CN113249223A - Application of leishmania transformed with expression plasmid in promotion of DC maturation, method for promoting DC maturation, and expression plasmid - Google Patents
Application of leishmania transformed with expression plasmid in promotion of DC maturation, method for promoting DC maturation, and expression plasmid Download PDFInfo
- Publication number
- CN113249223A CN113249223A CN202110521400.7A CN202110521400A CN113249223A CN 113249223 A CN113249223 A CN 113249223A CN 202110521400 A CN202110521400 A CN 202110521400A CN 113249223 A CN113249223 A CN 113249223A
- Authority
- CN
- China
- Prior art keywords
- expression plasmid
- leishmania
- maturation
- tumor
- eno1
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 239000013613 expression plasmid Substances 0.000 title claims abstract description 77
- 241000222722 Leishmania <genus> Species 0.000 title claims abstract description 65
- 230000035800 maturation Effects 0.000 title claims abstract description 39
- 238000000034 method Methods 0.000 title claims abstract description 18
- 230000001737 promoting effect Effects 0.000 title claims description 15
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 51
- 108091026890 Coding region Proteins 0.000 claims abstract description 36
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 30
- 239000000427 antigen Substances 0.000 claims abstract description 25
- 108091007433 antigens Proteins 0.000 claims abstract description 25
- 102000036639 antigens Human genes 0.000 claims abstract description 25
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 25
- 102000004196 processed proteins & peptides Human genes 0.000 claims abstract description 24
- 229920001184 polypeptide Polymers 0.000 claims abstract description 23
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 23
- 102100038910 Alpha-enolase Human genes 0.000 claims description 38
- 101000882335 Homo sapiens Alpha-enolase Proteins 0.000 claims description 38
- VLMZMRDOMOGGFA-WDBKCZKBSA-N festuclavine Chemical compound C1=CC([C@H]2C[C@H](CN(C)[C@@H]2C2)C)=C3C2=CNC3=C1 VLMZMRDOMOGGFA-WDBKCZKBSA-N 0.000 claims description 38
- 101001133056 Homo sapiens Mucin-1 Proteins 0.000 claims description 20
- 102100034256 Mucin-1 Human genes 0.000 claims description 20
- 101100346932 Mus musculus Muc1 gene Proteins 0.000 claims description 7
- 230000001131 transforming effect Effects 0.000 claims description 6
- 210000000987 immune system Anatomy 0.000 abstract description 12
- 210000004881 tumor cell Anatomy 0.000 abstract description 11
- 230000000694 effects Effects 0.000 abstract description 8
- 230000002708 enhancing effect Effects 0.000 abstract description 4
- 230000008685 targeting Effects 0.000 abstract 1
- 239000013612 plasmid Substances 0.000 description 22
- 210000004443 dendritic cell Anatomy 0.000 description 16
- 208000015181 infectious disease Diseases 0.000 description 11
- 108020004414 DNA Proteins 0.000 description 10
- 230000003213 activating effect Effects 0.000 description 10
- 230000015572 biosynthetic process Effects 0.000 description 10
- 238000003786 synthesis reaction Methods 0.000 description 10
- 238000011529 RT qPCR Methods 0.000 description 9
- 210000001744 T-lymphocyte Anatomy 0.000 description 7
- 210000004027 cell Anatomy 0.000 description 6
- 238000001502 gel electrophoresis Methods 0.000 description 6
- 238000000684 flow cytometry Methods 0.000 description 5
- 230000002757 inflammatory effect Effects 0.000 description 5
- YJQCOFNZVFGCAF-UHFFFAOYSA-N Tunicamycin II Natural products O1C(CC(O)C2C(C(O)C(O2)N2C(NC(=O)C=C2)=O)O)C(O)C(O)C(NC(=O)C=CCCCCCCCCC(C)C)C1OC1OC(CO)C(O)C(O)C1NC(C)=O YJQCOFNZVFGCAF-UHFFFAOYSA-N 0.000 description 4
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 4
- 229960000723 ampicillin Drugs 0.000 description 4
- 238000003776 cleavage reaction Methods 0.000 description 4
- 238000010276 construction Methods 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- 230000007017 scission Effects 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- ZHSGGJXRNHWHRS-VIDYELAYSA-N tunicamycin Chemical compound O([C@H]1[C@@H]([C@H]([C@@H](O)[C@@H](CC(O)[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C(NC(=O)C=C2)=O)O)O1)O)NC(=O)/C=C/CC(C)C)[C@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1NC(C)=O ZHSGGJXRNHWHRS-VIDYELAYSA-N 0.000 description 4
- MEYZYGMYMLNUHJ-UHFFFAOYSA-N tunicamycin Natural products CC(C)CCCCCCCCCC=CC(=O)NC1C(O)C(O)C(CC(O)C2OC(C(O)C2O)N3C=CC(=O)NC3=O)OC1OC4OC(CO)C(O)C(O)C4NC(=O)C MEYZYGMYMLNUHJ-UHFFFAOYSA-N 0.000 description 4
- 238000011144 upstream manufacturing Methods 0.000 description 4
- 241000894006 Bacteria Species 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- 108010074328 Interferon-gamma Proteins 0.000 description 3
- 241000222727 Leishmania donovani Species 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 238000012258 culturing Methods 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 238000001976 enzyme digestion Methods 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 239000013598 vector Substances 0.000 description 3
- 238000001262 western blot Methods 0.000 description 3
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- 102100032202 Cornulin Human genes 0.000 description 2
- 101000920981 Homo sapiens Cornulin Proteins 0.000 description 2
- 102100037850 Interferon gamma Human genes 0.000 description 2
- 241000222724 Leishmania amazonensis Species 0.000 description 2
- 230000006052 T cell proliferation Effects 0.000 description 2
- 108010044965 UDP-N-acetylglucosamine-lysosomal-enzyme N-acetylglucosaminephosphotransferase Proteins 0.000 description 2
- 230000003698 anagen phase Effects 0.000 description 2
- 210000000612 antigen-presenting cell Anatomy 0.000 description 2
- 230000011748 cell maturation Effects 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 239000013604 expression vector Substances 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 239000001963 growth medium Substances 0.000 description 2
- 230000028993 immune response Effects 0.000 description 2
- 239000002609 medium 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
- 238000003753 real-time PCR Methods 0.000 description 2
- 230000003248 secreting effect Effects 0.000 description 2
- 238000012163 sequencing technique Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 101800000535 3C-like proteinase Proteins 0.000 description 1
- 101800002396 3C-like proteinase nsp5 Proteins 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 238000011740 C57BL/6 mouse Methods 0.000 description 1
- -1 CD86 Proteins 0.000 description 1
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- 101150015836 ENO1 gene Proteins 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 description 1
- 101710088172 HTH-type transcriptional regulator RipA Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 101000596741 Homo sapiens Testis-specific protein TEX28 Proteins 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 241000222740 Leishmania braziliensis Species 0.000 description 1
- 241000222697 Leishmania infantum Species 0.000 description 1
- 241000222732 Leishmania major Species 0.000 description 1
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 101000746372 Mus musculus Granulocyte-macrophage colony-stimulating factor Proteins 0.000 description 1
- 238000009004 PCR Kit Methods 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 239000002033 PVDF binder Substances 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 230000006044 T cell activation Effects 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- 102100035104 Testis-specific protein TEX28 Human genes 0.000 description 1
- 108091005906 Type I transmembrane proteins Proteins 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000001464 adherent effect Effects 0.000 description 1
