CN112076309A - Application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection - Google Patents
Application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection Download PDFInfo
- Publication number
- CN112076309A CN112076309A CN202010740606.4A CN202010740606A CN112076309A CN 112076309 A CN112076309 A CN 112076309A CN 202010740606 A CN202010740606 A CN 202010740606A CN 112076309 A CN112076309 A CN 112076309A
- Authority
- CN
- China
- Prior art keywords
- csa
- chbp
- injury
- cells
- protein
- 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
- 230000006378 damage Effects 0.000 title claims abstract description 45
- 208000027418 Wounds and injury Diseases 0.000 title claims abstract description 38
- 208000014674 injury Diseases 0.000 title claims abstract description 38
- 102000003951 Erythropoietin Human genes 0.000 title claims abstract description 31
- 108090000394 Erythropoietin Proteins 0.000 title claims abstract description 31
- 229940105423 erythropoietin Drugs 0.000 title claims abstract description 31
- OXCMYAYHXIHQOA-UHFFFAOYSA-N potassium;[2-butyl-5-chloro-3-[[4-[2-(1,2,4-triaza-3-azanidacyclopenta-1,4-dien-5-yl)phenyl]phenyl]methyl]imidazol-4-yl]methanol Chemical compound [K+].CCCCC1=NC(Cl)=C(CO)N1CC1=CC=C(C=2C(=CC=CC=2)C2=N[N-]N=N2)C=C1 OXCMYAYHXIHQOA-UHFFFAOYSA-N 0.000 title claims abstract description 30
- 230000004224 protection Effects 0.000 title claims abstract description 28
- 206010061481 Renal injury Diseases 0.000 title claims abstract description 27
- 108090000765 processed proteins & peptides Proteins 0.000 title claims abstract description 25
- 125000004122 cyclic group Chemical group 0.000 title claims abstract description 24
- 208000037806 kidney injury Diseases 0.000 title claims abstract description 24
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 title claims abstract description 22
- 229930105110 Cyclosporin A Natural products 0.000 title claims abstract description 21
- 108010036949 Cyclosporine Proteins 0.000 title claims abstract description 21
- 229960001265 ciclosporin Drugs 0.000 title claims abstract description 20
- 108090000397 Caspase 3 Proteins 0.000 claims abstract description 58
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 47
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 44
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 claims abstract description 40
- 210000003734 kidney Anatomy 0.000 claims abstract description 30
- 238000011282 treatment Methods 0.000 claims abstract description 28
- 229940109239 creatinine Drugs 0.000 claims abstract description 20
- 241000699670 Mus sp. Species 0.000 claims abstract description 19
- 108020004459 Small interfering RNA Proteins 0.000 claims abstract description 19
- 108010088751 Albumins Proteins 0.000 claims abstract description 17
- 102000009027 Albumins Human genes 0.000 claims abstract description 17
- 230000006907 apoptotic process Effects 0.000 claims abstract description 17
- 108700010013 HMGB1 Proteins 0.000 claims abstract description 14
- 101000775678 Burkholderia pseudomallei (strain K96243) Protein-glutamine deamidase Cif Proteins 0.000 claims abstract description 13
- 101100339431 Arabidopsis thaliana HMGB2 gene Proteins 0.000 claims abstract description 12
- 101150021904 HMGB1 gene Proteins 0.000 claims abstract description 12
- 210000002700 urine Anatomy 0.000 claims abstract description 10
- 210000000683 abdominal cavity Anatomy 0.000 claims abstract description 9
- 239000004006 olive oil Substances 0.000 claims abstract description 7
- 235000008390 olive oil Nutrition 0.000 claims abstract description 7
- 230000002792 vascular Effects 0.000 claims abstract description 7
- 210000002966 serum Anatomy 0.000 claims abstract description 4
- 210000002919 epithelial cell Anatomy 0.000 claims abstract description 3
- 102000003952 Caspase 3 Human genes 0.000 claims abstract 8
- 230000003203 everyday effect Effects 0.000 claims abstract 2
- 210000004027 cell Anatomy 0.000 claims description 86
- 230000001640 apoptogenic effect Effects 0.000 claims description 26
- 108020004999 messenger RNA Proteins 0.000 claims description 21
- 238000003753 real-time PCR Methods 0.000 claims description 16
- 230000037396 body weight Effects 0.000 claims description 13
- 108090000672 Annexin A5 Proteins 0.000 claims description 12
- 102000004121 Annexin A5 Human genes 0.000 claims description 12
- 206010016654 Fibrosis Diseases 0.000 claims description 11
- 230000004761 fibrosis Effects 0.000 claims description 11
- 102100033215 DNA nucleotidylexotransferase Human genes 0.000 claims description 9
- 108010008286 DNA nucleotidylexotransferase Proteins 0.000 claims description 9
- 210000004369 blood Anatomy 0.000 claims description 9
- 239000008280 blood Substances 0.000 claims description 9
- 238000010839 reverse transcription Methods 0.000 claims description 9
- 238000001262 western blot Methods 0.000 claims description 9
- 238000000034 method Methods 0.000 claims description 8
