 Glutathione S-transferase P Human genes 0.000 description 2
- 108090000288 Glycoproteins Proteins 0.000 description 2
- 102000003886 Glycoproteins Human genes 0.000 description 2
- 101001010139 Homo sapiens Glutathione S-transferase P Proteins 0.000 description 2
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 2
- XQFRJNBWHJMXHO-RRKCRQDMSA-N IDUR Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(I)=C1 XQFRJNBWHJMXHO-RRKCRQDMSA-N 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 102000008070 Interferon-gamma Human genes 0.000 description 2
- 108010044467 Isoenzymes Proteins 0.000 description 2
- 102000003960 Ligases Human genes 0.000 description 2
- 108090000364 Ligases Proteins 0.000 description 2
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 2
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 2
- 108700001237 Nucleic Acid-Based Vaccines Proteins 0.000 description 2
- 108700026244 Open Reading Frames Proteins 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 108700008625 Reporter Genes Proteins 0.000 description 2
- 229920002684 Sepharose Polymers 0.000 description 2
- 108091081024 Start codon Proteins 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- 241000244030 Toxocara canis Species 0.000 description 2
- 206010056091 Varices oesophageal Diseases 0.000 description 2
- 230000001464 adherent effect Effects 0.000 description 2
- 238000001042 affinity chromatography Methods 0.000 description 2
- 239000011543 agarose gel Substances 0.000 description 2
- 150000001413 amino acids Chemical group 0.000 description 2
- 230000005875 antibody response Effects 0.000 description 2
- 230000002238 attenuated effect Effects 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000027455 binding Effects 0.000 description 2
- 108010006025 bovine growth hormone Proteins 0.000 description 2
- 230000036755 cellular response Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 239000013599 cloning vector Substances 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 238000005520 cutting process Methods 0.000 description 2
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 2
- 231100000676 disease causative agent Toxicity 0.000 description 2
- 208000024170 esophageal varices Diseases 0.000 description 2
- 201000010120 esophageal varix Diseases 0.000 description 2
- 239000011521 glass Substances 0.000 description 2
- 208000010758 granulomatous inflammation Diseases 0.000 description 2
- 239000001963 growth medium Substances 0.000 description 2
- 244000000013 helminth Species 0.000 description 2
- 230000001744 histochemical effect Effects 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 239000012678 infectious agent Substances 0.000 description 2
- 230000001524 infective effect Effects 0.000 description 2
- 230000002757 inflammatory effect Effects 0.000 description 2
- 206010022000 influenza Diseases 0.000 description 2
- 229960003130 interferon gamma Drugs 0.000 description 2
- 101150066555 lacZ gene Proteins 0.000 description 2
- 231100000518 lethal Toxicity 0.000 description 2
- 230000001665 lethal effect Effects 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 238000003670 luciferase enzyme activity assay Methods 0.000 description 2
- 239000012139 lysis buffer Substances 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 239000002609 medium Substances 0.000 description 2
- 210000001087 myotubule Anatomy 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 230000003472 neutralizing effect Effects 0.000 description 2
- 230000009871 nonspecific binding Effects 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 230000003000 nontoxic effect Effects 0.000 description 2
- 108020004707 nucleic acids Proteins 0.000 description 2
- 102000039446 nucleic acids Human genes 0.000 description 2
- 150000007523 nucleic acids Chemical class 0.000 description 2
- 210000005259 peripheral blood Anatomy 0.000 description 2
- 239000011886 peripheral blood Substances 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 208000007232 portal hypertension Diseases 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- 239000011591 potassium Substances 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000002685 pulmonary effect Effects 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 238000001179 sorption measurement Methods 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 238000004611 spectroscopical analysis Methods 0.000 description 2
- 210000003699 striated muscle Anatomy 0.000 description 2
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 2
- 230000008961 swelling Effects 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- 230000017105 transposition Effects 0.000 description 2
- 241001529453 unidentified herpesvirus Species 0.000 description 2
- 239000013598 vector Substances 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- 210000000264 venule Anatomy 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 230000003612 virological effect Effects 0.000 description 2
- 238000012800 visualization Methods 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- RDEIXVOBVLKYNT-VQBXQJRRSA-N (2r,3r,4r,5r)-2-[(1s,2s,3r,4s,6r)-4,6-diamino-3-[(2r,3r,6s)-3-amino-6-(1-aminoethyl)oxan-2-yl]oxy-2-hydroxycyclohexyl]oxy-5-methyl-4-(methylamino)oxane-3,5-diol;(2r,3r,4r,5r)-2-[(1s,2s,3r,4s,6r)-4,6-diamino-3-[(2r,3r,6s)-3-amino-6-(aminomethyl)oxan-2-yl]o Chemical compound OS(O)(=O)=O.O1C[C@@](O)(C)[C@H](NC)[C@@H](O)[C@H]1O[C@@H]1[C@@H](O)[C@H](O[C@@H]2[C@@H](CC[C@@H](CN)O2)N)[C@@H](N)C[C@H]1N.O1C[C@@](O)(C)[C@H](NC)[C@@H](O)[C@H]1O[C@@H]1[C@@H](O)[C@H](O[C@@H]2[C@@H](CC[C@H](O2)C(C)N)N)[C@@H](N)C[C@H]1N.O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N RDEIXVOBVLKYNT-VQBXQJRRSA-N 0.000 description 1
- LINMATFDVHBYOS-MBJXGIAVSA-N (2s,3r,4s,5r,6r)-2-[(5-bromo-1h-indol-3-yl)oxy]-6-(hydroxymethyl)oxane-3,4,5-triol Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1OC1=CNC2=CC=C(Br)C=C12 LINMATFDVHBYOS-MBJXGIAVSA-N 0.000 description 1
- IQFYYKKMVGJFEH-OFKYTIFKSA-N 1-[(2r,4s,5r)-4-hydroxy-5-(tritiooxymethyl)oxolan-2-yl]-5-methylpyrimidine-2,4-dione Chemical compound C1[C@H](O)[C@@H](CO[3H])O[C@H]1N1C(=O)NC(=O)C(C)=C1 IQFYYKKMVGJFEH-OFKYTIFKSA-N 0.000 description 1
- FSVJFNAIGNNGKK-UHFFFAOYSA-N 2-[cyclohexyl(oxo)methyl]-3,6,7,11b-tetrahydro-1H-pyrazino[2,1-a]isoquinolin-4-one Chemical compound C1C(C2=CC=CC=C2CC2)N2C(=O)CN1C(=O)C1CCCCC1 FSVJFNAIGNNGKK-UHFFFAOYSA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- PCDWFBFHIIKIPM-UHFFFAOYSA-N 3-ethyl-2h-1,3-benzothiazole-2-sulfonic acid Chemical compound C1=CC=C2N(CC)C(S(O)(=O)=O)SC2=C1 PCDWFBFHIIKIPM-UHFFFAOYSA-N 0.000 description 1
- QFVHZQCOUORWEI-UHFFFAOYSA-N 4-[(4-anilino-5-sulfonaphthalen-1-yl)diazenyl]-5-hydroxynaphthalene-2,7-disulfonic acid Chemical compound C=12C(O)=CC(S(O)(=O)=O)=CC2=CC(S(O)(=O)=O)=CC=1N=NC(C1=CC=CC(=C11)S(O)(=O)=O)=CC=C1NC1=CC=CC=C1 QFVHZQCOUORWEI-UHFFFAOYSA-N 0.000 description 1
- 101150064522 60 gene Proteins 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 240000003291 Armoracia rusticana Species 0.000 description 1
- 108010077805 Bacterial Proteins Proteins 0.000 description 1
- BTBUEUYNUDRHOZ-UHFFFAOYSA-N Borate Chemical compound [O-]B([O-])[O-] BTBUEUYNUDRHOZ-UHFFFAOYSA-N 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 238000011740 C57BL/6 mouse Methods 0.000 description 1
- 210000001239 CD8-positive, alpha-beta cytotoxic T lymphocyte Anatomy 0.000 description 1
- 241000244203 Caenorhabditis elegans Species 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 108020004705 Codon Proteins 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- 238000011537 Coomassie blue staining Methods 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 241000283715 Damaliscus lunatus Species 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 108090000331 Firefly luciferases Proteins 0.000 description 1
- 208000005422 Foreign-Body reaction Diseases 0.000 description 1
- 229930182566 Gentamicin Natural products 0.000 description 1
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 description 1
- 102100023541 Glutathione S-transferase omega-1 Human genes 0.000 description 1
- 108010034145 Helminth Proteins Proteins 0.000 description 1
- 208000006968 Helminthiasis Diseases 0.000 description 1
- 208000032843 Hemorrhage Diseases 0.000 description 1
- 206010019663 Hepatic failure Diseases 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 208000001718 Immediate Hypersensitivity Diseases 0.000 description 1
- 108010002350 Interleukin-2 Proteins 0.000 description 1
- 108090000978 Interleukin-4 Proteins 0.000 description 1
- 101710172804 K protein Proteins 0.000 description 1
- 108010074338 Lymphokines Proteins 0.000 description 1
- 102000008072 Lymphokines Human genes 0.000 description 1
- 108090000542 Lymphotoxin-alpha Proteins 0.000 description 1
- 102000004083 Lymphotoxin-alpha Human genes 0.000 description 1
- 241000282567 Macaca fascicularis Species 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 102000014171 Milk Proteins Human genes 0.000 description 1
- 108010011756 Milk Proteins Proteins 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 101001044384 Mus musculus Interferon gamma Proteins 0.000 description 1
- 101000960966 Mus musculus Interleukin-5 Proteins 0.000 description 1
- 208000029549 Muscle injury Diseases 0.000 description 1
- 241000187479 Mycobacterium tuberculosis Species 0.000 description 1
- 238000011887 Necropsy Methods 0.000 description 1
- 206010067482 No adverse event Diseases 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 101710194807 Protective antigen Proteins 0.000 description 1
- 206010037391 Pulmonary granuloma Diseases 0.000 description 1
- 101000702488 Rattus norvegicus High affinity cationic amino acid transporter 1 Proteins 0.000 description 1
- 208000035415 Reinfection Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 208000002848 Schistosomiasis mansoni Diseases 0.000 description 1
- 229920005654 Sephadex Polymers 0.000 description 1
- 239000012507 Sephadex™ Substances 0.000 description 1
- 238000003639 Student–Newman–Keuls (SNK) method Methods 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 239000008049 TAE buffer Substances 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- 206010045240 Type I hypersensitivity Diseases 0.000 description 1
- 206010047504 Visceral Larva Migrans Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- HGEVZDLYZYVYHD-UHFFFAOYSA-N acetic acid;2-amino-2-(hydroxymethyl)propane-1,3-diol;2-[2-[bis(carboxymethyl)amino]ethyl-(carboxymethyl)amino]acetic acid Chemical compound CC(O)=O.OCC(N)(CO)CO.OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O HGEVZDLYZYVYHD-UHFFFAOYSA-N 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 238000000246 agarose gel electrophoresis Methods 0.000 description 1
- 229940103272 aluminum potassium sulfate Drugs 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000000347 anti-schistosomal effect Effects 0.000 description 1
- 230000007503 antigenic stimulation Effects 0.000 description 1
- 208000010216 atopic IgE responsiveness Diseases 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 238000013320 baculovirus expression vector system Methods 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- SIEYLFHKZGLBNX-UHFFFAOYSA-N bupivacaine hydrochloride (anhydrous) Chemical compound [Cl-].CCCC[NH+]1CCCCC1C(=O)NC1=C(C)C=CC=C1C SIEYLFHKZGLBNX-UHFFFAOYSA-N 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 210000004323 caveolae Anatomy 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 239000003593 chromogenic compound Substances 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- 239000007979 citrate buffer Substances 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 239000012141 concentrate Substances 0.000 description 1
- 210000002808 connective tissue Anatomy 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- NKLPQNGYXWVELD-UHFFFAOYSA-M coomassie brilliant blue Chemical compound [Na+].C1=CC(OCC)=CC=C1NC1=CC=C(C(=C2C=CC(C=C2)=[N+](CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=2C=CC(=CC=2)N(CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=C1 NKLPQNGYXWVELD-UHFFFAOYSA-M 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- ATDGTVJJHBUTRL-UHFFFAOYSA-N cyanogen bromide Chemical compound BrC#N ATDGTVJJHBUTRL-UHFFFAOYSA-N 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 239000007857 degradation product Substances 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 230000008021 deposition Effects 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-J diphosphate(4-) Chemical compound [O-]P([O-])(=O)OP([O-])([O-])=O XPPKVPWEQAFLFU-UHFFFAOYSA-J 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 229910000397 disodium phosphate Inorganic materials 0.000 description 1
- 235000019800 disodium phosphate Nutrition 0.000 description 1
- 238000002224 dissection Methods 0.000 description 1
- 210000000883 ear external Anatomy 0.000 description 1
- 239000012039 electrophile Substances 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 238000012869 ethanol precipitation Methods 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 230000035558 fertility Effects 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000000706 filtrate Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000013505 freshwater Substances 0.000 description 1
- 229960002518 gentamicin Drugs 0.000 description 1
- 239000003365 glass fiber Substances 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 210000002443 helper t lymphocyte Anatomy 0.000 description 1
- 206010019847 hepatosplenomegaly Diseases 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 238000010562 histological examination Methods 0.000 description 1
- 239000000710 homodimer Substances 0.000 description 1
- 230000008348 humoral response Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 230000002134 immunopathologic effect Effects 0.000 description 1
- 230000007365 immunoregulation Effects 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 229940100602 interleukin-5 Drugs 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 230000000622 irritating effect Effects 0.000 description 1
- BPHPUYQFMNQIOC-NXRLNHOXSA-N isopropyl beta-D-thiogalactopyranoside Chemical compound CC(C)S[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O BPHPUYQFMNQIOC-NXRLNHOXSA-N 0.000 description 1
- 229930027917 kanamycin Natural products 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- SBUJHOSQTJFQJX-NOAMYHISSA-N kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 1
- 229930182823 kanamycin A Natural products 0.000 description 1
- 230000021633 leukocyte mediated immunity Effects 0.000 description 1
- 208000007903 liver failure Diseases 0.000 description 1
- 231100000835 liver failure Toxicity 0.000 description 1
- 229940106885 marcaine Drugs 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 108010077055 methylated bovine serum albumin Proteins 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 235000021239 milk protein Nutrition 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 230000011278 mitosis Effects 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 238000000491 multivariate analysis Methods 0.000 description 1
- 210000000663 muscle cell Anatomy 0.000 description 1
- 231100000302 myotoxic Toxicity 0.000 description 1
- 230000003630 myotoxic effect Effects 0.000 description 1
- 230000001338 necrotic effect Effects 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000001473 noxious effect Effects 0.000 description 1
- 239000012038 nucleophile Substances 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 229960000462 oxamniquine Drugs 0.000 description 1
- XCGYUJZMCCFSRP-UHFFFAOYSA-N oxamniquine Chemical compound OCC1=C([N+]([O-])=O)C=C2NC(CNC(C)C)CCC2=C1 XCGYUJZMCCFSRP-UHFFFAOYSA-N 0.000 description 1
- 208000014837 parasitic helminthiasis infectious disease Diseases 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 230000009120 phenotypic response Effects 0.000 description 1
- 230000003114 pinocytic effect Effects 0.000 description 1
- 238000013492 plasmid preparation Methods 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 210000003240 portal vein Anatomy 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- GRLPQNLYRHEGIJ-UHFFFAOYSA-J potassium aluminium sulfate Chemical compound [Al+3].[K+].[O-]S([O-])(=O)=O.[O-]S([O-])(=O)=O GRLPQNLYRHEGIJ-UHFFFAOYSA-J 0.000 description 1
- 229960002957 praziquantel Drugs 0.000 description 1
- 238000002810 primary assay Methods 0.000 description 1
- 230000001902 propagating effect Effects 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 238000010791 quenching Methods 0.000 description 1
- 230000000171 quenching effect Effects 0.000 description 1
- 238000010992 reflux Methods 0.000 description 1
- 239000012465 retentate Substances 0.000 description 1
- 238000011808 rodent model Methods 0.000 description 1
- 210000000518 sarcolemma Anatomy 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 239000013605 shuttle vector Substances 0.000 description 1
- 210000001057 smooth muscle myoblast Anatomy 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 238000007614 solvation Methods 0.000 description 1
- 238000002943 spectrophotometric absorbance Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 239000000021 stimulant Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 230000008719 thickening Effects 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 108700012359 toxins Proteins 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000005030 transcription termination Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 1
- 229960001005 tuberculin Drugs 0.000 description 1
- 238000005199 ultracentrifugation Methods 0.000 description 1
- 241000712461 unidentified influenza virus Species 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 238000012795 verification Methods 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/10—Transferases (2.)
