CN117257837A - Application of exosome in preparing medicine for treating muscular atrophy caused by type 2 diabetes - Google Patents
Application of exosome in preparing medicine for treating muscular atrophy caused by type 2 diabetes Download PDFInfo
- Publication number
- CN117257837A CN117257837A CN202310759269.7A CN202310759269A CN117257837A CN 117257837 A CN117257837 A CN 117257837A CN 202310759269 A CN202310759269 A CN 202310759269A CN 117257837 A CN117257837 A CN 117257837A
- Authority
- CN
- China
- Prior art keywords
- exosome
- exosomes
- diabetes
- umbilical cord
- mesenchymal stem
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 210000001808 exosome Anatomy 0.000 title claims abstract description 93
- 201000000585 muscular atrophy Diseases 0.000 title claims abstract description 61
- 208000001072 type 2 diabetes mellitus Diseases 0.000 title claims abstract description 29
- 239000003814 drug Substances 0.000 title claims abstract description 25
- 210000002901 mesenchymal stem cell Anatomy 0.000 claims abstract description 62
- 206010028289 Muscle atrophy Diseases 0.000 claims abstract description 55
- 210000003954 umbilical cord Anatomy 0.000 claims abstract description 47
- 230000020763 muscle atrophy Effects 0.000 claims abstract description 39
- 208000008589 Obesity Diseases 0.000 claims abstract description 20
- 230000001404 mediated effect Effects 0.000 claims abstract description 20
- 235000020824 obesity Nutrition 0.000 claims abstract description 20
- 230000004900 autophagic degradation Effects 0.000 claims abstract description 19
- 108010014380 Autophagy-Related Protein-1 Homolog Proteins 0.000 claims abstract description 16
- 230000002708 enhancing effect Effects 0.000 claims abstract description 14
- 238000002360 preparation method Methods 0.000 claims abstract description 12
- 102100036009 5'-AMP-activated protein kinase catalytic subunit alpha-2 Human genes 0.000 claims abstract 7
- 101000783681 Homo sapiens 5'-AMP-activated protein kinase catalytic subunit alpha-2 Proteins 0.000 claims abstract 7
- 102100039988 Serine/threonine-protein kinase ULK1 Human genes 0.000 claims abstract 3
- 238000000034 method Methods 0.000 claims description 34
- 210000001087 myotubule Anatomy 0.000 claims description 20
- 210000004027 cell Anatomy 0.000 claims description 15
- 239000002609 medium Substances 0.000 claims description 14
- 239000006228 supernatant Substances 0.000 claims description 14
- 210000003875 slow muscle fiber Anatomy 0.000 claims description 12
- 230000026731 phosphorylation Effects 0.000 claims description 10
- 238000006366 phosphorylation reaction Methods 0.000 claims description 10
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 claims description 10
- 210000001519 tissue Anatomy 0.000 claims description 10
- 238000012258 culturing Methods 0.000 claims description 7
- 108091003079 Bovine Serum Albumin Proteins 0.000 claims description 6
- 239000012091 fetal bovine serum Substances 0.000 claims description 6
- 229930182555 Penicillin Natural products 0.000 claims description 5
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 claims description 5
- 239000001963 growth medium Substances 0.000 claims description 5
- 238000004519 manufacturing process Methods 0.000 claims description 5
- 229940049954 penicillin Drugs 0.000 claims description 5
- 229960005322 streptomycin Drugs 0.000 claims description 5
- 230000004927 fusion Effects 0.000 claims description 3
- 230000000694 effects Effects 0.000 abstract description 10
- 229940079593 drug Drugs 0.000 abstract description 3
- 241000699670 Mus sp. Species 0.000 description 40
- 102000014156 AMP-Activated Protein Kinases Human genes 0.000 description 32
- 108010011376 AMP-Activated Protein Kinases Proteins 0.000 description 32
- 239000002953 phosphate buffered saline Substances 0.000 description 25
- 210000003205 muscle Anatomy 0.000 description 24
- 238000012360 testing method Methods 0.000 description 22
- 238000002347 injection Methods 0.000 description 20
- 239000007924 injection Substances 0.000 description 20
- 241000699666 Mus <mouse, genus> Species 0.000 description 17
- 238000004458 analytical method Methods 0.000 description 17
- 230000000052 comparative effect Effects 0.000 description 17
- 206010012601 diabetes mellitus Diseases 0.000 description 16
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 16
- 238000010172 mouse model Methods 0.000 description 16
- 235000009200 high fat diet Nutrition 0.000 description 14
- 102000016956 Autophagy-Related Protein-1 Homolog Human genes 0.000 description 13
- 102100040669 F-box only protein 32 Human genes 0.000 description 12
- 101710191029 F-box only protein 32 Proteins 0.000 description 12
- IPCSVZSSVZVIGE-UHFFFAOYSA-M hexadecanoate Chemical compound CCCCCCCCCCCCCCCC([O-])=O IPCSVZSSVZVIGE-UHFFFAOYSA-M 0.000 description 12
- 238000003119 immunoblot Methods 0.000 description 12
- 108020004459 Small interfering RNA Proteins 0.000 description 11
- 238000007912 intraperitoneal administration Methods 0.000 description 11
- FSASIHFSFGAIJM-UHFFFAOYSA-N 3-methyladenine Chemical compound CN1C=NC(N)=C2N=CN=C12 FSASIHFSFGAIJM-UHFFFAOYSA-N 0.000 description 10
- 238000012744 immunostaining Methods 0.000 description 10
- 230000001965 increasing effect Effects 0.000 description 10
- 108090000623 proteins and genes Proteins 0.000 description 10
- 230000002829 reductive effect Effects 0.000 description 10
- 102100025014 E3 ubiquitin-protein ligase TRIM63 Human genes 0.000 description 9
- 101710164910 E3 ubiquitin-protein ligase TRIM63 Proteins 0.000 description 9
- 102100024178 Microtubule-associated proteins 1A/1B light chain 3A Human genes 0.000 description 9
- 230000014509 gene expression Effects 0.000 description 9
- 239000000243 solution Substances 0.000 description 9
- ZPBYVFQJHWLTFB-UHFFFAOYSA-N 3-methyl-7H-purin-6-imine Chemical compound CN1C=NC(=N)C2=C1NC=N2 ZPBYVFQJHWLTFB-UHFFFAOYSA-N 0.000 description 8
- 102000004877 Insulin Human genes 0.000 description 8
- 108090001061 Insulin Proteins 0.000 description 8
- 229940125396 insulin Drugs 0.000 description 8
- 230000004069 differentiation Effects 0.000 description 7
- 102000004169 proteins and genes Human genes 0.000 description 7
- 210000003462 vein Anatomy 0.000 description 7
- 238000007446 glucose tolerance test Methods 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 210000003141 lower extremity Anatomy 0.000 description 6
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 239000008103 glucose Substances 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 230000002503 metabolic effect Effects 0.000 description 5
- 210000002027 skeletal muscle Anatomy 0.000 description 5
- 206010003694 Atrophy Diseases 0.000 description 4
- 206010022489 Insulin Resistance Diseases 0.000 description 4
- 238000003559 RNA-seq method Methods 0.000 description 4
- 239000000427 antigen Substances 0.000 description 4
- 102000036639 antigens Human genes 0.000 description 4
- 108091007433 antigens Proteins 0.000 description 4
- 230000037444 atrophy Effects 0.000 description 4
- 239000012822 autophagy inhibitor Substances 0.000 description 4
- 230000007747 autophagy signaling pathway Effects 0.000 description 4
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 4
- 230000000903 blocking effect Effects 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 210000003414 extremity Anatomy 0.000 description 4
- 239000007758 minimum essential medium Substances 0.000 description 4
- 239000000203 mixture Substances 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 201000006938 muscular dystrophy Diseases 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 230000003827 upregulation Effects 0.000 description 4
- 241000283707 Capra Species 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 238000011529 RT qPCR Methods 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000001737 promoting effect Effects 0.000 description 3
- 210000002966 serum Anatomy 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 2
- HSTOKWSFWGCZMH-UHFFFAOYSA-N 3,3'-diaminobenzidine Chemical compound C1=C(N)C(N)=CC=C1C1=CC=C(N)C(N)=C1 HSTOKWSFWGCZMH-UHFFFAOYSA-N 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 102100025222 CD63 antigen Human genes 0.000 description 2
- 102100027221 CD81 antigen Human genes 0.000 description 2
- 102100037904 CD9 antigen Human genes 0.000 description 2
- 101100447432 Danio rerio gapdh-2 gene Proteins 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 101150112014 Gapdh gene Proteins 0.000 description 2
- 101000934368 Homo sapiens CD63 antigen Proteins 0.000 description 2
- 101000914479 Homo sapiens CD81 antigen Proteins 0.000 description 2
- 101000738354 Homo sapiens CD9 antigen Proteins 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 239000002033 PVDF binder Substances 0.000 description 2
- 229930040373 Paraformaldehyde Natural products 0.000 description 2
- 108091030071 RNAI Proteins 0.000 description 2
- 229910000831 Steel Inorganic materials 0.000 description 2
- 102100029709 Tripartite motif-containing protein 54 Human genes 0.000 description 2
- 101710154817 Tripartite motif-containing protein 54 Proteins 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 230000000692 anti-sense effect Effects 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 230000004908 autophagic flux Effects 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 230000009977 dual effect Effects 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 238000000799 fluorescence microscopy Methods 0.000 description 2
