CN115074307B - Method for reprogramming fibroblast induced by small molecule compound into testis support cell and application thereof - Google Patents
Method for reprogramming fibroblast induced by small molecule compound into testis support cell and application thereof Download PDFInfo
- Publication number
- CN115074307B CN115074307B CN202110275603.2A CN202110275603A CN115074307B CN 115074307 B CN115074307 B CN 115074307B CN 202110275603 A CN202110275603 A CN 202110275603A CN 115074307 B CN115074307 B CN 115074307B
- Authority
- CN
- China
- Prior art keywords
- cell
- cells
- ciscs
- testis
- support
- 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.)
- Active
Links
- 210000004027 cell Anatomy 0.000 title claims abstract description 226
- 210000001550 testis Anatomy 0.000 title claims abstract description 57
- 210000002950 fibroblast Anatomy 0.000 title claims abstract description 45
- 238000000034 method Methods 0.000 title claims abstract description 33
- 230000008672 reprogramming Effects 0.000 title claims abstract description 25
- -1 small molecule compound Chemical class 0.000 title description 8
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 51
- 230000002381 testicular Effects 0.000 claims abstract description 24
- 239000002609 medium Substances 0.000 claims description 38
- 210000000717 sertoli cell Anatomy 0.000 claims description 28
- 230000014509 gene expression Effects 0.000 claims description 24
- 102000004169 proteins and genes Human genes 0.000 claims description 20
- SHGAZHPCJJPHSC-YCNIQYBTSA-N all-trans-retinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-YCNIQYBTSA-N 0.000 claims description 18
- 229930002330 retinoic acid Natural products 0.000 claims description 18
- 229960001727 tretinoin Drugs 0.000 claims description 18
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 claims description 17
- 108091007911 GSKs Proteins 0.000 claims description 14
- 230000006698 induction Effects 0.000 claims description 13
- 238000002054 transplantation Methods 0.000 claims description 13
- 108091003079 Bovine Serum Albumin Proteins 0.000 claims description 12
- 239000012091 fetal bovine serum Substances 0.000 claims description 12
- 210000002863 seminiferous tubule Anatomy 0.000 claims description 12
- 238000000338 in vitro Methods 0.000 claims description 11
- FTALBRSUTCGOEG-UHFFFAOYSA-N Riluzole Chemical group C1=C(OC(F)(F)F)C=C2SC(N)=NC2=C1 FTALBRSUTCGOEG-UHFFFAOYSA-N 0.000 claims description 10
- 239000003112 inhibitor Substances 0.000 claims description 10
- 229960004181 riluzole Drugs 0.000 claims description 10
- 230000008827 biological function Effects 0.000 claims description 9
- VUVUVNZRUGEAHB-CYBMUJFWSA-N 7-(3,5-dimethyl-4-isoxazolyl)-8-methoxy-1-[(1R)-1-(2-pyridinyl)ethyl]-3H-imidazo[4,5-c]quinolin-2-one Chemical group C1([C@@H](C)N2C3=C4C=C(C(=CC4=NC=C3NC2=O)C2=C(ON=C2C)C)OC)=CC=CC=N1 VUVUVNZRUGEAHB-CYBMUJFWSA-N 0.000 claims description 8
- 101150084041 WT1 gene Proteins 0.000 claims description 8
- 229940125763 bromodomain inhibitor Drugs 0.000 claims description 7
- 238000012258 culturing Methods 0.000 claims description 6
- 210000004602 germ cell Anatomy 0.000 claims description 6
- 102000034615 Glial cell line-derived neurotrophic factor Human genes 0.000 claims description 5
- 108091010837 Glial cell line-derived neurotrophic factor Proteins 0.000 claims description 5
- 230000012010 growth Effects 0.000 claims description 5
- 230000001506 immunosuppresive effect Effects 0.000 claims description 5
- 108010001857 Cell Surface Receptors Proteins 0.000 claims description 4
- 101150105763 FSHR gene Proteins 0.000 claims description 4
- 102100027627 Follicle-stimulating hormone receptor Human genes 0.000 claims description 4
- 101000862396 Homo sapiens Follicle-stimulating hormone receptor Proteins 0.000 claims description 4
- 101150078927 KRT18 gene Proteins 0.000 claims description 4
- 230000002776 aggregation Effects 0.000 claims description 4
- 238000004220 aggregation Methods 0.000 claims description 4
- 210000002889 endothelial cell Anatomy 0.000 claims description 4
- 101150082979 gdnf gene Proteins 0.000 claims description 4
- 230000001939 inductive effect Effects 0.000 claims description 4
- 239000000893 inhibin Substances 0.000 claims description 4
- ZPNFWUPYTFPOJU-LPYSRVMUSA-N iniprol Chemical compound C([C@H]1C(=O)NCC(=O)NCC(=O)N[C@H]2CSSC[C@H]3C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@H](C(N[C@H](C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC=4C=CC=CC=4)C(=O)N[C@@H](CC=4C=CC(O)=CC=4)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CC=4C=CC=CC=4)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC2=O)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CC=2C=CC=CC=2)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]2N(CCC2)C(=O)[C@@H](N)CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N2[C@@H](CCC2)C(=O)N2[C@@H](CCC2)C(=O)N[C@@H](CC=2C=CC(O)=CC=2)C(=O)N[C@@H]([C@@H](C)O)C(=O)NCC(=O)N2[C@@H](CCC2)C(=O)N3)C(=O)NCC(=O)NCC(=O)N[C@@H](C)C(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@H](C(=O)N[C@@H](CC=2C=CC=CC=2)C(=O)N[C@H](C(=O)N1)C(C)C)[C@@H](C)O)[C@@H](C)CC)=O)[C@@H](C)CC)C1=CC=C(O)C=C1 ZPNFWUPYTFPOJU-LPYSRVMUSA-N 0.000 claims description 4
- 238000000926 separation method Methods 0.000 claims description 4
- 230000001228 trophic effect Effects 0.000 claims description 4
- 102100021337 Gap junction alpha-1 protein Human genes 0.000 claims description 3
- 201000010208 Seminoma Diseases 0.000 claims description 3
- 230000008859 change Effects 0.000 claims description 3
- 230000002829 reductive effect Effects 0.000 claims description 3
- 238000007920 subcutaneous administration Methods 0.000 claims description 3
- 101000711846 Homo sapiens Transcription factor SOX-9 Proteins 0.000 claims description 2
- 206010062016 Immunosuppression Diseases 0.000 claims description 2
- 102100034204 Transcription factor SOX-9 Human genes 0.000 claims description 2
- 230000010261 cell growth Effects 0.000 claims description 2
- 230000012292 cell migration Effects 0.000 claims description 2
- 210000002919 epithelial cell Anatomy 0.000 claims description 2
- 230000028327 secretion Effects 0.000 claims description 2
- 102000004103 Glycogen Synthase Kinases Human genes 0.000 claims 4
- 230000004927 fusion Effects 0.000 claims 1
- 102000006240 membrane receptors Human genes 0.000 claims 1
- 238000011160 research Methods 0.000 abstract description 8
- 239000003814 drug Substances 0.000 abstract description 5
- 230000001172 regenerating effect Effects 0.000 abstract description 4
- 238000009509 drug development Methods 0.000 abstract description 3
- 208000017443 reproductive system disease Diseases 0.000 abstract description 2
- 230000008093 supporting effect Effects 0.000 description 17
- 238000012360 testing method Methods 0.000 description 13
- 241000699666 Mus <mouse, genus> Species 0.000 description 11
- 210000004698 lymphocyte Anatomy 0.000 description 11
- 230000001105 regulatory effect Effects 0.000 description 11
- 102000038624 GSKs Human genes 0.000 description 10
- 241000699670 Mus sp. Species 0.000 description 9
- 238000002474 experimental method Methods 0.000 description 9
- 210000001519 tissue Anatomy 0.000 description 9
- 239000003636 conditioned culture medium Substances 0.000 description 8
- 230000001850 reproductive effect Effects 0.000 description 8
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 7
- 229920000936 Agarose Polymers 0.000 description 7
- 230000001640 apoptogenic effect Effects 0.000 description 7
- 239000006285 cell suspension Substances 0.000 description 7
- 239000011521 glass Substances 0.000 description 7
- 239000012528 membrane Substances 0.000 description 7
- 239000000203 mixture Substances 0.000 description 7
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 150000001875 compounds Chemical class 0.000 description 6
- 238000001514 detection method Methods 0.000 description 6
- 230000005012 migration Effects 0.000 description 6
- 238000013508 migration Methods 0.000 description 6
- 150000003384 small molecules Chemical class 0.000 description 6
- 239000006228 supernatant Substances 0.000 description 6
- 238000011529 RT qPCR Methods 0.000 description 5
- 238000003501 co-culture Methods 0.000 description 5
- 238000010276 construction Methods 0.000 description 5
- 238000003125 immunofluorescent labeling Methods 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 230000004073 interleukin-2 production Effects 0.000 description 5
- 108020004999 messenger RNA Proteins 0.000 description 5
- 238000002156 mixing Methods 0.000 description 5
- 239000000047 product Substances 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 230000000920 spermatogeneic effect Effects 0.000 description 5
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 4
- 238000012449 Kunming mouse Methods 0.000 description 4
- 239000006180 TBST buffer Substances 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 239000011324 bead Substances 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- 238000011156 evaluation Methods 0.000 description 4
- 239000001963 growth medium Substances 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 108010082117 matrigel Proteins 0.000 description 4
- 230000035755 proliferation Effects 0.000 description 4
- 230000008439 repair process Effects 0.000 description 4
- 238000010186 staining Methods 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- 231100000027 toxicology Toxicity 0.000 description 4
- 238000005406 washing Methods 0.000 description 4
- 229920001817 Agar Polymers 0.000 description 3
- 102000000844 Cell Surface Receptors Human genes 0.000 description 3
- 238000000116 DAPI staining Methods 0.000 description 3
- 102000009024 Epidermal Growth Factor Human genes 0.000 description 3
- 101800003838 Epidermal growth factor Proteins 0.000 description 3
- 102000012673 Follicle Stimulating Hormone Human genes 0.000 description 3
- 108010079345 Follicle Stimulating Hormone Proteins 0.000 description 3