- C12N9/1085—Transferases (2.) transferring alkyl or aryl groups other than methyl groups (2.5)
- C12N9/1088—Glutathione transferase (2.5.1.18)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2799/00—Uses of viruses
- C12N2799/02—Uses of viruses as vector
- C12N2799/021—Uses of viruses as vector for the expression of a heterologous nucleic acid
- C12N2799/026—Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- the present invention relates generally to the fields of parasitology and molecular biology. More specifically, the present invention relates to a naked DNA vaccine for parasitic diseases.
- Schistosomiasis is a disease that affects over 250 million people worldwide, primarily in underdeveloped countries and approximately one percent of those who contract schistosomiasis will die annually from hemorrhage and/or liver failure.
- the causative agent is the human blood fluke, Schistosoma manson i which lives in the inferior mesenteric venules. In these venules, the male and female worms live, mate, and produce large numbers of eggs each day which ultimately accumulate in the liver and elicit a characteristic granulomatous inflammation.
- the ensuing disease is characterized by hepatosplenomegaly, portal hypertension and esophageal varices, the latter often being fatal.
- praziquantel ® and oxamniquine® are used to treat schistosomiasis but reinfection occurs with return to fresh water (where the snail intermediate hosts of S. mansoni live) and there is evidence accumulating that the worms are becoming resistant to these agents .
- lymphocyte proliferation assay i.e., the lymphocyte transformation assay or LTA
- PHA and Con A T cell mitogens
- Schistosoma mansoni egg antigens 21 -24.
- DNA when taken up by skeletal muscle, remains circular and neither integrates into chromosomal DNA nor replicates (5, 6).
- Naked DNA techniques 1 1
- intramuscular injection of plasmid DNA in particular have been used to immunize (i.e., vaccinate) experimental animals, and direct injection has been shown to elicit both cellular and humoral immune responses ( 1, 8- 10).
- Mice have been successfully immunized by intramuscular injection of plasmid DNA carrying an influenza gene as determined by their ability to survive a lethal challenge infection (9, 10).
- Using the same polynucleotide vaccine in mice revealed that 1 ) a single injection elicited both cellular and humoral responses that persisted for 1 year and 2) the cellular response was associated with the CD8+ subpopulation of T cells (i.e., killer T cells) (12).
- mice were given plasmid DNA injections by one of three routes (iv, ip, or im) and subjected to an influenza virus- challenge.
- gene vaccination resulted in 60% protection which was attributed to use of a gene from a heterologous virus (i.e., a virus related to the influenza virus that normally infects chickens but differing by 16% in amino acid sequence).
- Mice which have been vaccinated with plasmid DNA encoding a bovine herpes glycoprotein have gone on to make significant amounts of antiglycoprotein antibody and the sera from these mice contained significant amounts of viral neutralizing activity as compared to mice receiving the plasmid platform lacking the herpes gene (28).
- the prior art is deficient in the lack of effective means of treating Schistosomiasis using naked DNA vaccination technology.
- the present invention fulfills this longstanding need and desire in the art.
- the present invention discloses several eukaryotic expression plasmids containing genes derived from S. mansoni that serve as the basis of a polynucleotide vaccine.
- no adjuvants are required, no infectious agents (either killed or attenuated) are introduced into the patient during the immunization protocol, and no exogenous contaminants of any kind are introduced which could elicit spurious immune responses.
- gene products produced by skeletal muscle are presented to the immune system in such a way as to elicit cell-mediated immunity which makes this novel technique ideally suited for development into a human vaccine.
- the present invention uses naked DNA vaccine technology to establish the feasibility of preventing or ameliorating the effects of infection with the human intravascular blood fluke, Schistosoma mansoni.
- the present invention characterizes both cellular and humoral immune responses of mice vaccinated with
- Experimental development of naked DNA vaccination technology is important, not only for the control of the parasite causing schistosomiasis in 250 million people, but for establishing a new way to vaccinate against almost any infectious helminth in manner that does not require live or attenuated organisms or the addition to the vaccine of noxious chemicals in the form of adjuvants such as peanut oil or alum.
- the present invention determines ( 1 ) the effect of time and dose on the development of the cellular immune response of mice vaccinated with naked DNA encoding Schistosoma mansoni glutathione S-transferase; (2) the effect of time and dose on the development of the humoral immune response of mice vaccinated with naked DNA encoding Schistosoma mansoni glutathione S-transferase; (3) the effect of the H-2 haplotype on the cellular and humoral immune responses of mice vaccinated with naked DNA encoding Schistosoma mansoni glutathione S-transferase.
- the present invention uses selected strains of inbred mice which are known permissive hosts for the entire life cycle of S. mansoni.
- lymphocyte transformation assays in response to parasite glutathione S-transferase and granuloma formation around glutathione S-transferase-coated beads are monitored while humoral immune responses (i.e. , antibody production) are evaluated using a glutathione S-transferase specific enzyme-linked immunosorbent assay (ELISA).
- ELISA glutathione S-transferase specific enzyme-linked immunosorbent assay
- composition of matter comprising a naked DNA vaccine for the parasitic worm, Schistosoma mansoni or human blood fluke.
- novel plasmids and expression vectors for use in the methods of the present invention.
- a method of protecting against Sch is tos o m a manson i comprising administering to an animal in need of such treatment a pharmacologically effective dose of a naked DNA vaccine.
- Figure 1 shows a schematic drawing of the deletion of CMV promoter and replacement with ⁇ S A pro m oter .
- Inset miniprep of pRc ⁇ SA cut with (S)ma I and (P)vu II.
- Figure 2 shows the histochemical localization of E. coli ⁇ gal activity in mouse skeletal muscle 3 weeks after injection of 100 ⁇ l saline of b) saline with 50 ⁇ g pRSV-LacZ DNA. Orig. 400x.
- FIG. 4 shows that SmGST expression by HEK 293 cells 48 hours after CaPO4 transfection.
- (-) no DNA.
- CDNB; chloro-dinitrobenzene used as substrate; n number of dishes assayed.
- Figure 5 shows the splenic mass 8 weeks after no injection, intramuscular injection of 10 ⁇ g pSmGST3, 50 ⁇ g pSmGST3, or a subcutaneous injection of 25 ⁇ g of pure SmGST mixed with alum .
- pSmGST3 and pCMV-SmGST3 are used interchangeably and are of equal rank in identification.
- 130%- line significance.
- n number of spleens.
- Figure 6 shows an electrophoretogram showing purification of a 28 kDa protein encoding the S . manson i glutathione S-transferase.
- Figure 6A shows bacteria transduced with pCMV-GST3.
- Figure 6B shows control bacteria transduced with pCMV-Lux encoding an irrelevant protein.
- Lane 1 molecular weight markers X 1000;
- Lane 2 bacerial lysate (starting material) which was applied to a glutathione agarose affinity column;
- Lane 3 last wash prior to eluting the column;
- Lane 4 shows the concentrate of eluate from glutathione-agarose affinity column.
- Figure 7A and 7B show the nucleotide sequence of plasmids pRc/ASK8-SmGST3 and pCMV-SmGST3.
- Figure 8 shows the SDS-PAGE ( Figure 8A) and Western Blot (Figure 8B) of uninfected or Bac-SmGST-3-infected Sf21 cells.
- Figure 8B The gel in Figure 8A was electroblotted to a nitrocellulose membrane and immunostained with polyclonal rabbit anti-S.
- Figure 9 shows a SDS-PAGE of various stages during the isolation procedure of recombinant SmGST-3 protein from Bac-
- Figure 10 shows a Western Blot confirming the immunoreactive property of the 28 kDa protein band following concentration from pooled fractions 4 through 12 depicted in
- the present invention provides a vaccine comprising a non-infectiou s , non-inte grating DNA sequence encoding Schistosoma mansoni glutathione S-transferase.
- the DNA comprises a plasmid encoding a protein, polypeptide or peptide which is operably linked to a promoter.
- the plasmid is pCMVSmGST-3.
- the plasmid is selected from the group consisting of pBsCMV- 16.4 DNA and pRc/ASK8-SmGST3.
- the present invention is also directed to a method of protecting against infection by Schistosoma mansoni comprising the step of: administering to an animal in need of such treatment a pharmacologically effective dose of a vaccine composition comprising a non-infectious, non-integrating DNA sequence encoding Sch isto soma manson i glutathione S -transferase .
- the vaccine composition comprises a plasmid encoding a protein, polypeptide or peptide which is operably linked to a promoter.
- Representative examples of plasmids useful in this method of the present invention include pCMVSmGST-3, pBsCMV- 16.4 DNA and pRc/ASK8-SmGST3.
- the plasmid is administered in an animal in a dose of from about 25 ⁇ g to about 200 ⁇ g.
- the plasmid is administered into muscle although it is possible it could be adminstered into another site in the body.
- the plasmid would be administered between 1 and 6 times.
- the present invention is also directed to a method of developing cell mediated immunity to glutathione-S-transferase of Schistosoma mansoni comprising the step of: administering to an animal in need of such treatment a immunologically effective dose of a plasmid of the present invention.
- Representative examples of plasmids useful in this method of the present invention include pCMVSmGST-3 , pBsCMV- 16.4 DNA and pRc/ASK8-SmGST3.
- plasmid is administered in an animal in a dose of from about 25 ⁇ g to about 200 ⁇ g.
- the plasmid is administered into muscle although it is possible it could be adminstered into another site in the body.
- the plasmid would be administered between 1 and 6 times.
- a technique for injecting naked DNA directly into skeletal muscle using a reporter plasmid in which the lacZ gene was inserted 3' to an RSV promoter in a eukaryotic expression plasmids engineered was used.
- muscles were removed, frozen sections prepared, the sections stained for ⁇ -gal activity and the histochemical localization was compared to control muscles injected with saline.
- All lacZ DNA-injected muscles stained intensely indicating the presence of the plasmid and expression of the bacterial gene by the mouse muscle cells while all controls were completely devoid of reaction product (See Figure 1 ).
- the cDNA encoding the SmGST-3 isoenzyme was obtained.
- the cDNA was excised from a pGEX plasmid and ligated into a eukaryotic expression plasmids 3' to an immediate early CMV promoter.
- HEK 293 cells in tissue culture were transfected by the CaPO4 method. Plates at the time of transfection were approximately 50% of confluency and after transfection were allowed to incubate at 37°C/5% CO2 in air for another 48 hr. At this time the cells were confluent which indicates that the cells tolerated the foreign DNA well.
- Soluble extracts of the monolayers were shown (Figure 2) to overexpress GST activity when using chlorodinitrobenzene (CDNB) as the electrophilic substrate in the assay described by Habig and Jakoby (20) as compared to cells transfected with an irrelevant plasmid (which was also determined to be expressed, but is not shown in Fig. 2).
- CDNB chlorodinitrobenzene
- BALB/c mice have been vaccinated with various quantities of either pCMVSmGST-3 or an irrelevant plasmid (pRSV- Lux) and after two weeks the mice were infected with 200 cercariae of S. mansoni.
- FIG. 3 based on splenic mass increase (which correlates with lymphocyte proliferation in the white pulp) illustrates that mice receiving the pCMVSmGST-3 DNA are affected by the treatment.
- Glutathione S-transferases (EC 2.5.1.18) catalyze the conjugation of both endogenous and exogenous electrophiles with the major cellular nucleophile, reduced glutathione (GSH) (29).
- GSTs are found widely distributed in nature in both invertebrates such as Schistosoma mansoni and vertebrates such as mice, rats and humans ( 18) . Most organisms contain multiple GST isoenzymes that are characterized by distinct but often overlapping substrate specificities ( 18).
- Adult S. Glutathione S-transferases
- mansoni contain at least five forms of GSH S-transferase, three of which (designated SmGST- 1 , SmGST-2 and SmGST-3) account for 2-4% of the soluble ⁇ protein in the male worm (19, 20).
- GSH S-transferases from vertebrates that have been characterized are dimeric proteins composed of either identical (homodimer) or nonidentical (heterodimer) subunits ( 16, 17).
- the polypeptide subunits are catalytically independent and the quaternary structure is thought to be necessary for enzyme stability ( 17).
- three cytosolic forms of S. mansoni G S T were purified and found to have very similar catalytic, physicochemical, and immunological properties. However, the most striking feature is that they are catalytically active monomers rather than dimers ( 19).
- the parasite GST is a 28 kDa protein in contrast to vertebrate GSTs which range from 44 to 50 kDa and b) that the parasite proteins do not immunologically cross react with vertebrate (i.e. host) GSTs which makes the present invention herein feasible because immune responses against parasite GSTs will not immunoneutralize the hosts GSTs.
- This protein was found to have a Mr of 28 KDa and cDNA encoding the 28 K protein was subsequently cloned in E. coli and the recombinant protein used to immunize rats and BALB/c mice. Good immunoprotection against 5. mansoni in several host species (i.e., approximately 50% in the mice) has been demonstrated using Sm28 and either Freund's Complete or an alum adjuvant (31).
- This plasmid known as pRc/CMV was obtained from the Invitrogen Corporation and contains the immediate early enhancer-promotor sequence from cytomegalovirus (CMV) , polyadenylation signal and transcription termination sequences from the bovine growth hormone (BGH) gene and SV40 origin.
- CMV cytomegalovirus
- BGH bovine growth hormone
- This plasmid and its host species of permissive E. coli, TOP10F', were used for all subsequent experiments.
- the CMV promoter was first removed from the pRc/CMV plasmid and replaced with the chicken ⁇ -skeletal actin promoter sequence. This is shown schematically in Figure 1 and was performed as follows.
- the CMV promoter sequence is flanked 5' by an Nru I site and 3' by a Hind III site just inside the polycloning sequence.
- a Hind III restriction enzyme (RE) cut was performed on purified pRc/CMV DNA followed by a Klenow-mediated blunt end fill-in of the cut site.
- the linearized, cut DNA was ethanol precipitated and then cut with Nru I. Because Nru I is a blunt cutter, the pRc backbone was now ready for blunt end ligation.
- a 6.2 Kb p ⁇ SK fragment of genomic DNA was obtained carrying the chicken a -skeletal actin ( ⁇ SA) gene and its entire promoter (nb: skeletal actin promoters are highly conserved and have been shown to function in mammalian cells).
- the 675 bp promoter sequence as shown in Figure 1 was contained between an Eco RV and Nae I site.
- a sequential RE digest was performed and the 675 bp ⁇ SA fragment isolated from a 1 % agarose gel.
- the 5 Kb pRc plasmid and the 675 bp ⁇ SA fragment were ligated using T4 ligase. The ligation products were then transfected into TOP10F' and 22 colonies were screened for plasmid DNA.
- plasmids were cut with Sma I which cuts asymmetrically one time within the ⁇ SA sequence and once within the remaining pRc sequence. If the ⁇ SA sequence is in the correct 5'— >3' orientation with respect to the polycloning sites then two bands of 1800 bp and 4000 bp should be seen upon agarose gel electrophoresis. One such clone designated pRc ⁇ SA ⁇ was found (shown in the insert of Figure 1 ) and this orientation was confirmed by cutting with a different RE, Pvu II, which gives fragments of 81 1 , 1065 , 1 196, and 2400 bp.
- the plasmid DNA was isolated from a liter of bacteria. Glycerol stocks of TOPI OF' containing pRc ⁇ SA8 (pRcASK8 is of equal rank and used interchangeably) were prepared and stored at -80°C. A control plasmid for expression under the control of the ⁇ SA promoter sequence with the promoter sequence inserted in the 3'-->5' orientation was also stocked (pRc ⁇ SA 14) . The next stage was to insert the gene of interest into the polycloning site.
- the SmGST3 fragment was removed from the pCR3-SmGST3 TA cloning vector using Xba I and ligated into pRcASK ⁇ , which had also been cut with Xba I, using T4 ligase. The ligation products were then transformed into TOP10F' cells.
- the new construct, designated pRcASK8-SmGST3 was stocked in a glycerol solution and stored at -80°C.
- a eukaryotic reporter plasmid in which the E. coli ⁇ -galactosidase ( ⁇ - gal) gene is inserted into the polycloning site can be used to verify both properties.
- ⁇ - gal E. coli ⁇ -galactosidase
- mice were sacrificed after 1 , 2, or 3 weeks and the injected muscles removed, snap frozen in methyl butanol on dry ice, and 10 ⁇ m frozen sections prepared. The sections were stained with X-gal in the presence of potassium ferriferrocyanide. All muscles injected with the pBsRSV- ⁇ Gal plasmid had scattered myofibers which stained intensely blue- black indicating that the ⁇ -Gal gene product, ⁇ -galactosidase, was being expressed. None of the saline-injected muscles exhibited positive staining ( Figure 2).