- 230000030279 gene silencing Effects 0.000 description 2
- 230000009368 gene silencing by RNA Effects 0.000 description 2
- IPCSVZSSVZVIGE-UHFFFAOYSA-N hexadecanoic acid Chemical compound CCCCCCCCCCCCCCCC(O)=O IPCSVZSSVZVIGE-UHFFFAOYSA-N 0.000 description 2
- 238000012165 high-throughput sequencing Methods 0.000 description 2
- 238000003125 immunofluorescent labeling Methods 0.000 description 2
- 230000006872 improvement Effects 0.000 description 2
- 238000011503 in vivo imaging Methods 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 210000003098 myoblast Anatomy 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000002188 osteogenic effect Effects 0.000 description 2
- 229920002866 paraformaldehyde Polymers 0.000 description 2
- 239000008188 pellet Substances 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 239000002244 precipitate Substances 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 239000010959 steel Substances 0.000 description 2
- 210000000130 stem cell Anatomy 0.000 description 2
- 238000009168 stem cell therapy Methods 0.000 description 2
- 238000009580 stem-cell therapy Methods 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 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 1
- 102100022464 5'-nucleotidase Human genes 0.000 description 1
- 206010002027 Amyotrophy Diseases 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 108010082399 Autophagy-Related Proteins Proteins 0.000 description 1
- 238000011746 C57BL/6J (JAX™ mouse strain) Methods 0.000 description 1
- 102000034342 Calnexin Human genes 0.000 description 1
- 108010056891 Calnexin Proteins 0.000 description 1
- 208000017667 Chronic Disease Diseases 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 208000032131 Diabetic Neuropathies Diseases 0.000 description 1
- 101150073369 Fbxo32 gene Proteins 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 238000013218 HFD mouse model Methods 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 101710088172 HTH-type transcriptional regulator RipA Proteins 0.000 description 1
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 1
- 101000678236 Homo sapiens 5'-nucleotidase Proteins 0.000 description 1
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 1
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 1
- 239000012097 Lipofectamine 2000 Substances 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 239000012570 Opti-MEM I medium Substances 0.000 description 1
- 238000009004 PCR Kit Methods 0.000 description 1
- 235000021314 Palmitic acid Nutrition 0.000 description 1
- 102000003992 Peroxidases Human genes 0.000 description 1
- 101150101311 Sqstm1 gene Proteins 0.000 description 1
- -1 USA Proteins 0.000 description 1
- 102000006275 Ubiquitin-Protein Ligases Human genes 0.000 description 1
- 108010083111 Ubiquitin-Protein Ligases Proteins 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 210000001015 abdomen Anatomy 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- 239000006096 absorbing agent Substances 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000002293 adipogenic effect Effects 0.000 description 1
- 230000011759 adipose tissue development Effects 0.000 description 1
- 230000032683 aging Effects 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 210000000544 articulatio talocruralis Anatomy 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000002146 bilateral effect Effects 0.000 description 1
- 239000003012 bilayer membrane Substances 0.000 description 1
- 230000000975 bioactive effect Effects 0.000 description 1
- 230000005540 biological transmission Effects 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000001364 causal effect Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 1
- 230000037080 exercise endurance Effects 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000000706 filtrate Substances 0.000 description 1
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 1
- 210000002683 foot Anatomy 0.000 description 1
- 238000013238 high-fat diet model Methods 0.000 description 1
- 238000011532 immunohistochemical staining Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 235000015110 jellies Nutrition 0.000 description 1
- 239000008274 jelly Substances 0.000 description 1
- 210000000629 knee joint Anatomy 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 230000000512 lipotoxic effect Effects 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 230000003137 locomotive effect Effects 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 238000013227 male C57BL/6J mice Methods 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- WQEPLUUGTLDZJY-UHFFFAOYSA-N n-Pentadecanoic acid Natural products CCCCCCCCCCCCCCC(O)=O WQEPLUUGTLDZJY-UHFFFAOYSA-N 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 235000021590 normal diet Nutrition 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 238000003068 pathway analysis Methods 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 239000006223 plastic coating Substances 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 238000003762 quantitative reverse transcription PCR Methods 0.000 description 1
- 238000010814 radioimmunoprecipitation assay Methods 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 230000008844 regulatory mechanism Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000017423 tissue regeneration Effects 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 238000004627 transmission electron microscopy Methods 0.000 description 1
- 238000011870 unpaired t-test Methods 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 238000005303 weighing Methods 0.000 description 1
- 230000004584 weight gain Effects 0.000 description 1
- 235000019786 weight gain Nutrition 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
Classifications
-
- 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
- A61K35/28—Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Diabetes (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- General Chemical & Material Sciences (AREA)
- Cell Biology (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Developmental Biology & Embryology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Immunology (AREA)
- Hematology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Neurology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Physical Education & Sports Medicine (AREA)
- Biomedical Technology (AREA)
- Biotechnology (AREA)
- Epidemiology (AREA)
- Virology (AREA)
- Zoology (AREA)
- Emergency Medicine (AREA)
- Endocrinology (AREA)
- Obesity (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
The invention discloses an application of exosomes in preparing a medicament for treating muscular atrophy caused by type 2 diabetes mellitus, wherein the exosomes are derived exosomes of umbilical cord mesenchymal stem cells, and the exosomes improve the muscular atrophy caused by type 2 diabetes mellitus by enhancing the autophagy effect mediated by AMPK/ULK 1. The invention also provides application of the exosome in preparing medicines for treating muscular atrophy caused by obesity or disuse muscular atrophy. The invention verifies that the exosomes of the umbilical cord mesenchymal stem cells can improve the type 2 diabetes/obesity/disuse-induced muscle atrophy by enhancing the autophagy mediated by AMPK/ULK1, so the exosomes of the umbilical cord mesenchymal stem cells have good prospect in the preparation of medicaments for treating the type 2 diabetes/obesity/disuse-induced muscle atrophy.
Description
Technical Field
The invention relates to the field of biological medicine industry, in particular to application of exosomes in preparation of a medicament for treating amyotrophy caused by type 2 diabetes.
Background
Muscle atrophy refers to the decrease in muscle volume caused by the attenuation or even the disappearance of muscle fibers, and aging, braking, and some chronic diseases such as type 2 diabetes (T2 DM) and obesity all cause muscle atrophy. Type 2 diabetics often exhibit mild muscular atrophy in middle-aged years, with age and development of diabetic neuropathy becoming more severe. The causal role of obesity, diabetes and braking in muscle atrophy has been demonstrated in animal models. Muscle atrophy is caused by reduced protein synthesis and increased degradation rates. The ubiquitination-proteasome pathway and the autophagy-lysosomal pathway play an important role therein.
The stem cells have unique biological characteristics, extremely strong self-renewal capacity, multidirectional differentiation potential, secretion of various cytokines and other characteristics, and are the optimal seed cells for realizing organ tissue regeneration. The stem cell therapy can enable partial diabetics to stop insulin therapy or reduce the dosage of insulin, and the effectiveness and safety of the stem cell therapy for treating diabetes are also effectively verified. Exosomes (exosomes, EXOs) are extracellular vesicles secreted by cells containing bioactive molecules, including proteins, lipids, nucleic acids, and the like. At present, many researches at home and abroad show that the treatment based on mesenchymal stem cells (mesenchymal stem cell, MSC) plays an important role in muscle atrophy caused by various diseases.
The mesenchymal stem cells and the exosomes thereof have good application prospects in treating the muscular atrophy caused by the T2DM, however, the influence of the mesenchymal stem cells and the exosomes on the muscular atrophy of the T2DM patient is not clear.