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 3
- 102000014429 Insulin-like growth factor Human genes 0.000 description 3
- 102100020873 Interleukin-2 Human genes 0.000 description 3
- 108010002350 Interleukin-2 Proteins 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 229930040373 Paraformaldehyde Natural products 0.000 description 3
- 206010074268 Reproductive toxicity Diseases 0.000 description 3
- 230000009471 action Effects 0.000 description 3
- 239000008272 agar Substances 0.000 description 3
- 239000000872 buffer Substances 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 230000029087 digestion Effects 0.000 description 3
- 238000009826 distribution Methods 0.000 description 3
- 238000007877 drug screening Methods 0.000 description 3
- 229940116977 epidermal growth factor Drugs 0.000 description 3
- 229940028334 follicle stimulating hormone Drugs 0.000 description 3
- 238000007490 hematoxylin and eosin (H&E) staining Methods 0.000 description 3
- 238000010166 immunofluorescence Methods 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 150000002632 lipids Chemical class 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 230000000242 pagocytic effect Effects 0.000 description 3
- 229920002866 paraformaldehyde Polymers 0.000 description 3
- 231100000614 poison Toxicity 0.000 description 3
- BHMBVRSPMRCCGG-OUTUXVNYSA-N prostaglandin D2 Chemical compound CCCCC[C@H](O)\C=C\[C@@H]1[C@@H](C\C=C/CCCC(O)=O)[C@@H](O)CC1=O BHMBVRSPMRCCGG-OUTUXVNYSA-N 0.000 description 3
- BHMBVRSPMRCCGG-UHFFFAOYSA-N prostaglandine D2 Natural products CCCCCC(O)C=CC1C(CC=CCCCC(O)=O)C(O)CC1=O BHMBVRSPMRCCGG-UHFFFAOYSA-N 0.000 description 3
- 238000003753 real-time PCR Methods 0.000 description 3
- 230000007696 reproductive toxicity Effects 0.000 description 3
- 231100000372 reproductive toxicity Toxicity 0.000 description 3
- 238000002791 soaking Methods 0.000 description 3
- 210000001082 somatic cell Anatomy 0.000 description 3
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 3
- 102000015735 Beta-catenin Human genes 0.000 description 2
- 108060000903 Beta-catenin Proteins 0.000 description 2
- 102000001805 Bromodomains Human genes 0.000 description 2
- 108050009021 Bromodomains Proteins 0.000 description 2
- 102000029816 Collagenase Human genes 0.000 description 2
- 108060005980 Collagenase Proteins 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- 102000018233 Fibroblast Growth Factor Human genes 0.000 description 2
- 108050007372 Fibroblast Growth Factor Proteins 0.000 description 2
- 206010016654 Fibrosis Diseases 0.000 description 2
- 108010051975 Glycogen Synthase Kinase 3 beta Proteins 0.000 description 2
- 102100038104 Glycogen synthase kinase-3 beta Human genes 0.000 description 2
- 241000282414 Homo sapiens Species 0.000 description 2
- 238000011579 SCID mouse model Methods 0.000 description 2
- 230000006052 T cell proliferation Effects 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 238000007664 blowing Methods 0.000 description 2
- 238000001516 cell proliferation assay Methods 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 238000004140 cleaning Methods 0.000 description 2
- 229960002424 collagenase Drugs 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 230000013020 embryo development Effects 0.000 description 2
- 238000010195 expression analysis Methods 0.000 description 2
- 229940126864 fibroblast growth factor Drugs 0.000 description 2
- 230000004761 fibrosis Effects 0.000 description 2
- 238000000799 fluorescence microscopy Methods 0.000 description 2
- 230000006870 function Effects 0.000 description 2
- 238000010230 functional analysis Methods 0.000 description 2
- 238000010353 genetic engineering Methods 0.000 description 2
- 238000002513 implantation Methods 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 235000016709 nutrition Nutrition 0.000 description 2
- 230000005305 organ development Effects 0.000 description 2
- 239000002574 poison Substances 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 231100000091 reproductive toxicant Toxicity 0.000 description 2
- 238000012163 sequencing technique Methods 0.000 description 2
- 210000001626 skin fibroblast Anatomy 0.000 description 2
- 238000007711 solidification Methods 0.000 description 2
- 230000008023 solidification Effects 0.000 description 2
- 230000021595 spermatogenesis Effects 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 231100000167 toxic agent Toxicity 0.000 description 2
- 239000003440 toxic substance Substances 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 230000009278 visceral effect Effects 0.000 description 2
- 238000001262 western blot Methods 0.000 description 2
- 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
- 238000008940 Alkaline Phosphatase assay kit Methods 0.000 description 1
- 201000002909 Aspergillosis Diseases 0.000 description 1
- 208000036641 Aspergillus infections Diseases 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 229920000742 Cotton Polymers 0.000 description 1
- 101150027068 DEGS1 gene Proteins 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 1
- 238000008157 ELISA kit Methods 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 108091029865 Exogenous DNA Proteins 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000003957 Fibroblast growth factor 9 Human genes 0.000 description 1
- 108090000367 Fibroblast growth factor 9 Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 108010009202 Growth Factor Receptors Proteins 0.000 description 1
- 102000009465 Growth Factor Receptors Human genes 0.000 description 1
- 101710088172 HTH-type transcriptional regulator RipA Proteins 0.000 description 1
- 108010003272 Hyaluronate lyase Proteins 0.000 description 1
- 102000001974 Hyaluronidases Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 102000003960 Ligases Human genes 0.000 description 1
- 108090000364 Ligases Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 101100096242 Mus musculus Sox9 gene Proteins 0.000 description 1
- 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 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- NPGIHFRTRXVWOY-UHFFFAOYSA-N Oil red O Chemical compound Cc1ccc(C)c(c1)N=Nc1cc(C)c(cc1C)N=Nc1c(O)ccc2ccccc12 NPGIHFRTRXVWOY-UHFFFAOYSA-N 0.000 description 1
- TURHTASYUMWZCC-UHFFFAOYSA-N Olaquindox [BAN:INN] Chemical compound C1=CC=C2N([O-])C(C)=C(C(=O)NCCO)[N+](=O)C2=C1 TURHTASYUMWZCC-UHFFFAOYSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 241000009328 Perro Species 0.000 description 1
- 206010057249 Phagocytosis Diseases 0.000 description 1
- 238000003559 RNA-seq method Methods 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 108010087230 Sincalide Proteins 0.000 description 1
- 241000282898 Sus scrofa Species 0.000 description 1
- 206010043276 Teratoma Diseases 0.000 description 1
- 206010059271 Testicular dysplasia Diseases 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- 101100096235 Xenopus laevis sox9-a gene Proteins 0.000 description 1
- 101100096236 Xenopus laevis sox9-b gene Proteins 0.000 description 1
- 210000001015 abdomen Anatomy 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 230000003698 anagen phase Effects 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 229910052793 cadmium Inorganic materials 0.000 description 1
- 229940044194 cadmium Drugs 0.000 description 1
- BDOSMKKIYDKNTQ-UHFFFAOYSA-N cadmium atom Chemical compound [Cd] BDOSMKKIYDKNTQ-UHFFFAOYSA-N 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 238000010609 cell counting kit-8 assay Methods 0.000 description 1
- 239000006143 cell culture medium Substances 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 239000002771 cell marker Substances 0.000 description 1
- 230000009391 cell specific gene expression Effects 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000008668 cellular reprogramming Effects 0.000 description 1
- 230000036755 cellular response Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 230000003399 chemotactic effect Effects 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 230000005757 colony formation Effects 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 238000001816 cooling Methods 0.000 description 1
- 238000010219 correlation analysis Methods 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 230000010454 developmental mechanism Effects 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 230000010595 endothelial cell migration Effects 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 229940088598 enzyme Drugs 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 210000001723 extracellular space Anatomy 0.000 description 1
- 230000035558 fertility Effects 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- 230000035876 healing Effects 0.000 description 1
- 238000007417 hierarchical cluster analysis Methods 0.000 description 1
- 235000003642 hunger Nutrition 0.000 description 1
- 229960002773 hyaluronidase Drugs 0.000 description 1
- 239000000815 hypotonic solution Substances 0.000 description 1
- 230000008004 immune attack Effects 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 239000012982 microporous membrane Substances 0.000 description 1
- 238000010232 migration assay Methods 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 210000002894 multi-fate stem cell Anatomy 0.000 description 1
- 230000025020 negative regulation of T cell proliferation Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 230000035764 nutrition Effects 0.000 description 1
- 229950010210 olaquindox Drugs 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 206010033675 panniculitis Diseases 0.000 description 1
- 239000012466 permeate Substances 0.000 description 1
- 230000008782 phagocytosis Effects 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 238000010791 quenching Methods 0.000 description 1
- 230000000171 quenching effect Effects 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 238000010814 radioimmunoprecipitation assay Methods 0.000 description 1