- the cDNA was liberated with Eco RI and using a QIAEX gel extraction kit, the 782 bp fragment was isolated and ligated into an Eco RI site within the polycloning sequence of pB sCMV , a gene transfer vector engineered for eukaryotic gene expression.
- Orientation was determined by an asymmetrical cut with Mun I which gave two fragments of predicted sizes ( 818 and 4300 bp) while inverse orientation yielded expected fragments of 1538 and 3580 bp. As seen in Figure 3, five colonies contained inserts that were in the correct orientation and three colonies contained inversions. This plasmid was electroporated into electrocompetent DHlOb cells. Based on miniprep analysis of resultant overnight colonies, a liter of bacteria were grown overnight, and expression grade plasmid DNA was isolated by double alkaline lysis followed by double CsCl gradient ultracentrifugation.
- the resulting DNA was dialyzed against 4 liters of I X TAE buffer, ethanol precipitated and dissolved in 1.0 ml sterile, deionized, distilled H2O. Final DNA and RNA concentrations were determined spectrophotometrically at OD260/280. Two clones, designated 3.7 and 3.9, were prepared in parallel and the final DNA yields were 3.0 and 3.1 mg DNA per ml, respectively .
- TOP10F' cells carrying the pCMVGST3 plasmid were grown overnight and then collected by centrifugation. The cells were lysed and the soluble fraction collected and applied to a GSH- agarose affinity column to purify the overexpressed protein, SmGST3.
- a parallel culture of pCMV-Lux expressing the irrelevant protein, luciferase was used as a control. Affinity purified SmGST3 was eluted from the column with 10 mM glutathione. The recovery was monitored by coomassie blue staining of 12% polyacrylamide gel electrophoresis and examination of the 28 kDa band ( Figure 6).
- HEK 293 cells in tissue culture were transfected by the CaP ⁇ 4 method. Plates at the time of transfection were approximately 50% of confluency and after transfection were allowed to incubate at 37°C in 5% C02 in air for another 48 hours. At this time the cells were confluent which indicated that the cells tolerated the foreign DNA well.
- CDNB chloro-dinitrobenzene
- PCR primers were designed to add an Xba I restriction site to both the 5' and 3' ends of the fragment and to add a Kozak consensus sequence that would enhance the binding of the polymerase to the promoter sequence) to the 5' end.
- the PCR fragment was cloned directly from the thermal cycler into Invitrogen's TA cloning vector, pCR3. This vector is actually designed to serve as a eukaryotic expression vector and the cloned fragment is under the control of the immediate early CMV promoter.
- This construct was electroporated into DH lOb cells and yielded an 85% cloning efficiency. Tubes of stock bacteria carrying this construct (designated pCR3-SmGST3) were stored at -80°C.
- mice were injected intramuscularly (im) (vaccinated) with 10 or 50 ⁇ g of either pCMVGST-3 or an irrelevant plasmid (pRSV-Lux, containing the luciferase reporter gene). After two weeks the mice were challenge-infected with 200 cercariae of S. mansoni. An additional group was vaccinated in the conventional way with 25 ⁇ g of SmGST3 isolated from adult S . mansoni adsorbed on alum adjuvant as described by Mishell and Shiigi (Mishell, BB, et al.. Selected Methods in Cellular Immunology, W.H. Freeman, San Francisco, pp. 1 -486, ( 1980)).
- FIG. 5 demonstrates that mice receiving the pCMVSmGST-3 DNA responded to the treatment.
- the mice were sacrificed 6 weeks after infection and 8 weeks after vaccination.
- the portal vein was retrogradely perfused and the adult worms collected.
- the livers were fixed in formalin for histological analysis and the muscle that was injected with DNA was excised and placed on dry ice for luciferase assay. Sera were collected from all mice prior to vaccination, immediately prior to infection and at sacrifice.
- mice vaccinated with 10 ⁇ g of pCMV-GST3.7 DNA had 89% fewer eggs than mice receiving any of the other treatment regimens. This has been suggested as being the manner in which
- mice vaccinated with DNA or isolated GST protein Eggs per unit area of mice vaccinated with DNA or isolated GST protein
- Control 6 60.0 pRSV-Lux 5 69.7 (- 16%) p pBBssCCMMVVSSmmGGSSTT33 44 66..77 89%
- the lymphocyte proliferation assay (26, 35) used is as follows. Briefly, spleens removed asceptically are reduced to single cell suspensions in cold RPMI 1640 medium. The suspensions are filtered through 2 layers of sterile gauze and the passing debris allowed to settle for 10 minutes. The resulting suspension is decanted and centrifuged at 900 X g for 10 minutes at 4°C and resuspended in fresh RPMI 1640. The cell concentration is determined by electronic counting (Coulter Counter) and adjusted to 10? cells/ml.
- the cells are cultured at a density of 2 X 10 ⁇ cells/0.175 ml of RPMI 1640 containing 5% normal human, heat- inactivated serum and 20 ⁇ g of gentamicin sulfate.
- Unstimulated control cultures receive an additional 25 ⁇ l of RPMI 1640 and stimulated cultures 25 ⁇ l of RPMI 1640 containing either 20 ⁇ g/ml Con A (2.5 ⁇ g/200 ⁇ l culture); or 24 ⁇ g GSH-agarose affinity purified 5.
- mansoni GST about 70% of which is GST3 (3 ⁇ g/200 ⁇ l culture).
- the supernatant is removed by aspiration and the bead pellet resuspended in 1 ml of the purified GST protein in borate buffer (pH 8.3) at 1 -2 mg/ml.
- the protein-gel suspension is mixed overnight at 4°C on an end-over-end mixer.
- the gel suspension is again centrifuged as above and the pellet resuspended in 3 ml of 1 M ethanolamine, pH 8.0 at room temperature to block unbound sites on the beads.
- the beads are washed several times with sterile phosphate buffered saline (sPBS), pH 7.6, counted and adjusted to 10,000/ml.
- sPBS sterile phosphate buffered saline
- the ELISA procedure used is essentially as described (26) with modifications for use in detecting anti-SmGST antibodies. Briefly, the affinity purified SmGST is diluted in 0.05 M carbonate buffer at 3-5 ⁇ g/ml and 100 ⁇ l of the solution is added to each well and incubated at room temperature 1 -2 hours and then 4°C overnight. The plates are then rinsed 3 times with PBS and then nonspecific binding sites blocked with 2% bovine serum albumin diluted in PBS for 30 minutes at room temperature. The blocking solution is rinsed away by 3 washes of PBS containing 0.05% Tween 20. The plates can either be dried and stored or used as follows.
- Each mouse serum to be tested is diluted (see below) in PBS/Tween 20 and 100 ⁇ l of this dilution added to the precoated wells (each dilution done in duplicate) and the plate incubated at room temperature 1 hr. The plate is then washed 4 times with PBS/Tween 20 and 100 ⁇ l of horseradish peroxidase-labelled goat anti-mouse immunoglobulin (Heavy and light chains combined) diluted 1 : 100 in PBS/Tween 20 added to each well and incubated for 1 hour. The plate is then washed 5 times in PBS/Tween 20 and once in PBS.
- Each wel l receives 100 ⁇ l of the substrate solution, 0.4 mM 2,2'-azino-di(3 ethylbenzthiazoline sulfonic acid) and 2 mM H 2O 2 diluted to a final molarity in 0.05 M citrate buffer (pH 4.0). After optimal color development occurs ( 15-30 min. in the dark at room temperature) the reaction is stopped by adding 50 ⁇ l of 1.0 N
- Spectrophotometric absorbance is determined in individual wells with an enzyme immunoassay (BioTek 308) plate reader at 405 nM with a 490 nM reference.
- a standard curve is established by coating wells with 0.1 , 0.5, 1.0, 2.5, 5.0, and 10.0 ⁇ g of affinity-purified SmGST per well. The greatest dilution of a reference polyclonal rabbit anti-SmGST serum giving a linear recognition of the standard curve is used to determine what dilution of the test sera is examined in the ELISA assay.
- Plasmids were prepared by standard methods that included double alkaline lysis of the bacteria propagating the plasmids, two purifications on CsCl gradients, dialysis against 1 mM Tris-HCl (pH 7.4) and 0. 1 mM EDTA, ethanol precipitation, and solvation into distilled, deionized autoclaved water as described (4- 6). Approximately 5 mg of pCMV-SmGST3 was prepared. However, should the quantity of plasmid DNA need to be replenished, the bacteria containing this plasmid is in house in a frozen stock. In addition, the pRSV-LacZ reporter gene plasmid may be used.
- mice Each naked DNA injection has added to it 10 ⁇ g of pRSV-LacZ DNA prepared as above.
- This control serves two purposes. First, it provides knowledge that the DNA solution for vaccination was delivered to the muscle and secondly, it indicates that foreign genes were being expressed by the muscle in the case that no immunological response to SmGST3 is observed. Additionally, a monoclonal antibody, that specifically recognizes E. coli ⁇ -gal can be used to perform an ELISA in parallel with the GST ELISA to monitor host response to the transferred genes.
- mice are anesthetized with Metaphane until they are nonresponsive to irritative stimuli (whisker tug and paw pulling). While maintaining this level of anesthesia, a small incision through the skin perpendicular to the long axis of the quadriceps of the leg are made to expose the length of the muscle.
- a 100 ⁇ l injection of sterile saline containi ng the desired mass of expression-grade plasmid DNA is injected into the body of the quadriceps muscle using a 1 ml tuberculin syringe and a 27 g x 0.5 in needle. The needle is withdrawn slowly so as to prevent reflux, and the incision closed with 1 -2 sterile wound clips. The mice are allowed to regain consciousness and after 1 hour are returned to the vivarium. The mice are checked daily. This procedure has been performed on 40 mice with no adverse reactions (i.e., all lived, no infections, all expressed the transferred gene).
- T cell-mediated immunity Two measures of T cell-mediated immunity (CMI) are used.
- An in vitro correlate of T cell clonal expansion namely the lymphocyte proliferation assay in response to specific antigen with dividing cells allowed to incorporate ⁇ pr.j jR i n t 0 newly synthesized DNA, is the primary assay.
- mice 15 for each time period are established as follows: Negative Control Groups -- 1 ) untreated; 2) irrelevant pCMV (a plasmid with no gene inserted in the downstream polycloning site and therefore not expressing any gene product); Positive Control group — to make sure that there is one positive T cell response, this group is vaccinated with pRSV-LacZ (a plasmid encoding the E. coli ⁇ -gal actosidase enzyme) and the T cells stimulated with the commercially available enzyme.
- pRSV-LacZ a plasmid encoding the E. coli ⁇ -gal actosidase enzyme
- mice Three groups of mice are set up as follows: one group each is vaccinated with 10 ⁇ g, 50 ⁇ g or 100 ⁇ g of pCMV-SmGST3. Five mice from each treatment group are sacrificed on days 7, 14 and 28 after vaccination and spleen cells are removed and used in the lymphocyte proliferation assay . Lymphocytes from each animal's spleen are tested separately in a microwell assay requiring only 2.5 X 10 ⁇ cells (average spleen contains 5-7 X 10 ⁇ cells). Spleen cells are exposed to either concanavalin A (a known T cell mitogen), E. coli ⁇ -galactosidase, SmGST3, or maintained in complete medium only.
- concanavalin A a known T cell mitogen
- E. coli ⁇ -galactosidase SmGST3
- the cells are labelled with 0.5 ⁇ Ci of ⁇ H-TdR (specific activity: 2 Ci/mmol) and the cells harvested 8 hours later onto glass filter strips using an automated cell harvester. The radioactivity on individual filters are measured by liquid scintillation spectroscopy. All treatment groups are setup and tested in triplicate.
- lymphocyte proliferation assay To verify that the response seen in the lymphocyte proliferation assay is T cell-dependent, one aliquot of cells from each mouse is treated with anti-Thy 1 and complement (to destroy T cells) (35) and the remaining cells exposed to the stimulants. This treatment should abolish isotope incorporation if T cells mediate the response.
- I I significant lymphocyte proliferation is found following vaccination with DNA encoding SmGST3, the media from the proliferation cultures is assayed using Quantikine kits from Genzyme Corporation (Cambridge, MA) specific for mouse IFN- ⁇ and IL-5 to ascertain whether vaccination elicited a Thl or
- Th2 phenotypic response
- the second measure of CMI used is granuloma formation around beads coated with specific antigen. This is an in vivo correlate of the histopathological response that occurs in the liver of infected individuals in response to deposition of eggs of the parasite. Groups of mice are set up in an identical fashion as described above for the lymphocyte proliferation assay but they will not be the same mice. At 7, 14 and 28 days after naked DNA vaccination, 5 ,000 Sepharose 4B beads with covalently attached SmGST3 are injected into the lateral tail vein of mice. These beads embolize in the microvasculature of the lungs and if the animal has T cells which recognize the SmGST3 on the bead's surface, granulomatous inflammation will ensue.
- mice On days 13, 20, and 34 post vaccination (1 e., 6 days after bead embo zation) the mice are sacrificed, the lungs perfused with 10% buffered neutral formalin, and histological sections stained with hematoxy n and eosin prepared. Twenty five granulomas in each lung are measured in 2 perpendiculai p l ane s with a filar micrometer and an approximation oi the cross sectional area calculated by multiplying the two measures together (as an indication of the degree of floridness of the reaction).
- the reaction consists of several hundred cells and the predominant types are macrophages, lymphocytes and eosinophils.
- Granulomas are being measured in the lung because the granulomatous response is a systemic manifestation oi cell-mediated immunity and the reactions occuring in the lung around beads are more circumscribed and therefore easiei to measure and analyze than in liver.
- This experimental pai adigm has been used in studies of T cell immunopathological mechanisms in mu ⁇ ne toxocariasis (24, 25).
- T cell-mediated immunity has been implicated in two major aspects of the disease process.
- the best resistance against S. mansoni has come from the use of irradiated cercana but such material is too inflammatory to use in humans.
- dissection of the immune responses elicited by vaccination of mice with irradiated cercana indicates that the protective response is dependent on T and B lymphocyte function, but independent of complement, IgM or IgE-mediated immediate hypersensitivity (36).
- Mice vaccinated one time with irradiated cercaria require the presence of CD4+ T cells and an antibody and macrophages.
- the macrophages are activated by the lymphokine interferon- ⁇ (IFN- ⁇ ) which is produced by CD4+ T helper cells of the 1 st subset type ( designated Th l cells, which also secrete IL-2 and lymphotoxin).
- IFN- ⁇ lymphokine interferon- ⁇
- Th2 subset which produce IL-4 (mediating IgE production), IL-5 (mediating eosinophilia) and IL- 10 (which down regulates Th l cells) does not decrease resistance induced by irradiated cercaria.
- T cells from mice immunized one time with irradiated cercaria proliferated and produ c ed I F N - ⁇ upon subsequent chal lenge with S . ma n son i antigens during in vitro culture (37) and this finding forms a basis for the use of the lymphocyte proliferation assay to detect a response to naked DNA vaccination using a plasmid encoding S. mansoni GST3.
- SmGST3 been shown to be a protective antigen when the recombinant protein mixed with adjuvant was used to immunize mice, rats and primates against challenge infection (38). If T cell proliferation is seen, are the Thl vs Th2 cytokine assays used.
- immune interferon gamma IFN- ⁇
- IL-5 interleukin 5
- Granuloma formation has been found to be a T cell- mediated process (39) and, until recently, considered to be a Thl phenomenon. This conclusion was based in part on the observation that an intradermal injection of S. mansoni egg antigens into the footpad of infected mice elicited a delayed swelling and concomitant production of IFN- ⁇ . However, administration of anti- IFN- monoclonal antibody (mAb) did not have any effect on the size or cellular composition of egg granulomas in the liver while a comparable treatment with mAb against IL-5 abrogated bone marrow, peripheral blood and granuloma eosinophils in infected mice (33).
- mAb monoclonal antibody
- ELISA grade microwell plates are coated with purified S. mansoni glutathione S-transferase and used in an ELISA assay. Briefly, the ELISA assay works as follows: Microwell plates are coated with specific antigen (SmGST3) and nonspecific binding sites blocked with excess milk protein. Sera to be tested are serially diluted and added to the wells. Subsequently, unbound sera is washed away, and an antibody against mouse immunoglobulin is added to detect any mouse antibodies that bound to the fixed antigen.
- SmGST3 specific antigen
- This latter antibody is conjugated to alkaline phosphatase and when appropriate chromogenic substrate for the enzyme is added to the well, a colored product is produced which can be quantitated at a specific wavelength using an immunoassay microplate (ELISA) reader.