Disclosure of Invention
The present disclosure has been made in view of the above-mentioned prior art, and an object thereof is to provide an application of exosomes in preparing a medicament for treating muscular dystrophy caused by type 2 diabetes.
To this end, a first aspect of the present disclosure provides the use of an exosome, which is a derived exosome of umbilical cord mesenchymal stem cells, for the preparation of a medicament for treating muscle atrophy caused by type 2 diabetes, the exosome improving muscle atrophy caused by type 2 diabetes by enhancing AMPK/ULK 1-mediated autophagy. In the present disclosure, it was verified that exosomes of umbilical cord mesenchymal stem cells can improve type 2 diabetes-induced muscle atrophy by enhancing AMPK/ULK 1-mediated autophagy, and therefore, exosomes of umbilical cord mesenchymal stem cells have a good prospect in the preparation of drugs for treating type 2 diabetes-induced muscle atrophy.
In the application related to the disclosure, optionally, the preparation method of the exosome includes: obtaining human umbilical cord tissue; culturing human umbilical cord tissue by using an alpha-MEM culture medium until the cell reaches 80% of fusion; culturing in exosome-free culture medium, and collecting to obtain supernatant; and centrifuging the supernatant to obtain the exosome. Thereby, umbilical cord mesenchymal stem cell-derived exosomes can be obtained by extraction from human umbilical cord tissue.
In the application of the first aspect to which the present disclosure relates, optionally, the alpha-MEM medium contains 10% fetal bovine serum, 100U/mL penicillin and 100. Mu.g/mL streptomycin. Therefore, the umbilical cord mesenchymal stem cell-derived exosomes can be prepared.
In an application of the first aspect to which the present disclosure relates, optionally, the supernatant is filtered through a 0.22 μm filter and centrifuged. Therefore, the umbilical cord mesenchymal stem cell-derived exosomes can be prepared.
In the application of the first aspect to which the present disclosure relates, optionally, the supernatant is filtered through a 0.22 μm filter and centrifuged at 120000g for 70 minutes to obtain the exosome. Therefore, the umbilical cord mesenchymal stem cell-derived exosomes can be prepared.
In an application of the first aspect to which the present disclosure relates, optionally, the exosomes increase the cross-sectional area and number of muscle fibers to improve muscle atrophy caused by type 2 diabetes.
In an application of the first aspect to which the present disclosure relates, optionally, the exosomes regulate the ratio of fast and slow muscle fibers to improve muscle atrophy caused by type 2 diabetes.
In the use of the first aspect of the present disclosure, optionally, the exosomes promote phosphorylation of AMPK and ULK1 to enhance AMPK/ULK1 mediated autophagy.
In a second aspect, the present disclosure provides a use of an exosome, which is umbilical cord mesenchymal stem cell-derived exosome, for improving muscle atrophy caused by obesity by enhancing AMPK/ULK 1-mediated autophagy, in the preparation of a medicament for treating muscle atrophy caused by obesity. In the present disclosure, it was verified that exosomes of umbilical mesenchymal stem cells can improve obesity-induced muscle atrophy by enhancing AMPK/ULK 1-mediated autophagy, and thus, exosomes of umbilical mesenchymal stem cells have a good prospect in the preparation of drugs for treating muscle atrophy caused by obesity.
In a third aspect, the disclosure provides a use of an exosome in the manufacture of a medicament for treating disuse muscle atrophy, wherein the exosome is umbilical mesenchymal stem cell-derived exosome, the exosome improving disuse muscle atrophy by enhancing AMPK/ULK 1-mediated autophagy. In the present disclosure, it was verified that exosomes of umbilical mesenchymal stem cells can improve disuse muscle atrophy by enhancing AMPK/ULK 1-mediated autophagy, and therefore, exosomes of umbilical mesenchymal stem cells have a good prospect in the application of preparing a medicament for treating disuse muscle atrophy.
According to the present disclosure, there can be provided an application of a human umbilical cord mesenchymal stem cell-derived exosome in preparing a medicament for treating muscular dystrophy caused by type 2 diabetes and/or obesity, and an application of a human umbilical cord mesenchymal stem cell-derived exosome in preparing a medicament for treating disuse muscular dystrophy.
Drawings
FIG. 1 is a graph showing the identification results of hucMSCs according to the present invention.
Fig. 2 is a graph showing the results of the test of hucMSCs of example 1 and comparative example 1 of the present invention for improving muscular atrophy caused by diabetes.
Fig. 3 is a graph showing the results of the test of hucMSCs of example 2 and comparative example 2 for relieving muscular atrophy caused by obesity.
Fig. 4 is a graph showing test results of improved brake (IM) -induced muscular atrophy of hucMSCs in example 3 and comparative example 3 of the present invention.
Fig. 5 is a graph showing the first test results of example 4 and comparative example 4 according to the present invention.
FIG. 6 is a graph showing the second test results of example 4 and comparative example 4 according to the present invention.
Fig. 7 is a graph showing the results of a test for hucMSCs of example 5 of the present invention to inhibit PA-induced myotube atrophy by enhancing AMPK/autophagy signaling pathway.
FIG. 8 is a graph showing the results of a first test of improving diabetes-induced muscle atrophy by MSC-EXO according to example 6 of the present invention.
FIG. 9 is a graph showing the second test result of improving diabetes-induced muscle atrophy by MSC-EXO according to example 6 of the present invention.
FIG. 10 is a graph showing the results of a test of MSC-EXO in example 7 of the present invention to inhibit PA-induced myotube atrophy by enhancing the AMPK/autophagy signaling pathway.
Detailed Description
Hereinafter, preferred embodiments of the present disclosure will be described in detail with reference to the accompanying drawings. In the following description, the same members are denoted by the same reference numerals, and overlapping description thereof is omitted. In addition, the drawings are schematic, and the ratio of the sizes of the components to each other, the shapes of the components, and the like may be different from actual ones.
It should be noted that the terms "comprises" and "comprising," and any variations thereof, such as a process, method, system, article, or apparatus that comprises or has a list of steps or elements is not necessarily limited to those steps or elements expressly listed or inherent to such process, method, article, or apparatus, but may include or have other steps or elements not expressly listed or inherent to such process, method, article, or apparatus.
In addition, headings and the like referred to in the following description of the invention are not intended to limit the scope or content of the invention, but rather are merely indicative of reading. Such subtitles are not to be understood as being used for segmenting the content of the article, nor should the content under the subtitle be limited only to the scope of the subtitle.
Proper noun abbreviations and resolution to which the present disclosure relates:
type 2 diabetes mellitus2 diabetes mellitus, T2 DM;
MSC/MSCs Mesenchymal stem cell/cells mesenchymal stem cells;
hucMSCs human umbilical cord MSCs human umbilical cord mesenchymal stem cells;
MSC-EXO, hucMC-derived exosomes derived from human umbilical cord mesenchymal stem cells;
HFD High-fat diet;
IM hindlimb immobilization hind limb braking;
PBS Phosphate-buffered saline;
IPGTT Intraperitoneal glucose tolerance test glucose tolerance test in abdominal cavity;
IPITT Intraperitoneal insulin tolerance test intraperitoneal insulin resistance test.
Reference will now be made in detail to embodiments of the present invention, examples of which are illustrated in the accompanying drawings, wherein like or similar reference numerals refer to like or similar elements throughout or elements having like or similar functionality. The embodiments described below by way of the drawings are exemplary only and should not be construed as limiting the invention.
It will be understood by those skilled in the art that, unless otherwise defined, all terms (including technical and scientific terms) used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
It will be further understood that terms, such as those defined in commonly used dictionaries, should be interpreted as having a meaning that is consistent with their meaning in the context of the prior art and will not be interpreted in an idealized or overly formal sense unless expressly so defined herein.
As used herein, the singular forms "a", "an", "the" and "the" are intended to include the plural forms as well, unless expressly stated otherwise, as understood by those skilled in the art. It will be further understood that the terms "comprises" and/or "comprising," when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, and/or groups thereof.
In the description of the present specification, a description referring to terms "one embodiment," "some embodiments," "examples," "specific examples," or "some examples," etc., means that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the present invention. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, the different embodiments or examples described in this specification and the features of the different embodiments or examples may be combined and combined by those skilled in the art without contradiction.