- 238000012764 semi-quantitative analysis Methods 0.000 claims description 8
- 241000699666 Mus <mouse, genus> Species 0.000 claims description 7
- 238000011534 incubation Methods 0.000 claims description 7
- 239000000523 sample Substances 0.000 claims description 7
- 102000000412 Annexin Human genes 0.000 claims description 6
- 108050008874 Annexin Proteins 0.000 claims description 6
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 claims description 6
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 claims description 6
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 claims description 6
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 claims description 6
- 241001465754 Metazoa Species 0.000 claims description 6
- 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 6
- 238000000540 analysis of variance Methods 0.000 claims description 6
- 238000004113 cell culture Methods 0.000 claims description 6
- 239000002299 complementary DNA Substances 0.000 claims description 6
- 230000001054 cortical effect Effects 0.000 claims description 6
- 230000010339 dilation Effects 0.000 claims description 6
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 claims description 6
- WEXRUCMBJFQVBZ-UHFFFAOYSA-N pentobarbital Chemical compound CCCC(C)C1(CC)C(=O)NC(=O)NC1=O WEXRUCMBJFQVBZ-UHFFFAOYSA-N 0.000 claims description 6
- 102000013415 peroxidase activity proteins Human genes 0.000 claims description 6
- 108040007629 peroxidase activity proteins Proteins 0.000 claims description 6
- 238000010186 staining Methods 0.000 claims description 6
- 238000007619 statistical method Methods 0.000 claims description 6
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 5
- 108020004414 DNA Proteins 0.000 claims description 5
- 239000013642 negative control Substances 0.000 claims description 5
- 230000002441 reversible effect Effects 0.000 claims description 5
- 210000001519 tissue Anatomy 0.000 claims description 5
- 210000005239 tubule Anatomy 0.000 claims description 5
- 238000011725 BALB/c mouse Methods 0.000 claims description 4
- 108091081021 Sense strand Proteins 0.000 claims description 4
- 238000004458 analytical method Methods 0.000 claims description 4
- 230000000692 anti-sense effect Effects 0.000 claims description 4
- 238000009835 boiling Methods 0.000 claims description 4
- 238000001514 detection method Methods 0.000 claims description 4
- 238000011065 in-situ storage Methods 0.000 claims description 4
- 230000008595 infiltration Effects 0.000 claims description 4
- 238000001764 infiltration Methods 0.000 claims description 4
- 210000004969 inflammatory cell Anatomy 0.000 claims description 4
- 239000007788 liquid Substances 0.000 claims description 4
- 238000002156 mixing Methods 0.000 claims description 4
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 claims description 4
- DHRLEVQXOMLTIM-UHFFFAOYSA-N phosphoric acid;trioxomolybdenum Chemical compound O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.O=[Mo](=O)=O.OP(O)(O)=O DHRLEVQXOMLTIM-UHFFFAOYSA-N 0.000 claims description 4
- 239000002504 physiological saline solution Substances 0.000 claims description 4
- 239000003531 protein hydrolysate Substances 0.000 claims description 4
- 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 4
- KZDCMKVLEYCGQX-UDPGNSCCSA-N 2-(diethylamino)ethyl 4-aminobenzoate;(2s,5r,6r)-3,3-dimethyl-7-oxo-6-[(2-phenylacetyl)amino]-4-thia-1-azabicyclo[3.2.0]heptane-2-carboxylic acid;hydrate Chemical compound O.CCN(CC)CCOC(=O)C1=CC=C(N)C=C1.N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 KZDCMKVLEYCGQX-UDPGNSCCSA-N 0.000 claims description 3
- BZTDTCNHAFUJOG-UHFFFAOYSA-N 6-carboxyfluorescein Chemical compound C12=CC=C(O)C=C2OC2=CC(O)=CC=C2C11OC(=O)C2=CC=C(C(=O)O)C=C21 BZTDTCNHAFUJOG-UHFFFAOYSA-N 0.000 claims description 3
- 108010085238 Actins Proteins 0.000 claims description 3
- 108091003079 Bovine Serum Albumin Proteins 0.000 claims description 3
- 108010035532 Collagen Proteins 0.000 claims description 3
- 102000008186 Collagen Human genes 0.000 claims description 3
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 claims description 3
- 108010067770 Endopeptidase K Proteins 0.000 claims description 3
- 108700039887 Essential Genes Proteins 0.000 claims description 3
- 206010030113 Oedema Diseases 0.000 claims description 3
- 239000002033 PVDF binder Substances 0.000 claims description 3
- 238000011529 RT qPCR Methods 0.000 claims description 3
- 238000000692 Student's t-test Methods 0.000 claims description 3
- 108010006785 Taq Polymerase Proteins 0.000 claims description 3
- 239000000872 buffer Substances 0.000 claims description 3
- 239000003153 chemical reaction reagent Substances 0.000 claims description 3
- 229920001436 collagen Polymers 0.000 claims description 3
- 238000012937 correction Methods 0.000 claims description 3
- 230000002354 daily effect Effects 0.000 claims description 3
- 230000008021 deposition Effects 0.000 claims description 3
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 claims description 3