- ELISA immunoassay microplate
- a standard curve of increasing concentrations of GST is established in 6-8 wells and a reference polyclonal rabbit serum used in place of the mouse sera.
- SmGST3 is an invertebrate GST with immunologically different properties (speaking strictly from the amino acid sequence point of view) the human or rodent host would be expected to recognize the protein as a foreign one and therefore, SmGST3 should be immunogenic and elicit an antibody response.
- Mice and rats which have been passively administered a mAb against SmGST3 and then challenged with infective cercaria were found to have reduced worm burdens, the female worms released fewer eggs, and the eggs that were released had poorer viability than worms recovered from mice not receiving the mAb
- mice with different major histocompatibility complex (MHC) H-2 haplotypes are given a single dose of naked SmGST3 plasmid DNA and all the tests are conducted at the single time after vaccination for which the responses were maximal.
- the following strai ns of mice (with their H-2 haplotypes in parentheses ) used C57BL/6 (H-2b)--this mouse strain is considered a high responder strain (42, 43); CBA (H-2k)— this strain is considered a moderate to low responder strain (42, 43); SJL (H-2s)--this strain exhibits high levels of eosinophils in response to tissue invasive helminth infections but makes virtually no IgE antibody in these infections (44); and P strain (H-2p), the only mouse strain known to not develop resistance to infection with S.
- mice are either untreated or vaccinated with 10, 50, or 100 ⁇ g pCMV-SmGST3 DNA. At the time of maximal response mice are bled for serum and sacrificed. In those mice receiving SmGST3-coated beads, the lungs are perfused and processed for histological examination. The spleens of the remaining mice are used to establish individual lymphocyte proliferation assays stimulated with either the T cell mitogen, Con A, or the specific antigen, SmGST3. The sera collected is tested in the SmGST3 ELISA to determine relative antibody levels. Humans do not respond to antigenic stimulation as uniformly as do inbred strains of mice.
- Baculovirus expression vector which encodes the 16.4 SmGST-3 insert is descri bed.
- a 6X his(tidine) tag was inserted at the 5' end of the coding sequence which permits metal affinity isolation using a Ni-NTA matrix.
- the Baculovirus Expression Vector System (GIBCO-BRL Cat. #10584-027) was used per the vendors instructions.
- a set of custom po lymeras e chain reaction (PCR) primers were commercially synthesized to the 5' and 3' ends of the cDNA encoding the S. mansoni GST-3 contained in the pBsCMV- 16.4 plasmid.
- the primers were designed so that an EcoR I restriction site was inserted 5' to the ATG start codon while a Hind III site was added immediately after the stop codon in the open reading frame.
- the final construct uses an ATG start codon located 5' to a 6X his(tidine) tag and aligned in such a manner that the inserted
- PCR product was maintained in the open reading frame.
- the resulting 65 1 bp amplified fragment was then ligated into the pFastBac HTb shuttle vector which had been linearized by cutting with EcoRI and Hind III.
- the ligated product was next transformed into DH5 ⁇ F' competent cells and amplified.
- the plasmid DNA was isolated using a Qiagen Spin Prep column. Following visualization on a an agarose gel, 1 ng of plasmid DNA was used to transform DH l OBac cells. These cells contain a bacmid with a mini-attTn7 target site and a helper plasmid.
- the mini-Tn7 element on the pFastBac plasmid can transpose to the mini-attTn7 target site on the bacmid in the presence of transposition proteins provided by the helper plasmid.
- the transposition was carried out by incubation on LB plates containing kanamycin (50 ⁇ g/m l ; gentamicin (7 ⁇ g/ml ); tetracycline (10 ⁇ g/ml), and 100 ⁇ g/ml Bluo- gal plus 40 ⁇ g/ml IPTG. White colonies containing the recombinant bacmid were identified by blue-white screening. High molecular weight DNA was isolated by alkaline lysis from mini prep cultures.
- bacmid DNA containing the SmGST-3 cDNA insert was used to transfcct Sf21 insect ovary cells in the presence of CellFECTIN.
- the culture medium from this transfection was collected to use as a source of Baculovirus-SmGST-3 (Bac-SmGST-3) pending verification that the Sf21 cells were producing the S . mansoni glutathione S-transferase .
- the cells were lysed with 5X lysis buffer (50 mM Tris, pH 7.5 ; 650 mM NaCl; 5% Triton X- 100; 50 mM NaF, 50 mM Na2HPO4; and 50 mM Na pyrophosphate).
- the proteinase inhibitor, PMSF 100 mM stock in absolute ethanol
- the lysis product was centrifuged at 17100 X g for 10 min at room temperature and the soluble phase separated from the pellet.
- the pellet and supernatant were separated on a 12.5% SDS-PAGE gel and the resulting isolated proteins transblotted to nitrocellulose for immunoblotting.
- the nitrocellulose membranes were blocked with 3 % nonfat dry milk solution, and then stained with a 1 : 160,000 dilution of a rabbit anti-S. mansoni GST-3 polyclonal antibody (provided by Dr. James Tracy, U. Wise. Vet School).
- the membranes were washed and a goat anti-rabbit IgG conjugated to horseradish peroxidase (Sigma Chemical Co., St. Louis, MO) diluted 1 : 10.000 was applied. Following several additional washes, the bands were visualized using the ECL chemiluminescent kit from Amersham.
- the chemiluminescent signal was collected on X-ray film and following development, the Western blot image as well as the pretransfer, Coomassie Brilliant Blue-stained replicate SDS-PAGE gel were scanned into computer storage using an HP ScanJet IICX at identical scale to facilitate alignment. The results are shown below as Figure 8.
- the immunodetection of a strong signal at 28 kDa is proof that the Baculovirus-infected cells are indeed producing the SmGST-3 protein. It is noteworthy that little, if any, of the protein is in the pellet fraction and little, if any, is in the extracellular medium. Having verified that the cells can produce the recombinant protein, it one can purify the protein to near homogeneity by passing the lysis supernatant over a Ni-NTA column. The Ni matrix has a strong affinity for the 6X his tag which was cloned 5' to the SmGST-3 molecule.
- Ni-NTA was purchased from Qiagen (Chatsworth, CA) and used as per the vendor's instructions.
- the gel slurry was washed in Buffer A and incubated overnight with 1 ml of Sf21 cell extract (i.e., the resulting lysis supernatant).
- the unbound material was washed away by pelleting the slurry and removing the supernatant (which was retained for subsequent analysis and to verify that the 28 kDa band was removed).
- Buffer A containing 20 mM imidazole
- adherent protein was eluted by washing the column with Buffer C containing 250 mM imidazole and 2 ml fractions collected. The presence or absence of the immunoreactive 28 kDa protein in each of the fractions was again confirmed by SDS-PAGE and chemiluminescent western blotting.
- Figure 9 The results of one such experiment are shown in Figure 9 which suggests that the protein is being recovered in fractions 4 through 12. These fractions were next pooled, dialyzed against phosphate buffered saline and concentrated using an Amicon Centriprep with a nominal Molecular Weight Cutoff of 10 kDa.
- Figure 10 depicts a chemiluminescent Western blot indicating that the protein was concentrated and retained immunoreactivity.
- 100 to 200 ⁇ g of rSmGST-3 from a single T-75 flask of Sf21 cells infected at 80 to 85 confluency can be produced routinely and harvested after 3 days of infection. The resulting protein was next used to skin test mice which had been intramuscularly vaccinated 2 weeks previously.
- One test used to evaluate the immune status of vaccinated mice is a simple skin test in which an antigen used to induce the vaccinated state is subsequently injected intradermally to see if the foreign protein is recognized.
- the end point of the skin test is the quantification of the inflammation (here taken as increased thickness which represents the swelling due to the accumulation of an inflammatory infiltrate) present 24 hr after the skin test is begun.
- the significance of the duration of the test over a twenty four hour period is that inflammation occurring during the first six hours is considered to be indicative of the immediate hypersensiti ve state (mediated by IgE and mast cells) while inflammation occurring at 24 hours represents the delayed hypersensiti ve state .
- Delayed hypersensitive responses are manifestations of cell-mediated immunity and are the desired outcome of any vaccination against an infectious agent such as
- mice vaccinated with naked DNA encoding an S. mansoni-deri ved gene manifest cell-mediated immunity groups of mice were vaccinated with either pBsCMV- 16.4 DNA (specific lest) or pBsRSV-lux (nonspecific, irrelevant DNA encoding firefly luciferase).
- pBsCMV- 16.4 DNA specific lest
- pBsRSV-lux nonspecific, irrelevant DNA encoding firefly luciferase
- mice vaccinated with pBsCMV-16.4 DNA were compared to the SET values of mice vaccinated with pBsRSV-lux.
- the results of a typical experiment are shown in the table below.
- mice vaccinated with the pBsCMV-16.4 DNA and skin tested 14 days later have statistically significant cell mediated immunity against the SmGST-3 antigen.
- the following references were cited herein:
Landscapes
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Molecular Biology (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
A vaccine comprising a non-infectious, non-integrating DNA sequence encoding Schistosoma mansoni glutathione S-transferase and a method of protection against said parasitic infection.
Description
DNA ENCODING 28 kDa GLUTATHIONE S-TRANSFERASE OF SCHISTOSOMA MANSONI AND USES THEREOF
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention relates generally to the fields of parasitology and molecular biology. More specifically, the present invention relates to a naked DNA vaccine for parasitic diseases.
Description of the Related Art
Schistosomiasis is a disease that affects over 250 million people worldwide, primarily in underdeveloped countries and approximately one percent of those who contract schistosomiasis will die annually from hemorrhage and/or liver failure. The causative agent is the human blood fluke, Schistosoma manson i which lives in the inferior mesenteric venules. In these venules, the male and female worms live, mate, and produce large numbers of eggs each day which ultimately accumulate in the liver and elicit a characteristic granulomatous inflammation. In a significant number of infected individuals, the ensuing disease is characterized by hepatosplenomegaly, portal hypertension and esophageal varices, the latter often being fatal. Currently, praziquantel® and oxamniquine® are used to treat schistosomiasis but reinfection occurs with return to fresh water (where the snail intermediate hosts of S. mansoni live) and there is evidence accumulating that the worms are becoming resistant to these agents .
Studies have examined the effects of addition of mitomycin C-treated cells from either infected or normal mice added into lymphocytes undergoing lymphocyte proliferation (i.e.,
the lymphocyte transformation assay or LTA) in response to T cell mitogens (PHA and Con A) or Schistosoma mansoni egg antigens (21 -24). These studies established that unstimulated spleen cells from infected mice, but not from normal mice, induced incorporation of almost 3 times more 3H-TdR than did normal cells (22). This activity was subsequently shown to be dependent on the presence of macrophages in the cultures (23). These studies form the basis of using the lymphocyte proliferation assay as indicative of a cell-mediated immune (CMI) response in mice vaccinated against a specific schistosome protein.
Immunoregulation of pulmonary granuloma formation in toxocariasis (mice infected with the larval stage of the canine roundworm, Toxocara canis, the causative agent of human visceral larva migrans) was established. This model system correlated T cells and their subsets, which were monitored in the LTA, with the development of granulomas forming around T. cam's- anti gen coated CNBr-4B Sephadex beads embolized into the pulmonary microvasculature. This work forms the basis for the remaining CMI assay. A T. cams-specific ELISA to measure parasite-specific antibodies was also developed. This procedure has been modified slightly to measure anti-S. mansoni glutathione S-transferase (SmGST).
Injection of genetic constructs directly into striated muscle in vivo results in expression and, in many instances, secretion of the gene product into the circulation (4-6). Following intramuscular injection, plasmid DNA is found distributed throughout the muscle and is able to diffuse throughout muscle- associated connective tissues. Foreign transgenes have been expressed for up to 19 months but the efficiency of transfer is relatively low (2% of treated muscle) (5). Localization studies at the electron microscopic level using plasmid DNA conjugated to colloidal gold and compared to gold-tagged polyethylene glycol have shown that nucleic acids (but not the polyethylene glycol) are able to traverse the external lamina and enter myofibers through the T-tubule system, apparently associated with caveolae in the sarcolemma (7).
Since the original observations by Wolff et al. (4) that striated muscle can express injected transgenes or mRNAs, many of
the variables of the technique have been clarified although many questions still remain. Of particular interest is the observation that the level of expression of transgenes tends not to be predictable although in most cases the degree of expression seems to be proportional to the amount of nucleic acid taken up. For example, a 100 gram injection into the mouse quadriceps muscle resulted in approximately 1.5 % of the fibers expressing β -gal activity as demonstrated by enzyme histochemistry 7 days after the injection. Staining could be observed up to 400 microns from the injection site. At present, the best evidence available indicates that plasmid
DNA, when taken up by skeletal muscle, remains circular and neither integrates into chromosomal DNA nor replicates (5, 6).
The expression of naked transgenes has been found to vary substantially following injection into different mice under identical conditions of injection ( 14). Subsequent studies (6) indicated that the injection technique per se is not responsible for this observed variation in transgene expression. The volume of fluid containing the genetic construct does not appear to be critical in determining uptake or expression. Likewise, neither the rate of injection (less than 30 seconds to 5 minutes) or the type of needle used to administer it (27 gauge with 0.2 cm limiting collar compared with glass capillary), seem to affect the outcome. However, conditions that lead to increased degradation of DNA can result in less uptake and expression. Interestingly, DNA dissolved in sterile saline results in 5-fold to 10-fold greater transgene activity than the same DNA dissolved in 20% sucrose. An injection of 25% sucrose 15 to 30 minutes prior to DNA injection was reported to reduce the variability in the technique (14). This presumably results from the fact that saline causes less muscle damage than the sucrose vehicle in the former case and may actually stimulate pinocytotic uptake in the latter instance. Animals which received multiple injections of the same construct over several weeks expressed less activity than did mice receiving a single injection. Moreover, naked DNA taken up by skeletal muscle tends to persist for substantial periods of time and better expression can be expected with fewer administrations. The choice of promoter sequence to drive gene transcription can influence gene expression by a factor of 1 ,000 or more ( 14). More recently,
the drug Marcaine® has been found to increase uptake and expression dramatically when given a few days prior to the DNA injection (27). The drug is slightly myotoxic and satellite cells surrounding necrotic fibers undergo mitosis which may be account for the enhanced uptake and expression.
Naked DNA techniques ( 1 1 ), and intramuscular injection of plasmid DNA in particular, have been used to immunize (i.e., vaccinate) experimental animals, and direct injection has been shown to elicit both cellular and humoral immune responses ( 1, 8- 10). Mice have been successfully immunized by intramuscular injection of plasmid DNA carrying an influenza gene as determined by their ability to survive a lethal challenge infection (9, 10). Using the same polynucleotide vaccine in mice revealed that 1 ) a single injection elicited both cellular and humoral responses that persisted for 1 year and 2) the cellular response was associated with the CD8+ subpopulation of T cells (i.e., killer T cells) (12). Similarly, chickens were given plasmid DNA injections by one of three routes (iv, ip, or im) and subjected to an influenza virus- challenge. In these studies, gene vaccination resulted in 60% protection which was attributed to use of a gene from a heterologous virus (i.e., a virus related to the influenza virus that normally infects chickens but differing by 16% in amino acid sequence). Mice which have been vaccinated with plasmid DNA encoding a bovine herpes glycoprotein have gone on to make significant amounts of antiglycoprotein antibody and the sera from these mice contained significant amounts of viral neutralizing activity as compared to mice receiving the plasmid platform lacking the herpes gene (28). Cattle have also been injected intramuscularly with plasmids carrying bovine herpesvirus genes and these cattle exhibited significantly elevated titres of anti- bovine herpesvirus glycoprotein antibody and the treated animals shed significantly less virus than the control animals following infectious challenge (28) . Recently, non-human primates
(cynomolgus macaques) were injected intramuscularly with a plasmid expressing the gp l 60 gene of HIV. Animals were inoculated three times and serum collected two weeks later. This regimen led to seroconversion of all test animals and the sera was shown to contain neutralizing activity as determined by the
reduction in number of microsyncytia forming in cultures of T cells exposed to HIV-I ( 1 , 13).
The prior art is deficient in the lack of effective means of treating Schistosomiasis using naked DNA vaccination technology. The present invention fulfills this longstanding need and desire in the art.
SUMMARY OF THE INVENTION
The present invention discloses several eukaryotic expression plasmids containing genes derived from S. mansoni that serve as the basis of a polynucleotide vaccine. In this system, no adjuvants are required, no infectious agents (either killed or attenuated) are introduced into the patient during the immunization protocol, and no exogenous contaminants of any kind are introduced which could elicit spurious immune responses. Lastly, there is solid evidence that gene products produced by skeletal muscle are presented to the immune system in such a way as to elicit cell-mediated immunity which makes this novel technique ideally suited for development into a human vaccine.