The present disclosure relates to:
application of Mesenchymal Stem Cells (MSCs) in preparing medicament for treating muscular dystrophy caused by type 2 diabetes;
use of mesenchymal stem cell-derived exosomes (MSC-EXO) for the preparation of a medicament for treating muscle atrophy caused by type 2 diabetes;
use of Mesenchymal Stem Cells (MSCs) in the manufacture of a medicament for treating muscle atrophy caused by obesity;
use of mesenchymal stem cell-derived exosomes (MSC-EXO) for the preparation of a medicament for treating muscle atrophy caused by obesity;
use of Mesenchymal Stem Cells (MSCs) in the manufacture of a medicament for the treatment of disuse muscle atrophy;
use of mesenchymal stem cell-derived exosomes (MSC-EXO) for the preparation of a medicament for disuse muscle atrophy.
In the present disclosure, in some examples, the mesenchymal stem cells may be human umbilical cord mesenchymal stem cells (hucMSCs), and the mesenchymal stem cell-derived exosomes may be human umbilical cord mesenchymal stem cell-derived exosomes. In this case, umbilical cord is used as a material source of mesenchymal stem cells and exosomes thereof, and has advantages of reduced harm to human body and easy collection when collecting compared with other materials such as bone marrow, liver, etc., and umbilical cord-derived mesenchymal stem cells have stronger proliferation and differentiation ability.
In some examples, human umbilical cord mesenchymal stem cells and their exosomes may be obtained from fresh umbilical cord tissue of a neonate.
In some examples, human umbilical cord mesenchymal stem cells may be obtained from umbilical cord tissue by culturing. In some examples, human umbilical cord tissue may be cultured using alpha-MEM medium. The medium may contain 10% fetal bovine serum, 100U/mL penicillin and 100. Mu.g/mL streptomycin. Thus, human umbilical cord mesenchymal stem cells can be obtained by culture.
In some examples, human umbilical cord mesenchymal stem cell-derived exosomes may be isolated after culturing human umbilical cord tissue using a culture medium. In some examples, the supernatant after cultivation may be filtered using a 0.22 μm filter, whereby the larger particle size component of the supernatant can be removed and the exosomes can be retained in the supernatant as much as possible. In some examples, the filtered supernatant may be centrifuged and the centrifuged precipitate is the exosome. The filtered supernatant may be centrifuged at 120000g for 70 minutes and the pellet after centrifugation is the exosome. Thus, the exosomes derived from the human umbilical mesenchymal stem cells can be prepared.
The present disclosure evaluates the efficacy of human umbilical mesenchymal stem cells and their derived exosomes for diabetes and/or obesity-induced muscle atrophy and disuse muscle atrophy, and studies the molecular mechanism of specific actions. Human umbilical cord mesenchymal stem cells or their derived exosomes have an ameliorating effect on muscular atrophy caused by diabetes and/or obesity and disuse muscular atrophy, and human umbilical cord mesenchymal stem cells or their derived exosomes ameliorate muscular atrophy by enhancing AMPK/ULK 1-mediated autophagy. In some examples, human umbilical mesenchymal stem cells or exosomes derived therefrom may increase the cross-sectional area and number of muscle fibers to improve muscle atrophy, in some examples by adjusting the ratio of fast and slow muscle fibers.
In order that the invention may be readily understood, a further description of the invention will be rendered by reference to specific embodiments that are illustrated in the appended drawings and are not to be construed as limiting embodiments of the invention.
It will be appreciated by those skilled in the art that the drawings are merely schematic representations of examples and that the elements of the drawings are not necessarily required to practice the invention.
Examples
In embodiments of the present disclosure, hucMSCs or hucMSCs-derived exosomes (MSC-EXO) interventions were performed on various mouse models (diabetic db/db mice, high Fat Diet (HFD) mice, hindlimb brake (IM) mice), C2C12 myotubes, and skeletal muscle strength and locomotor performance were tested using grip testing and running machine running, muscle mass was assessed using body composition, muscle weight and muscle fiber cross-sectional area (CSA), potential regulatory mechanisms were explored by RNA-seq analysis of the tibialis anterior muscle (TA) and Western blot (immunoblot) detection of related signal pathways to explore the efficacy and mechanism of hucMSCs or hucMSCs-derived exosomes (MSC-EXO) on diabetes-induced muscle atrophy.
In this example, all of the test materials used were conventional in the art and were commercially available.
The experimental method comprises the following steps:
obtaining hucMSCs and its exosomes
(1) Fresh human umbilical cord tissue was obtained from healthy newborns, after washing the umbilical cord and removing blood vessels, the Wharton's jelly was cut into small pieces and placed in a cell culture flask containing an alpha-MEM medium containing 10% fetal bovine serum, 100U/mL penicillin and 100. Mu.g/mL streptomycin. The medium was changed every 3 days until the cells reached 80% confluence.
(2) 3 rd to 5 th generation cells for flow cytometry analysis, differentiation induction, drug administration and Transwell TM Co-culturing. After 48 hours of culture in exosome-free medium, the cell supernatant was collected, centrifuged to remove cells and cell debris, and then filtered through a 0.22 μm sterile filter. Subsequently, the filtrate was ultracentrifuged at 120000×g at 4℃for 70 minutes, and the precipitate was an exosome. The exosome-containing pellet was resuspended in PBS and stored at-80 ℃ for subsequent experiments.
The PARTICLE size of the exosomes was detected by ZetaVIEWS/N17-310 (PARTICLE METRIX, germany). The morphology of the exosomes was detected by transmission electron microscopy. The expression of the exosome markers CD9, CD63, CD81 was detected by immunoblotting.
Establishing animal model
(1) Establishment of T2 DM-related muscle-wasting mouse model
A4-week-old male diabetic db/db mouse (Kwanes laboratory animal Co., ltd.) was obtained, fed with a normal diet, and placed in an environment with a light/dark cycle at a temperature of 22℃to 25℃and a humidity of 55.+ -. 5% for 12 hours, and the blood glucose in the empty abdomen was measured 2 times continuously to be 16.7mmol/L or more, and model of type 2 diabetic (T2 DM) mice was successful.
(2) Establishing obesity-related muscle-wasting mouse model
Male C57BL/6J mice (Shandong university model animal study center) were obtained at 6 weeks of age, and fed on a High Fat Diet (HFD) for 30 weeks from 8 weeks of age, to establish an obesity-related muscle-wasting-reduced mouse model.
For human umbilical cord mesenchymal stem cells (hucMSCs), human umbilical cord mesenchymal stem cells (1×10 6 cells/mouse) were suspended in PBS buffer and injected into both mouse models via tail vein every 7 days for 8 cycles.
For human umbilical cord mesenchymal stem cell-derived exosomes (MSC-EXO), human umbilical cord mesenchymal stem cell-derived exosomes were injected every 3 days into both mouse models by tail vein at 200 μg for 8 weeks.
(3) Establishing a model of disuse muscle-wasting mice
The left hind limb of a 6-week old male C57BL/6J mouse was fixed with a soft plastic coated steel wire for 2 weeks as follows: the steel wire tie with the soft plastic coating is cut into a length of 20-25 cm and is used for fixing the left hind limb of the mouse from the foot to the thigh, so that the knee joint is in an extending state, and the ankle joint is in a plantar flexion state, thereby establishing and obtaining the model of the disuse muscle-wasting mouse.
Then, human umbilical cord mesenchymal stem cells (1×10 in PBS buffer solution 6 cells/mouse) was injected into tibialis anterior and soleus muscles once every 7 days for a total of 4 cycles, resulting in an im+hucmscs treated group. The control group (im+pbs treated group) was then given the same volume of PBS buffer injected without human umbilical cord mesenchymal stem cells.
Method 1: metabolic testing and in vivo muscle performance analysis
The body composition of each mouse model was measured using a dual energy X-ray absorber. The muscle mass ratio represents the percentage of muscle mass composition to body composition.
Mice were subjected to an intraperitoneal glucose tolerance test (IPGTT) and an intraperitoneal insulin tolerance test (IPITT), and glucose and insulin resistance of the mice were measured.
The running machine is used for measuring the running distance, and the running distance is specifically: after two days of acclimation, the motor ability of the mice was determined by measuring their ability to run to fatigue, and the mice were first allowed to run at 8 m/min for 20min. Thereafter the speed of the treadmill was increased by 0.2m/min. Fatigue is defined as the hind limb contacting the electric grid for more than 10 seconds (electric grid) is the device that punishes for false responses in animal learning experiments and gives electric shock to animals.
The grip strength is measured by using an electronic grip dynamometer, specifically: the mice were trained to grasp the horizontal grid with the grip dynamometer attached to each limb and pull back gently in a horizontal direction parallel to the grip dynamometer, the force applied to the grid each time before the mice lost grip was recorded, 3 grip trials were performed per mouse, 3 measurements were recorded and the average was taken.