- 238000002474 experimental method Methods 0.000 claims description 3
- 239000012091 fetal bovine serum Substances 0.000 claims description 3
- 238000001943 fluorescence-activated cell sorting Methods 0.000 claims description 3
- 238000007710 freezing Methods 0.000 claims description 3
- 230000008014 freezing Effects 0.000 claims description 3
- 230000004927 fusion Effects 0.000 claims description 3
- 238000003384 imaging method Methods 0.000 claims description 3
- 239000002609 medium Substances 0.000 claims description 3
- 239000012528 membrane Substances 0.000 claims description 3
- 230000004060 metabolic process Effects 0.000 claims description 3
- 239000008267 milk Substances 0.000 claims description 3
- 210000004080 milk Anatomy 0.000 claims description 3
- 235000013336 milk Nutrition 0.000 claims description 3
- 230000001338 necrotic effect Effects 0.000 claims description 3
- 230000007935 neutral effect Effects 0.000 claims description 3
- 229910052757 nitrogen Inorganic materials 0.000 claims description 3
- 239000008188 pellet Substances 0.000 claims description 3
- 229940056360 penicillin g Drugs 0.000 claims description 3
- 229960001412 pentobarbital Drugs 0.000 claims description 3
- 229920002401 polyacrylamide Polymers 0.000 claims description 3
- 229920002981 polyvinylidene fluoride Polymers 0.000 claims description 3
- 230000010410 reperfusion Effects 0.000 claims description 3
- 239000000758 substrate Substances 0.000 claims description 3
- 238000012353 t test Methods 0.000 claims description 3
- 208000037999 tubulointerstitial fibrosis Diseases 0.000 claims description 3
- 210000003934 vacuole Anatomy 0.000 claims description 3
- 101710088172 HTH-type transcriptional regulator RipA Proteins 0.000 claims description 2
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 claims description 2
- 230000000903 blocking effect Effects 0.000 claims description 2
- 239000007853 buffer solution Substances 0.000 claims description 2
- 238000007865 diluting Methods 0.000 claims description 2
- 239000011521 glass Substances 0.000 claims description 2
- 238000011068 loading method Methods 0.000 claims description 2
- 239000000137 peptide hydrolase inhibitor Substances 0.000 claims description 2
- 238000011002 quantification Methods 0.000 claims description 2
- 238000010814 radioimmunoprecipitation assay Methods 0.000 claims description 2
- 239000011535 reaction buffer Substances 0.000 claims description 2
- 229960005322 streptomycin Drugs 0.000 claims description 2
- 239000006228 supernatant Substances 0.000 claims description 2
- 238000010257 thawing Methods 0.000 claims description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 2
- 102000007469 Actins Human genes 0.000 claims 1
- 102100037907 High mobility group protein B1 Human genes 0.000 claims 1
- 238000003304 gavage Methods 0.000 claims 1
- 239000007928 intraperitoneal injection Substances 0.000 claims 1
- 239000012460 protein solution Substances 0.000 claims 1
- 102000055207 HMGB1 Human genes 0.000 abstract description 13
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 abstract description 3
- 230000008859 change Effects 0.000 abstract description 2
- 230000031146 intracellular signal transduction Effects 0.000 abstract 1
- 239000011780 sodium chloride Substances 0.000 abstract 1
- 102100029855 Caspase-3 Human genes 0.000 description 50
- 230000002485 urinary effect Effects 0.000 description 13
- 230000003247 decreasing effect Effects 0.000 description 12
- 230000000694 effects Effects 0.000 description 8
- 230000001154 acute effect Effects 0.000 description 7
- 230000007246 mechanism Effects 0.000 description 6
- 230000001684 chronic effect Effects 0.000 description 5
- 241000269331 Ambystoma Species 0.000 description 4
- 108010051975 Glycogen Synthase Kinase 3 beta Proteins 0.000 description 4
- 102100038104 Glycogen synthase kinase-3 beta Human genes 0.000 description 4
- 206010029155 Nephropathy toxic Diseases 0.000 description 4
- 102100023085 Serine/threonine-protein kinase mTOR Human genes 0.000 description 4
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 4
- 231100000417 nephrotoxicity Toxicity 0.000 description 4
- 230000007694 nephrotoxicity Effects 0.000 description 4
- 210000000056 organ Anatomy 0.000 description 4
- 238000002054 transplantation Methods 0.000 description 4
- 210000004926 tubular epithelial cell Anatomy 0.000 description 4
- 108010013238 70-kDa Ribosomal Protein S6 Kinases Proteins 0.000 description 3
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 3
- 239000000090 biomarker Substances 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000002243 precursor Substances 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 102100022900 Actin, cytoplasmic 1 Human genes 0.000 description 2
- 208000034706 Graft dysfunction Diseases 0.000 description 2
- 241000205573 Jeffersonia Species 0.000 description 2
- 102000001253 Protein Kinase Human genes 0.000 description 2