- 238000000246 agarose gel electrophoresis Methods 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 230000005975 antitumor immune response Effects 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 208000036815 beta tubulin Diseases 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 238000002619 cancer immunotherapy Methods 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 230000000139 costimulatory effect Effects 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 230000036425 denaturation Effects 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 239000002158 endotoxin Substances 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 230000000762 glandular Effects 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 210000000777 hematopoietic system Anatomy 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 239000000203 mixture Substances 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 230000000242 pagocytic effect Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 238000012257 pre-denaturation Methods 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 239000003531 protein hydrolysate Substances 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 238000010814 radioimmunoprecipitation assay Methods 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 229920000785 ultra high molecular weight polyethylene Polymers 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4713—Autoimmune diseases, e.g. Insulin-dependent diabetes mellitus, multiple sclerosis, rheumathoid arthritis, systemic lupus erythematosus; Autoantigens
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4727—Mucins, e.g. human intestinal mucin
-
- 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
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
-
- 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
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0639—Dendritic cells, e.g. Langherhans cells in the epidermis
-
- 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
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/998—Proteins not provided for elsewhere
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Biotechnology (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- General Engineering & Computer Science (AREA)
- Biophysics (AREA)
- Toxicology (AREA)
- Microbiology (AREA)
- Gastroenterology & Hepatology (AREA)
- Medicinal Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Hematology (AREA)
- Physics & Mathematics (AREA)
- Diabetes (AREA)
- Plant Pathology (AREA)
- Rehabilitation Therapy (AREA)
- Rheumatology (AREA)
- Cell Biology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
The invention discloses application of Leishmania transformed with an expression plasmid in promotion of DC maturation, a DC maturation promotion method and the expression plasmid, wherein the expression plasmid contains at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence. Leishmania transformed with the expression plasmid can infect DC in a targeting way and promote the maturation of the DC, thereby enhancing the effect of killing tumor cells by an immune system of an organism.
Description
Technical Field
The invention relates to the technical field of tumor immunology, in particular to application of Leishmania transformed with an expression plasmid in promotion of DC maturation, a DC maturation promotion method and the expression plasmid.
Background
Most antigens need to be taken up, processed, internalized and presented by Antigen Presenting Cells (APCs) to be recognized by T cell receptors and thus promote T cell activation and proliferation. In recent years, Dendritic Cells (DCs) have been the focus of much attention in the field of tumor therapy as the most antigen-presenting APC, the only one that activates naive T cells. Most of DC in human body are in immature state, the DC in immature state expresses low level of costimulatory factor and adhesion factor, and the capability of inducing organism to generate immune response is low. However, immature DC have a very strong antigen phagocytic ability, and can be activated by inflammatory factors or internalized antigens, and the like, and become mature. The matured DCs express surface histocompatibility complex molecules (MHC-I/II) at a high level, and the taken antigen peptides are presented to T cells through MHC-I/II antigen presenting molecules, thereby activating the immune system of the body to kill tumor cells. Therefore, if immature DCs in the body are extracted to activate the maturation of the DCs, and then the mature DCs are returned to the body, the efficiency of the DCs for activating T cells is improved, and a stronger anti-tumor immune response is generated. However, the prior art has poor effect of activating DC maturation, thereby reducing the effect of DC activating immune system to kill tumor cells.
The above is only for the purpose of assisting understanding of the technical solutions of the present invention, and does not represent an admission that the above is the prior art.
Disclosure of Invention
The invention mainly aims to provide application of Leishmania transformed with expression plasmids in promotion of DC maturation, a DC maturation promotion method and expression plasmids, and aims to improve the effect of activating DC maturation and further enhance the effect of killing tumor cells by an immune system.
In order to achieve the above objects, the present invention provides, in a first aspect, the use of Leishmania transformed with an expression plasmid for promoting DC maturation.
Optionally, the expression plasmid comprises at least one of a tumor associated protein coding sequence, a tumor associated polypeptide coding sequence, a tumor specific antigen coding sequence.
Optionally, the expression plasmid comprises an ORF of ENO1 or an ORF of MUC 1.
Alternatively, the ORF sequence of ENO1 is as set forth in SEQ ID No: 1, the ORF sequence of the MUC1 is shown as SEQ ID No: 2, respectively.
In a second aspect, the present invention also provides a method for promoting DC maturation, comprising the steps of:
(1) transforming the expression plasmid into leishmania;
(2) infecting said leishmania infected DCs with said expression plasmid;
wherein, the expression plasmid contains at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence.
Optionally, the expression plasmid comprises an ORF of ENO1 or an ORF of MUC 1.
Alternatively, the ORF sequence of ENO1 is as set forth in SEQ ID No: 1, the ORF sequence of the MUC1 is shown as SEQ ID No: 2, respectively.
In a third aspect, the present invention also provides an expression plasmid for transforming leishmania, wherein the leishmania transformed with the expression plasmid promotes DC maturation, wherein the expression plasmid comprises at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence, and a tumor-specific antigen coding sequence.
Optionally, the expression plasmid comprises an ORF of ENO1 or an ORF of MUC 1.
Alternatively, the ORF sequence of ENO1 is as set forth in SEQ ID No: 1, the ORF sequence of the MUC1 is shown as SEQ ID No: 2, respectively.
The invention provides application of Leishmania transformed with an expression plasmid in promotion of DC maturation, a DC maturation promotion method and the expression plasmid, wherein the expression plasmid contains at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence. Thus, leishmania targetedly infects DC, DC is stimulated by leishmania to produce a large amount of inflammatory factors, meanwhile, expression plasmids in the leishmania body express tumor-associated proteins, tumor-associated polypeptides or tumor-specific antigens at high levels, immature DC take up and internalize expressed proteins/polypeptides in large amounts, inflammatory factors and DC internalized proteins/polypeptides induce DC maturation; presenting tumor-related protein/polypeptide or specific antigen to T cells by the mature DC, secreting Th1 type cell factor IFN-gamma, inducing and activating T cells to clone, and further activating the immune system of an organism to kill tumor cells; thus, immature DCs are efficiently induced to mature by Leishmania transformed with the expression plasmid, enhancing the effect of the body's immune system in eliminating tumor cells.