The present invention uses naked DNA vaccine technology to establish the feasibility of preventing or ameliorating the effects of infection with the human intravascular blood fluke, Schistosoma mansoni. The present invention characterizes both cellular and humoral immune responses of mice vaccinated with
DNA encoding a gene whose product is a good candidate for vaccine use when admixed with adjuvants and given to mice, rats, and primates. Experimental development of naked DNA vaccination technology is important, not only for the control of the parasite causing schistosomiasis in 250 million people, but for establishing a new way to vaccinate against almost any infectious helminth in manner that does not require live or attenuated organisms or the addition to the vaccine of noxious chemicals in the form of adjuvants such as peanut oil or alum. Vaccination of susceptible hosts using intramuscularly injected naked DNA encoding a parasite-specific protein elicits immunological responses necessary for inducing resistance to infection and/or ameliorating the disease in such a way as to alter
subsequent transmission of the disease. The present invention determines ( 1 ) the effect of time and dose on the development of the cellular immune response of mice vaccinated with naked DNA encoding Schistosoma mansoni glutathione S-transferase; (2) the effect of time and dose on the development of the humoral immune response of mice vaccinated with naked DNA encoding Schistosoma mansoni glutathione S-transferase; (3) the effect of the H-2 haplotype on the cellular and humoral immune responses of mice vaccinated with naked DNA encoding Schistosoma mansoni glutathione S-transferase. The present invention uses selected strains of inbred mice which are known permissive hosts for the entire life cycle of S. mansoni. As measures of cellular immunity, lymphocyte transformation assays in response to parasite glutathione S-transferase and granuloma formation around glutathione S-transferase-coated beads are monitored while humoral immune responses (i.e. , antibody production) are evaluated using a glutathione S-transferase specific enzyme-linked immunosorbent assay (ELISA).
In one embodiment of the present invention, there is provided a composition of matter comprising a naked DNA vaccine for the parasitic worm, Schistosoma mansoni or human blood fluke. In another embodiment of the present invention, there is provided novel plasmids and expression vectors for use in the methods of the present invention. In yet another embodiment of the present invention, there is provided a method of protecting against Sch is tos o m a manson i comprising administering to an animal in need of such treatment a pharmacologically effective dose of a naked DNA vaccine. Other and further aspects, features, and advantages of the present invention will be apparent from the following description of the presently preferred embodiments of the invention given for the purpose of disclosure.
BRIEF DESCRIPTION OF THE DRAWINGS
So that the matter in which the above-recited features, advantages and objects of the invention, as well as others which
will become clear, are attained and can be understood in detail, more particular descriptions of the invention briefly summarized above may be had by reference to certain embodiments thereof which are illustrated in the appended drawings. These drawings form a part of the specification. It is to be noted, however, that the appended drawings illustrate preferred embodiments of the invention and therefore are not to be considered limiting in their scope.
Figure 1 shows a schematic drawing of the deletion of CMV promoter and replacement with α S A pro m oter . Inset=miniprep of pRcαSA cut with (S)ma I and (P)vu II.
Figure 2 shows the histochemical localization of E. coli β gal activity in mouse skeletal muscle 3 weeks after injection of 100 μl saline of b) saline with 50 μg pRSV-LacZ DNA. Orig. 400x. Figure 3 shows a photograph of a gel showing RE orientation analysis of pCMVGST3 clones. Lane 1 = Hind Ill/lambda markers , 818 and 4300 bp lanes are correct orientation.
Figure 4 shows that SmGST expression by HEK 293 cells 48 hours after CaPO4 transfection. (-) = no DNA. CDNB; chloro-dinitrobenzene used as substrate; n=number of dishes assayed.
Figure 5 shows the splenic mass 8 weeks after no injection, intramuscular injection of 10 μ g pSmGST3, 50 μg pSmGST3, or a subcutaneous injection of 25 μg of pure SmGST mixed with alum . (pSmGST3 and pCMV-SmGST3 are used interchangeably and are of equal rank in identification.) 130%- line=significance. n=number of spleens.
Figure 6 shows an electrophoretogram showing purification of a 28 kDa protein encoding the S . manson i glutathione S-transferase. Figure 6A shows bacteria transduced with pCMV-GST3. Figure 6B shows control bacteria transduced with pCMV-Lux encoding an irrelevant protein. Lane 1 = molecular weight markers X 1000; Lane 2 = bacerial lysate (starting material) which was applied to a glutathione agarose affinity column; Lane 3 = last wash prior to eluting the column; Lane 4 shows the concentrate of eluate from glutathione-agarose affinity column. Note the over expressed 28 kDa band in lane 2A
and the recovery of a discrete band at 28 kDa in lane 4A (arrow) which is completely absent in lane 4B. Gel was composed of 12% polyacrylamide and was stained with coomassie blue, destained, dried and scanned with a HP Scanjet. Figure 7A and 7B show the nucleotide sequence of plasmids pRc/ASK8-SmGST3 and pCMV-SmGST3.
Figure 8 shows the SDS-PAGE (Figure 8A) and Western Blot (Figure 8B) of uninfected or Bac-SmGST-3-infected Sf21 cells. A 12.5% SDS-PAGE was run as follows: Lane M = molecular weight Markers (in kDa) indicated at left of image; Lane U = uninfected Sf21 cell lysate; Lanes 3, 4, and 5 = Sf21 cell lysates harvested at 3, 4 or 5 days after infection with Bac-SmGST-3. Note the absence of the 28 kDa band in lane U and its over expression in lanes 3, 4, and 5. (Figure 8B) The gel in Figure 8A was electroblotted to a nitrocellulose membrane and immunostained with polyclonal rabbit anti-S. mansoni GST-3 followed by staining with goat anti-rabbit IgG conjugated to horseradish peroxidase. Immunolocalization was observed using an ECL kit from Amersham followed by image capture on X-ray film. Both Figures 8A and Figure 8B are computer scans of the original images. Note the strong band at 28 kDa indicative of the production of SmGST-3. The faint images below the band are degradation products of the 28 kDa protein.
Figure 9 shows a SDS-PAGE of various stages during the isolation procedure of recombinant SmGST-3 protein from Bac-
SmGST-3 infected Sf21 cell lysates. Lanes contain the following: W = whole unfractionated Bac-SmGST-3 infected Sf21 cell-lysate; A = same lysate after adsorption with Ni-NTA slurry (en batch); 1 , 2, and 3 = washes with Buffer A or B to remove weakly adherent proteins; MR = 10 kDa molecular weight markers; Fractions 1 - 4 =
2 ml fractions were collected while eluting the Ni-NTA slurry with Buffer C containing 250 mM imidazole. Note the presence of the 28 kDa bands in fractions 4 through 12.
Figure 10 shows a Western Blot confirming the immunoreactive property of the 28 kDa protein band following concentration from pooled fractions 4 through 12 depicted in
Figure 9. The image represents lanes loaded with the following:
Lane 1 = whole lysate (compare with Lane W in Figure 9); Lane 2 =
post adsorption fraction showing depletion of immunoreactive band; Lane 3 = 10 kDa ladder (position determined by comparison with SDS-PAGE used for blotting); Lane 4 = filtrate from 10 kDa cutoff Centriprep concentrator indicating that 28 kDa protein was partitioned out of this solution; Lane 5 = retentate from 10 kDa cutoff Centriprep concentrator indicating that 28 kDa protein was partitioned into this fraction.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a vaccine comprising a non-infectiou s , non-inte grating DNA sequence encoding Schistosoma mansoni glutathione S-transferase. Preferably, the DNA comprises a plasmid encoding a protein, polypeptide or peptide which is operably linked to a promoter. In one embodiment, the plasmid is pCMVSmGST-3. In other embodiments, the plasmid is selected from the group consisting of pBsCMV- 16.4 DNA and pRc/ASK8-SmGST3.
The present invention is also directed to a method of protecting against infection by Schistosoma mansoni comprising the step of: administering to an animal in need of such treatment a pharmacologically effective dose of a vaccine composition comprising a non-infectious, non-integrating DNA sequence encoding Sch isto soma manson i glutathione S -transferase . Preferably, the vaccine composition comprises a plasmid encoding a protein, polypeptide or peptide which is operably linked to a promoter. Representative examples of plasmids useful in this method of the present invention include pCMVSmGST-3, pBsCMV- 16.4 DNA and pRc/ASK8-SmGST3. Generally, a person having ordinary skill in this art would be able to determine optimal dosage of the compositions of the present invention to achieve the desired outcome. Generally, the plasmid is administered in an animal in a dose of from about 25 μg to about 200 μg. Preferably, the plasmid is administered into muscle although it is possible it could be adminstered into another site in the body. Generally, the plasmid would be administered between 1 and 6 times.
The present invention is also directed to a method of developing cell mediated immunity to glutathione-S-transferase of
Schistosoma mansoni comprising the step of: administering to an animal in need of such treatment a immunologically effective dose of a plasmid of the present invention. Representative examples of plasmids useful in this method of the present invention include pCMVSmGST-3 , pBsCMV- 16.4 DNA and pRc/ASK8-SmGST3. Generally, a person having ordinary skill in this art would be able to determine optimal dosage of the compositions of the present invention to achieve the desired outcome. Generally, the plasmid is administered in an animal in a dose of from about 25 μg to about 200 μ g. Preferably, the plasmid is administered into muscle although it is possible it could be adminstered into another site in the body. Generally, the plasmid would be administered between 1 and 6 times.
In the present invention, a technique for injecting naked DNA directly into skeletal muscle using a reporter plasmid in which the lacZ gene was inserted 3' to an RSV promoter in a eukaryotic expression plasmids engineered was used. At weekly intervals muscles were removed, frozen sections prepared, the sections stained for β-gal activity and the histochemical localization was compared to control muscles injected with saline. All lacZ DNA-injected muscles stained intensely indicating the presence of the plasmid and expression of the bacterial gene by the mouse muscle cells while all controls were completely devoid of reaction product (See Figure 1 ). Next, the cDNA encoding the SmGST-3 isoenzyme was obtained. The cDNA was excised from a pGEX plasmid and ligated into a eukaryotic expression plasmids 3' to an immediate early CMV promoter.
To establish that this construct was both non-toxic to mammalian cells and to verify that cells transfected with pCMV- SmGST-3 expressed the GST gene, HEK 293 cells in tissue culture were transfected by the CaPO4 method. Plates at the time of transfection were approximately 50% of confluency and after transfection were allowed to incubate at 37°C/5% CO2 in air for another 48 hr. At this time the cells were confluent which indicates that the cells tolerated the foreign DNA well. Soluble extracts of the monolayers were shown (Figure 2) to overexpress GST activity when using chlorodinitrobenzene (CDNB) as the electrophilic substrate in the assay described by Habig and Jakoby
(20) as compared to cells transfected with an irrelevant plasmid (which was also determined to be expressed, but is not shown in Fig. 2). Finally, BALB/c mice have been vaccinated with various quantities of either pCMVSmGST-3 or an irrelevant plasmid (pRSV- Lux) and after two weeks the mice were infected with 200 cercariae of S. mansoni. Figure 3 based on splenic mass increase (which correlates with lymphocyte proliferation in the white pulp) illustrates that mice receiving the pCMVSmGST-3 DNA are affected by the treatment. Glutathione S-transferases (GST) (EC 2.5.1.18) catalyze the conjugation of both endogenous and exogenous electrophiles with the major cellular nucleophile, reduced glutathione (GSH) (29). GSTs are found widely distributed in nature in both invertebrates such as Schistosoma mansoni and vertebrates such as mice, rats and humans ( 18) . Most organisms contain multiple GST isoenzymes that are characterized by distinct but often overlapping substrate specificities ( 18). Adult S. mansoni contain at least five forms of GSH S-transferase, three of which (designated SmGST- 1 , SmGST-2 and SmGST-3) account for 2-4% of the soluble ■ protein in the male worm (19, 20).
All GSH S-transferases from vertebrates that have been characterized are dimeric proteins composed of either identical (homodimer) or nonidentical (heterodimer) subunits ( 16, 17). The polypeptide subunits are catalytically independent and the quaternary structure is thought to be necessary for enzyme stability ( 17). In 1988, three cytosolic forms of S. mansoni G S T were purified and found to have very similar catalytic, physicochemical, and immunological properties. However, the most striking feature is that they are catalytically active monomers rather than dimers ( 19). It has been established that a) the parasite GST is a 28 kDa protein in contrast to vertebrate GSTs which range from 44 to 50 kDa and b) that the parasite proteins do not immunologically cross react with vertebrate (i.e. host) GSTs which makes the present invention herein feasible because immune responses against parasite GSTs will not immunoneutralize the hosts GSTs.
GSTs are good candidates for use in vaccines against S. mansoni (29-32). In the rodent model of S. mansoni infection a Mr
28,000 molecule (Sm28 or P28) was identified as a major surface protein (where it may mediate the conjugation of GSH to antischistosomal drugs) on larval schistosomes (schistosomules) and was subsequently identified as a GST (32). From a routine screening of S. mansoni RNA translation products using immune serum from infected mice, a protein was identified which could be detected on the surface of the immature S. mansoni worm and the adult. This protein was found to have a Mr of 28 KDa and cDNA encoding the 28 K protein was subsequently cloned in E. coli and the recombinant protein used to immunize rats and BALB/c mice. Good immunoprotection against 5. mansoni in several host species (i.e., approximately 50% in the mice) has been demonstrated using Sm28 and either Freund's Complete or an alum adjuvant (31).
Determination of whether vaccination with a polynucleotide (i.e. naked DNA) will yield a better immunization than the more conventional vaccination using a pathogen-derived protein with an adjuvant such as aluminum potassium sulfate (alum, which is acceptable for human and veterinary use) is desirable. The genetic constructs were made and several milligrams of expression-grade DNA were stored at -80°C. To demonstrate that cellular immunity in murine schistosomiasis is induced, lymphocyte proliferation to specific S. mansoni antigen (SmGST-3), an already documented manifestation of cellular immunity and pulmonary bead granuloma formation (34) are used. Likewise, the development of mouse anti-SmGST antibodies (i.e., the humoral immune response) are monitored by ELISA.
The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion.
EXAMPLE 1 Modification of a eukaryotic expression vector
This plasmid known as pRc/CMV was obtained from the Invitrogen Corporation and contains the immediate early enhancer-promotor sequence from cytomegalovirus (CMV) , polyadenylation signal and transcription termination sequences from the bovine growth hormone (BGH) gene and SV40 origin. This plasmid and its host species of permissive E. coli, TOP10F',
were used for all subsequent experiments. The CMV promoter was first removed from the pRc/CMV plasmid and replaced with the chicken α -skeletal actin promoter sequence. This is shown schematically in Figure 1 and was performed as follows. The CMV promoter sequence is flanked 5' by an Nru I site and 3' by a Hind III site just inside the polycloning sequence. First, a Hind III restriction enzyme (RE) cut was performed on purified pRc/CMV DNA followed by a Klenow-mediated blunt end fill-in of the cut site. The linearized, cut DNA was ethanol precipitated and then cut with Nru I. Because Nru I is a blunt cutter, the pRc backbone was now ready for blunt end ligation. From a stock of custom fragments, a 6.2 Kb pαSK fragment of genomic DNA was obtained carrying the chicken a -skeletal actin (α SA) gene and its entire promoter (nb: skeletal actin promoters are highly conserved and have been shown to function in mammalian cells). The 675 bp promoter sequence as shown in Figure 1 was contained between an Eco RV and Nae I site. A sequential RE digest was performed and the 675 bp αSA fragment isolated from a 1 % agarose gel. The 5 Kb pRc plasmid and the 675 bp αSA fragment were ligated using T4 ligase. The ligation products were then transfected into TOP10F' and 22 colonies were screened for plasmid DNA. Because blunt end ligations can result in the fragment being ligated in either direction, plasmids were cut with Sma I which cuts asymmetrically one time within the αSA sequence and once within the remaining pRc sequence. If the α SA sequence is in the correct 5'— >3' orientation with respect to the polycloning sites then two bands of 1800 bp and 4000 bp should be seen upon agarose gel electrophoresis. One such clone designated pRcαSAδ was found (shown in the insert of Figure 1 ) and this orientation was confirmed by cutting with a different RE, Pvu II, which gives fragments of 81 1 , 1065 , 1 196, and 2400 bp. The plasmid DNA was isolated from a liter of bacteria. Glycerol stocks of TOPI OF' containing pRcα SA8 (pRcASK8 is of equal rank and used interchangeably) were prepared and stored at -80°C. A control plasmid for expression under the control of the α SA promoter sequence with the promoter sequence inserted in the 3'-->5' orientation was also stocked (pRcαSA 14) .