Method 2: histological and immunostaining
(1) After anesthesia of the mice, the bilateral Tibialis Anterior (TA) was dissected and weighed. One muscle was treated with liquid nitrogen and stored in a-80 ℃ refrigerator, the other was fixed in 4% paraformaldehyde and hematoxylin-eosin (H & E) staining and immunostaining were performed.
(2) A 4% paraformaldehyde fixed TA muscle sample was embedded in paraffin and sectioned at 5 μm thickness in maximum cross section. After dewaxing, the sections were H & E stained according to standard procedures.
(3) For immunohistochemical staining, antigen retrieval was performed using antigen retrieval liquid after section dewaxing. Endogenous peroxidase was inactivated with 3% hydrogen peroxide for 15min, and after blocking in blocking solution (10% goat serum) for 30min, anti-fast and slow muscle fiber antibodies were added dropwise and incubated overnight at 4 ℃. The following day sections were incubated with secondary antibody for 60min at room temperature and then stained with 3,3' -Diaminobenzidine (DAB) solution. Images were taken using a microscope and the cross-sectional area (CSA) of the muscle fibers and the ratio of fast to slow muscle fibers were calculated using Image-Pro Plus software.
(4) For immunofluorescent staining, antigen retrieval was performed using antigen retrieval solution after section dewaxing. After blocking in blocking solution (10% goat serum) at room temperature for 30min, LC3 antibody was added dropwise and incubated overnight at 4 ℃. The next day was incubated with goat anti-rabbit FITC secondary at room temperature for 60min, followed by staining with DAPI for 5min. Fluorescent images were observed and captured using a fluorescent microscope. The proportion of LC3 positive myofibers was analyzed and calculated using ImageJ software.
Method 3: exosome tracing
For ex vivo tracking, MSC-EXO was labeled with Cy7 and injected into mice via tail vein. Mice were imaged using an In Vivo Imaging System (IVIS) 12 hours after injection. For in vitro exosome tracing, MSC-EXO was labeled using PKH67 green fluorescent cell ligation kit, then incubated with fully differentiated myotubes for 24 hours, and fluorescent signals were detected and captured by fluorescence microscopy.
Method 4: cell lines and RNAi
Human Embryonic Lung Fibroblasts (HELFs) were obtained from the China center for cell culture (Shanghai, china) and were cultured at 37℃with 5% CO 2 In the incubator (C), the culture was performed using a high-sugar DMEM medium supplemented with 10% fetal bovine serum, 100U/mL penicillin and 100. Mu.g/mL streptomycin.
Mouse C2C12 myoblasts were obtained from the chinese infrastructure cell resource pool and cultured using high sugar DMEM medium supplemented with 10% fetal bovine serum and antibiotics.
After the degree of fusion reached 80% -90%, the medium was replaced with a differentiation medium consisting of dmem+2% heat inactivated horse serum and antibiotics, with the medium being refreshed every 2 days for 4 days. Fully differentiated myotubes were stimulated with palmitic acid (0.6 mM) for 24 hours and then passed through a Transwell TM The system was co-cultured with or incubated with derived exosomes (25 μg/ml) of human umbilical cord mesenchymal stem cells for 24h.
To further explore the mechanisms of human umbilical mesenchymal stem cells and their derived exosomes, myotubes were pre-treated with small interfering RNAs (siRNA) or autophagy inhibitors 3-methyladenine (3-MA, 2 mM) prior to hucMSCs and MSC-EXO treatment.
siRNA oligonucleotides were synthesized by GenePharma, inc. (Shanghai, china). Negative Control (NC) siRNA sequences were:
sense5'-UUCUCCGAACGUGUCACGUTT-3'(SEQ ID NO.1)、
antisense 5'-ACGUGACACGUUCGGAGAATT-3'(SEQ ID NO.2)。
the AMPK alpha 2 siRNA sequence is:
sense 5'-CCCAGAUGAACGCUAAGAUTT-3'(SEQ ID NO.3)、
antisense5'-AUCUUAGCGUUCAUCUGGGTT-3'(SEQ ID NO.4)。
ampkα2 siRNA transfection was performed on C2C12 myotubes using Lipofectamine2000 transfection reagent. Briefly, C2C12 myoblasts were plated on 6-well plates and induced to fully differentiate, then the medium was removed and transfected with Opti-MEM I medium containing siRNA-AMPKα2 (125 nM) and Lipo2000 for 6 hours. Then replaced with differentiation medium and treated with hucMSCs or MSC-EXO. After 24h, myotubes were collected for subsequent experiments.
Method 5: immunoblot analysis
The mouse tibialis anterior muscle samples were lysed with RIPA lysate and protein concentration was measured by BCA method. Subsequently, the proteins were isolated and transferred onto polyvinylidene fluoride (PVDF) membranes (IPVH 00010,0.45 μm microwells), the membranes were blocked with 5% skim milk for 1 hour at room temperature, and incubated overnight in a specific primary antibody at 4 ℃. After incubation of horseradish peroxidase-conjugated secondary antibodies for 1 hour at room temperature the next day, protein bands were detected using chemiluminescence. The main antibodies are: atrogin1 (protein, china, cat No. 67172-1-Ig, 1:5000), muRF1 (protein, cat No. 55456-1-AP, 1:1000), AMP-activated protein kinase (AMPK, USA, CST, cat No. 5831S, 1:1000), AMP-activated protein kinase (Thr 172) (CST, cat No.2535S, 1:1000), ULK1 (Bioss, china, cat No. bs-3602R, 1:500), phospho- (p-) ULK1 (Immunoway, USA, cat No. YP1542, 1:1000), LC3 (protein, cat No. 14600-1-AP, 1:1000), p62 (Abcam, USA, cat No. 91526, 1:1000), GADPH (Boster, china, cat No. 3:500).
Method 6: RNA sequencing (RNA-seq) and real-time quantitative PCR analysis
Total RNA was isolated using Trizol reagent (Invitrogen, USA). Library construction and sequencing work was done by Shenzhen megagene Inc. STAR (v.2.5) was used to index the mouse reference genome (mm 10) and align the resulting fastq files. Differential expression analysis was performed using DESeq2 (v1.28.1). KEGG pathway enrichment was performed by cluster analyzer clusterifier (v3.10.1). 1. Mu.g of RNA was reverse transcribed using a reverse transcription kit (Cat. No. RR047A; takara, japan). Primers were chemically synthesized by Shanghai Ji Ma pharmaceutical technologies.
The primer sequences were as follows:
fbxo32: sense 5'-GGGGTCACCCTGCAGCTTTGC-3' (SEQ ID NO. 5) and anti 5'-GGGGAAAGTGAGACGGAGCAGC-3' (SEQ ID NO. 6);
trim63: sense5'-ATGGACCGGCACGGGGTGTA-3' (SEQ ID NO. 7) and anti 5'-GCACATCGGGTGGCTGCCTT-3' (SEQ ID NO. 8);
ulk1: sense 5'-AAGTTCGAGTTCTCTCGCAAG-3' (SEQ ID NO. 9) and anti 5'-CGATGTTTTCGTGCTTTAGTTCC-3' (SEQ ID NO. 10);
map1lc3b: sense 5'-TTATAGAGCGATACAAGGGGGAG-3' (SEQ ID NO. 11) and anti 5'-CGCCGTCTGATTATCTTGATGAG-3' (SEQ ID NO. 12);
sqstm1: sense 5'-AGGATGGGGACTTGGTTGC-3' (SEQ ID NO. 13) and anti 5'-TCACAGATCACATTGGGGTGC-3' (SEQ ID NO. 14);
gapdh: sense 5'-AAGGGCTCATGACCACAGTC-3' (SEQ ID NO. 15) and anti-sense 5'-CAGGGATGATGTTCTGGGCA-3' (SEQ ID NO. 16).
The real-time polymerase chain reaction was performed using SYBR Green PCR kit (Cat.No. RR420A; takara), the gene expression change was determined based on the relative quantification method comparing Ct values, and normalized quantification was performed by using Gapdh as a control.
Statistical analysis
All data are expressed as mean ± standard error. The differences between the two groups were tested statistically using either unpaired t-test or one-way anova, followed by GraphPad Prism 8 software. Differences were considered statistically significant at P < 0.05.
Analysis of results
Example 1: hucMSCs treatment group
hucMSCs were injected by tail vein every 7d (1×10) using mouse model 1 6 Individual/mouse, suspended in 200 μl PBS) to db/db mice at 5 weeks of age, 8 injections total.