- 210000004994 reproductive system Anatomy 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 108090000064 retinoic acid receptors Proteins 0.000 description 1
- 102000003702 retinoic acid receptors Human genes 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 239000004017 serum-free culture medium Substances 0.000 description 1
- IZTQOLKUZKXIRV-YRVFCXMDSA-N sincalide Chemical compound C([C@@H](C(=O)N[C@@H](CCSC)C(=O)NCC(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(N)=O)NC(=O)[C@@H](N)CC(O)=O)C1=CC=C(OS(O)(=O)=O)C=C1 IZTQOLKUZKXIRV-YRVFCXMDSA-N 0.000 description 1
- 238000009999 singeing Methods 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 239000012192 staining solution Substances 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 210000004304 subcutaneous tissue Anatomy 0.000 description 1
- 230000003319 supportive effect Effects 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 230000034512 ubiquitination Effects 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 229910021642 ultra pure water Inorganic materials 0.000 description 1
- 239000012498 ultrapure water Substances 0.000 description 1
- 210000001835 viscera Anatomy 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
- 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
- C12N5/0681—Cells of the genital tract; Non-germinal cells from gonads
- C12N5/0683—Cells of the male genital tract, e.g. prostate, epididymis; Non-germinal cells from testis, e.g. Leydig cells, Sertoli cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/48—Reproductive organs
- A61K35/52—Sperm; Prostate; Seminal fluid; Leydig cells of testes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
- A61P15/08—Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5014—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5044—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
-
- 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
- C12N2501/06—Anti-neoplasic drugs, anti-retroviral drugs, e.g. azacytidine, cyclophosphamide
-
- 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
- C12N2501/30—Hormones
- C12N2501/38—Hormones with nuclear receptors
- C12N2501/385—Hormones with nuclear receptors of the family of the retinoic acid recptor, e.g. RAR, RXR; Peroxisome proliferator-activated receptor [PPAR]
-
- 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
- C12N2501/70—Enzymes
- C12N2501/72—Transferases [EC 2.]
- C12N2501/727—Kinases (EC 2.7.)
-
- 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
- C12N2503/00—Use of cells in diagnostics
- C12N2503/02—Drug screening
-
- 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
- C12N2506/00—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
- C12N2506/13—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
- C12N2506/1307—Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from adult fibroblasts
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2500/00—Screening for compounds of potential therapeutic value
- G01N2500/10—Screening for compounds of potential therapeutic value involving cells
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Reproductive Health (AREA)
- Cell Biology (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Urology & Nephrology (AREA)
- Molecular Biology (AREA)
- Hematology (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Zoology (AREA)
- Biochemistry (AREA)
- Organic Chemistry (AREA)
- Toxicology (AREA)
- Wood Science & Technology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Pathology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- General Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Gynecology & Obstetrics (AREA)
- Virology (AREA)
- Developmental Biology & Embryology (AREA)
- General Engineering & Computer Science (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Endocrinology (AREA)
Abstract
The present invention provides a method for reprogramming mammalian fibroblasts into testicular support cells (CiSCs), testicular support cells (CiSCs) obtained by the method and uses thereof. The method is easy to operate, does not introduce exogenous genes, is hopeful to be developed into a widely applied method for obtaining the non-testis-source support cells, not only provides a cell model and a theoretical basis for basic research and drug development of reproductive system diseases, but also brings wide prospects for application of testis support cells in the field of regenerative medicine.
Description
Technical Field
The invention relates to the technical fields of cell biology and tissue engineering, in particular to a method for reprogramming fibroblasts into testis support cells by using a small molecular compound and application thereof.
Background
Testis support cells (Sertoli cells) are one of the important somatic cells in testis, are an important component of seminiferous tubules, can provide physical support and stable microenvironment for spermatogenesis, provide nutritional support for spermatogenesis, and have unique immune-free characteristics so as to protect germ cells from body immune attack and foreign matter interference. The Sertoli cell has very wide application in the fields of basic research and clinical treatment, can be used as a model cell for in-vitro reproductive system related mechanism research and reproductive toxicology evaluation, and can be used as a seed cell for treating testicular dysplasia through participating in tissue reconstruction. In addition, the Sertoli cells play an important role in the field of regenerative medicine, and can provide nutrition and immune protection for non-testis cells after transplantation, thereby being beneficial to the survival of transplanted cells and reducing the rejection reaction of organisms. However, mature Sertoli cells are mitotically inactive, and long-term culture in vitro can lead to loss of their functional characteristics, and human Sertoli cells are very limited in availability, making large-scale culture in vitro difficult. Therefore, it is necessary to find an effective strategy to obtain replacement cells for Sertoli cells independent of donor testis cells.
Cell reprogramming is a research hotspot in the fields of tissue engineering and regenerative medicine, and the advent of this technology created new opportunities for drug screening, disease modeling, artificial organ development and cell therapies. There have been a number of studies demonstrating the overexpression of lineage specific transcription factors in somatic cells to induce direct transformation of differentiated cells into target cells, and although these studies open up promising approaches to obtain functional support cells, the risk of genomic insertion of foreign DNA sequences poses safety issues for their future use, which requires elimination of reliance on genetic manipulation. In recent years, more and more research has shown that the role and advantage of small molecules in cell fate reprogramming can overcome the limitations of genetic manipulation. And the small molecule compounds are easy to manufacture, preserve and standardize. However, there has been no report on the conversion of fibroblasts into supporting cells using a chemical reprogramming strategy.
Disclosure of Invention
The invention aims to provide a method for inducing fibroblast to be reprogrammed into testis support cells by using a small molecular compound, which has simple operation and high induction efficiency and provides an effective solution for the problems of insufficient testis support cell sources and the like in clinic.
Specifically, the present invention provides the following:
1. a method of reprogramming/inducing mammalian fibroblasts into testicular-like support cells (CiSCs), the method comprising the steps of:
(1) Treating the isolated mammalian fibroblasts with a BET bromodomain inhibitor; and
(2) The cells from step (1) are then further treated with a glycogen synthase kinase (GSK-3) inhibitor and Retinoic Acid (RA), thereby obtaining the testicular support cells (CiSCs).
The BET bromodomain inhibitors are a class of highly specific and potent BET inhibitors that function by competing binding to the bromodomain and BRD domain of the external terminal (BET) family of proteins. The BET family protein can be specifically combined with an activated chromatin structural domain, so that a cell specific gene expression mode is maintained, and the inhibition of the BET family protein can lead to the inactivation of a fibrosis growth factor receptor, so that a fibrosis transcription factor is inhibited, and the reprogramming of cells is facilitated.
The glycogen synthase kinase (GSK-3) inhibitor is a type of inhibitor which can reduce the targeted ubiquitination degradation of beta-catenin by inhibiting glycogen synthase kinase-3 beta (GSK-3 beta), so as to strengthen the Wnt/beta-catenin signal path, wherein the signal path can participate in regulating and controlling cell self-renewal, embryo development and organogenesis and plays a key role in maintaining pluripotency and in the reprogramming process of somatic cells.
Retinoic Acid (RA) is an agonist of retinoic acid receptor, and can improve the reprogramming Cheng Xiaolv and guide target cells to differentiate germ cells by regulating retinoic acid signal channels to participate in the processes of embryo development, germ system differentiation and the like.
2. The method according to 1 above, wherein the BET bromodomain inhibitor is I-BET151 and/or the glycogen synthase kinase (GSK-3) inhibitor is Riluzole (Riluzole).
3. The method according to 1 above, wherein in step (1) the BET bromodomain inhibitor is added during the logarithmic growth phase of the fibroblasts, the treatment time is preferably about 0.5 hours to 10 days, more preferably about 1-5 days, most preferably about 4 days; and/or wherein said step (2) is performed for about 10 to 40 days, preferably about 25 days.