The next stage was to insert the gene of interest into the polycloning site. The SmGST3 fragment was removed from the pCR3-SmGST3 TA cloning vector using Xba I and ligated into pRcASKδ, which had also been cut with Xba I, using T4 ligase. The ligation products were then transformed into TOP10F' cells. The new construct, designated pRcASK8-SmGST3 was stocked in a glycerol solution and stored at -80°C.
EXAMPLE 2 In vivo gene transfer of pBsRSV-βGal and visualization of gene expression
In order to verify that transferred genes are in the intended tissues and that the foreign genes are being expressed, a eukaryotic reporter plasmid in which the E. coli β-galactosidase (β- gal) gene is inserted into the polycloning site can be used to verify both properties. By staining frozen sections for β-gal activity, fibers in which the transgene are being expressed will stain positively while all other fibers fail to stain. Groups of mice were injected in the quadriceps muscle with either saline or 50 μg of purified pBsRSV-βGal DNA. Mice were sacrificed after 1 , 2, or 3 weeks and the injected muscles removed, snap frozen in methyl butanol on dry ice, and 10 μm frozen sections prepared. The sections were stained with X-gal in the presence of potassium ferriferrocyanide. All muscles injected with the pBsRSV-βGal plasmid had scattered myofibers which stained intensely blue- black indicating that the β-Gal gene product, β-galactosidase, was being expressed. None of the saline-injected muscles exhibited positive staining (Figure 2).
EXAMPLE 3
Fabrication of pBsCMV-GST3
A plasmid, pGT16.4, carrying the cDNA encoding the 28 kDa glutathione S-transferase (= SmGST3) situated between two Eco RI sites was obtained from Dr. Jim Tracy (University of Wisconsin Veterinary School). The cDNA was liberated with Eco RI and using a QIAEX gel extraction kit, the 782 bp fragment was isolated and ligated into an Eco RI site within the polycloning sequence of pB sCMV , a gene transfer vector engineered for
eukaryotic gene expression. Orientation was determined by an asymmetrical cut with Mun I which gave two fragments of predicted sizes ( 818 and 4300 bp) while inverse orientation yielded expected fragments of 1538 and 3580 bp. As seen in Figure 3, five colonies contained inserts that were in the correct orientation and three colonies contained inversions. This plasmid was electroporated into electrocompetent DHlOb cells. Based on miniprep analysis of resultant overnight colonies, a liter of bacteria were grown overnight, and expression grade plasmid DNA was isolated by double alkaline lysis followed by double CsCl gradient ultracentrifugation. The resulting DNA was dialyzed against 4 liters of I X TAE buffer, ethanol precipitated and dissolved in 1.0 ml sterile, deionized, distilled H2O. Final DNA and RNA concentrations were determined spectrophotometrically at OD260/280. Two clones, designated 3.7 and 3.9, were prepared in parallel and the final DNA yields were 3.0 and 3.1 mg DNA per ml, respectively .
EXAMPLE 4
Isolation of recombinant SmGST3 protein by GSH-agarose affinity chromatography
TOP10F' cells carrying the pCMVGST3 plasmid were grown overnight and then collected by centrifugation. The cells were lysed and the soluble fraction collected and applied to a GSH- agarose affinity column to purify the overexpressed protein, SmGST3. A parallel culture of pCMV-Lux expressing the irrelevant protein, luciferase was used as a control. Affinity purified SmGST3 was eluted from the column with 10 mM glutathione. The recovery was monitored by coomassie blue staining of 12% polyacrylamide gel electrophoresis and examination of the 28 kDa band (Figure 6).
EXAMPLE 5
Transfer of pBsCMV-GST3.7 in to HEK 292 cells
To establish that this construct was both non-toxic to mammalian cells and to verify that cells transfected with pBsCMV-
GST3.7 expressed the GST gene, HEK 293 cells in tissue culture were transfected by the CaPθ4 method. Plates at the time of transfection were approximately 50% of confluency and after transfection were allowed to incubate at 37°C in 5% C02 in air for another 48 hours. At this time the cells were confluent which indicated that the cells tolerated the foreign DNA well. Soluble extracts of the monolayers over expressed GST activity when using chloro-dinitrobenzene (CDNB) as the electrophilic substrate in the assay described by Habig and Jakoby (34) as compared to cells transfected with an irrelevant plasmid (See Figure 4). This demonstrated that the construct was not toxic and suggested that the plasmid could be used in a gene transfer protocol for live animal experiments.
EXAMPLE 6
Addition of Xba I restriction sites to either end of SmGST3
To make it easier to work with the cDNA encoding SmGST3, (PCR) primers were designed to add an Xba I restriction site to both the 5' and 3' ends of the fragment and to add a Kozak consensus sequence that would enhance the binding of the polymerase to the promoter sequence) to the 5' end. The PCR fragment was cloned directly from the thermal cycler into Invitrogen's TA cloning vector, pCR3. This vector is actually designed to serve as a eukaryotic expression vector and the cloned fragment is under the control of the immediate early CMV promoter. This construct was electroporated into DH lOb cells and yielded an 85% cloning efficiency. Tubes of stock bacteria carrying this construct (designated pCR3-SmGST3) were stored at -80°C.
EXAMPLE 7
Vaccinate- and-challenge experiment
BALB/c mice were injected intramuscularly (im) (vaccinated) with 10 or 50 μ g of either pCMVGST-3 or an irrelevant plasmid (pRSV-Lux, containing the luciferase reporter gene). After two weeks the mice were challenge-infected with 200 cercariae of S. mansoni. An additional group was vaccinated in the
conventional way with 25 μg of SmGST3 isolated from adult S . mansoni adsorbed on alum adjuvant as described by Mishell and Shiigi (Mishell, BB, et al.. Selected Methods in Cellular Immunology, W.H. Freeman, San Francisco, pp. 1 -486, ( 1980)). Results are shown in Figure 5 and were based on the splenic mass increase which correlates with lymphocyte proliferation in the white pulp. Figure 5 demonstrates that mice receiving the pCMVSmGST-3 DNA responded to the treatment. The mice were sacrificed 6 weeks after infection and 8 weeks after vaccination. The portal vein was retrogradely perfused and the adult worms collected. The livers were fixed in formalin for histological analysis and the muscle that was injected with DNA was excised and placed on dry ice for luciferase assay. Sera were collected from all mice prior to vaccination, immediately prior to infection and at sacrifice. To confirm that plasmid DNA was retained and expressed in skeletal muscle for the 8 week duration of the experiment, muscles that had been removed at necropsy and snap frozen were subjected to the luciferase assay. The data from this assay are shown in TABLE I, and based on the pRSV-Lux group, indicate that DNA injected into these mice persisted for the duration of the experiment. Worm burdens did not appear to be affected by polynucleotide vaccination (Table II).
TABLE I
Luciferase luminometry on skeletal muscle from mice injected with either pRSV-Lux or pBsCMV-GST3.7 DNA eight weeks previously
TREATMENT n Lux units: mean±S E
Lux Std 1 unit 1 1 0,75 8
Lux Std 5 unit 1 57 ,765
None-Inf. only 4 484±99 pRSV-Lux 10 μg 5 53 ,952±25 , 889 pSmGST3 10 μg 4 833±308 pSmGST3 50 μg 4 468 ±42
TABLE II
Portal perfusion of mice 6.5 weeks after injection with 200 S. mansoni cercariae
Recoveries Control pRSV-Lux pBsCMVGST pBaCMVGST SmGST+Alum
10 μg 50 μg
Males 4 0 50 26 65 90
Females 4 0 72 26 69 67
Total 8 0 122 52 135 147
(n) 6 5 4 5 5
WWoorrmmss// 1133..33 24.4 13 27 29.4
Mouse
The luciferase data at 8 weeks after DNA injection into these mice demonstrate that the injected DNA did persist for the duration of the experiment consistent with the findings of others. However, worm burdens did not appear to be affected by polynucleotide vaccination. However, when the number of S . mansoni eggs per unit area of liver was examined in H & E stained sections, mice vaccinated with 10 μg of pCMV-GST3.7 DNA had 89% fewer eggs than mice receiving any of the other treatment regimens. This has been suggested as being the manner in which
SmGST-induced vaccination manifests itself. It is worth pointing out that the number of worms per mouse in the control group and the pCMV-GST3 group in Table II is almost identical indicating that different numbers of worms can not account for the reduced egg counts in the SmGST3 group. This is compelling evidence that naked DNA vaccination is an effective modality to control schistosomiasis. A decrease in the number of eggs deposited per female results in less morbidity per patient (less liver involvement) and, more importantly, fewer eggs to reach the environment and spread the infection to new snails.
TABLE III
Eggs per unit area of mice vaccinated with DNA or isolated GST protein
Treatmen t n Eggs/100 mm2. liver % Reduction
Control 6 60.0 pRSV-Lux 5 69.7 (- 16%) p pBBssCCMMVVSSmmGGSSTT33 44 66..77 89%
SmGST + Alum 5 46.0 23%
Table III: Eggs pi εr unit are f liver of mice vaccinated with DNA or isolated GST protein. Eggs (25/mo.) counted in H&E stained, 5 μm sections taken at 125 μm intervals. n=number of mice.
EXAMPLE 8
Lymphocyte Proliferation Assay
The lymphocyte proliferation assay (26, 35) used is as follows. Briefly, spleens removed asceptically are reduced to single cell suspensions in cold RPMI 1640 medium. The suspensions are filtered through 2 layers of sterile gauze and the passing debris allowed to settle for 10 minutes. The resulting suspension is decanted and centrifuged at 900 X g for 10 minutes at 4°C and resuspended in fresh RPMI 1640. The cell concentration is determined by electronic counting (Coulter Counter) and adjusted to 10? cells/ml. The cells are cultured at a density of 2 X 10^ cells/0.175 ml of RPMI 1640 containing 5% normal human, heat- inactivated serum and 20 μg of gentamicin sulfate. Unstimulated control cultures receive an additional 25 μl of RPMI 1640 and stimulated cultures 25 μl of RPMI 1640 containing either 20 μg/ml Con A (2.5 μ g/200 μ l culture); or 24 μ g GSH-agarose affinity purified 5. mansoni GST (about 70% of which is GST3) (3 μg/200 μl culture). Cultures are incubated in 96 well flat bottom plates with loose-fitting covers in a 37°C incubator gassed with 5% CO2 in air with saturated humidity. On the morning of the 5th day (4.67
days of culture) 0.5 μCi of tritiated thymidine (^H-TdR, Sp. Act. = 2.0 Ci/mmol) is added in 25 μ l of RPMI 1640 and the cultures incubated an additional 8 hours. The cells are then harvested onto glass fiber filter strips using an automated cell harvester (BRANDEL M24V-Special). The amount of isotope incorporated into nascent DNA is determined by liquid scintillation spectroscopy after immersing the individual filters into vials of Econofluor LSC cocktail. All LSC data is corrected for quenching and expressed as disintegrations per minute (DPM).
EXAMPLE 9
Bead Granuloma Formation
The method for studying bead granuloma formation was used exactly as described previously (24, 25) except that GSH- agarose affinity-purified SmGST is substituted for methylated bovine serum albumin or T. canis antigens. Briefly, freeze-dried CNBr-activated Sepharose 4B beads (0.5 g) will be hydrated for 15 minutes in 100 ml o 1 mM HC1, sized using 41 and 52 μm Spectromesh macro filtration screens to achieve a uniform bead population (46 ± 8 μ m ) which is then concentrated by centrifugation at 1 ,000 X g (4 times) for 10 minutes. The supernatant is removed by aspiration and the bead pellet resuspended in 1 ml of the purified GST protein in borate buffer (pH 8.3) at 1 -2 mg/ml. The protein-gel suspension is mixed overnight at 4°C on an end-over-end mixer. The gel suspension is again centrifuged as above and the pellet resuspended in 3 ml of 1 M ethanolamine, pH 8.0 at room temperature to block unbound sites on the beads. The beads are washed several times with sterile phosphate buffered saline (sPBS), pH 7.6, counted and adjusted to 10,000/ml. Intravenous injection of 0.5 ml of the bead suspension via the lateral tail vein of mice anesthetized with Metaphane initiates bead granuloma formation. Six days later the mice are sacrificed, the lungs perfused with 2.5 ml of 10% buffered neutral formalin, and histological sections cut and stained with H & E. Granulomata forming around the beads are quantitated by planimetry .
EXAMPLE 10
ELISA
The ELISA procedure used is essentially as described (26) with modifications for use in detecting anti-SmGST antibodies. Briefly, the affinity purified SmGST is diluted in 0.05 M carbonate buffer at 3-5 μg/ml and 100 μl of the solution is added to each well and incubated at room temperature 1 -2 hours and then 4°C overnight. The plates are then rinsed 3 times with PBS and then nonspecific binding sites blocked with 2% bovine serum albumin diluted in PBS for 30 minutes at room temperature. The blocking solution is rinsed away by 3 washes of PBS containing 0.05% Tween 20. The plates can either be dried and stored or used as follows. Each mouse serum to be tested is diluted (see below) in PBS/Tween 20 and 100 μ l of this dilution added to the precoated wells (each dilution done in duplicate) and the plate incubated at room temperature 1 hr. The plate is then washed 4 times with PBS/Tween 20 and 100 μl of horseradish peroxidase-labelled goat anti-mouse immunoglobulin (Heavy and light chains combined) diluted 1 : 100 in PBS/Tween 20 added to each well and incubated for 1 hour. The plate is then washed 5 times in PBS/Tween 20 and once in PBS. Each wel l receives 100 μl of the substrate solution, 0.4 mM 2,2'-azino-di(3 ethylbenzthiazoline sulfonic acid) and 2 mM H 2O 2 diluted to a final molarity in 0.05 M citrate buffer (pH 4.0). After optimal color development occurs ( 15-30 min. in the dark at room temperature) the reaction is stopped by adding 50 μl of 1.0 N
NaF to each well. Spectrophotometric absorbance is determined in individual wells with an enzyme immunoassay (BioTek 308) plate reader at 405 nM with a 490 nM reference. A standard curve is established by coating wells with 0.1 , 0.5, 1.0, 2.5, 5.0, and 10.0 μg of affinity-purified SmGST per well. The greatest dilution of a reference polyclonal rabbit anti-SmGST serum giving a linear recognition of the standard curve is used to determine what dilution of the test sera is examined in the ELISA assay.
EXAMPLE 11
Plasmid Preparation and Naked DNA Vaccination
Plasmids were prepared by standard methods that included double alkaline lysis of the bacteria propagating the
plasmids, two purifications on CsCl gradients, dialysis against 1 mM Tris-HCl (pH 7.4) and 0. 1 mM EDTA, ethanol precipitation, and solvation into distilled, deionized autoclaved water as described (4- 6). Approximately 5 mg of pCMV-SmGST3 was prepared. However, should the quantity of plasmid DNA need to be replenished, the bacteria containing this plasmid is in house in a frozen stock. In addition, the pRSV-LacZ reporter gene plasmid may be used. Each naked DNA injection has added to it 10 μg of pRSV-LacZ DNA prepared as above. At the time the mice are sacrificed, the quadriceps muscle that was injected, as well as the contralateral quadriceps i s dissected out, snap frozen in methyl butanol on dry ice, and sectioned on a cryostat for demonstration of the bacterial enzyme, β -galactosidase (2% X-Gal in potassium ferriferrocyanide). This control serves two purposes. First, it provides knowledge that the DNA solution for vaccination was delivered to the muscle and secondly, it indicates that foreign genes were being expressed by the muscle in the case that no immunological response to SmGST3 is observed. Additionally, a monoclonal antibody, that specifically recognizes E. coli β-gal can be used to perform an ELISA in parallel with the GST ELISA to monitor host response to the transferred genes.
To perform the naked DNA vaccination, mice are anesthetized with Metaphane until they are nonresponsive to irritative stimuli (whisker tug and paw pulling). While maintaining this level of anesthesia, a small incision through the skin perpendicular to the long axis of the quadriceps of the leg are made to expose the length of the muscle. A 100 μl injection of sterile saline containi ng the desired mass of expression-grade plasmid DNA is injected into the body of the quadriceps muscle using a 1 ml tuberculin syringe and a 27 g x 0.5 in needle. The needle is withdrawn slowly so as to prevent reflux, and the incision closed with 1 -2 sterile wound clips. The mice are allowed to regain consciousness and after 1 hour are returned to the vivarium. The mice are checked daily. This procedure has been performed on 40 mice with no adverse reactions (i.e., all lived, no infections, all expressed the transferred gene).