Drawings
In order to more clearly illustrate the embodiments of the present invention or the technical solutions in the prior art, the drawings used in the description of the embodiments or the prior art will be briefly described below, it is obvious that the drawings in the following description are only some embodiments of the present invention, and for those skilled in the art, other drawings can be obtained according to the structures shown in the drawings without creative efforts.
FIG. 1 is a plasmid map of p6.5MCS plasmid;
FIG. 2 is a set of graphs showing the results of gel electrophoresis of ENO1 in example I;
FIG. 3 is a graph showing the results of qRT-PCR of ENO1 in example two;
FIG. 4 is a graph of the qRT-PCR results of MUC1 in example two;
FIG. 5 is a graph showing Western blotting results of ENO1 in example two;
FIG. 6 is a graph showing the results of flow cytometry detection of ENO1 in example three;
FIG. 7 is a graph comparing the results of the MFI analysis of ENO1 in example III;
FIG. 8 is a graph comparing the results of MFI analysis of the deactivated group of ENO1 in example III;
FIG. 9 is a graph showing the results of flow cytometry analysis of MUC1 in example three.
The implementation, functional features and advantages of the objects of the present invention will be further explained with reference to the accompanying drawings.
Detailed Description
It should be noted that if the description of "first", "second", etc. is provided in the embodiment of the present invention, the description of "first", "second", etc. is only for descriptive purposes and is not to be construed as indicating or implying relative importance or implicitly indicating the number of indicated technical features. Thus, a feature defined as "first" or "second" may explicitly or implicitly include at least one such feature. In addition, the meaning of "and/or" appearing throughout is to include three juxtapositions, exemplified by "A and/or B" including either scheme A, or scheme B, or a scheme in which both A and B are satisfied.
The invention provides an application of Leishmania transformed with an expression plasmid in promoting DC maturation.
In the application of the invention, the expression plasmid can shuttle to the Escherichia coli and propagate in the Escherichia coli in a large amount. The expression plasmid can also be shuttled to leishmania and express foreign genes at high levels within the leishmania. The exogenous gene can be at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence. The kind of the expression plasmid is various. For example, the expression plasmid may be at least one of p6.5MCS plasmid, pX63NEO (Hyg) plasmid, pHM plasmid, pTEX plasmid, cLNEO (Hyg) plasmid, and pALT-NEO plasmid. The Leishmania may be at least one of Leishmania amazonensis LV78(MPRO/BR/72/M1845), Leishmania major (LV39), Leishmania donovani (LV39), Leishmania brasiliensis and Leishmania infantum. Any Leishmania that can target infection of DCs and can be transferred into expression plasmids is within the scope of the present invention.
The invention provides application of Leishmania transformed with expression plasmids in promotion of DC maturation. The expression plasmid contains at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence. Thus, leishmania targetedly infects DC, DC is stimulated by leishmania to produce a large amount of inflammatory factors, meanwhile, expression plasmids in the leishmania body express tumor-associated proteins, tumor-associated polypeptides or tumor-specific antigens at high levels, immature DC take up and internalize expressed proteins/polypeptides in large amounts, inflammatory factors and DC internalized proteins/polypeptides induce DC maturation; presenting tumor-related protein/polypeptide or specific antigen to T cells by the mature DC, secreting Th1 type cell factor IFN-gamma, inducing and activating T cells to clone, and further activating the immune system of an organism to kill tumor cells; thus, immature DCs are efficiently induced to mature by Leishmania transformed with the expression plasmid, enhancing the effect of the body's immune system in eliminating tumor cells.
The types of the tumor-associated proteins, tumor-associated polypeptides, and tumor-specific antigens are very diverse. Optionally, the expression plasmid comprises an ORF of ENO1 or an ORF of MUC 1. ENO1 is an important protein in the process of tumor metastasis, can cause specific immune response in tumors, and is a potential target for cancer immunotherapy. ENO1 belongs to an autoantigen, and healthy people have only low or no levels of ENO1 antibodies. Clinically, however, high levels of autoantibodies against ENO1 can be detected in the blood of many neoplastic and autoimmune disease patients. The ENO1 can induce the activation of the immune system of the body and generate antigen-specific CD4+And CD8+T cell responses. MUC1 is a type I transmembrane protein, normally expressed predominantly in the abluminal or glandular surface of epithelial cells, expressed apically, and distributed in polarity. MUC1 is also expressed in various cells of the hematopoietic system (e.g., T, B, DC). In most epithelial malignant tumors, the expression of MUC1 is abnormal, the expression level is increased by more than 100 times, the distribution of MUC1 protein on the cell surface is changed, the expression polarity is lost, the protein glycosylation is not complete, and new sugar chains and peptide epitopes are presented. The present study demonstrated that MUC1 antigen can specifically induce T cell proliferation and interferon gamma secretion. Thus, MUC1 is considered as a target for immunotherapy.
Alternatively, the ORF sequence of ENO1 is as set forth in SEQ ID No: 1, the ORF sequence of the MUC1 is shown as SEQ ID No: 2, respectively. The ORF sequence of ENO1 may also differ from SEQ ID No: 1, and the ORF sequence of MUC1 may also differ from the sequence of SEQ ID No: 2.