- 230000009693 chronic damage Effects 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 230000001434 glomerular Effects 0.000 description 2
- 230000028993 immune response Effects 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 230000002757 inflammatory effect Effects 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 210000001985 kidney epithelial cell Anatomy 0.000 description 2
- 239000004816 latex Substances 0.000 description 2
- 229920000126 latex Polymers 0.000 description 2
- 230000003589 nefrotoxic effect Effects 0.000 description 2
- 231100000381 nephrotoxic Toxicity 0.000 description 2
- 230000009979 protective mechanism Effects 0.000 description 2
- 230000008439 repair process Effects 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 208000009304 Acute Kidney Injury Diseases 0.000 description 1
- 102100021809 Chorionic somatomammotropin hormone 1 Human genes 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 101800001586 Ghrelin Proteins 0.000 description 1
- 102400000442 Ghrelin-28 Human genes 0.000 description 1
- 206010048748 Graft loss Diseases 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 208000033626 Renal failure acute Diseases 0.000 description 1
- 208000007536 Thrombosis Diseases 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000009692 acute damage Effects 0.000 description 1
- 201000011040 acute kidney failure Diseases 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 229940046731 calcineurin inhibitors Drugs 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000014564 chemokine production Effects 0.000 description 1
- 208000020832 chronic kidney disease Diseases 0.000 description 1
- 238000011284 combination treatment Methods 0.000 description 1
- 230000002301 combined effect Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 230000007850 degeneration Effects 0.000 description 1
- 230000003412 degenerative effect Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 208000028208 end stage renal disease Diseases 0.000 description 1
- 201000000523 end stage renal failure Diseases 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 238000001415 gene therapy Methods 0.000 description 1
- GNKDKYIHGQKHHM-RJKLHVOGSA-N ghrelin Chemical compound C([C@H](NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)CN)COC(=O)CCCCCCC)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CO)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1N=CNC=1)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C1=CC=CC=C1 GNKDKYIHGQKHHM-RJKLHVOGSA-N 0.000 description 1
- 108010011901 glutaminyl-glutamyl-glutaminyl-leucyl-glutamyl-arginyl-alanyl-leucyl-asparagyl-seryl-serine Proteins 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 239000000710 homodimer Substances 0.000 description 1
- 210000004276 hyalin Anatomy 0.000 description 1
- 229960003444 immunosuppressant agent Drugs 0.000 description 1
- 230000001861 immunosuppressant effect Effects 0.000 description 1
- 229940125721 immunosuppressive agent Drugs 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 108010069754 innate repair receptor Proteins 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 208000028867 ischemia Diseases 0.000 description 1
- 230000003907 kidney function Effects 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 230000004792 oxidative damage Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000021419 recognition of apoptotic cell Effects 0.000 description 1
- 230000008929 regeneration Effects 0.000 description 1
- 238000011069 regeneration method Methods 0.000 description 1
- 210000005084 renal tissue Anatomy 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/04—Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
- A61K38/12—Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K49/00—Preparations for testing in vivo
- A61K49/0004—Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
- A61K49/0008—Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/12—Drugs for disorders of the urinary system of the kidneys
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/87—Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
- C12N15/88—Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6844—Nucleic acid amplification reactions
- C12Q1/6851—Quantitative amplification
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N1/00—Sampling; Preparing specimens for investigation
- G01N1/28—Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
- G01N1/30—Staining; Impregnating ; Fixation; Dehydration; Multistep processes for preparing samples of tissue, cell or nucleic acid material and the like for analysis
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N21/00—Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
- G01N21/84—Systems specially adapted for particular applications
-
- 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
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/573—Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
-
- 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