4. The method according to 1 above, wherein the fibroblast is a primary fibroblast, preferably an embryonic fibroblast, visceral fibroblast or skin fibroblast; and/or the mammal is a human, mouse, rat, monkey, dog, cat, cow, rabbit, horse or pig.
5. The method according to 1 above, wherein the step (1) is performed in DMEM medium containing FBS; and/or said step (2) is performed in a testis-supporting cell induction medium, wherein said testis-supporting cell induction medium is DMEM medium comprising FBS, insulin-like growth factor, epidermal growth factor, fibroblast growth factor 9, follicle stimulating hormone and prostaglandin D2.
6. The method of any one of claims 1-5 above, further comprising:
1) Identifying expression of a testis support cell-related gene in said testis-like support cells (CiSCs), wherein said testis support cell-related gene is selected from the group consisting of Wt1, krt18, fshr, ar, gdnf and Amh;
2) Identifying expression of a testicular support cell Sertoli cell specific protein in the testicular support cell (CiSCs), wherein the testicular support cell Sertoli cell specific protein is selected from the group consisting of SOX9, WT1, ZO-1, and CX43;
3) Identifying expression of the Sertoli cell surface receptor protein FSHR and/or AR in said testicular support cells (CiSCs);
4) Identifying secretion of trophic factors GDNF and INHIBIN in said testicular support cells (CiSCs); and/or
5) Identifying a testis support cell-related biological function of the testis support cell (CiSCs), wherein the testis support cell-related biological function is selected from the group consisting of cell migration ability, cell aggregation ability, ability to recruit endothelial cells, ability to induce cells to form tubular structures, ability to construct a seminiferous tubule structure in vitro, ability to provide a growth microenvironment for seminoma cells, ability to phagocytose germ cells, immunosuppressive ability (inhibition of T cell proliferation and IL-2 production), ability to produce tubular structures in subcutaneous grafts, and role in cell testis transplantation.
7. Testis-like support cells (CiSCs) obtainable by the method according to any of the above 1-6.
8. Use of testicular support cells (CiSCs) according to 7 above in research of reproductive developmental mechanisms, reproductive toxicology assessment, drug development and/or in vivo transplantation therapy.
9. A kit for reprogramming mammalian fibroblasts to testicular-like support cells (CiSCs), comprising:
1) A BET bromodomain inhibitor, preferably I-BET151;
2) Glycogen synthase kinase (GSK-3) inhibitors, preferably Riluzole (Riluzole); and
3) Retinoic Acid (RA).
Use of a bet bromodomain inhibitor, a glycogen synthase kinase (GSK-3) inhibitor, and Retinoic Acid (RA) in the preparation of a kit or pharmaceutical composition for reprogramming mammalian fibroblasts to testicular-like support cells (CiSCs).
More specifically, the invention utilizes three small molecule compounds I-BET151, riluzole, and RA (IRR) to induce fibroblast reprogramming into testis support cells, comprising the following steps: the primary fibroblast obtained by separation is passaged into a 6-hole plate, and the number of cells in each hole is 1-3 multiplied by 10 5 Culturing in DMEM medium containing Fetal Bovine Serum (FBS) at volume concentration of 10%, and treating fibroblast with 10 μm I-BET151 for 4 days after cell growth to logarithmic phase, wherein cell morphology is changed from original long spindle shape to flat shape, and cell gap is reduced to show epithelial cell characteristics; then the medium was replaced with a Sertoli cell induction medium containing 3. Mu.M Retinoic Acid (RA) and 10. Mu.M Riluzole, after which the medium was replaced every 3 days for induction for 25 days to obtain testis-like support cells having testis support cell characteristics.
The primary fibroblasts may be extracted from animals and selected from embryonic fibroblasts, visceral fibroblasts or skin fibroblasts. Methods for isolating fibroblasts are routine in the art.
The Sertoli cell induction culture is preferably DMEM medium containing 10% FBS,50ng/mL insulin-like growth factor, 50ng/mL epidermal growth factor, 50ng/mL fibroblast growth factor 9,0.1 μg/mL follicle stimulating hormone and 0.5 μg/mL prostaglandin D2.
The invention detects testis Support Cell (SCs) marker genes and protein expression levels of testis Support Cell (SCs) obtained by reprogramming for 25 days, and evaluates biological functions of the CiSCs, thereby comprehensively identifying the development state of the CiSCs. The results indicate that CiSCs are not only capable of expressing supporting cell marker genes and proteins, but also have similar biological functions as Sertoli cells, including the ability to migrate, aggregate, spontaneously form a tubular network structure, maintain seminoma survival in vitro, phagocytize apoptotic spermatogenic cells, and immunosuppression. CiSCs are transplanted into the testes and subcutaneously of mice and can be involved in the formation of seminiferous tubules of the testes and in the reconstruction of ectopic tubular structures. The above results indicate that CiSCs exhibit primary testicular support cell status.
The reprogramming testis-like support cell obtained by the invention can be used as a cell model for researching reproductive development mechanism, evaluating reproductive toxicology and researching and developing medicines related to Sertoli cells, and can also be used as a seed cell for in vivo transplantation related to Sertoli.
The invention demonstrates that the fibroblast chemistry reprogramming is carried out to construct a testicle supporting cell model and the functions thereof, and has the following advantages:
1) The invention utilizes three small molecular compounds to induce the fibroblast to be reprogrammed into the testis support cell, avoids the exogenous DNA caused by gene operation from interfering with genome, and has simple operation and high induction efficiency.
2) The reprogramming method provided by the invention is direct reprogramming, does not pass through a multipotent stem cell stage, and avoids the teratoma risk and ethical problems caused by stem cells.
3) The reprogrammed testis-like supporting cell obtained by the invention is a functional testis supporting cell, has an expression profile and biological function similar to those of an endogenous supporting cell, can be used for drug screening and reproductive toxicity evaluation by taking the testis supporting cell as a target cell, and has a wide application prospect in clinical transplantation treatment related to Sertoli cells.
Drawings
FIG. 1, schematic representation of small molecule compound-induced reprogramming of fibroblasts into testis-supporting cells, and morphology of testis-like supporting cells after reprogramming (MEFs-mouse fibroblasts; ciSCs-testis-like supporting cells; SCs-testis supporting cells).
FIG. 2, ciSCs express Sertoli cell-specific genes and proteins. (A) q RT-PCR detects the expression of Sertoli cell-associated mRNA in CiSCs, MEFs and SCs, including Wt1, krt18, fshr, ar, gdnf and Amh. (B) Immunofluorescent staining examined the expression of Sertoli cell-specific proteins (SOX 9, WT1, ZO-1 and CX 43) in CiSCs, MEFs and SCs. Scale bar = 20 μm. (C) Western Blot analysis of the expression of Sertoli cell surface receptor proteins (FSHR and AR) in CiSCs. (D) ELISA analysis showed that CiSCs can secrete trophic factors including GDNF and INHIBIN.
FIG. 3, migration capability of CiSCs. (A-B) CiSCs healed scratches rapidly in the scratch test and had higher mobility within 12 hours and 24 hours. (C-D) Transwell migration experiments demonstrated that CiSCs had a greater number of transmembrane cells within 24 hours. Membrane pore size = 8 μm.
Fig. 4, ciSCs, have the ability to aggregate, attract endothelial cell migration and spontaneously form a tubular network. (A) Soft agar colony formation experiments verify the aggregation capacity of CiSCs. (B-C) mobility determination of HUVECs incubated in conditioned Medium collected from MEFs, ciSCs and SCs. (D) Ability of MEFs, ciSCs and SCs to form a network after 4 hours of incubation on Matrigel. (E-G) CiSCs spontaneously form 3D tubule-like structures in vitro. (E) in vitro tubular structure construction experiment figures. (F) Bright field and H & E staining from MEFs, ciSCs and SCs reconstructed tissue. Scale bar = 10 μm. (G) immunofluorescent staining of reconstructed tissue sections. ZO-1, green; DAPI, blue. Scale bar = 10 μm.
FIG. 5, ciSCs, are capable of supporting germ cell survival in vitro, phagocytic apoptotic germ cells, and inhibiting T cell proliferation and IL-2 production. (A-B) MIFARE and MIFARE cell number statistics after 6 days co-culture with primary isolated mouse MIFARE cells with MEFs, ciSCs and SCs as feeder layers. Scale bar, 100 microns. (C) Representative images of lipid droplets in MEFs, ciSCs and SCs 3 days after co-culture with apoptotic sperm producing cells. The ORO staining was used to assess the ability to phagocytose apoptotic cells. Scale bar = 20 μm. (D-E) analysis of proliferation levels of primary mouse lymphocytes treated with MEFs, ciSCs and SCs conditioned Medium, respectively, and ELISA was used to detect IL-2 production.