SEQ ID No: 1 is as follows:
GGATCCATGTCTATTCTCAGGATCCACGCCAGAGAGATCTTTGACTCCCGTGGGAATCCCACTGTTGAGGTCGATCTGTACACCGCAAAAGGTCTCTTCCGAGCTGCGGTGCCCAGCGGTGCGTCCACTGGCATCTACGAGGCCCTAGAACTCCGAGACAATGATAAGACCCGCTTCATGGGGAAGGGTGTCTCACAGGCTGTTGAGCACATCAATAAAACTATTGCGCCTGCTCTGGTTAGCAAGAAAGTGAATGTTGTGGAGCAAGAGAAGATTGACAAGCTGATGATCGAGATGGACGGCACAGAGAATAAATCTAAATTTGGTGCAAATGCCATCCTGGGAGTGTCCCTGGCTGTCTGCAAAGCTGGTGCCGTGGAAAAGGGGGTGCCCCTTTACCGCCACATTGCTGACTTGGCCGGCAACCCTGAAGTCATCCTGCCTGTCCCGGCTTTCAATGTGATCAACGGTGGTTCTCATGCTGGCAACAAGCTGGCCATGCAAGAGTTCATGATCCTGCCTGTGGGGGCATCCAGCTTCCGGGAAGCCATGCGCATTGGAGCAGAGGTTTACCACAACCTGAAGAACGTGATCAAGGAGAAGTACGGGAAGGACGCCACCAATGTGGGTGATGAGGGTGGATTCGCACCTAACATCCTGGAGAACAAAGAAGCACTGGAGCTGCTCAAGACTGCAATCGCAAAGGCCGGCTACACTGACCAGGTTGTCATTGGCATGGATGTGGCTGCCTCCGAGTTCTACAGGTCTGGCAAGTATGACCTGGACTTCAAGTCTCCGGATGACCCCAGCAGGTACATCACTCCCGACCAGCTGGCTGATCTGTACAAGTCCTTCGTCCAGAACTACCCAGTGGTGTCCATCGAAGATCCCTTTGACCAGGACGACTGGGGCGCCTGGCAGAAGTTCACGGCTAGTGCGGGCATCCAGGTGGTGGGCGATGACCTCACAGTGACCAACCCTAAGCGGATTGCCAAGGCTGCGAGCGAGAAGTCCTGCAACTGCCTCTTGCTCAAAGTGAACCAGATCGGCTCTGTGACCGAATCCCTGCAGGCGTGTAAGCTGGCCCAATCCAATGGCTGGGGCGTCATGGTGTCCCACCGATCTGGGGAAACTGAGGACACTTTCATCGCAGACCTGGTGGTGGGGCTCTGCACTGGGCAGATCAAGACTGGTGCCCCTTGCCGATCCGAGCGCCTGGCCAAGTACAATCAGATCCTCAGAATTGAGGAAGAGCTGGGCAGCAAAGCCAAGTTTGCTGGCAGGTCCTTCAGGAACCCCCTGGCCAAATAAGGATCC
SEQ ID No: 2 is as follows:
ATGACCCCGGGCATTCGGGCTCCTTTCTTCCTGCTGCTACTTCTAGCAAGTCTAAAAGGTTTTCTTGCCCTTCCAAGTGAGGAAAACAGTGTCACCTCATCTCAGGACACCAGCAGTTCCTTAGCATCGACTACCACTCCAGTCCACAGCAGCAACTCAGACCCAGCCACCAGACCTCCAGGGGACTCCACCAGCTCTCCAGTCCAGAGTAGCACCTCTTCTCCAGCCACCAGAGCTCCTGAAGACTCTACCAGTACTGCAGTCCTCAGTGGCACCTCCTCCCCAGCCACCACAGCTCCAGTGAACTCCGCCAGCTCTCCAGTAGCCCATGGTGACACCTCTTCCCCAGCCACTAGCCTTTCAAAAGACTCCAACAGCTCTCCAGTAGTCCACAGTGGCACCTCTTCAGCTCCGGCCACCACAGCTCCAGTGGATTCCACCAGCTCTCCAGTAGTCCACGGTGGTACCTCGTCCCCAGCCACCAGCCCTCCAGGGGACTCCACCAGCTCTCCAGACCATAGTAGCACCTCTTCTCCAGCCACCAGAGCTCCCGAAGACTCTACCAGTACTGCAGTCCTCAGTGGCACCTCCTCCCCAGCCACCACAGCTCCAGTGGACTCCACCAGCTCTCCAGTAGCCCATGATGACACCTCTTCCCCAGCCACTAGCCTTTCAGAAGACTCCGCCAGCTCTCCAGTAGCCCACGGTGGCACCTCTTCTCCAGCCACCAGCCCTCTAAGGGACTCCACCAGTTCTCCAGTCCACAGTAGTGCCTCCATCCAAAACATCAAGACTACATCAGACTTAGCTAGCACTCCAGACCACAATGGCACCTCAGTCACAACTACCAGCTCTGCACTGGGCTCAGCCACCAGTCCAGACCACAGTGGTACCTCAACTACAACTAACAGCTCTGAATCAGTCTTGGCCACCACTCCAGTTTACAGTAGCATGCCATTCTCTACTACCAAAGTGACGTCAGGCTCAGCTATCATTCCAGACCACAATGGCTCCTCGGTGCTACCTACCAGTTCTGTGTTGGGCTCAGCTACCAGTCTAGTCTATAATACCTCTGCAATAGCTACAACTCCAGTCAGCAATGGCACTCAGCCTTCAGTGCCAAGTCAATACCCTGTTTCTCCTACCATGGCCACCACCTCCAGCCACAGCACTATTGCCAGCAGCTCTTACTATAGCACAGTACCATTTTCTACCTTCTCCAGTAACAGTTCACCCCAGTTGTCTGTTGGGGTCTCCTTCTTCTTCTTGTCTTTTTACATTCAAAACCACCCATTTAATTCTTCTCTGGAAGACCCCAGCTCCAACTACTACCAAGAACTGAAGAGGAACATTTCTGGATTGTTTCTGCAGATTTTTAACGGAGATTTTCTGGGGATCTCTAGCATCAAGTTCAGGTCAGGCTCCGTGGTGGTAGAATCGACTGTGGTTTTCCGGGAGGGTACTTTTAGTGCCTCTGACGTGAAGTCACAGCTTATACAGCATAAGAAGGAGGCAGATGACTATAATCTGACTATTTCAGAAGTCAAAGTGAATGAGATGCAGTTCCCTCCCTCTGCCCAGTCCCGGCCGGGGGTACCAGGCTGGGGCATTGCCCTGCTGGTGCTGGTCTGTATTTTGGTTGCTTTGGCTATCGTCTATTTCCTTGCCCTGGCAGTGTGCCAGTGCCGCCGAAAGAGCTATGGGCAGCTGGACATCTTTCCAACCCAGGACACCTACCATCCTATGAGTGAATACCCTACCTACCACACTCACGGACGCTACGTGCCCCCTGGCAGTACCAAGCGTAGCCCCTATGAGGAGGTTTCGGCAGGTAATGGCAGTAGCAGTCTCTCTTATACCAACCCAGCTGTGGTGACCACTTCTGCCAACTTGTAG
the invention also provides a method for promoting DC maturation, which comprises the following steps: (1) transforming the expression plasmid into leishmania; (2) infecting said leishmania infected DCs with said expression plasmid; wherein, the expression plasmid contains at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence.
The specific steps of the method of the present invention refer to the following embodiments, which are not described herein. The method for promoting DC maturation can effectively induce DC maturation, thereby effectively activating the immune system of an organism to kill tumor cells and enhancing the effect of the immune system of the organism to eliminate the tumor cells.
The invention also provides an expression plasmid, which is used for transforming leishmania, and the leishmania transformed with the expression plasmid promotes DC maturation, wherein the expression plasmid comprises at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence. The expression plasmid of the invention can freely shuttle to Leishmania and can express tumor-associated protein, polypeptide or specific antigen at high level. The protein/polypeptide expressed by the expression plasmid can be greatly taken up and internalized by immature DC, so that the maturation of the DC is effectively induced, and the immune system of an organism is activated to kill tumor cells.
Example one construction of expression plasmid
The expression plasmid is constructed by adopting p6.5MCS plasmid. Referring to FIG. 1, the total length of p6.5MCS plasmid is 9185bp, and includes a partial sequence of pBluescript plasmid, a Multiple Cloning Site (MCS) and a partial gene sequence of Leishmania. The part sequence of the pBluescript plasmid contains an Ampicillin resistant (Ampicillin) gene sequence. The partial gene sequence of Leishmania contains N-acetylglucosamine-1-phosphotransferase (NAGT) sequence, which is used for expressing and mediating leishmania to generate resistance to Tunicamycin (Tunicamycin).