- G01N33/58—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
- G01N33/581—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with enzyme label (including co-enzymes, co-factors, enzyme inhibitors or substrates)
-
- 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
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6863—Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/90—Enzymes; Proenzymes
- G01N2333/914—Hydrolases (3)
- G01N2333/916—Hydrolases (3) acting on ester bonds (3.1), e.g. phosphatases (3.1.3), phospholipases C or phospholipases D (3.1.4)
- G01N2333/922—Ribonucleases (RNAses); Deoxyribonucleases (DNAses)
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- General Health & Medical Sciences (AREA)
- Biomedical Technology (AREA)
- Urology & Nephrology (AREA)
- Biotechnology (AREA)
- Physics & Mathematics (AREA)
- Biochemistry (AREA)
- Organic Chemistry (AREA)
- Hematology (AREA)
- Genetics & Genomics (AREA)
- Pathology (AREA)
- Medicinal Chemistry (AREA)
- Analytical Chemistry (AREA)
- Microbiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Physics & Mathematics (AREA)
- Zoology (AREA)
- Animal Behavior & Ethology (AREA)
- Cell Biology (AREA)
- Wood Science & Technology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- General Engineering & Computer Science (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Food Science & Technology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biophysics (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Rheumatology (AREA)
- Endocrinology (AREA)
- Plant Pathology (AREA)
- Diabetes (AREA)
Abstract
The invention discloses an application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection, wherein a control group comprises the following components: exposing the abdominal cavity and renal pedicles; IR: two renal pedicles were separated, clamped closed with vascular clamps for 30 minutes to allow the kidneys to change color, and then perfused for 2 or 8 weeks; IR + CsA: dissolving CsA in olive oil, and intragastrically administering to IR mice every day; IR + CHBP: dissolving CHBP in saline, and injecting the IR mice into the abdominal cavity once every 3 days; IR + CsA + CHBP: simultaneous treatment of IR mice with CsA and CHBP; urine albumin/creatinine, serum creatinine and histology, apoptosis, caspase-3 and HMGB1 were evaluated, and intracellular signaling pathways were screened through a protein chip; a renal epithelial cell model is established, renal injury is simulated, and the influence of CsA, CHBP and/or caspase-3 siRNA on TCMK-1 is researched.
Description
Technical Field
The invention belongs to the field of medicines, and particularly relates to application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection.
Background
Kidney transplantation is the primary treatment for patients with end-stage renal disease, but graft dysfunction and donor shortages remain major concerns of widespread concern today. Ischemia-reperfusion (IR) injury is closely related to delayed function of transplanted organs, acute rejection and subsequent chronic graft dysfunction. Cyclosporin a (Cyclosporine a, CsA) is the most commonly used immunosuppressant after renal transplantation, but its nephrotoxicity is not negligible.
IR injury is an inevitable injury in kidney transplants that can trigger immune responses, oxidative damage, inflammatory responses and cell death, manifested by Tubular Epithelial Cell (TEC) death, inflammatory cell infiltration, cytokine and chemokine production, caspase-3 activation. TECs are most susceptible to IR damage, but are also involved in regeneration, which may be relevant to reduce damage and promote repair. Surviving TECs dedifferentiate and enter the cell cycle within hours after injury, initiating proliferation and maintaining homeostasis.
The 32kD caspase-3 precursor cleaves to the 17kD active subunit, playing apoptotic and inflammatory roles in renal IR-related injury. HMGB1 is an injury-associated molecule that is rapidly released from the nucleus into the extracellular domain, mediating apoptosis and inflammation in acute kidney injury and subsequent fibrosis. Our previous studies showed that caspase-3 and HMGB1 levels correlated with renal IR injury and the degree of fibrosis. Various treatments, including caspase-3 siRNA, improved renal injury and reduced caspase-3 and HMGB1 expression.
CsA is one of the most important immunosuppressive agents in many organ transplants, including the kidney. CsA belongs to the group of calcineurin inhibitors and has a great effect in increasing organ and patient survival rates. The use of CsA reduces the incidence of acute rejection and early graft loss after renal transplantation. However, CsA does not improve the long-term survival of transplanted kidneys due to its nephrotoxicity. CsA nephrotoxicity is manifested by interstitial fibrosis and hyaline degenerative degeneration of the arteriolar duct wall, which can lead to chronic graft injury and progressive renal function impairment.