FIG. 6, ciSCs in vivo transplantation. (A) CiSCs xenografts produce tubule-like structures. Cell clusters of MEFs, ciSCs and SCs were transplanted subcutaneously into NOD/SCID mice, respectively. Three weeks after injection, the grafts were removed, fixed and H & E stained. (B) After testicular transplantation, ciSCs form a seminiferous tubule in cooperation with endogenous support cells. Fluorescently labeled cells (CFDA-SE) were injected into testes of 2 week old mice. When the mice were adult, testes were removed and frozen sections were made. Fluorescent cells were observed for distribution by fluorescence microscopy after DAPI staining. Scale bar = 20 μm.
FIG. 7, ciSCs expression profiling. (A) Venn diagram analysis of the subset of genes shared by CiSCs and SCs. (B) Statistics of the number of differentially expressed genes in MEFs, ciSCs and SCs (DEGs, FPKM values, fold change)>2). (C) Pearson correlation metric was used to compare the similarity of transcriptional profiles of MEFs, ciSCs and SCs. (D) RNA-seq data analysis of heat maps of differentially expressed genes in MEFs, ciSCs and SCs. Color scale represents log of gene expression 2 A scale. Red and blue represent up-and down-regulated genes, respectively. (E) The scatter plot shows that in CiSCs, most of the support cell markers are activated and the fibroblast markers are silenced. Normalization of FPKM values, i.e., log, of sample differential genes 2 (FPKM+1). (F) GO terms of the first 10 GO functional analyses in CISC compared to MEF. (G) qRT-PCR analysis of expression of Sertoli-related genes and fibroblast-related genes in MEFs, ciSCs and SCs.
FIG. 8, immunofluorescence assay to evaluate the response of CiSCs to reproductive toxicants. CiSC and SCs were treated with known reproductive poisons (IL-1 a, busulfan, cadmium and Olaquindox) or controls (0.1% dmso), respectively, for 48 hours. Immunofluorescent staining detects disruption of the reticulation and supporting cell-specific protein ZO-1 to assess reproductive toxicity. DAPI, blue; ZO-1, green. Scale bar = 100 μm.
Detailed Description
Unless otherwise indicated, terms used herein have the ordinary technical meaning as understood by those skilled in the art.
The invention is further illustrated in the following examples by reference to the accompanying drawings. These examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The reagents used in the following are all commercially available products unless otherwise indicated.
Example 1: small molecule compounds induce reprogramming of mouse fibroblasts into Sertoli cells
(1) Preparation of mouse fibroblasts (MEFs): selecting 13.5 day mouse embryo (Kunming mouse, purchased from Guangdong province animal center), removing head, tail, limbs and viscera, cutting the rest tissue, digesting with 0.25% trypsin for 10min, adding double volume of DMEM medium containing 10% FBS to stop digestion, repeatedly blowing to give single cell suspension, inoculating cell suspension into 10cm culture dish at 37deg.C, and 5% CO 2 Culturing for 4 hours, then replacing fresh culture medium for continuous culture, and carrying out passage once after 2 days to finally obtain the fibroblast (MEFs).
(2) Small molecule compounds induce reprogramming of mouse fibroblasts into testis support cells: mouse fibroblasts (MEFs) were passaged into 6-well plates with a cell number of 3X 10 per well 5 In this case, the cells were grown after 24 hours in DMEM medium containing 10% FBS by volume and grown to the logarithmic phase using 10. Mu.M of I-BET151 (C 23 H 21 N 5 O 3 GSK1210151A, available from Selleck company) treated fibroblasts for 4 days, then the medium was replaced with a Sertoli cell induction medium (containing 10% FBS,50ng/mL insulin-like growth factor, 50ng/mL epidermal growth factor, 50ng/mL fibroblast growth factor 9,0.1. Mu.g/mL follicle stimulating hormone and 0.5. Mu.g/mL prostaglandin D2 medium) containing 3. Mu.M Retinoic Acid (RA) (available from Selleck company) and 10. Mu.M Riluzole (Riluzole) (available from Selleck company), after which testicular support cells (CiSCs) having testicular support cell characteristics were obtained by replacing the medium once every 3 days for induction for 25 days.
The method for inducing mouse fibroblast to reprogram into testis support cell by small molecule compound is shown in figure 1, and the induced testis support cells are arranged in a rope shape with clear boundary, which is similar to the form of primary support cell SCs (separation method is described below).
Isolation of primary support cell SCs in mice: male Kunming mice were sacrificed at about 4 weeks of birth, and sterilized by soaking in 75% alcohol for 10min. The testis (without damaging the testis white membrane) was removed and placed in sterile PBS (containing 1% diab) pre-chilled in advance, and washed four times with PBS repeatedly; then, the testis membrane was torn off using replaced sterile forceps, the seminiferous tubules were removed, placed in a new bacterial dish containing sterile PBS, and repeatedly pulled with forceps to loosen the seminiferous tubules. The seminiferous tubules were then placed in a 50mL centrifuge bowl, 0.5mg/mL collagenase IV (purchased from Sigma) was added, digested with shaking at 37℃for 15min, and mixed well. After the tissue subsides naturally, the supernatant is discarded to remove the mesenchymal cells. Then, 5mL of collagenase IV (purchased from Sigma) at a concentration of 0.5mg/mL was added, and the mixture was allowed to stand and digest for 20 minutes, and the mixture was passed through a 100 μm filter membrane and washed with sterile PBS multiple times to remove myoid cells. Finally, the scraped tissue mass was placed in a 15mL centrifuge tube, 1mg/mL hyaluronidase 4mL was added, and the seminiferous tubules were completely dispersed by repeated blowing using a glass pipette. Digestion is carried out at 37℃for 5min and centrifugation is carried out at 1000rpm for 5min. The supernatant was discarded and resuspended in 5mL of DMEM medium containing 10% serum and diabody. The cell suspension was transferred to a 90mm cell culture dish, continuously cultured in an incubator at 37℃for 48 hours, the medium was discarded, and 0.1M Tris-HCl hypotonic solution was added for 2 minutes to remove sperm-like cells. Fresh DMEM complete medium is replaced, the culture is continued for 48 hours at 37 ℃, and the culture medium can be used for experiments after the cells grow fully.
Example 2: qRT-PCR detection of testicular support cell gene expression induced by small molecule compound
Mouse fibroblasts were induced to reprogram into testis-like support cells according to the procedure of example 1, ciSCs cells were collected, total RNA was extracted from the cells using HiPure Total RNA Mini Kit (R4111-02, magen), and 1 μg of RNA was reverse transcribed into cDNA using RT Master Mix (RR 036A, taKaRa), then cDNA samples were mixed with 2× ChamQ SYBR qPCR Green Master Mix (Q311-02, vazyme) and real-time quantitative PCR analysis was performed using CFX Connect real-time PCR detection system (Bio-rad). PCR was performed for 40 cycles using the primers described in Table 1. The relative expression levels were normalized to those of the internal control (GAPDH) and confirmed in at least 3 separate assays.
As shown in FIG. 2A, ciSCs are capable of expressing testis support cell-associated genes, including Wt1, krt18, fshr, ar, gdnf and Amh.
Example 3 Induction of expression of testis-like supporting cell proteins by Small molecules
Immunofluorescence detection of the seltoli cell-characteristic protein: testis-like support cells were obtained as in example 1, MEFs, ciSCs and SCs were spread on cell slide plates, respectively, and after incubation for 24 hours, the cell slide plates were fixed with 4% formaldehyde (PFA) for 10min, immersed in PBS buffer for 5min X3 times, 0.2% Triton X-100 was allowed to permeate for 30min, blocking buffer (P0102, beyotide) was incubated at room temperature for 1 hour, primary antibody (see Table 2) was added, incubated overnight at 4℃in a wet box, then washed with PBS buffer for 7min X3 times, and secondary antibody (see Table 2) (Goat Anti-Rabbit IgG H)&L(Alexa488 Available from Abcam corporation), incubated for 1 hour at room temperature, and observed by fluorescence microscopy.
As shown in FIG. 2B, ciSCs were able to express testis-supporting cell characteristic proteins, including WT1 (NP-659032.3), sox9 (NP-035578.3), ZO-1 ((NP-033412.2) and CX43 (NP-034418.1).
Western blot detection of Sertoli cell characteristic protein: ciSCs were obtained as in example 1, cellular proteins were extracted as usual, run on 12% sds-PAGE, and then transfer to a TBST buffer of 5% skim milk powder after transfer, blocking for 1 hour at room temperature, washing 5min×3 times with TBST buffer, adding primary antibody (from Abcam corporation), incubating overnight at 4 ℃, then washing 5min×3 times with TBST buffer, adding secondary antibody (from Bioworld corporation), incubating for 1 hour at room temperature, washing 5min×3 times with TBST buffer, and photographing with developer.
As shown in FIG. 2C, ciSCs were able to express testicular support cell surface receptor proteins, including FSHR (NP 038551.3) and AR (NP 038504.1).