EXAMPLE 12
Analysis of Data
All data deri ved from experiments containing multiple groups are analyzed by multivariate analysis of variance (MANOVA) with post hoc testing (Duncan's multiple range test or Student-Newman-Keuls multiple range test). Significance is assessed at the p < 0.05 level of confidence in all cases. All data are analyzed using the NWA Statpak software (Northwest Analytical, Inc . , Portland, OR) running on a laboratory PC. Comparisons between two groups only are performed using the Student's T test.
EXAMPLE 12
The detailed sequences of the plasmids used herein are provided in Figures 7 A & 7B . SmGST3 plasmids in their bacterial host have been frozen down as glycerol stocks.
EXAMPLE 13
Development of cellular immune responses of mice vaccinated with naked DNA encodi n g Sch istosoma manson i glutathione S- tran sferase Two measures of T cell-mediated immunity (CMI) are used. An in vitro correlate of T cell clonal expansion, namely the lymphocyte proliferation assay in response to specific antigen with dividing cells allowed to incorporate ^ pr.j jR int0 newly synthesized DNA, is the primary assay. To demonstrate that naked DNA vaccination stimulates a T cell-mediated immune response groups of 15 mice (5 for each time period) are established as follows: Negative Control Groups -- 1 ) untreated; 2) irrelevant pCMV (a plasmid with no gene inserted in the downstream polycloning site and therefore not expressing any gene product); Positive Control group — to make sure that there is one positive T cell response, this group is vaccinated with pRSV-LacZ (a plasmid encoding the E. coli β -gal actosidase enzyme) and the T cells stimulated with the commercially available enzyme.
Three groups of mice are set up as follows: one group each is vaccinated with 10 μg, 50 μg or 100 μg of pCMV-SmGST3. Five mice from each treatment group are sacrificed on days 7, 14 and 28 after vaccination and spleen cells are removed and used in the lymphocyte proliferation assay . Lymphocytes from each animal's spleen are tested separately in a microwell assay requiring only 2.5 X 10^ cells (average spleen contains 5-7 X 10^ cells). Spleen cells are exposed to either concanavalin A (a known T cell mitogen), E. coli β-galactosidase, SmGST3, or maintained in complete medium only. After 4.67 days of culture, the cells are labelled with 0.5 μCi of ^ H-TdR (specific activity: 2 Ci/mmol) and the cells harvested 8 hours later onto glass filter strips using an automated cell harvester. The radioactivity on individual filters are measured by liquid scintillation spectroscopy. All treatment groups are setup and tested in triplicate.
To verify that the response seen in the lymphocyte proliferation assay is T cell-dependent, one aliquot of cells from each mouse is treated with anti-Thy 1 and complement (to destroy T cells) (35) and the remaining cells exposed to the stimulants. This treatment should abolish isotope incorporation if T cells mediate the response. I I significant lymphocyte proliferation is found following vaccination with DNA encoding SmGST3, the media from the proliferation cultures is assayed using Quantikine kits from Genzyme Corporation (Cambridge, MA) specific for mouse IFN-γ and IL-5 to ascertain whether vaccination elicited a Thl or
Th2 phenotypic response.
The second measure of CMI used is granuloma formation around beads coated with specific antigen. This is an in vivo correlate of the histopathological response that occurs in the liver of infected individuals in response to deposition of eggs of the parasite. Groups of mice are set up in an identical fashion as described above for the lymphocyte proliferation assay but they will not be the same mice. At 7, 14 and 28 days after naked DNA vaccination, 5 ,000 Sepharose 4B beads with covalently attached SmGST3 are injected into the lateral tail vein of mice. These beads embolize in the microvasculature of the lungs and if the animal has T cells which recognize the SmGST3 on the bead's surface, granulomatous inflammation will ensue. On days 13, 20, and 34
post vaccination (1 e., 6 days after bead embo zation) the mice are sacrificed, the lungs perfused with 10% buffered neutral formalin, and histological sections stained with hematoxy n and eosin prepared. Twenty five granulomas in each lung are measured in 2 perpendiculai p l ane s with a filar micrometer and an approximation oi the cross sectional area calculated by multiplying the two measures together (as an indication of the degree of floridness of the reaction). To account for sections through the edges of the bead, the cross sectional area of the bead are subtracted from the areas of the granulomas Using granuloma area of the untreated mice harboring SmGST3 coated beads as the denominator, a Granuloma Index for all other groups of mice are calculated using the formula GI = ( [Test Grp-Untreated Grp]/Untreated Grp) Besides measuring the approximate area of the granulomatous l eaction, the types of cells found in the reaction are noted. If the animal does not recognize the SmGST3 on the beads, a foreign body reaction occurs in which the beads are surrounded by only a few cells most of which are fibroblasts. If the SmGST3 antigen is recognized, the reaction consists of several hundred cells and the predominant types are macrophages, lymphocytes and eosinophils. Granulomas are being measured in the lung because the granulomatous response is a systemic manifestation oi cell-mediated immunity and the reactions occuring in the lung around beads are more circumscribed and therefore easiei to measure and analyze than in liver. This experimental pai adigm has been used in studies of T cell immunopathological mechanisms in muπne toxocariasis (24, 25).
In schistosomiasis mansoni, T cell-mediated immunity has been implicated in two major aspects of the disease process. The mechanism of protection against infection following immunization by pπoi exposure to parasite antιgen(s) and the granulomatous response to eggs which accumulate in the host tissues both have been shown to be T cell-dependent reactions. To date the best resistance against S. mansoni has come from the use of irradiated cercana but such material is too inflammatory to use in humans. Howevei , dissection of the immune responses elicited by vaccination of mice with irradiated cercana indicates that the protective response is dependent on T and B lymphocyte function,
but independent of complement, IgM or IgE-mediated immediate hypersensitivity (36). Mice vaccinated one time with irradiated cercaria require the presence of CD4+ T cells and an antibody and macrophages. The macrophages are activated by the lymphokine interferon-γ (IFN-γ) which is produced by CD4+ T helper cells of the 1 st subset type ( designated Th l cells, which also secrete IL-2 and lymphotoxin). Depletion of cells of the Th2 subset, which produce IL-4 (mediating IgE production), IL-5 (mediating eosinophilia) and IL- 10 (which down regulates Th l cells) does not decrease resistance induced by irradiated cercaria. T cells from mice immunized one time with irradiated cercaria proliferated and produ c ed I F N -γ upon subsequent chal lenge with S . ma n son i antigens during in vitro culture (37) and this finding forms a basis for the use of the lymphocyte proliferation assay to detect a response to naked DNA vaccination using a plasmid encoding S. mansoni GST3. SmGST3 been shown to be a protective antigen when the recombinant protein mixed with adjuvant was used to immunize mice, rats and primates against challenge infection (38). If T cell proliferation is seen, are the Thl vs Th2 cytokine assays used. Because naked DNA vaccination has been used to vaccinate laboratory and agricultural animals against several viral i nfections where Th l -mediated immunity is absolutely essential , immune interferon gamma (IFN-γ) as indicative of a Th l response and interleukin 5 (IL-5 ) as representative of a Th2-mediated response is quantified. While it is expected that the naked DNA vaccination will give a Thl response, knowing whether or not it does, permits a modification of the vaccine protocol wherein a plasmid encoding IL-12 which drives ThO cells to become Thl cells, can be incorporated into the antigen-encoding DNA to yield a multigenic vaccine.
Granuloma formation has been found to be a T cell- mediated process (39) and, until recently, considered to be a Thl phenomenon. This conclusion was based in part on the observation that an intradermal injection of S. mansoni egg antigens into the footpad of infected mice elicited a delayed swelling and concomitant production of IFN-γ. However, administration of anti- IFN- monoclonal antibody (mAb) did not have any effect on the size or cellular composition of egg granulomas in the liver while a
comparable treatment with mAb against IL-5 abrogated bone marrow, peripheral blood and granuloma eosinophils in infected mice (33). These experiments were done on acutely infected mice at a time when the granulomatous response is maximal, and so too, are eosinophilia and IgE levels. These later responses depend on Th2 lymphokines (33). This suggests that Th2, and not Thl , cells are mediating granuloma formation in acute schistosomiasis. From the perspective of vaccine development, granuloma formation represents a quantifiable aspect of host response to the parasite (i.e, number of eggs per gram liver). If vaccination can reduce the number of eggs released per female worm (as measured by the number of granulomas around eggs in the livers) then the vaccinated population would be putting out fewer eggs into the environment and this should have a major effect on the subsequent transmission of the disease.
EXAMPLE 14
Development of humoral immune responses of mice vaccinated with naked DNA encoding Schistosoma mansoni glutathione S- transferase
In order to measure anti-SmGST3 antibody, peripheral blood sera are collected from each mouse prior to being randomized into a treatment group and then again immediately prior to being infected. To determine whether mice are making anti-SmGST3 antibody, ELISA grade microwell plates are coated with purified S. mansoni glutathione S-transferase and used in an ELISA assay. Briefly, the ELISA assay works as follows: Microwell plates are coated with specific antigen (SmGST3) and nonspecific binding sites blocked with excess milk protein. Sera to be tested are serially diluted and added to the wells. Subsequently, unbound sera is washed away, and an antibody against mouse immunoglobulin is added to detect any mouse antibodies that bound to the fixed antigen. This latter antibody is conjugated to alkaline phosphatase and when appropriate chromogenic substrate for the enzyme is added to the well, a colored product is produced which can be quantitated at a specific wavelength using an immunoassay microplate (ELISA) reader. To make this assay semiquantitative, a standard curve of increasing concentrations of
GST is established in 6-8 wells and a reference polyclonal rabbit serum used in place of the mouse sera. Thus, the present invention determines if vaccination elicits a specific antibody response and the relationship of this response to the amount of DNA administered and the length of time after administration.
Because SmGST3 is an invertebrate GST with immunologically different properties (speaking strictly from the amino acid sequence point of view) the human or rodent host would be expected to recognize the protein as a foreign one and therefore, SmGST3 should be immunogenic and elicit an antibody response. Mice and rats which have been passively administered a mAb against SmGST3 and then challenged with infective cercaria were found to have reduced worm burdens, the female worms released fewer eggs, and the eggs that were released had poorer viability than worms recovered from mice not receiving the mAb
(41). These properties of a vaccine would go a long way towards disrupting the transmission of this disease by reducing the infective egg burden released into the water. This in turn would protect the snail population from participating in the obligatory asexual phase of the life cycle. Additionally, it is the egg deposited in the liver that elicits the granulomatous response and it is this very response, which serves to sequester S. mansoni- d eτi v ed hepatocellular toxins, that ultimately destroys the liver, and generates the portal hypertension and potentially lethal esophageal varices. Fewer eggs would ameliorate this aspect of the infection. These are properties that increase the value of targeting S. mansoni GST with a vaccine. Also, the presence of anti-SmGST3 antibodies in the serum validates the expression of the polynucleotide injected and gives a clinical basis for knowing a priori whether a person was in fact, immunized.
EXAMPLE 15
Effect of the H-2 haplotype on immune responses of mice vaccinated with naked DNA encoding S. mansoni glutathione S- transferase
Mice with different major histocompatibility complex (MHC) H-2 haplotypes are given a single dose of naked SmGST3
plasmid DNA and all the tests are conducted at the single time after vaccination for which the responses were maximal. The following strai ns of mice (with their H-2 haplotypes in parentheses ) used : C57BL/6 (H-2b)--this mouse strain is considered a high responder strain (42, 43); CBA (H-2k)— this strain is considered a moderate to low responder strain (42, 43); SJL (H-2s)--this strain exhibits high levels of eosinophils in response to tissue invasive helminth infections but makes virtually no IgE antibody in these infections (44); and P strain (H-2p), the only mouse strain known to not develop resistance to infection with S. mansoni (45). Mice are either untreated or vaccinated with 10, 50, or 100 μg pCMV-SmGST3 DNA. At the time of maximal response mice are bled for serum and sacrificed. In those mice receiving SmGST3-coated beads, the lungs are perfused and processed for histological examination. The spleens of the remaining mice are used to establish individual lymphocyte proliferation assays stimulated with either the T cell mitogen, Con A, or the specific antigen, SmGST3. The sera collected is tested in the SmGST3 ELISA to determine relative antibody levels. Humans do not respond to antigenic stimulation as uniformly as do inbred strains of mice. While most inbred strains that have been studied respond to vaccination with irradiated cercaria by developing moderate to high levels of protective immunity against a challenge infection, the P strain always fails to do so following vaccination (45). Peak levels of IgG to larval schistosome surface antigens produced by P strain mice are equivalent to those in high responder strains of mice and are of the same isotypes which suggests that antibody titers alone are not what determines protective immunity. However, antibody against SmGST3 may decrease transmission by reducing fecundity and egg viability.
EXAMPLE 16
Production of Baculovirus Expression Vector for Production of Recombinant SmGST-3 Protein in Insect Cells
In order to determine whether a vaccination "takes" and to evaluate the relative strength of the immune responses of
animals vaccinated with polynucleotides, ample quantity of the protein which is encoded by the DNA used to vaccinate was necessary. Isolation of the natural protein from the parasite itself is tedious, time consuming, expensive, and difficult. The use of gene transfer technology can obviate this difficulty by inserting the gene for the protein into a virus which can infect insect cells that can be maintained in tissue culture. Such cells can be lysed and the protein easily purified. The spent culture medium can be used as a source of new virus to infect subsequent cultures of insect cells and thus, an unlimited quantity of recombinant protein can be obtained for use in immunoassays. The production of a Baculovirus expression vector which encodes the 16.4 SmGST-3 insert is descri bed. To facilitate the purification of the recombinant protein, a 6X his(tidine) tag was inserted at the 5' end of the coding sequence which permits metal affinity isolation using a Ni-NTA matrix.
EXAMPLE 17
Construction of A Baculovirus expressing the SmGST-3 ( 16.41 cDNA
The Baculovirus Expression Vector System (GIBCO-BRL Cat. #10584-027) was used per the vendors instructions. A set of custom po lymeras e chain reaction (PCR) primers were commercially synthesized to the 5' and 3' ends of the cDNA encoding the S. mansoni GST-3 contained in the pBsCMV- 16.4 plasmid. The primers were designed so that an EcoR I restriction site was inserted 5' to the ATG start codon while a Hind III site was added immediately after the stop codon in the open reading frame. The final construct uses an ATG start codon located 5' to a 6X his(tidine) tag and aligned in such a manner that the inserted
PCR product was maintained in the open reading frame. The resulting 65 1 bp amplified fragment was then ligated into the pFastBac HTb shuttle vector which had been linearized by cutting with EcoRI and Hind III. The ligated product was next transformed into DH5 α F' competent cells and amplified. The plasmid DNA was isolated using a Qiagen Spin Prep column. Following visualization on a an agarose gel, 1 ng of plasmid DNA was used to transform DH l OBac cells. These cells contain a bacmid
with a mini-attTn7 target site and a helper plasmid. The mini-Tn7 element on the pFastBac plasmid can transpose to the mini-attTn7 target site on the bacmid in the presence of transposition proteins provided by the helper plasmid. The transposition was carried out by incubation on LB plates containing kanamycin (50 μ g/m l ; gentamicin (7 μg/ml ); tetracycline (10 μg/ml), and 100 μg/ml Bluo- gal plus 40 μg/ml IPTG. White colonies containing the recombinant bacmid were identified by blue-white screening. High molecular weight DNA was isolated by alkaline lysis from mini prep cultures. Finally, 5 μg of bacmid DNA containing the SmGST-3 cDNA insert was used to transfcct Sf21 insect ovary cells in the presence of CellFECTIN. The culture medium from this transfection was collected to use as a source of Baculovirus-SmGST-3 (Bac-SmGST-3) pending verification that the Sf21 cells were producing the S . mansoni glutathione S-transferase .