Comparative example 1: PBS treatment group
The hucMSCs solution of example 1 was replaced with PBS buffer and injected into db/db mice at 5 weeks of age.
After 1 week of the 8 th injection of example 1 and comparative example 1, example 1 and comparative example 1 were tested using metabolic testing and in vivo muscle performance analysis methods (method 1) and histological and immunostaining (method 2), respectively, as follows:
(1) The isolated hucMSCs were identified by flow cytometry, adipogenesis and osteogenic induced differentiation, and the results are shown in FIG. 1, wherein the hucMSCs showed positive expression of CD105 and CD73 on the surface, negative expression of CD34 and HLA-DR, and the hucMSCs had adipogenic and osteogenic multipotential differentiation ability, and were in line with the characteristics of stem cells.
(2) Intraperitoneal glucose tolerance test (IPGTT) and intraperitoneal insulin tolerance test (IPITT) were performed, and the results are shown in part A-B of FIG. 2, and hucMSCs improved glucose and insulin resistance.
(2) The grip strength was measured using an electronic grip dynamometer, and the results are shown in part C of FIG. 2, where the muscle strength of db/db mice was reduced compared to db/m mice, and hucMSCs injection was improved.
(3) The dual energy X-ray examination of the mouse muscle proportion, the isolated muscle weighing, and as shown in the D-F part of fig. 2, the injection of hucMSCs had no effect on body weight, but improved the muscle tissue proportion and the quality of Tibialis Anterior (TA).
(4) Histological H & E staining, immunostaining and molecular index detection were performed, resulting in G-L fractions as shown in fig. 2, db/db mice muscle fiber cross-sectional area (CSA), including fast and slow muscle fibers, significantly less than db/m mice; the hucMSCs can relieve the decrease of muscle fiber CSA caused by diabetes, increase the proportion of slow muscle fiber and promote the formation of muscle fiber; as shown in part M of FIG. 2, injection of hucMSCs also reduced the expression of E3-ubiquitin ligase Atrogin1 and MuRF 1.
The above results indicate that injection of hucMSCs can reduce diabetes-induced db/db mice muscle atrophy without affecting body weight. Thus, hucMSCs can alleviate muscle atrophy caused by diabetes.
Example 2: HFD+hucMSCs treatment group
With the use of the mouse model 2,high fat diet feeding was started at 8 weeks of age, and hucMSCs (1×10) were injected by tail vein at 38 weeks of age 6 Individual/mouse, suspended in 200 μl PBS) to High Fat Diet (HFD) fed mice, were injected every 7 days for a total of 8 injections.
Comparative example 2: HFD+PBS treatment group
The hucMSCs solution in example 2 was replaced with PBS and 200 μl of PBS was injected into High Fat Diet (HFD) fed mice at 38 weeks of age by tail vein every 7 days for a total of 8 injections.
After 1 week from the last administration of example 2 and comparative example 2, example 2 and comparative example 2 were tested using metabolic testing and in vivo muscle performance analysis methods (method 1) and histological and immunostaining (method 2), respectively, as follows:
(1) Exercise tolerance and grip strength were measured by intraperitoneal glucose tolerance test (IPGTT) and intraperitoneal insulin tolerance test (IPITT) using a treadmill and an electronic grip meter, and as a result, hucMSCs improved glucose and insulin tolerance and enhanced exercise tolerance and grip strength as shown in parts a-D of fig. 3.
(2) The results were measured by histological and immunostaining methods as follows:
as in parts E-F of fig. 3, hucMSCs reduced weight gain induced by high fat diets and increased muscle mass fraction; as in part G of fig. 3, injection of hucMSCs can increase the quality of the mouse tibialis anterior.
Meanwhile, as shown in the H-M part of fig. 3, injection of hucMSCs can also increase the cross-sectional area of muscle fibers, particularly fast muscle fibers, and can increase the percentage of slow muscle fibers and the number of muscle fibers; as in part N of FIG. 3, injection of hucMSCs inhibited the upregulation of muscle atrophy-associated Atrogin1 and MuRF1 protein levels.
The above results indicate that injection of hucMSCs can reduce obesity-induced muscle atrophy.
Example 3: im+hucmscs treatment group
hucMSCs (1×10) were injected locally into the braked limb of mouse model 3 6 Mice/mouse suspended in 200 μl PBS), 1 injection every 7 days for 4 times.
Comparative example 3: im+pbs treatment group
The hucMSCs solution in example 3 was replaced with PBS and the braked limb was locally injected with 200 μl of PBS 1 every 7 days for a total of 4 times.
Example 3 and comparative example 3 were tested using metabolic testing and in vivo muscle performance analysis methods (method 1) and histological and immunostaining (method 2), respectively, as follows:
(1) The grip strength of the mice was measured using a treadmill and an electronic grip dynamometer, and as shown in section a-B of fig. 4, the exercise endurance and grip strength of the mice injected with hucMSCs of example 3 were improved compared to comparative example 3.
(2) The results were measured by histological and immunostaining methods as follows:
as in the C-D section of FIG. 4, hucMSCs were able to increase the ratio of the muscle mass and the mass of the tibialis anterior muscle in mice.
As in the E-K portion of FIG. 4, hucMSCs also increased the cross-sectional area of mouse muscle fibers, including the fast and slow muscle fibers of IM mice, increased the percentage of slow muscle fibers and the number of muscle fibers, and reduced brake-induced upregulation of Atrogin1 and MuRF1 expression.
The above results indicate that hucMSCs can ameliorate muscle atrophy induced by braked limbs (disuse muscle atrophy), further indicating that the anti-muscular atrophy effect of hucMSCs may be due to its direct effect on muscles, rather than an indirect effect by improving body metabolism.
Example 4: db/db+hucmscs treated group, hfd+hucmscs treated group, im+hucmscs treated group
The injection of hucMSCs (1×10) was performed on mouse model 1, mouse model 2 and mouse model 3, respectively 6 Individual/mouse, suspended in 200 μl PBS).
Comparative example 4: db/db+pbs treated group, hfd+pbs treated group, im+pbs treated group
The hucMSCs solution in example 4 was replaced with PBS, and mice model 1, 2 and 3 were treated with 200 μl PBS, respectively.
The tibialis anterior muscle of model 1 mice injected with PBS buffer or hucMSCs in example 4 and comparative example 4, respectively, was tested using high throughput sequencing technology (RNA-seq) and real-time quantitative PCR analysis (method 6), as follows:
(1) High throughput sequencing showed 1426 Differentially Expressed Genes (DEGs), as shown in fig. 5 a, with 627 genes up-regulated and 799 genes down-regulated in model 1 mice injected with hucMSCs relative to control injected with PBS buffer; KEGG pathway analysis showed that DEGs was enriched in AMPK and autophagy signaling pathways as shown in B of fig. 5.
(2) Model 1 mice were analyzed by RT-qPCR and as shown in section C-D of fig. 5, injection of hucMSCs was able to down-regulate muscle wasting-related genes Fbxo32 (Atrogin 1), trim63 (MuRF 1) and autophagy-related genes Ulk1, map1lc3b and Sqstm1.
(3) In db/db mice, the levels of AMP-activated protein kinase (AMPK) (T172) and downstream ULK1 (S555) phosphorylation were lower than in db/m mice, as shown in part A of FIG. 6, and the levels of phosphorylation could be up-regulated after hucMSCs injection, while the ratio of LC3-II to LC3-I and p62 content were significantly higher in db/db mice than in db/m mice, but could be significantly down-regulated in hucMSCs dry prognosis.
Furthermore, immunofluorescent staining of LC3 showed that injection of hucMSCs reduced the proportion of LC3 positive myofibers, indicating that hucMSCs increased autophagy levels of skeletal muscle and restored autophagy flux, as shown in part B of fig. 6.
(4) Analysis of model 2 high fat diet mice and model 3 brake mice demonstrated that hucMSCs intervention also promotes phosphorylation of AMPK (T172) and ULK1 (S555) and reduces LC3-II to LC3-I ratio, p62 content and LC3 positive myofibers ratio as in the C-F section of fig. 6.
The above results indicate that hucMSCs are capable of activating the AMPK/ULK1 signaling pathway, promoting autophagy of skeletal muscle.
Example 5: pa+hucmscs treatment group
Myotube model induced in vitro with C2C12, treated with 0.6mM Palmitate (PA) induced myotube atrophy. PA can mimic lipotoxicity and induce myotube atrophy.