Elisa assay Sertoli cell-associated secreted proteins: ciSCs were obtained as in example 1, and the supernatant of the cell culture was collected and subjected to lyophilization, followed by detection with GDNF test kit (from Jiangsu Nanjing institute of biological engineering, cat# H071) and INHB test kit (from Jiangsu Nanjing institute of biological engineering, cat# H320-2).
As a result, as shown in FIG. 2D, ciSCs are able to secrete trophic factors GDNF (NP 001288286.1) and INHIBIN (NP 032407.1).
Example 4 identification of testis support cell-related biological Functions of small molecule-induced testis-like support cells
Testis-like support cells (CiSCs) were obtained as in example 1 for identification of testis support cell-related biological functions.
1) Scratch experiments detect the migration ability of cells: the day before the experiment, a marker was used to draw parallel lines on the back of the 6-well plate to ensure that three parallel lines were run through each well. After the experimental cells were digested into single cell suspensions, the foregoing CiSCs, MEFs and SCs were mixed at 1X 10 6 The individual cell/well concentrations were cultured overnight in 6-well plates. The next day, cells were scraped with 200 μl of parallel lines drawn perpendicular to the bottom of the plate, washed twice with PBS to remove cell debris, and then placed in serum-free medium. Photography was performed at 0 and 24 hours after incubation, and the average distance of scratches at each time point was measured.
As a result, as shown in FIG. 3 at A, B, at 12h post-scratch, the scratch width of CiSCs was significantly narrowed relative to MEFs and the healing rate was relatively close to that of SCs, with MEFs scratch width did not change much as the experiment was extended to 24h, while the scratches of CiSCs and SCs were substantially healed.
2) Transwell migration experiments further verify the migration ability of CiSCs: taking logarithmic growth cells, discarding the culture medium, washing twice with PBS, adding a serum-free culture medium, and starving for culturing for 12-24 hours; after digestion, the cells were washed 1 to 2 times with PBS and resuspended in 1% BSA-containing medium at a cell concentration of 1 to 5X 10 5 100. Mu.L (2X 10) of the sample was taken out per mL 4 ) The cell suspension was added to the upper chamber of the Transwell chamber, and 500. Mu.L of a cell culture medium containing 10% FBS was added as the lower chamber to the 24-well plate, and cultured in an incubator at 37℃for 48 hours; taking out the cell, sucking off culture medium in the upper and lower chambers, wiping off cells inside the upper chamber with cotton bud, adding 500 μl of 4% paraformaldehyde into the lower chamber, fixing cells at room temperature for 15min, discarding the fixing solution, soaking in PBS, cleaning for 5min, adding into the lower chamberAdding 500 mu L of DAPI staining solution, staining for 10min at room temperature in dark, soaking and cleaning with ultrapure water for 5min, repeating for 3 times, dripping anti-fluorescence quenching agent on the glass slide, placing the cell on the glass slide, shooting under a fluorescence microscope, and calculating the cell number.
As a result, as shown in FIG. 3 at C, D, the number of migrating cells of CiSCs and SCs was significantly higher than MEFs, and the number of migrating cells of CiSCs was 3 times that of MEFs with statistically significant differences when three groups of transmembrane cells were counted.
3) Soft agar clone formation verifies cell aggregation ability: mixing sterile 5% agarose at 40deg.C with DMEM medium preheated at 37deg.C at a ratio of 1:9, adding the mixture into 12-well plate with a volume of 1mL per well, standing at room temperature for solidification, mixing single cell suspension with 0.5% agarose preheated at 40deg.C at equal volume, adding cell-agarose mixture into 12-well plate pre-coated with agar, and collecting cell number of 1×10 per well 4 After standing at room temperature until complete solidification, plates were placed in an incubator for cultivation for 24 hours, 200. Mu.L of medium was added to each well and replaced every other day, and cultivation was continued until colony appeared, and the colonies were stained with an alkaline phosphatase assay kit (available from Shanghai Biyun biotechnology Co., ltd., cat# product No. P0321S) and photographed to calculate the number of clones.
As shown in FIG. 4A, ciSCs and primary support cells formed spheroid aggregates, but MEFs failed to form cell aggregates.
4) Transwell assays induce the ability of cells to recruit endothelial cells: will be 2X 10 5 CiSCs, MEFs and SCs were grown in 1 well of a 6-well plate, and after 24 hours of culture, the original medium was replaced with serum-free DMEM, and the cells were further cultured for 48 hours, and then the supernatants were collected as conditioned medium, respectively. Will be 1X 10 4 Individual umbilical endothelial cells (HUVEC) (laboratory preservation) were seeded on a microporous membrane (8 mm) Boyden chamber containing serum-free DMEM in the upper part and 50% conditioned medium (600 μl total) in the lower part and incubated for 12 hours. Subsequently, cells in the inner chamber were wiped off while fixing the cells migrating through the membrane with 4% formaldehyde (PFA) at Room Temperature (RT) for 15 minutes and stained with DAPI at room temperature for 10 minutes. Migration measurement by computing DAPI blue signal for each imageAnd (3) cells.
As a result, as shown in FIG. 4 at B, C, after 24 hours of co-culture, the number of HUVECs attracted by SCs and CiSCs and passing through the underside of the membrane was 2-fold higher than MEFs.
5) Demonstration of the ability of induced cells to form tubular structures: matrigel (available from BD Co., U.S. under the trade name 356234) was mixed with DMEM medium containing 5% FBS in equal volume, spread on a pre-chilled 24-well plate, and coagulated in a 37℃incubator for 1 hr to obtain 1X 10 gel 6 The individual cells were seeded onto the coagulated matrigel, allowed to stand in an incubator for 4 hours, and the network structure was observed with a microscope and photographed.
As a result, both CiSCs and SCs can form a network on matrigel consistent with the onset of tubular formation, as shown in FIG. 4D.
6) 3D culture in vitro construction of a similar aspergillosis seminiferous tube structure: the cell suspensions were dropped onto the lid of the petri dish and incubated upside down at 34℃for 2 days, wherein each drop of suspension contained 1X 10 6 Individual cells/20. Mu.L, while mixing 20ml of sterile 0.75% agarose solution pre-heated at 40℃with an equal volume of 2 XDMEM medium, pouring into a petri dish and cooling to room temperature, and slicing the solidified mixture into 1cm 3 Is transferred to wells of a 24-well plate, the agarose blocks are immersed in DMEM complete medium, placed overnight in a 37 ℃ incubator to exchange nutrients in the blocks, then the cell clusters cultured in the suspension droplets for 48 hours are transferred to the surface of the agarose blocks, fresh medium is replaced to ensure that the liquid level is parallel to the upper surface of the agarose blocks, further cultured in a 34 ℃ incubator, the medium is updated every other day during which time, the cell clusters are collected every 14 days, 4% paraformaldehyde is fixed for 10 minutes at room temperature, then frozen sections of 10 μm thickness are made, HE staining observes the internal morphology of the cell clusters, immunofluorescent staining (agat Anti-rabit IgG H&L(Alexa488, available from Abcam corporation) detects the expression of ZO-1 (np_ 033412.2) in the cell cluster.
As a result, as shown in FIG. 4 at E, F, G, ciSCs and SCs show a pronounced tendency for cell mass growth relative to the radioactive, loose structure of MEFs and develop a relatively compact sphere structure. After HE staining, it was observed that the mass formed by CiSCs appeared to have a relatively smooth and dense exogenous profile, the internal cell shape had a chemotactic cord structure, and a distinct network structure was observed by enlarging the cord, which was quite similar to the state of SCs. Upon immunofluorescent staining, cell regularity interconnections can be observed in the cell mass composed of CiSCs, and ZO-1 protein forms a basal membrane around the cell chain.
7) CiSCs co-culture with spermatogonia: ciSCs, MEFs and SCs were treated with medium containing 10. Mu.g/ml mitomycin for 3 hours and the treated cells were used as feeder cells at 5X 10 5 The number of individual cells/well was inoculated into 6-well plates coated with 1% gelatin and cultured for 24 hours. The sperm cells separated for 24 hours were then isolated according to 1X 10 6 The ratio of/well was seeded into 6-well plates with feeder cells laid out, and the culture was continued in an incubator during which the medium was refreshed every 2 days, and after 6 days of culture, the growth of the sperm cells was observed and photographed in a bright field.
8) Separation of spermatogonia: in example 1, primary supporting cells were isolated and cultured, after 48 hours of primary culture, spermatogenic cells suspended in the culture solution were collected, transferred to a new cell culture dish, and cultured at 37℃for 2 days to obtain spermatogenic cells;
as a result, as shown in FIG. 5 at A, B, after 6 days of co-culture with sperm cells, a greater number of spermatogenic cell clusters were observed in CiSCs than in MEFs, and there was no significant difference in the number of cell clusters between CiSCs and endogenous supporting cells. The results demonstrate that CiSCs can provide a better microenvironment for the survival of sperm cells than MEFs.