EXAMPLE 18
Screening of Sf2 1 Cells for Production of SmGST-3 Recombinant Protein frSmGST-3 )
At either 3, 4, or 5 days after infecting Sf21 cells with Bac-SmGST-3, the cells were lysed with 5X lysis buffer (50 mM Tris, pH 7.5 ; 650 mM NaCl; 5% Triton X- 100; 50 mM NaF, 50 mM Na2HPO4; and 50 mM Na pyrophosphate). The proteinase inhibitor, PMSF ( 100 mM stock in absolute ethanol), was added to the lysis buffer i mmediately prior to use (5 mM final concentration). The lysis product was centrifuged at 17100 X g for 10 min at room temperature and the soluble phase separated from the pellet. The pellet and supernatant were separated on a 12.5% SDS-PAGE gel and the resulting isolated proteins transblotted to nitrocellulose for immunoblotting. The nitrocellulose membranes were blocked with 3 % nonfat dry milk solution, and then stained with a 1 : 160,000 dilution of a rabbit anti-S. mansoni GST-3 polyclonal antibody (provided by Dr. James Tracy, U. Wise. Vet School). The membranes were washed and a goat anti-rabbit IgG conjugated to horseradish peroxidase (Sigma Chemical Co., St. Louis, MO) diluted 1 : 10.000 was applied. Following several additional washes, the bands were visualized using the ECL chemiluminescent
kit from Amersham. The chemiluminescent signal was collected on X-ray film and following development, the Western blot image as well as the pretransfer, Coomassie Brilliant Blue-stained replicate SDS-PAGE gel were scanned into computer storage using an HP ScanJet IICX at identical scale to facilitate alignment. The results are shown below as Figure 8.
The immunodetection of a strong signal at 28 kDa is proof that the Baculovirus-infected cells are indeed producing the SmGST-3 protein. It is noteworthy that little, if any, of the protein is in the pellet fraction and little, if any, is in the extracellular medium. Having verified that the cells can produce the recombinant protein, it one can purify the protein to near homogeneity by passing the lysis supernatant over a Ni-NTA column. The Ni matrix has a strong affinity for the 6X his tag which was cloned 5' to the SmGST-3 molecule.
EXAMPLE 19
Purification of 6X-his tagged rSmGST-3 Using Ni-NTA Metal Affinity Chromatography
Ni-NTA was purchased from Qiagen (Chatsworth, CA) and used as per the vendor's instructions. The gel slurry was washed in Buffer A and incubated overnight with 1 ml of Sf21 cell extract (i.e., the resulting lysis supernatant). Next the unbound material was washed away by pelleting the slurry and removing the supernatant (which was retained for subsequent analysis and to verify that the 28 kDa band was removed). Following several washes in Buffer A (containing 20 mM imidazole) to remove weakly binding products from the gel, the slurry was transferred to a mini column and washed again, in Buffer B followed by another wash with Buffer A. Finally, the adherent protein was eluted by washing the column with Buffer C containing 250 mM imidazole and 2 ml fractions collected. The presence or absence of the immunoreactive 28 kDa protein in each of the fractions was again confirmed by SDS-PAGE and chemiluminescent western blotting.
The results of one such experiment are shown in Figure 9 which suggests that the protein is being recovered in fractions 4
through 12. These fractions were next pooled, dialyzed against phosphate buffered saline and concentrated using an Amicon Centriprep with a nominal Molecular Weight Cutoff of 10 kDa. Figure 10 depicts a chemiluminescent Western blot indicating that the protein was concentrated and retained immunoreactivity. Now 100 to 200 μg of rSmGST-3 from a single T-75 flask of Sf21 cells infected at 80 to 85 confluency can be produced routinely and harvested after 3 days of infection. The resulting protein was next used to skin test mice which had been intramuscularly vaccinated 2 weeks previously.
EXAMPLE 20
Skin Testing of Mice Vaccinated with pBsCMV- 16.4 DNA Using the Recombinant GST Protein
One test used to evaluate the immune status of vaccinated mice is a simple skin test in which an antigen used to induce the vaccinated state is subsequently injected intradermally to see if the foreign protein is recognized. The end point of the skin test is the quantification of the inflammation (here taken as increased thickness which represents the swelling due to the accumulation of an inflammatory infiltrate) present 24 hr after the skin test is begun. The significance of the duration of the test over a twenty four hour period is that inflammation occurring during the first six hours is considered to be indicative of the immediate hypersensiti ve state (mediated by IgE and mast cells) while inflammation occurring at 24 hours represents the delayed hypersensiti ve state . Delayed hypersensitive responses are manifestations of cell-mediated immunity and are the desired outcome of any vaccination against an infectious agent such as
Mycobacterium tuberculosis or S. mansoni.
To evaluate whether mice vaccinated with naked DNA encoding an S. mansoni-deri ved gene manifest cell-mediated immunity, groups of mice were vaccinated with either pBsCMV- 16.4 DNA (specific lest) or pBsRSV-lux (nonspecific, irrelevant DNA encoding firefly luciferase). Fourteen days after vaccination the thickness of the outer ear was measured with a precision pocket micrometer (Mi tutoyo Instruments #7309) just prior to the
intradermal injection of approximately 30 μl of 100 μg/ml of recombinant SmGST3 protein in buffered saline. The ear was measured again at twenty four hours and specific ear thickening (SET) in micrometers was calculated as: SET = (Thickness at 24 hr minus Thickness at 0 hr)
To interpret the data, the SET values of mice vaccinated with pBsCMV-16.4 DNA were compared to the SET values of mice vaccinated with pBsRSV-lux. The results of a typical experiment are shown in the table below.
Vaccine DNA no of mice SET Value±SD p value*
50 μg 16 3 73.3 ± 5.8 < 0.05
100 μg 16.4 3 76.7 ± 15.3 <0.05 lux 3 46.7 ± 15.3
* single tail Students t test comparing 16.4 DNA vs. lux value
These results indicate that mice vaccinated with the pBsCMV-16.4 DNA and skin tested 14 days later have statistically significant cell mediated immunity against the SmGST-3 antigen. The following references were cited herein:
1. Wang, B., et al., DNA-Cell Biol. 12:799-805, (1993).
2. Barr, E., et al., Science 254:1507-1509, (1991).
3. Dhawan, J., et al.. Science 254:1509-1512, (1991). 4. Wolff, J. A., et al., Science 247:1465-1468, (1990).
5. Wolff, J. A., et al., Human Mol. Genet. 1:363-369, (1992).
6. Wolff, J. A., et al., Biotechniques 11:474-485, (1991).
7. Wolff, J. A., et al., J. Cell Sci. 103:1249-1259, (1992).
8. Fynan, E. F., et al., DNA-Cell Biol.12:785-789, (1993). 9. Montgomery, D. L., et al., DNA-Cell Biol. 12:777-783, (1993).
10. Ulmer, J. B., et al.. Science 259:1745-1749, (1993).
11. Tang, D-C, et al.. Nature 356:152-154, (1992).
12. Yankauckas, M. A., et al., DNA-Cell Biol. 12:771-776, (1993).
13. Wang, B.. et al., Proc. Natl. Acad. Sci. USA 90:4156-4160, (1993).
14. Davis, H. L., et al., Hum. Gene Ther.4:151-159, (1993).
15. Danko, I., et al.. Gene Ther. 1:1-8. (1993).
16. Mannervik, B. In: Adv. in Enzymy. Meister, A., Ed. Vol. 57:357-
417. Wiley, New York, NY., (1985).
17. Jaffe, J. J., et al.. Mol. Biochem. Parasitol.20:199-206, (1986). 18. O'Leary, K. A., et al., Exp. Parasitol.75:47-55, 1992).
19. O'Leary, K. A., et al., Arch. Biochem. Biophys.264:1-12, (1988).
20. Habig, W. H., et al., Meth. Enzymol. 77:398-405, (1981).
21. Kayes, S. G., et al., J. Immunol. 122:2340-2344, (1979).
22. Kayes, S. G., el al., Cell Immunol. 83:152-160, (1984). 23. Kayes, S. G., et al., Cell Immunol. 83:161-169, (1984).
24. Kayes, S. G., Clin. Immunol. Immunopathol. 41:55-65, (1986).
25. Kayes, S. G., et al., J. Parasitol.74:950-956, (1988).
26. Kayes, S. G., el al., Infect. Immun.48:697-703, (1985).
27. Danko, I., et al.. Gene Therapy 1:1-8, (1993). 28. Cox, G. J., et al.. J. Virol.67:5664-5667, (1993).
29. Smith, D. B., et al., Proc. Natl. Acad. Sci. USA 83:8703-8708, (1986).
30. Mitchell, G. F., In Progress in Immunology (VI), Cinader, B., and
G. Miller, Eds.; p.798, Academic Press, FL.,(1986).
31. Balloul. J. M., ci al., Nature 326:149-153, (1987).
32. Taylor, J. B., et al.. EMBO J.7:465-472, (1988).
33. James et al., Curr. Top. Microbiol. Immunol. 155:21-31, (1990).
34. Boros, D. L., ct al., J. Immunol. 110:1118-1128, (1973). 35. Kayes, S. G., J. Parasitol.70:522-529, (1984).
36. Sher, A., et al.. J. Immunol. 128:1880-1884, (1982).
37. James, S. L., et al., Infect. Immun.44:569-575, (1984).
38. Capron, A., et al.. Annu. Rev. Med. 43:209-218, (1992).
39. Warren, K. S., el al.. Am. J. Pathol.51:735-745, (1967). 40. Jones, R. E., et al.. Exp. Parasitol.78:64-75, (1994).
41. Xu, C.-B., et al.. Eur. J. Immunol. 21:1801-1807, (1991).
42. Richter, D., et al., Infect. Immun. 61:3003-3011, (1993).
43. Richter, D., et al.. Infect. Immun. 61:146-154, (1993).
44. Watanabe et al.. Proc. Natl. Acad. Sci. USA 85:4460-4464, (1988).
45. James, S. L.. et al.. J. Immunol. 133:1587-1593, (1984).
Claims
1. A vaccine comprising a non-infectious, non- integrating DNA sequence encoding Schistosoma mansoni glutathione S-transferase.
2. The DNA sequence of claim 1, wherein said DNA comprises a plasmid encoding a protein, polypeptide or peptide which is operably linked to a promoter.
3. The DNA sequence of claim 1, wherein said plasmid is pCMVSmGST-3.
4. The DNA sequence of claim 1, wherein said plasmid is selected from the group consisting of pBsCMV-16.4 DNA and pRc/ASK8-SmGST3.
5. A method of protecting against infection by Schistosoma mansoni comprising the step of: administering to an animal in need of such treatment a pharmacologically effective dose of a vaccine composition comprising a non-infectious, non-integrating DNA sequence encoding Schistosoma mansoni glutathione S-transferase.
6. The method of claim 5, wherein said vaccine composition comprises a plasmid encoding a protein, polypeptide or peptide which is operably linked to a promoter.
7. The method of claim 6, wherein said plasmid is pCMVSmGST-3.
8. The method of claim 6, wherein said plasmid is selected from the group consisting of pBsCMV-16.4 DNA and pRc/ASK8-SmGST3.
9. The method of claim 6, wherein said plasmid is administered in an animal in a dose of from about 25 μg to about 200 μg .
1 0. The method of claim 6, wherein said plasmid is administered into muscle.
1 1 . The method of claim 6, wherein said plasmid is administered between 1 and 6 times.
1 2. A method of developing cell mediated immunity to glutathione-S-transferase of Schistosoma mansoni comprising the step of: administering to an animal in need of such treatment a immunologically effective dose of the plasmid of claim 6.
1 3. The method of claim 12, wherein said plasmid is pCMVSmGST-3.
14. The method of claim 12, wherein said plasmid is pBsCMV- 16.4 DNA.
1 5 . The method of claim 12, wherein said plasmid is administered in an animal in a dose of from about 25 μg to about 200 μg .
1 6. The method of claim 12, wherein said plasmid is administered into m uscle.
1 7. The method of claim 12, wherein said plasmid is administered between 1 and 6 times.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU20786/97A AU2078697A (en) | 1996-03-13 | 1997-03-13 | Dna encoding 28 kda glutathione s-transferase of schistosoma mansoni and uses thereof |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US1332196P | 1996-03-13 | 1996-03-13 | |
US60/013,321 | 1996-03-13 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO1997033610A1 true WO1997033610A1 (en) | 1997-09-18 |
WO1997033610A9 WO1997033610A9 (en) | 1997-11-20 |
Family
ID=21759366
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US1997/003977 WO1997033610A1 (en) | 1996-03-13 | 1997-03-13 | DNA ENCODING 28 kDa GLUTATHIONE S-TRANSFERASE OF SCHISTOSOMA MANSONI AND USES THEREOF |
Country Status (2)
Country | Link |
---|---|
AU (1) | AU2078697A (en) |
WO (1) | WO1997033610A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP1074612A1 (en) * | 1999-08-04 | 2001-02-07 | Pasteur Merieux Serums Et Vaccins | Use of vegetal peptones for DNA vaccine production |
US8734807B1 (en) | 2013-04-06 | 2014-05-27 | Gabriel Langlois-Rahme | Preventing and curing Schistosomiasis mansoni by inhibiting Trk receptors on female Schistosoma |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5583011A (en) * | 1994-07-25 | 1996-12-10 | Board Of Trustees Of The Leland Stanford University | Compositions, treatments, and diagnostics for Schistosomiasis and related diseases |
-
1997
- 1997-03-13 WO PCT/US1997/003977 patent/WO1997033610A1/en active Application Filing
- 1997-03-13 AU AU20786/97A patent/AU2078697A/en not_active Abandoned
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5583011A (en) * | 1994-07-25 | 1996-12-10 | Board Of Trustees Of The Leland Stanford University | Compositions, treatments, and diagnostics for Schistosomiasis and related diseases |
Non-Patent Citations (6)
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP1074612A1 (en) * | 1999-08-04 | 2001-02-07 | Pasteur Merieux Serums Et Vaccins | Use of vegetal peptones for DNA vaccine production |
US8734807B1 (en) | 2013-04-06 | 2014-05-27 | Gabriel Langlois-Rahme | Preventing and curing Schistosomiasis mansoni by inhibiting Trk receptors on female Schistosoma |
Also Published As
Publication number | Publication date |
---|---|
AU2078697A (en) | 1997-10-01 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Estunningsih et al. | Evaluation of antigens of Fasciola gigantica as vaccines against tropical fasciolosis in cattle | |
DE3856586T2 (en) | Respiratory syncytial virus, vaccines and diagnostic assays | |
Darcy et al. | Induction of a protective antibody‐dependent response against toxoplasmosis by in vitro excreted/secreted antigens from tachyzoites of Toxoplasma gondii | |
KR970011149B1 (en) | Recombinant apox virus | |
CA1340538C (en) | Recombinant coccidiosis vaccines | |
EP1383795B1 (en) | A recombinant newcastle disease virus nucleoprotein mutant as a marker vaccine | |
KR20000065266A (en) | Recombinant Protein Containing C-terminal Fragment of Malaria Protozoal MSP-1 | |
AU2002224815A1 (en) | A recombinant newcastle disease virus nucleoprotein mutant as a marker vaccine | |
US20110159024A1 (en) | Subunits of the adenovirus fiber protein and uses thereof as vaccines | |
Osorio et al. | Recombinant raccoon pox vaccine protects mice against lethal plague | |
US9228002B2 (en) | Leishmania vaccine using sand fly salivary immunogen | |
JP3450018B2 (en) | Coccidiosis vaccine | |
WO1997033610A1 (en) | DNA ENCODING 28 kDa GLUTATHIONE S-TRANSFERASE OF SCHISTOSOMA MANSONI AND USES THEREOF | |
WO1996032479A1 (en) | Hookworm vaccine | |
WO1997033610A9 (en) | DNA ENCODING 28 kDa GLUTATHIONE S-TRANSFERASE OF SCHISTOSOMA MANSONI AND USES THEREOF | |
CA2256710A1 (en) | Vaccine containing a peroxiredoxin and/or a .beta.-tubulin | |
AU676828B2 (en) | Antigens protective against (echinococcus granulosus) infection and vaccines containing such antigens | |
DE69132404T2 (en) | VARICELLA ZOSTER VIRUSANTIGEN | |
Arasu et al. | Brugia malayi: recombinant antigens expressed by genomic DNA clones | |
US5599692A (en) | Antigenic polypeptides of Taenia ovis | |
US5492695A (en) | Vaccinating cats against Dirofilaria immitis with an L4 homogenate | |
JP3428666B2 (en) | Recombinant Marek's disease virus and its production | |
WO1999025826A1 (en) | Method and kit for determining severe inflammatory reactions to mosquito bites | |
Wilson et al. | The development of a schistosome vaccine. | |
US20040156866A1 (en) | Vaccine to protect animals against leishmania |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A1 Designated state(s): AU BR CA |
|
AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE |
|
COP | Corrected version of pamphlet |
Free format text: PAGES 1/17-17/17,DRAWINGS,REPLACED BY NEW PAGES 1/21-21/21;DUE TO LATE TRANSMITTAL BY THE RECEIVINGOFFICE |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
NENP | Non-entry into the national phase |
Ref country code: CA |
|
122 | Ep: pct application non-entry in european phase |