The construction of the expression plasmid is as follows (using ENO1 as an example):
(1) carrying out enzyme digestion on the purified p6.5MCS plasmid by using BamHI enzyme, and carrying out enzyme digestion at 37 ℃ overnight;
(2) carrying out agarose gel electrophoresis detection on the enzyme digestion product, and recovering a target band to obtain a linearized p6.5MCS vector (9185 bp);
(3) extracting RNA of a mouse, carrying out reverse transcription, PCR amplification and purification to obtain an ORF fragment (1344bp) of the murine ENO1 gene;
(4) connecting the ENO1 and the digested p6.5MCS plasmid by using the seamless cloning kit to obtain an expression vector p6.5MCS-ENO 1;
(6) transferring the expression plasmid into escherichia coli (adding ampicillin into the culture medium);
(5) PCR identification of positive clone of bacterial liquid;
(6) sequencing positive clones, and comparing and analyzing a sequencing result with a target gene sequence;
(7) the positive clones were cultured overnight at 37 ℃ (ampicillin was added to the medium);
(8) carrying out PCR identification on positive clones by using the bacterium liquid again;
(9) and extracting the plasmid from the positive clone bacterium liquid without endotoxin to obtain p6.5MCS-ENO1 expression plasmid.
The invention also constructs the p6.5MCS-MUC1 expression plasmid, and the construction process is similar to that of the p6.5MCS-ENO1 expression plasmid and is not repeated. Wherein, the sequences of the upstream primer and the downstream primer of the ORF of the ENO1 amplified by PCR are shown as SEQ ID No: 3 and SEQ ID No: 4 is shown in the specification; the sequences of the upstream primer and the downstream primer for amplifying the ORF of MUC1 are shown as SEQ ID No: 5 and SEQ ID No: 6, see table 1 for specific sequences. The PCR reaction system is referred to the PCR kit instruction. The PCR reaction conditions were pre-denaturation at 95 ℃ for 2min, replication (denaturation at 95 ℃ for 30s, annealing at 65 ℃ for 30s, and extension at 72 ℃ for 90s) for 38 cycles, and final extension at 72 ℃ for 5 min.
TABLE 1 PCR primer sequences
Referring to FIG. 2, panel A in FIG. 2 is a gel electrophoresis of linearized p6.5MCS plasmid (9185 bp); FIG. 2B is a gel electrophoresis of PCR amplified ORF of ENO1 (1305 bp); FIG. 2C is a gel electrophoresis image of PCR identification of positive clone bacteria liquid; FIG. 2D is a gel electrophoresis image of EcoRI and BglII double-digested plasmid after extraction. In diagram D of FIG. 2, the number 1 indicates the cleavage result of the p6.5MCS empty plasmid control, the p6.5MCS vector has only an EcoRI cleavage site, does not contain a BglII cleavage site, and the EcoRI and BglII double-cleaved p6.5MCS vector has only one band; the plasmid p6.5MCS-ENO1 was digested at accession number 2, and the ORF of ENO1 contained a BglII cleavage site, so that the plasmid p6.5MCS-ENO1 was digested with EcoRI and BglII to generate two fragments of 3844bp and 6652 bp. The results in FIG. 2 demonstrate that the p6.5MCS-ENO1 plasmid has been successfully constructed. The gel electrophoresis result of the construction process of p6.5MCS-MUC1 plasmid is not shown.
Example II expression plasmid transfer into Leishmania
The two expression plasmids constructed in example one were separately transferred into Leishmania.
The specific steps are as follows (taking ENO1 as an example as well):
(1) the number of Leishmania amazonensis (LV78) in the logarithmic growth phase was counted to 1.2X 107Then, the mixture was collected and washed twice with PBS;
(2) adding 100 mu l of transfection solution into p6.5MCS-ENO1 expression plasmid solution containing 10 mu g, re-suspending and transferring the solution into an electric transfer cup, putting the electric transfer cup into a Lonza Nucleofector nuclear transfer system, and carrying out transfection by using a program U-033;
(3) taking out the electric rotating cup from the Nucleofector device, immediately adding 3ml of M199 complete culture medium, and culturing for 24 hours;
(4) adding Tunicamycin (Tunicamycin) 5ug/ml into the above solution, and culturing for 3 weeks;
(5) performing DNase I treatment (Takara) using TRIzol reagent solution (ThermoFisher), and isolating total RNA from the harvested Leishmania;
(6) designing primers, and synthesizing cDNA by using a ReverTra Ace q-PCR RT kit (Toyobo);
(7) adding the cDNA into SYBR Green Real-time PCR premix solution, and analyzing the expression level of ORF of ENO1 in the expression vector by Real-time quantitative PCR;
(8) RIPA lysis buffer (containing 1mM protease inhibitor PMSF) lyses cells;
(9) centrifuging at 0-4 ℃ and 12,000rpm for 15min, collecting the supernatant of the protein lysate, heating at 100 ℃ for 10 min, and carrying out electrophoretic separation on polyacrylamide gel containing SDS;
(10) the electrophoresis gel was then transferred to a PVDF membrane (Millipore), detection was performed using a diabody, blotting was developed with a chemiluminescent reagent, and the results were observed.
In this example, the sequences of the upstream and downstream primers of ENO1 were amplified by real-time quantitative PCR as shown in SEQ ID No: 7 and SEQ ID No: 8 is shown in the specification; the sequences of the upstream primer and the downstream primer for amplifying the MUC1 are shown as SEQ ID No: 9 and SEQ ID No: 10, see table 2. For the setting of the real-time quantitative PCR reaction system and the reaction program, please refer to the description of the real-time quantitative PCR kit. The diabodies used in this example were anti-ENO 1 (or anti-MUC 1 protein) and anti-beta-tubulin (control).
TABLE 2 real-time quantitative PCR primer sequences
FIG. 3 shows the results of qRT-PCR of ENO1, FIG. 4 shows the results of qRT-PCR of MUC1, and the control is Leishmania (LV78) not transferred into the expression plasmid. The results in FIGS. 3 and 4 demonstrate that the foreign gene on the expression plasmid can be expressed at high levels in the Leishmania. FIG. 5 is a Western blot result of ENO1, indicating that p6.5MCS-ENO1 has successfully expressed the ENO1 protein in the Leishmania. Western blot results for p6.5MCS-MUC1 were consistent with those for p6.5MCS-ENO1 and are not shown.
EXAMPLE III Leishmania infected DC transformed with expression plasmid
Culture of immature DC: bone marrow of C57BL/6 mouse was cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum, recombinant mouse GM-CSF (31.5ng/ml) for 3 days; culture was continued to day seven by addition of fresh medium containing GM-CSF to yield large numbers of loosely adherent immature DCs (imDCs).