8) Phagocytic capacity identification of CiSCs: the primary support cells were isolated 24 hours after, the spermatogenic cells suspended in the medium were collected and cultured for 3 days in the absence of feeder layers to induce spontaneous apoptosis, and the apoptotic cells were washed with 1 XPBS and resuspended in DMEM for phagocytosis assay. CiSCs, MEFs and SCs were combined at 5X 10 4 Individual cells/well were seeded on glass slides in 24 well plates and after 24 hours of incubation, apoptotic sperm cells were grown at 1X 10 6 Adding individual cells/wells into well plate, and co-culturing spermatogonial cells with CiSCsAfter 3 days of culture, apoptotic cells were removed and cells plated on glass coverslips were stained with ORO.
As a result, as shown in FIG. 5C, a large number of red-colored lipid droplets appeared in CiSCs and SCs after oil red O staining, while few lipid droplets were observed in MEFs, indicating that the phagocytic activity of CiSCs was significantly higher than that of MEFs.
9) Identification of immunosuppressive Capacity of CiSCs: the immunosuppressive ability of cells was assessed by inhibiting lymphocyte proliferation and interleukin-2 production. Will be 2X 10 5 CiSCs, MEFs and SCs were planted in one well of a 6-well plate, cultured in an incubator at 37℃for 24 hours, replaced with serum-free DMEM, and after further culturing for 48 hours, the supernatants were collected as conditioned medium, respectively. For lymphocyte proliferation assays, conditioned medium was diluted to 20%, 40%, 60% and 80% with DMEM medium, and 10% pbs was added, respectively. Lymphocytes were isolated from spleens of male Kunming mice (purchased from the animal center in Guangdong province) of 6 to 8 weeks old, mixed with a conditioned medium of prescribed concentration, inoculated into 96-well plates at 2X 10 cells/well, placed at 37℃for further 48 hours, and proliferation of lymphocytes was measured by CCK-8 kit (purchased from MCE Co., ltd., cat# HY-K030). The production of interleukin-2 in lymphocytes was analyzed by mixing lymphocytes with respective 40% medium under different cell conditions at a concentration of 1X 10 6 Individual cells/wells were seeded in 6-well plates and cultured for 2 days, lymphocytes were collected and lysed with RIPA cell lysate, and the concentration of IL-2 in the lymphocyte lysate was determined by ELISA kit (ex institute of bioengineering, build H003, from singeing, south of the Jiangsu).
The results are shown in figure 5 as D, E; compared to MEFs conditioned medium, the medium from CiSCs or SCs was able to significantly inhibit lymphocyte proliferation and the IL-2 levels in lymphocytes were low, suggesting that CiSCs could inhibit T cell proliferation and IL-2 production in vitro.
Example 5: in vivo transplantation of small molecule-induced testicular support cells
Testicular support cells (CiSCs) were obtained as in example 1 for in vivo transplantation experiments.
(1) The creation of tubular structures within subcutaneous implants: MEFs, ciSCs and SCs clusters were subcutaneously injected into the backs of NOD/SCID mice (purchased from animal centers, guangdong province) and kept for three weeks, subcutaneous tissue blocks were removed, 4% paraformaldehyde was fixed, frozen sections were prepared, and tissue structures were observed by HE staining.
As a result, as shown in FIG. 6A, the formation of tubular structures was observed in grafts of CiSCs and SCs 3 weeks after transplantation, as compared with MEFs.
(2) Cell testis transplantation: cells were transplanted into testes of new 10 day old Kunming mice (purchased from animal centers in Guangdong province) to identify whether the cells were involved in the construction of testis seminiferous tubules. Using CFSE cell proliferation assay and tracking kit (Kaiyi, cat# KGA 318) fluorescence labeled MEFs, ciSCs and SCs, after anesthetizing the mammalian mice, a wound of about 1cm was excised from the lower abdomen of the mammalian mice, and the left testis was gently pulled out with forceps, under a stereo microscope, 1X 10 was performed with glass microneedles 6 Fluorescently labeled cells were injected into the testes, the testes injected with labeled cells were placed back into the abdominal cavity, then the incision was sutured and sterilized with iodophor, and when recipient mice were awake, they were placed back in the cage and housed with the master mice. One month after implantation, the recipient mice developed to maturity, testes were obtained and frozen into sections, and after DAPI staining, the distribution of green fluorescent cells in the testes was observed and photographed.
As a result, as shown in FIG. 6B, two weeks after implantation, ciSCs integrated into and colonized the basal portion of the recipient's seminiferous tubules. In contrast, MEFs are distributed between the seminiferous tubules.
EXAMPLE 6 expression profiling
Testis-like supporting cells (CiSCs) were obtained as in example 1, cells were collected, total RNA was extracted by Trizol method, and the quality and concentration of the samples were checked using Nano Drop ND-1000RNA (Invitrogen, cat# 15596018), requiring A260/A280 to be 1.8 or more and A260/A230 to be 1.5 or more in quality control, ensuring that the samples were free from contamination, and checking RIN value of the samples to determine RNA integrity, wherein RIN to be 7.0 indicates that the sample integrity was good. Determination of RNA DNA from Total RNA was digested with DNase I and the digested product was purified with magnetic beads. For eukaryotes, mRNA was enriched with Oligo (dT) beads and for prokaryotes, rRNA was removed with a kit to obtain enriched mRNA. Uniformly mixing mRNA with a proper amount of breaking reagent, breaking under the action of high temperature, and carrying out reverse transcription by using segmented mRNA as a template and using a six-base random primer to synthesize double-stranded cDNA; end repair, polyA addition, sequencing linker addition: the end repair complex enzyme performs end repair on the double-stranded cDNA. The repair product was purified with magnetic beads, end-added with A under the action of polymerase and ligated with sequencing adaptors under the action of ligase. The ligation products were amplified by configuring a PCR reaction system and purified using magnetic beads, and quality and yield were detected by Agilent 2100 Bioanalyzer and real-time fluorescent quantitative PCR, and sequenced on-machine until the library construction was completed.
The results are shown in FIG. 7: the Venn diagram analysis showed that 16982 genes were shared between CiSCs and SCs (FIG. 7, A). The results of differential gene number analysis showed that a total of 4552 genes were differentially expressed between CiSCs and MEFs, including 2337 genes up-regulated and 2175 genes down-regulated, while 4818 genes were differentially expressed between SCs and MEFs, including 2618 up-regulated and 2200 down-regulated genes (fig. 7, b). Person correlation and hierarchical clustering analysis showed that CiSCs aggregated with endogenous Sertoli cells (fig. 7, c). Compared with MEFs, 466 genes in CiSCs are up-regulated, 748 genes are down-regulated, and the expression level is changed by more than 2 times. The global gene expression pattern of CiSCs is more similar to that of adult support cells (7,D). The scatter plot shows that genes up-regulated in CiSCs are significantly enriched in SCs, while down-regulated genes are enriched in MEFs (fig. 7, e). GO functional analysis showed that differential genes are associated with supporting cellular function such as cell surface, extracellular space, extracellular matrix and cell attachment (figure 7,F). Consistent with the expression profile, qRT-PCR further showed that the Sertoli cell-associated gene was strongly activated in CiSCs, while the amount of fibroblast marker gene expression was significantly reduced, and the global gene expression pattern of CiSCs was similar to that of mature support cells, unlike that of primary fibroblasts (fig. 7,G).
Example 7 application of small molecule induced testis-like support cells in evaluation of reproduction toxicity
According to realityEXAMPLE 1 procedure Induction of reprogramming of mouse fibroblasts into testis-like support cells at 1X 10 5 The individual cells/wells were seeded on glass slides in 24 well plates, placed in a 37℃incubator for 24 hours, the original medium was replaced with medium containing different reproductive toxicants, and the culture was continued for 48 hours, the supernatant was discarded, and the cell response to reproductive toxic substances was judged by immunofluorescence detection of the expression of Zona Occludes 1 protein (ZO-1) in the cells. The final solvent concentration was consistent under all conditions and all compounds tested were reported to have reproductive toxicity. Details of the test compounds are listed in table 3.
As shown in FIG. 8, ciSCs showed a broken ZO-1 distribution pattern after treatment with a reproduction toxic substance, and ZO-1 expression was significantly disrupted with no significant difference in the degree of disruption of the primary testicular supportive cytotoxicant. These findings indicate that induced class of support cells (CiSCs) are sensitive to reproductive poisons and can be an alternative model for evaluating the deleterious effects of substances on male fertility.
It will be appreciated by persons skilled in the art that although the invention has been specifically described with reference to the above embodiments, the invention is not limited to these specific embodiments. Based on the methods and technical solutions taught by the present invention, those skilled in the art can make appropriate modifications or improvements without departing from the spirit of the present invention, and the equivalent embodiments thus obtained are within the scope of the present invention.