Example 5 was tested using autophagy inhibitor 3-MA or AMPK targeted siRNA (method 4), immunoblotting (method 5), and specifically as follows:
(1) The results are shown in fig. 7a by immunoblot analysis:
at this dose, PA treatment significantly promoted the expression of Atrogin1 and MuRF1, while hucMSCs treatment was able to eliminate PA-induced increases in Atrogin1 and MuRF1 expression, while PA reduced the phosphorylation levels of AMPK (T172) and ULK1 (S555), increasing LC3-II to LC3-I ratio and p62 content in myotubes. The use of hucMSCs treatment was able to significantly rescue PA-induced AMPK/ULK1 signaling of C2C12 myotubes, reducing the content of LC3II/LC3I and p62, indicating an autophagy process and an increase in autophagy flux.
Meanwhile, as shown in fig. 7B, the hucMSCs treatment can also increase the diameter of myotubes.
Thus supporting the direct cellular autonomy of hucMSCs on myotubes.
(2) Transfection assays were performed using targeted siRNA for autophagy inhibitor 3-MA or AMPK, and immunoblot analysis results are shown in figure 7, section C, with hucMSCs-mediated upregulation of ULK1 phosphorylation and reduction of LC3II/LC3I and p62 levels in the presence of 3-MA being partially eliminated; as shown in part D of FIG. 7, 3-MA was able to reduce the hucMSCs-mediated decline in the levels of Atrogin1 and MuRF1 and increase in myotube diameter.
(3) As shown in E-F of FIG. 7, after silencing AMPK by siRNA, the effect of hucMSCs on improvement of p-AMPK/p-ULK1, LC3/p62, atrogin1/MuRF1 and myotube diameter was reduced.
The above results indicate that hucMSCs alleviate PA-induced muscle atrophy by promoting AMPK-mediated autophagy in the C2C12 myotubes.
Example 6: MSC-EXO treatment group
MSC-EXO (200. Mu.g in 200. Mu.l PBS) was injected into db/db mice of mouse model 1 by tail vein every 3d for 8 weeks.
Comparative example 6: PBS treatment group
The hucMSCs solution in example 6 was replaced with PBS and 200 μl PBS was injected into db/db mice of mouse model 1 every 3d for 8 weeks.
Example 6 was tested using metabolic testing and in vivo muscle performance analysis methods (method 1) and histological and immunostaining (method 2), exosome tracking (method 3), immunoblotting (method 5), as follows:
(1) As shown in A-B of FIG. 8, MSC-EXO with a diameter of about 130nm isolated from hucMSCs culture supernatant was in a circular bilayer membrane structure with complete coating by nanoparticle size analysis and Transmission Electron Microscope (TEM) analysis.
Immunoblotting results are shown in FIG. 8C, CD9, CD63 and CD81 were enriched in MSC-EXO, while endoplasmic reticulum marker Calnexin was enriched in hucMSCs and deleted in MSC-EXO.
(2) The distribution of MSC-EXO in vivo was detected after 12h by performing exosome tracking using Cy 7-labeled MSC-EXO and injecting into mice. The results are shown in FIGS. 8D-E, where mice were imaged by an In Vivo Imaging System (IVIS), which images show that MSC-EXO can reach skeletal muscle.
(3) The grip strength was measured by intraperitoneal glucose tolerance test (IPGTT) and intraperitoneal insulin tolerance test (IPITT) using an electronic grip dynamometer, and as a result, MSC-EXO injection improved glucose and insulin resistance and grip strength as shown in A-C of FIG. 9.
(4) As measured by histological and immunostaining methods, as shown in D-F of fig. 9, MSC-EXO intervention had no effect on body weight, but increased the muscle mass fraction and increased the mass of tibialis anterior; as shown in G-L of FIG. 9, MSC-EXO also increases the cross-sectional area (CSA) of db/db mouse muscle fibers, including fast and slow muscle fibers, increasing the percentage of slow muscle fibers and the number of muscle fibers.
(5) As measured by immunoblotting, MSC-EXO was able to promote phosphorylation of AMPK (T172) and ULK1 (S555) while reducing the content of LC3II/LC3I and p62 and the proportion of LC3 positive myofibers, as shown in M-N of FIG. 9.
The above results indicate that injection of MSC-EXO can prevent diabetes-induced muscle atrophy and enhance the AMPK/autophagy signaling pathway, consistent with the effects of hucMSCs.
Example 7: PA+MSC-EXO treatment group
Fully differentiated C2C12 myotubes were treated with 25 μg/ml MSC-EXO for 24 hours in the presence and absence of targeted siRNA to autophagy inhibitor 3-MA or AMPK.
Example 7 was tested using exosome tracking (method 3), cell lines and RNAi (method 4), immunoblotting (method 5) respectively, as follows:
(1) For in vitro exosome tracing, MSC-EXO was labeled using PKH67, and fluorescent signals were detected and captured by fluorescence microscopy, as shown in fig. 10 a, and the labeled exosomes were found to be able to be taken up by the C2C12 myotubes.
(2) By immunoblotting analysis, as shown in FIG. 10B, MSC-EXO treatment was able to reduce PA-induced expression of Atrogin1 and MuRF1, and at the same time, was able to promote phosphorylation of AMPK (T172) and ULK1 (S555), and reduce the content of LC3II/LC3I and p 62.
(3) As shown in FIG. 10C, treatment of fully differentiated C2C12 myotubes with MSC-EXO can increase the diameter of the myotubes.
(4) By immunoblot analysis, the results are shown in FIG. 10D, where 3-MA is capable of partially eliminating MSC-EXO mediated upregulation of ULK1 phosphorylation levels and reduction of LC3II/LC3I and p62 levels; as shown in D-E of FIG. 10, 3-MA was able to reduce MSC-EXO mediated decreases in the levels of Atrogin1 and MuRF1 and increases in myotube diameter.
(5) As shown in F-G of FIG. 10, silencing AMPK with siRNA attenuated the effect of MSC-EXO on p-AMPK/p-ULK1, LC3/p62, atrogin1/MuRF1 and myotube diameter.
The above results indicate that MSC-EXO can alleviate muscle atrophy by promoting AMPK-mediated autophagy.
The foregoing description of the preferred embodiments of the present disclosure is provided only and not intended to limit the disclosure so that various modifications and changes may be made to the present disclosure by those skilled in the art. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present disclosure should be included in the protection scope of the present disclosure.
While the disclosure has been described in detail in connection with the drawings and embodiments, it should be understood that the foregoing description is not intended to limit the disclosure in any way. Modifications and variations of the present disclosure may be made as desired by those skilled in the art without departing from the true spirit and scope of the disclosure, and such modifications and variations fall within the scope of the disclosure.
Claims (10)
1. Use of an exosome in the manufacture of a medicament for treating muscle atrophy induced by type 2 diabetes, wherein the exosome is umbilical cord mesenchymal stem cell-derived exosome that ameliorates muscle atrophy induced by type 2 diabetes by enhancing AMPK/ULK 1-mediated autophagy.
2. The use according to claim 1, wherein the method of preparation of the exosomes comprises:
obtaining human umbilical cord tissue;
culturing human umbilical cord tissue by using an alpha-MEM culture medium until the cell reaches 80% of fusion;
culturing in exosome-free culture medium, and collecting to obtain supernatant;
and centrifuging the supernatant to obtain the exosome.
3. The use according to claim 2, wherein the alpha-MEM medium contains 10% fetal bovine serum, 100U/mL penicillin and 100 μg/mL streptomycin.
4. The use according to claim 2, characterized in that the supernatant is filtered through a 0.22 μm filter and centrifuged.
5. The use according to claim 4, wherein the supernatant is filtered through a 0.22 μm filter and centrifuged at 120000g for 70 minutes to obtain the exosomes.
6. The use according to claim 1, wherein the exosomes increase the cross-sectional area and number of muscle fibers to improve muscle atrophy caused by type 2 diabetes.
7. The use of claim 6, wherein the exosomes modulate the ratio of fast and slow muscle fibers to ameliorate muscle atrophy caused by type 2 diabetes.
8. The use of claim 1, wherein the exosomes promote phosphorylation of AMPK and ULK1 to enhance AMPK/ULK1 mediated autophagy.
9. Use of an exosome, which is umbilical cord mesenchymal stem cell-derived exosome, for the preparation of a medicament for treating muscle atrophy caused by obesity, the exosome improving muscle atrophy caused by obesity by enhancing AMPK/ULK 1-mediated autophagy.