Culturing Leishmania: l. lv78 leishmania promastigotes (capable of expressing GFP tag) were cultured in M199 medium containing 10% fetal bovine serum, penicillin and streptomycin until logarithmic growth phase was reached.
Infection: firstly, Leishmania promastigotes are used for infecting immature DCs, the infection complex number is 4 and 10 respectively, and the infection condition is 35 ℃ for 8 h. ② infecting immature DC with inactivated Leishmania promastigotes (incubated at 70 ℃ for 45 minutes), the multiplicity of infection is 4 and 10 respectively, and the infection condition is 35 ℃ for 8 h.
And (3) analysis: DC cells of control group (uninfected leishmania promastigotes) and experimental group (infected live/dead leishmania promastigotes) were subjected to flow cytometry analysis using monoclonal antibodies against CD80, CD86, and MHC II, respectively.
FIG. 6 shows the results of flow cytometry analysis of Leishmania promastigote infected DCs transformed with p6.5MCS-ENO1, and the results show that the positive rates of MHC II expression and CD86 of live Leishmania promastigote infected DCs were increased compared with the control group of uninfected Leishmania promastigotes, and the positive rate of group with a multiplicity of infection of 10 was more significant than the increase of group with a multiplicity of infection of 4. FIG. 7 shows that the trend of change in MFI (mean fluorescence intensity) of Leishmania promastigotes transformed with p6.5MCS-ENO1 is consistent with the positive ratio in FIG. 6, indicating that infection of DC with Leishmania promastigotes transformed with p6.5MCS-ENO1 promotes DC maturation and expression of MHC II and CD 86. Referring to FIG. 8, the positive rate of CD86 was slightly increased in the group of infected killed Leishmania promastigotes compared to the uninfected control group, indicating that although the inactivated Leishmania promastigotes were less effective in promoting DC cell maturation than in the live Leishmania promastigotes, they also had some induction of DC maturation and expression of CD 86.
FIG. 9 shows the results of flow cytometry analysis of Leishmania promastigotes infected DC transformed with p6.5MCS-MUC1, and the results show that MUC1 was consistent with those of ENO1, and that the positive rates of MHC II and CD86 of live Leishmania promastigotes were also increased compared with the uninfected control group, wherein the positive rate was more significant in the group with a multiplicity of infection of 10 than in the group with a multiplicity of infection of 4.
In combination with the above results, Leishmania transformed with the expression plasmid can effectively promote DC cell maturation. Therefore, the leishmania transformed with the expression plasmid provided by the invention has great potential value in promoting DC maturation, promoting DC maturation method and expression plasmid in tumor treatment.
The above description is only a preferred embodiment of the present invention, and is not intended to limit the scope of the present invention, and all modifications and equivalents of the present invention, which are made by the contents of the present specification and the accompanying drawings, or directly/indirectly applied to other related technical fields, are included in the scope of the present invention.
SEQUENCE LISTING
<110> Shenzhen Rotzmann international transformation medical research institute
<120> use of Leishmania transformed with expression plasmid for promoting DC maturation, method and table for promoting DC maturation
Daplasmid
<130>
<160> 10
<170> PatentIn version 3.3
<210> 1
<211> 1317
<212> DNA
<213> Artificial Synthesis
<400> 1
ggatccatgt ctattctcag gatccacgcc agagagatct ttgactcccg tgggaatccc 60
actgttgagg tcgatctgta caccgcaaaa ggtctcttcc gagctgcggt gcccagcggt 120
gcgtccactg gcatctacga ggccctagaa ctccgagaca atgataagac ccgcttcatg 180
gggaagggtg tctcacaggc tgttgagcac atcaataaaa ctattgcgcc tgctctggtt 240
agcaagaaag tgaatgttgt ggagcaagag aagattgaca agctgatgat cgagatggac 300
ggcacagaga ataaatctaa atttggtgca aatgccatcc tgggagtgtc cctggctgtc 360
tgcaaagctg gtgccgtgga aaagggggtg cccctttacc gccacattgc tgacttggcc 420
ggcaaccctg aagtcatcct gcctgtcccg gctttcaatg tgatcaacgg tggttctcat 480
gctggcaaca agctggccat gcaagagttc atgatcctgc ctgtgggggc atccagcttc 540
cgggaagcca tgcgcattgg agcagaggtt taccacaacc tgaagaacgt gatcaaggag 600
aagtacggga aggacgccac caatgtgggt gatgagggtg gattcgcacc taacatcctg 660
gagaacaaag aagcactgga gctgctcaag actgcaatcg caaaggccgg ctacactgac 720
caggttgtca ttggcatgga tgtggctgcc tccgagttct acaggtctgg caagtatgac 780
ctggacttca agtctccgga tgaccccagc aggtacatca ctcccgacca gctggctgat 840
ctgtacaagt ccttcgtcca gaactaccca gtggtgtcca tcgaagatcc ctttgaccag 900
gacgactggg gcgcctggca gaagttcacg gctagtgcgg gcatccaggt ggtgggcgat 960
gacctcacag tgaccaaccc taagcggatt gccaaggctg cgagcgagaa gtcctgcaac 1020
tgcctcttgc tcaaagtgaa ccagatcggc tctgtgaccg aatccctgca ggcgtgtaag 1080