Erythropoietin (EPO) is capable of protecting different organs including the brain, heart and kidneys from IR damage. This protection is achieved by a heterodimeric EPO receptor and a β -co-receptor (EPOR/β cR), also known as the innate repair receptor, which is pharmacologically distinct from the homodimer of Erythropoietin (EPOR). Our previous studies have shown that EPO can reduce tubular apoptosis but promote inflammatory apoptosis in IR injury. However, high doses of EPO often cause side effects such as hypertension and thrombosis. Therefore, researchers developed a Helical B Surface Peptide (HBSP) that interacts only with EPOR/β cR. HBSP consists of 11 amino acids derived from the surface of helix B in the EPO 3D structure (QEQLERALNSS), but has a short half-life of only a few minutes. To improve its plasma stability, a novel metabolically stable Cyclic HBSP (CHBP)) is further produced, with a prolonged half-life and potent tissue protection.
The present study further investigated the role and mechanism of CHBP in acute and chronic injury-related models. We hypothesized that CHBP protects kidney and TEC from IR and/or CsA induced damage. Renal protection of CHBP may involve several mechanisms of signaling, which necessitates further investigation of how the underlying protective mechanisms differ between 2 and 8 weeks after IR and/or CsA-induced injury, or whether CHBP has a comparable modulating effect on the immune response of CsA.
Disclosure of Invention
The technical problem to be solved is as follows: aiming at the technical problems that the high-dose EPO generally causes side effects, the half-life of the linear erythropoietin-derived peptide in vivo is short, and the like, the cyclic erythropoietin-derived peptide is selected to be used as a research object. Since the dose, efficiency, etc. of the cyclic ghrelin-derived peptide used in vivo are not clear, the present application preliminarily set the dose of 24nmol/kg body weight as the experimental dose in the preliminary experimental work. How to enter the body to target and act on the kidney by the cyclic erythropoietin-derived peptide is not known at present, and the cyclic erythropoietin-derived peptide is injected from the abdominal cavity and enters the systemic circulation to finally act on the kidney tissue. The invention aims to provide application of cyclic erythropoietin-derived peptide in acute and chronic kidney injury and cyclosporin A injury protection.
The technical scheme is as follows:
an application of cyclic erythropoietin-derived peptide in protecting kidney injury and cyclosporin A injury.
Further, the method comprises the following steps:
the first step is as follows: establishing a mouse kidney injury model: male BALB/c mice, 25-30g, were randomly divided into 5 groups of 6 mice each, i.e. n-6, with control groups: the abdominal cavity and renal pedicles were exposed and anesthetized with 0.01ml/g of 1% pentobarbital; IR group: separating two renal pedicles, clamping them for 30min with vascular clamps to discolor the kidneys, and then releasing the vascular clamps to allow reperfusion of the blood for 2 or 8 weeks; IR + CsA group: 2mg CsA was dissolved in 1ml olive oil and the IR group mice were gavaged daily at 35mg/kg body weight; IR + CHBP group: dissolving 0.11mg CHBP in 32ml 0.9% physiological saline, and injecting into IR group mice abdominal cavity according to 24nmol/kg body weight standard once every 3 days; IR + CsA + CHBP group: the IR group mice were treated simultaneously with CsA gastric lavage and CHBP once every 3 days;
the second step is that: collecting urine samples at weeks 2, 4, 6 and 8 using a metabolism cage, at week 2 or 8, sacrificing the animals, collecting blood samples and kidney samples, fixing the kidney samples with 10% (w/v) neutral formalin buffer solution, quick freezing in liquid nitrogen and storing at-80 ℃ for later use, measuring urine albumin/creatinine using an automated biochemical analyzer for urine samples, and measuring serum creatinine (SCr) using an automated biochemical analyzer for blood samples;
the third step: kidney specimens were paraffin-embedded sections, stained with hematoxylin and eosin (H & E), and semi-quantitative assessment of the degree of tubulointerstitial damage (TID) was scored from 0 to 4 points: the percentage of the damaged area is less than 5 percent, and 0 is obtained; 5% -25% to obtain 1 point; 25% -50% to obtain 2 points; 50% -75% to obtain 3 points; 4 points are obtained when the content exceeds 75 percent; the parameters evaluated include tubular dilation and vacuoles within the tubular lumen, interstitial dilation, inflammatory cell infiltration, protein casts, intraluminal cells or cell debris; each section was scored by randomly selecting 12 cortical fields at 200x magnification;
the fourth step: masson trichrome staining, detecting tubulointerstitial fibrosis, randomly selecting 20 cortical fields for each section under 400x magnification, and scanning using Image-Pro Plus software to assess collagen deposition;