TABLE 1 quantitative PCR primers
TABLE 2 antibody information
TABLE 3 information on reproduction toxic Compounds
Industrial applicability
The invention demonstrates the construction of the testicle support cell model prepared by the reprogramming method and the application thereof, and proves that the constructed cell model can be used as a substitute model for researching the physiological function of the testicle support cell, so as to be applied to mechanism research, drug screening and toxicology evaluation which take the testicle support cell as a target cell, and the constructed cell can also be used in the related transplantation treatment which takes the testicle support cell as a seed cell. The method is easy to operate and high in efficiency, does not introduce exogenous genes, is hopeful to be developed into a widely applied method for obtaining the non-testis-source support cells, not only provides a cell model and a theoretical basis for basic research and drug development of reproductive system diseases, but also brings wide prospects for application of testis support cells in the field of regenerative medicine.
Claims (2)
1. A method of reprogramming/inducing mammalian fibroblasts into testicular-like support cells (CiSCs), the method comprising the steps of:
(1) Treating isolated mammalian fibroblasts with a BET bromodomain inhibitor to change the morphology of the cells from original long fusions to flattened shapes; and
(2) Further treating the cells from step (1) with a glycogen synthase kinase (GSK-3) inhibitor and Retinoic Acid (RA) to thereby obtain said testicular support cells (CiSCs), wherein said BET bromodomain inhibitor is I-BET151 and said glycogen synthase kinase (GSK-3) inhibitor is Riluzole,
wherein the mammalian fibroblast is a mouse embryonic fibroblast,
the method comprises the following specific steps: the primary fibroblast obtained by separation is passaged into a 6-hole plate, and the number of cells in each hole is 1-3 multiplied by 10 5 Culturing in DMEM medium containing Fetal Bovine Serum (FBS) at volume concentration of 10%, and treating fibroblast with 10 μm I-BET151 for 4 days after cell growth to logarithmic phase, wherein cell morphology is changed from original long spindle shape to flat shape, and cell gap is reduced to show epithelial cell characteristics; then the medium was replaced with a Sertoli cell induction medium containing 3. Mu.M Retinoic Acid (RA) and 10. Mu.M Riluzole, after which the medium was replaced every 3 days for induction for 25 days to obtain testis-like support cells having testis support cell characteristics.
2. The method of claim 1, the method further comprising:
1) Identifying expression of a testis support cell-related gene in said testis-like support cells (CiSCs), wherein said testis support cell-related gene is selected from the group consisting of Wt1, krt18, fshr, ar, gdnf and Amh;
2) Identifying expression of a testicular support cell Sertoli cell specific protein in the testicular support cell (CiSCs), wherein the testicular support cell Sertoli cell specific protein is selected from the group consisting of SOX9, WT1, ZO-1, and CX43;
3) Identifying expression of the Sertoli cell surface receptor protein FSHR and/or AR in said testicular support cells (CiSCs);
4) Identifying secretion of trophic factors GDNF and INHIBIN in said testicular support cells (CiSCs); and/or
5) Identifying a testis support cell-related biological function of the testis support cell (CiSCs), wherein the testis support cell-related biological function is selected from the group consisting of cell migration ability, cell aggregation ability, ability to recruit endothelial cells, ability to induce cells to form tubular structures, ability to construct a seminiferous tubule structure in vitro, ability to provide a growth microenvironment for seminoma cells, ability to phagocytose germ cells, immunosuppression ability, ability to produce tubular structures in subcutaneous grafts, and role in cell testis transplantation.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202110275603.2A CN115074307B (en) | 2021-03-15 | 2021-03-15 | Method for reprogramming fibroblast induced by small molecule compound into testis support cell and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202110275603.2A CN115074307B (en) | 2021-03-15 | 2021-03-15 | Method for reprogramming fibroblast induced by small molecule compound into testis support cell and application thereof |
Publications (2)
Publication Number | Publication Date |
---|---|
CN115074307A CN115074307A (en) | 2022-09-20 |
CN115074307B true CN115074307B (en) | 2024-01-09 |
Family
ID=83241384
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202110275603.2A Active CN115074307B (en) | 2021-03-15 | 2021-03-15 | Method for reprogramming fibroblast induced by small molecule compound into testis support cell and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN115074307B (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN106636210A (en) * | 2016-10-09 | 2017-05-10 | 广州暨南大学医药生物技术研究开发中心 | Method for inducing transdifferentiation of fibroblast into similar testicular interstitial cells by combination of transcription factors |
CN109706114A (en) * | 2019-02-21 | 2019-05-03 | 吉林大学 | A kind of method that bull testis sertoli cell induces multi-potent stem cell |
WO2020006675A1 (en) * | 2018-07-03 | 2020-01-09 | 清华大学 | Method for in vitro reprogramming of fibroblasts into sertoli cells and application thereof |
Family Cites Families (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP6938154B2 (en) * | 2014-11-07 | 2021-09-22 | 国立大学法人大阪大学 | Differentiation-inducing cell population from which undifferentiated cells have been removed, its utilization and its production method |
KR101986830B1 (en) * | 2017-09-07 | 2019-06-07 | 차의과학대학교 산학협력단 | Sertoli-like cells from stem cells, a method for preparing thereof, and uses thereof |
-
2021
- 2021-03-15 CN CN202110275603.2A patent/CN115074307B/en active Active
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN106636210A (en) * | 2016-10-09 | 2017-05-10 | 广州暨南大学医药生物技术研究开发中心 | Method for inducing transdifferentiation of fibroblast into similar testicular interstitial cells by combination of transcription factors |
WO2020006675A1 (en) * | 2018-07-03 | 2020-01-09 | 清华大学 | Method for in vitro reprogramming of fibroblasts into sertoli cells and application thereof |
CN109706114A (en) * | 2019-02-21 | 2019-05-03 | 吉林大学 | A kind of method that bull testis sertoli cell induces multi-potent stem cell |
Non-Patent Citations (2)
Title |
---|
"培养原始生殖细胞的新方法——Sertoli细胞的应用";路欣 等;《首都医科大学学报》;第25卷(第1期);第15-18页 * |
"Reprogramming by De-bookmarking the Somatic Transcriptional Program through Targeting of BET Bromodomains";Zhicheng Shao 等;《Cell Reports》;第16卷;第3138–3145页 * |
Also Published As
Publication number | Publication date |
---|---|
CN115074307A (en) | 2022-09-20 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Zhang et al. | Long-term propagation of porcine undifferentiated spermatogonia | |
EP3223831B1 (en) | Methods for generation of podocytes from pluripotent stem cells and cells produced by the same | |
US9151744B2 (en) | Lung tissue model | |
Li et al. | Generation of offspring-producing 3D ovarian organoids derived from female germline stem cells and their application in toxicological detection | |
KR20100093513A (en) | Isolation, characterization and propagation of germline stem cells | |
Mohamadi et al. | Comparison of colony formation in adult mouse spermatogonial stem cells developed in Sertoli and STO coculture systems | |
US9468656B2 (en) | Stem cell preparations and methods of use | |
Lidgerwood et al. | Defined medium conditions for the induction and expansion of human pluripotent stem cell-derived retinal pigment epithelium | |
CN113767167A (en) | In vivo generation of functional and patient-specific thymus tissue from induced pluripotent stem cells | |
Mohammadzadeh et al. | Differentiation of spermatogonial stem cells by soft agar three-dimensional culture system | |
KR101562366B1 (en) | Scaffold for inducing myocardiocyte differentiation compring methacrylated gelatin | |
WO2017100498A1 (en) | Isolation of fusion-competent myoblasts and therapeutic applications thereof related to muscular dystrophy | |
Loffet et al. | Pluripotent stem cell derived intestinal organoids with an enteric nervous system | |
Hemadi et al. | Use of alginate hydrogel to improve long-term 3D culture of spermatogonial stem cells: stemness gene expression and structural features | |
JP7274683B2 (en) | METHOD FOR GENERATING KIDNEY CONSTRUCTION WITH DENDRITIC DRANGED COLLECTING ductS FROM PLIPOTENTIAL STEM CELLS | |
KR20120006386A (en) | Stem cell derived from first trimester placenta and cellular therapeutic agents comprising the same | |
Kwak et al. | Development of pluripotent stem cell-derived epidermal organoids that generate effective extracellular vesicles in skin regeneration | |
KR101760239B1 (en) | Method for isolating primary mesenchymal stem cells derived from human embryonic stem cells using cell insert culture system | |
CN115074307B (en) | Method for reprogramming fibroblast induced by small molecule compound into testis support cell and application thereof | |
CN111836884B (en) | Method for producing lacrimal gland tissue derived from stem cells | |
AU2014202292B2 (en) | Stem cell preparations and methods of use | |
Baazm et al. | Effects of different Sertoli cell types on the maintenance of adult spermatogonial stem cells in vitro | |
Johnen et al. | Presence of xenogenic mouse RNA in RPE and IPE cells cultured on mitotically inhibited 3T3 fibroblasts | |
Yang et al. | Small‐Molecule‐Driven Direct Reprogramming of Fibroblasts into Functional Sertoli‐Like Cells as a Model for Male Reproductive Toxicology | |
US20220195395A1 (en) | Physiologic growth of cultured intestinal tissue |
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 | ||
GR01 | Patent grant | ||
GR01 | Patent grant |