ctggcccaat ccaatggctg gggcgtcatg gtgtcccacc gatctgggga aactgaggac 1140
actttcatcg cagacctggt ggtggggctc tgcactgggc agatcaagac tggtgcccct 1200
tgccgatccg agcgcctggc caagtacaat cagatcctca gaattgagga agagctgggc 1260
agcaaagcca agtttgctgg caggtccttc aggaaccccc tggccaaata aggatcc 1317
<210> 2
<211> 1896
<212> DNA
<213> Artificial Synthesis
<400> 2
atgaccccgg gcattcgggc tcctttcttc ctgctgctac ttctagcaag tctaaaaggt 60
tttcttgccc ttccaagtga ggaaaacagt gtcacctcat ctcaggacac cagcagttcc 120
ttagcatcga ctaccactcc agtccacagc agcaactcag acccagccac cagacctcca 180
ggggactcca ccagctctcc agtccagagt agcacctctt ctccagccac cagagctcct 240
gaagactcta ccagtactgc agtcctcagt ggcacctcct ccccagccac cacagctcca 300
gtgaactccg ccagctctcc agtagcccat ggtgacacct cttccccagc cactagcctt 360
tcaaaagact ccaacagctc tccagtagtc cacagtggca cctcttcagc tccggccacc 420
acagctccag tggattccac cagctctcca gtagtccacg gtggtacctc gtccccagcc 480
accagccctc caggggactc caccagctct ccagaccata gtagcacctc ttctccagcc 540
accagagctc ccgaagactc taccagtact gcagtcctca gtggcacctc ctccccagcc 600
accacagctc cagtggactc caccagctct ccagtagccc atgatgacac ctcttcccca 660
gccactagcc tttcagaaga ctccgccagc tctccagtag cccacggtgg cacctcttct 720
ccagccacca gccctctaag ggactccacc agttctccag tccacagtag tgcctccatc 780
caaaacatca agactacatc agacttagct agcactccag accacaatgg cacctcagtc 840
acaactacca gctctgcact gggctcagcc accagtccag accacagtgg tacctcaact 900
acaactaaca gctctgaatc agtcttggcc accactccag tttacagtag catgccattc 960
tctactacca aagtgacgtc aggctcagct atcattccag accacaatgg ctcctcggtg 1020
ctacctacca gttctgtgtt gggctcagct accagtctag tctataatac ctctgcaata 1080
gctacaactc cagtcagcaa tggcactcag ccttcagtgc caagtcaata ccctgtttct 1140
cctaccatgg ccaccacctc cagccacagc actattgcca gcagctctta ctatagcaca 1200
gtaccatttt ctaccttctc cagtaacagt tcaccccagt tgtctgttgg ggtctccttc 1260
ttcttcttgt ctttttacat tcaaaaccac ccatttaatt cttctctgga agaccccagc 1320
tccaactact accaagaact gaagaggaac atttctggat tgtttctgca gatttttaac 1380
ggagattttc tggggatctc tagcatcaag ttcaggtcag gctccgtggt ggtagaatcg 1440
actgtggttt tccgggaggg tacttttagt gcctctgacg tgaagtcaca gcttatacag 1500
cataagaagg aggcagatga ctataatctg actatttcag aagtcaaagt gaatgagatg 1560
cagttccctc cctctgccca gtcccggccg ggggtaccag gctggggcat tgccctgctg 1620
gtgctggtct gtattttggt tgctttggct atcgtctatt tccttgccct ggcagtgtgc 1680
cagtgccgcc gaaagagcta tgggcagctg gacatctttc caacccagga cacctaccat 1740
cctatgagtg aataccctac ctaccacact cacggacgct acgtgccccc tggcagtacc 1800
aagcgtagcc cctatgagga ggtttcggca ggtaatggca gtagcagtct ctcttatacc 1860
aacccagctg tggtgaccac ttctgccaac ttgtag 1896
<210> 3
<211> 19
<212> DNA
<213> Artificial Synthesis
<400> 3
ctactgccag aaattcgcc 19
<210> 4
<211> 18
<212> DNA
<213> Artificial Synthesis
<400> 4
agggatctcc ggtccatg 18
<210> 5
<211> 33
<212> DNA
<213> Artificial Synthesis
<400> 5
gctcgaattc agatcatgac cccgggcatt cgg 33
<210> 6
<211> 40
<212> DNA
<213> Artificial Synthesis
<400> 6
cactaaaggg agatcctaca agttggcaga agtggtcacc 40
<210> 7
<211> 23
<212> DNA
<213> Artificial Synthesis
<400> 7
tcctggagaa caaagaagca ctg 23
<210> 8
<211> 22
<212> DNA
<213> Artificial Synthesis
<400> 8
cttcgatgga caccactggg ta 22
<210> 9
<211> 20
<212> DNA
<213> Artificial Synthesis
<400> 9
ggctccgtgg tggtagaatc 20
<210> 10
<211> 20
<212> DNA
<213> Artificial Synthesis
<400> 10
Claims (10)
1. Use of Leishmania transformed with an expression plasmid to promote DC maturation.
2. The use of claim 1, wherein said expression plasmid comprises at least one of a tumor associated protein coding sequence, a tumor associated polypeptide coding sequence, a tumor specific antigen coding sequence.
3. The use of claim 2, wherein the expression plasmid comprises the ORF of ENO1 or the ORF of MUC 1.
4. The use of claim 3, wherein the ORF of ENO1 has the sequence shown in SEQ ID No: 1, the ORF sequence of the MUC1 is shown as SEQ ID No: 2, respectively.
5. A method of promoting DC maturation, comprising the steps of:
(1) transforming the expression plasmid into leishmania;
(2) infecting said leishmania infected DCs with said expression plasmid;
wherein, the expression plasmid contains at least one of a tumor-associated protein coding sequence, a tumor-associated polypeptide coding sequence and a tumor-specific antigen coding sequence.
6. The method of promoting DC maturation according to claim 5, wherein the expression plasmid comprises the ORF of ENO1 or the ORF of MUC 1.
7. The method for promoting DC maturation according to claim 6, wherein the ORF sequence of ENO1 is as set forth in SEQ ID No: 1, the ORF sequence of the MUC1 is shown as SEQ ID No: 2, respectively.
8. An expression plasmid for transforming leishmania, wherein leishmania transformed with the expression plasmid promotes DC maturation, wherein the expression plasmid comprises at least one of a tumor associated protein coding sequence, a tumor associated polypeptide coding sequence, and a tumor specific antigen coding sequence.