fifth, the sections were labeled in situ with terminal deoxynucleotidyl transferase (TdT) fragmented dna (isel) using the ApopTag peroxidase kit: digesting the section with 40 μ g/mL proteinase K at 37 ℃ for 15 minutes, then incubating with TdT and digoxigenin-dUTP at 37 ℃ for 60 minutes, adding digoxigenin-peroxidase complex for 30 minutes, developing the section with 3' -amino-9-ethylcarbazole (AEC) substrate, and selecting 20 fields of view to detect apoptotic cells of tubules, lumen and interstitial region in 200x field of view;
sixth step, western blot analysis: preparing 25 microgram of kidney protein for separation on 12-15% polyacrylamide denaturing gel, transferring it to PVDF membrane, blocking with 5% milk before incubation with caspase-3 antibody, HMGB1 antibody, internal reference beta-actin, after incubation with peroxidase-coupled secondary antibody, scanning and developing using molecular imager Chemi Doc XRS + system, and semi-quantitative analysis by Image Lab software;
seventh step, real-time quantitative PCR: detection of Caspase-3 mRNA, Caspase-3 and housekeeping gene GAPDH probes in kidney and TCMK-1 cells in the StepOne Plus real-time PCR System by Reverse Transcription (RT) real-time quantitative polymerase chain reaction (qPCR) were labeled with 6-carboxyfluorescein (FAM), 2. mu.g of total RNA extracted with Trizol reagent was used for reverse transcription into cDNA, Taq polymerase was used for qPCR reaction bufferMu.l of cDNA were amplified in solution, wherein the reaction system containing 900nM forward and reverse primers and 250nM probe was subjected to 40 cycles at 95 ℃ for 10 min, followed by 95 ℃ for 15 sec and 60 ℃ for 1 min, with the normal group as control and GAPDH as correction, using 2-ΔΔCtThe method calculates the expression of caspase-3 mRNA;
eighth step, protein chip analysis: the protein chip in the protein chip kit utilizes 50 mu g of protein to simultaneously detect 18 types of phosphorylated or cracked signal molecules, obtains images by exposing a glass slide to a molecular imaging system for a short time according to the instructions of the chip, and uses Alpha View software 3.3 to perform semi-quantitative analysis by scanning the volume density;
ninth step, TCMK-1 cell culture: TCMK-1 cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum, 100 units/ml penicillin G and 100. mu.g/ml streptomycin, and the cells were transfected with Caspase-3 siRNA into TCMK-1 cells using Lipofectamine @ RNAiMAX for 24 hours with/without CHBP fusion to 60-70% cell length, the sequence of the double stranded CASP-3siRNA being: sense strand 5'-GCUUCUUCAGAGGCGACUAtt-3' and antisense strand 5'-UAGUCGCCUCUGAAGAAGCta-3', wherein the negative control siRNA does not target any known mammalian gene, siRNA transfected cells were cultured for 4-6 hours, and CsA was added to cells with/without CHBP treatment;
step ten, determining apoptosis by a flow cytometer: collecting TCMK-1 cells attached to the wall, resuspending the Cell pellet in buffer, and incubating with annexin-V and PI away from light for 15 minutes, taking the incubation of annexin-V or PI only and the dye-free set as controls, analyzing the sample by BD FACS Calibur flow cytometer using Cell Quest research software, counting 10000 cells, and the result quadrant dot plot shows: viable cells (Annexin V-/PI-), early apoptotic cells (Annexin V +/PI-), late apoptotic cells (Annexin V +/PI +) or necrotic cells (Annexin V-/PI +), the number of each type of cell being expressed as a percentage of total gated cells;
eleventh, statistical analysis: statistical analysis was performed using GraphPad Prism 6.0 software, data expressed as mean ± Standard Error of Mean (SEM), analysis of variance (ANOVA) was used to compare results between three or more groups, two-sided t-test was used to compare results between two groups, P ≦ 0.05 was considered statistically significant, and all data from cell culture studies represent at least three independent experiments.
Further, in the first step, 2mg of CsA was dissolved in 1ml of olive oil, and the mice in the group IR were gavaged daily at 35mg/kg body weight, while 0.11mg of CHBP was dissolved in 32ml of 0.9% physiological saline every 3 days, and the mice in the group IR were intraperitoneally injected at 24nmol/kg body weight.
Further, the third step is interstitial expansion as edema or fibrosis.
Further, the sixth step produces 25 micrograms of kidney protein: taking out tissue from-80 deg.C, thawing, adding 1ml protein lysate (prepared by mixing protease inhibitor PMSF and protein lysate RIPA at a ratio of 1: 100) per 100mg tissue, homogenizing on ice homogenizer, mixing in ice bath for 30min, transferring into EP tube, centrifuging at 4 deg.C and 12000g for 15min, and collecting supernatant. And (3) determining the protein concentration by using a BCA method of a protein quantification kit, taking a certain amount of protein liquid, adding 6 Xprotein loading and diluting to 1X, boiling for 5 minutes in boiling water to denature the protein, and storing at 4 ℃.