10. Use of an exosome in the manufacture of a medicament for treating disuse muscle atrophy, wherein the exosome is umbilical cord mesenchymal stem cell-derived exosome that ameliorates disuse muscle atrophy by enhancing AMPK/ULK 1-mediated autophagy.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202310759269.7A CN117257837A (en) | 2023-06-26 | 2023-06-26 | Application of exosome in preparing medicine for treating muscular atrophy caused by type 2 diabetes |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202310759269.7A CN117257837A (en) | 2023-06-26 | 2023-06-26 | Application of exosome in preparing medicine for treating muscular atrophy caused by type 2 diabetes |
Publications (1)
Publication Number | Publication Date |
---|---|
CN117257837A true CN117257837A (en) | 2023-12-22 |
Family
ID=89218541
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202310759269.7A Pending CN117257837A (en) | 2023-06-26 | 2023-06-26 | Application of exosome in preparing medicine for treating muscular atrophy caused by type 2 diabetes |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN117257837A (en) |
Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20070114449A (en) * | 2006-05-29 | 2007-12-04 | 연세대학교 산학협력단 | A method for isolating and culturing mesenchymal stem cell derived from umbilical cord blood |
WO2013124815A2 (en) * | 2012-02-22 | 2013-08-29 | Brainstem Biotec Ltd. | Mesenchymal stem cells for in vitro modeling and cell-based therapy of human diseases and banks thereof |
KR20160137864A (en) * | 2015-05-22 | 2016-12-01 | 차의과학대학교 산학협력단 | Pharmaceutical Compositions for Improving or Treating Muscle Atrophy Comprising Conditioned Medium of Stem Cells Derived from Umbilical Cord |
CN107557332A (en) * | 2017-09-28 | 2018-01-09 | 吉林省拓华生物科技有限公司 | CD29+People's Mesenchymal Stem Cells from Umbilical Cord and its purposes in skeletal muscle atrophy medicine under preparing treatment high glucose and high fat environment |
CN113215094A (en) * | 2021-05-17 | 2021-08-06 | 山东大学齐鲁医院 | Mesenchymal stem cell exosome for reversing dedifferentiation of islet beta cells of type 2diabetes, and preparation method and application thereof |
WO2023027317A1 (en) * | 2021-08-23 | 2023-03-02 | ㈜셀라토즈테라퓨틱스 | Composition for treating muscle loss-related diseases comprising exosomes derived from tonsil mesenchymal stem cells |
-
2023
- 2023-06-26 CN CN202310759269.7A patent/CN117257837A/en active Pending
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR20070114449A (en) * | 2006-05-29 | 2007-12-04 | 연세대학교 산학협력단 | A method for isolating and culturing mesenchymal stem cell derived from umbilical cord blood |
WO2013124815A2 (en) * | 2012-02-22 | 2013-08-29 | Brainstem Biotec Ltd. | Mesenchymal stem cells for in vitro modeling and cell-based therapy of human diseases and banks thereof |
KR20160137864A (en) * | 2015-05-22 | 2016-12-01 | 차의과학대학교 산학협력단 | Pharmaceutical Compositions for Improving or Treating Muscle Atrophy Comprising Conditioned Medium of Stem Cells Derived from Umbilical Cord |
CN107557332A (en) * | 2017-09-28 | 2018-01-09 | 吉林省拓华生物科技有限公司 | CD29+People's Mesenchymal Stem Cells from Umbilical Cord and its purposes in skeletal muscle atrophy medicine under preparing treatment high glucose and high fat environment |
CN113215094A (en) * | 2021-05-17 | 2021-08-06 | 山东大学齐鲁医院 | Mesenchymal stem cell exosome for reversing dedifferentiation of islet beta cells of type 2diabetes, and preparation method and application thereof |
WO2023027317A1 (en) * | 2021-08-23 | 2023-03-02 | ㈜셀라토즈테라퓨틱스 | Composition for treating muscle loss-related diseases comprising exosomes derived from tonsil mesenchymal stem cells |
Non-Patent Citations (3)
Title |
---|
JIA SONG等: "Mesenchymal stromal cells ameliorate diabetes-induced muscle atrophy through exosomes by enhancing AMPK/ULK1-mediated autophagy", J CACHEXIA SARCOPENIA MUSCLE, vol. 14, no. 2, 27 January 2023 (2023-01-27), pages 923 * |
姚之肖: "人脐带间充质干细胞外泌体对肌腱修复的作用及相关机制研究", 中国博士学位论文全文数据库(电子期刊)医药卫生科技辑, no. 1, 15 January 2022 (2022-01-15), pages 066 - 86 * |
黄勇彬等: "间充质干细胞促进肌肉组织修复的应用前景", 中国组织工程研究, vol. 28, no. 1, 2 March 2023 (2023-03-02), pages 107 - 112 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Yang et al. | MiR-124 enriched exosomes promoted the M2 polarization of microglia and enhanced hippocampus neurogenesis after traumatic brain injury by inhibiting TLR4 pathway | |
Zhao et al. | Dose-effect relationship and molecular mechanism by which BMSC-derived exosomes promote peripheral nerve regeneration after crush injury | |
Hu et al. | Exosomes from human umbilical cord blood accelerate cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis and fibroblast function | |
Xu et al. | Exosomes derived from pro‐inflammatory bone marrow‐derived mesenchymal stem cells reduce inflammation and myocardial injury via mediating macrophage polarization | |
Lai et al. | MicroRNA-221/222 mediates ADSC-exosome-induced cardioprotection against ischemia/reperfusion by targeting PUMA and ETS-1 | |
Song et al. | Mesenchymal stromal cells ameliorate diabetes‐induced muscle atrophy through exosomes by enhancing AMPK/ULK1‐mediated autophagy | |
Xu et al. | Inhibition of exosomal miR‐24‐3p in diabetes restores angiogenesis and facilitates wound repair via targeting PIK3R3 | |
Zhu et al. | Enhanced angiogenesis promoted by human umbilical mesenchymal stem cell transplantation in stroked mouse is Notch1 signaling associated | |
Wang et al. | Nicotinamide riboside enhances endothelial precursor cell function to promote refractory wound healing through mediating the Sirt1/AMPK pathway | |
Li et al. | Bone morphogenetic protein 4 inhibits liposaccharide‐induced inflammation in the airway | |
Huang et al. | Extracellular vesicle-derived miR-511–3p from hypoxia preconditioned adipose mesenchymal stem cells ameliorates spinal cord injury through the TRAF6/S1P axis | |
Amundson et al. | Removal of COVID-19 spike protein, whole virus, exosomes, and exosomal micrornas by the hemopurifier® lectin-affinity cartridge in critically Ill patients with COVID-19 infection | |
Zhang et al. | M2 macrophage exosome-derived lncRNA AK083884 protects mice from CVB3-induced viral myocarditis through regulating PKM2/HIF-1α axis mediated metabolic reprogramming of macrophages | |
Wang et al. | A long-term anti-inflammation markedly alleviated high-fat diet-induced obesity by repeated administrations of overexpressing IL10 human umbilical cord-derived mesenchymal stromal cells | |
Xu et al. | TNF‐α Enhances the Therapeutic Effects of MenSC‐Derived Small Extracellular Vesicles on Inflammatory Bowel Disease through Macrophage Polarization by miR‐24‐3p | |
CN117257837A (en) | Application of exosome in preparing medicine for treating muscular atrophy caused by type 2 diabetes | |
EP4149498A1 (en) | Treatment of complications caused by infections of severe acute respiratory syndrome coronavirus | |
CN110646621B (en) | Application of CHRNA4 in preparation of drug for treating non-alcoholic steatohepatitis | |
CN114668844B (en) | Application of Nr-CWS-treated MSCs-derived exosome in promotion of skin wound healing | |
Beng et al. | Effects of R-salbutamol on the inflammatory response and acute lung injury in endotoxemic mice | |
CN115404205A (en) | Novel exosome and preparation method and application thereof | |
Gu | METTL3 Modulates Radiation-Induced Cardiac Fibrosis via the Akt/mTOR Pathway | |
Yu et al. | Extracellular Vesicles in Lung Health, Disease, and Therapy: Adipose-derived exosomes protect the pulmonary endothelial barrier in ventilator-induced lung injury by inhibiting the TRPV4/Ca2+ signaling pathway | |
CN113005093A (en) | CD73 modified human umbilical cord blood-derived exosome and application thereof | |
Fu et al. | The ameliorative effect of C-Kit pos hepatic endothelial Mertk deficiency on nonalcoholic steatohepatitis |
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 |