9. The expression plasmid of claim 8, wherein the expression plasmid comprises an ORF of ENO1 or an ORF of MUC 1.
10. The expression plasmid of claim 9, wherein the ORF sequence of ENO1 is as set forth in SEQ ID No: 1, the ORF sequence of the MUC1 is shown as SEQ ID No: 2, respectively.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202110521400.7A CN113249223A (en) | 2021-05-13 | 2021-05-13 | Application of leishmania transformed with expression plasmid in promotion of DC maturation, method for promoting DC maturation, and expression plasmid |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202110521400.7A CN113249223A (en) | 2021-05-13 | 2021-05-13 | Application of leishmania transformed with expression plasmid in promotion of DC maturation, method for promoting DC maturation, and expression plasmid |
Publications (1)
Publication Number | Publication Date |
---|---|
CN113249223A true CN113249223A (en) | 2021-08-13 |
Family
ID=77183313
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202110521400.7A Pending CN113249223A (en) | 2021-05-13 | 2021-05-13 | Application of leishmania transformed with expression plasmid in promotion of DC maturation, method for promoting DC maturation, and expression plasmid |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN113249223A (en) |
Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060228342A1 (en) * | 2002-05-28 | 2006-10-12 | Robinson Ramirez-Pineda | Method for generating antigen-presenting cells |
CN101516199A (en) * | 2006-07-21 | 2009-08-26 | 加州理工学院 | Targeted gene delivery for dendritic cell vaccination |
US20120288524A1 (en) * | 2011-05-10 | 2012-11-15 | Rosalind Franklin University Of Medicine And Science | Leishmania-based carrier for vaccine delivery |
CN103372205A (en) * | 2012-04-26 | 2013-10-30 | 上海复和泰生物技术有限公司 | Preparation method of tumor dendritic cell vaccine sensitized by glycosylated MUC1 (mucoprotein 1) antigen |
CN106687583A (en) * | 2014-06-23 | 2017-05-17 | Jw可瑞基因株式会社 | Method for preparing dendritic cells with increased specific gene expression, and composition for treating or preventing autoimmune diseases, containing dendritic cells prepared using same |
CN111587288A (en) * | 2017-07-14 | 2020-08-25 | 生物克隆专利有限公司 | Maturation of dendritic cells |
US20200376100A1 (en) * | 2019-05-31 | 2020-12-03 | Rosalind Franklin Univ. of Medicine and Science | Immunotherapy of solid tumors by vaccination with transgenic leishmania expressing cancer vaccines and with recombinant vaccines with leishmania as adjuvants |
CN112458058A (en) * | 2020-11-24 | 2021-03-09 | 康九生物科技(长春)有限公司 | DC cell over-expressing TRAF6, DC cell vaccine, construction method and application |
-
2021
- 2021-05-13 CN CN202110521400.7A patent/CN113249223A/en active Pending
Patent Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20060228342A1 (en) * | 2002-05-28 | 2006-10-12 | Robinson Ramirez-Pineda | Method for generating antigen-presenting cells |
CN101516199A (en) * | 2006-07-21 | 2009-08-26 | 加州理工学院 | Targeted gene delivery for dendritic cell vaccination |
US20120288524A1 (en) * | 2011-05-10 | 2012-11-15 | Rosalind Franklin University Of Medicine And Science | Leishmania-based carrier for vaccine delivery |
CN103372205A (en) * | 2012-04-26 | 2013-10-30 | 上海复和泰生物技术有限公司 | Preparation method of tumor dendritic cell vaccine sensitized by glycosylated MUC1 (mucoprotein 1) antigen |
CN106687583A (en) * | 2014-06-23 | 2017-05-17 | Jw可瑞基因株式会社 | Method for preparing dendritic cells with increased specific gene expression, and composition for treating or preventing autoimmune diseases, containing dendritic cells prepared using same |
CN111587288A (en) * | 2017-07-14 | 2020-08-25 | 生物克隆专利有限公司 | Maturation of dendritic cells |
US20200376100A1 (en) * | 2019-05-31 | 2020-12-03 | Rosalind Franklin Univ. of Medicine and Science | Immunotherapy of solid tumors by vaccination with transgenic leishmania expressing cancer vaccines and with recombinant vaccines with leishmania as adjuvants |
CN112458058A (en) * | 2020-11-24 | 2021-03-09 | 康九生物科技(长春)有限公司 | DC cell over-expressing TRAF6, DC cell vaccine, construction method and application |
Non-Patent Citations (6)
Title |
---|
BRETON M 等: "Live nonpathogenic parasitic vector as a candidate vaccine against visceral leishmaniasis", 《INFECT IMMUN》 * |
DIEGO A 等: "Leishmania braziliensis infection induces dendritic cell activation, ISG15 transcription, and the generation of protective immune responses", 《JOURNAL OF IMMUNOLOGY》 * |
XIN L 等: "Down-regulation of dendritic cell signaling pathways by Leishmania amazonensis amastigotes", 《MOL IMMUNOL》 * |
余鹏 等: "利什曼原虫感染树突状细胞早期基因表达差异分析及关键基因筛选", 《中国免疫学杂志》 * |
无: "Accession number: BC039179.1,Mus musculus enolase 1, alpha non-neuron, mRNA (cDNA clone MGC:25104 IMAGE:4501041), complete cds", 《GENBANK》 * |
无: "Accession number: M84683.1,Mus musculus episialin (Muc1) mRNA, complete cds", 《GENBANK》 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2018207281B2 (en) | Modulating expression of polypeptides via new gene switch expression systems | |
AU2015286820B2 (en) | Stabilization of poly(A) sequence encoding DNA sequences | |
Bonehill et al. | Messenger RNA-electroporated dendritic cells presenting MAGE-A3 simultaneously in HLA class I and class II molecules | |
EP2817330B1 (en) | Use of icos-based cars to enhance antitumor activity and car persistence | |
US20200165302A1 (en) | Modified vsv-g and vaccines thereof | |
US20180002720A1 (en) | Cell for use in immunotherapy which contains modified nucleic acid construct encoding wilms tumor gene product or fragment thereof, method for producing said cell, and said nucleic acid construct | |
WO2019080538A1 (en) | Chimeric antigen receptor, nk cell modified by same, coding dna, mrna, expression vector, preparation method and application | |
CN105802909B (en) | T cell preparation with HER2 specific TCR and uses thereof | |
WO2017101244A1 (en) | Method for preparing and using lentivirus expression vector, and method for preparing recombinant lentivirus | |
CN111019959B (en) | Nucleotide molecule for in vitro transcription of mRNA, presenting cell and application | |
JP5114403B2 (en) | SART3-derived peptides useful for cancer vaccine therapy for HLA-A3 supertype allele positive prostate cancer patients | |
CA2537161C (en) | Preventive cancer vaccine based on brother of regulator of imprinted sites molecule (boris) | |
JP5938143B2 (en) | Lentiviral vector containing MHC class I promoter | |
Liu et al. | Use and specificity of breast cancer antigen/milk protein BA46 for generating anti-self-cytotoxic T lymphocytes by recombinant adeno-associated virus-based gene loading of dendritic cells | |
CN102180961B (en) | Black-blue spotted puffer fish interferon IFN gamma 2 and preparation method and application thereof | |
RU2319741C2 (en) | Microorganisms as carriers of nucleotide sequences encoding cellular antigens for treatment of tumors | |
Fukuma et al. | Cancer prevention with semi-allogeneic ES cell-derived dendritic cells | |
CN102993292A (en) | AFP (Alpha Fetal Protein) recombinant protein and in-intro recombinant expression method | |
CN113249223A (en) | Application of leishmania transformed with expression plasmid in promotion of DC maturation, method for promoting DC maturation, and expression plasmid | |
CN113355362B (en) | Use of chemically modified CRISPR/Cpf1 complex | |
WO2017101243A1 (en) | Method for preparing and using lentivirus expression vector, and method for preparing recombinant lentivirus | |
RU2507265C2 (en) | RECOMBINANT PLASMID DNA pCI-UB-POLYEPI CONTAINING EPITOPES OF TUMOUR-ASSOCIATED ANTIGENS FOR COLORECTAL CANCER AND METHOD OF ITS APPLICATION FOR STIMULATION OF SPECIFIC ANTITUMOUR IMMUNE RESPONSE AGAINST COLORECTAL CANCER CELLS | |
CN109568574B (en) | Application of sPD1 protein and/or sPD1 gene as immune adjuvant | |
CN111254119B (en) | T cell with effect of improving immune response and preparation method and application thereof | |
WO2023082640A1 (en) | Method for enhancing durability of immune cell |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
RJ01 | Rejection of invention patent application after publication |
Application publication date: 20210813 |
|
RJ01 | Rejection of invention patent application after publication |