Further, the TCMK-1 cells in the ninth step are mouse kidney epithelial cell line CCL139TM。
Further, the CsA treatment in said ninth step was continued for 24 hours with CsA concentrations of 2.5, 5, 10, 20 and 40 μ g/ml, respectively.
Further, Caspase-3 siRNAs were transfected into TCMK-1 cells in the ninth step, where the CASP-3 siRNAs were 10, 20, 30 and 40 nM.
Further, the double-stranded CASP-3siRNA in the ninth step has the sequence: sense strand 5'-GCUUCUUCAGAGGCGACUAtt-3' and antisense strand 5'-UAGUCGCCUCUGAAGAAGCta-3'.
Further, the negative control siRNA in the ninth step is NCsiRNA, #4390843, Thermo Fisher Scientific.
Has the advantages that:
1. the novel cyclic erythropoietin-derived peptides (CHBPs) are used for protection against acute and chronic kidney injury, as well as against nephrotoxic injury from cyclosporin a (csa) during renal transplantation.
2. Establishing a stable mouse model of the toxic kidney injury of IR and CsA, researching the mechanism of acute and chronic kidney injury and observing the effect and mechanism of CHBP whether CsA treatment intervention exists; exploring the signaling pathways that the renal protection of CHBP may involve several mechanisms, it is necessary to further investigate how the potential protective mechanisms are different 2 and 8 weeks after IR and/or CsA-induced injury, in order to screen biological indicators for effective and low-invasive diagnostic and therapeutic monitoring; a novel specific gene therapy (caspase-3 siRNA) was designed to improve the survival of transplanted kidneys.
3. In the 2-8 week model, urinary albumin/creatinine was elevated following IR injury and further elevated following CsA intervention, but CHBP reversed the injury with a trend similar to tubulointerstitial injury, fibrosis, HMGB1 and active caspase-3 expression changes, but no corresponding change in SCr.
4. At weeks 2 and/or 8, IR leads to an increase in apoptotic cells and CsA and CHBP intervention can reduce apoptosis.
5. At both time points CsA treatment reduced the expression of p 70S 6 kinase, mTOR, S6ribosomal protein, GSK-3 β and caspase-3, with or without CHBP intervention, while the addition of CHBP to the IR + CsA group increased the expression of p53 and S6 RP.
6. The protection effect of CHBP is reflected in that the combined effect of the injury acute phase and CASP-3siRNA can reduce the CSA-induced TCMK-1 cell apoptosis.
7. The protection of CHBP during acute phase of renal injury is manifested by repair of IR injury, while during chronic injury, the protection of CHBP is manifested by protection of CsA nephrotoxic injury, with different underlying mechanisms. Urinary albumin/creatinine is a better biomarker compared to Scr. The CHBP and CASP-3siRNA synergistic effect can effectively reduce the CsA-induced tubular epithelial cell apoptosis.
8. CHBP decreased urinary albumin, and at weeks 2 and 4, the CsA treated group increased the urinary albumin/creatinine ratio (mg/. mu.mol) compared to the IR group (FIGS. 1A, B), and at weeks 6 and 8, the IR group increased the urinary albumin/creatinine ratio compared to the control group (FIGS. 1C, D). CHBP treatment in the IR group significantly reduced this ratio at all time points with/without CsA treatment (FIGS. 1A-D).
In addition, the dynamic distribution of urinary albumin/creatinine was higher in the IR group than in the control and CHBP-treated groups at all time points of the subsequent 2-8 weeks, and the urinary albumin/creatinine ratio was further increased by CsA (fig. 1E). However, the difference between the control and IR groups appeared to expand over time, while CHBP treatment with/without CsA reduced the difference between them. The trends for the two CsA groups with/without CHBP were unexpectedly similar, both peaking at week 4, declining at week 6, and then settling at week 8, despite the remote distance between the two.
However, at weeks 2 and 8, there was no significant difference in SCR levels in all groups (FIG. 1F, G).
9. CHBP improved tissue damage and semi-quantitative analysis of TID was performed in H & E stained sections. TID scores were significantly higher in the IR group at weeks 2 and 8 than in the control group (fig. 2A-D), and CsA was further higher at week 8 than in the IR group. Most interestingly, the IR group improved TID by CHBP treatment only at 2 weeks, and the CsA group improved TID in both time points.
Trichrome staining by Masson showed a significant increase in interstitial fibrosis in the IR group compared to the control group, but was improved by CHBP at both time points (fig. 3A-D). At week 8, CHBP in the CsA treated group significantly reduced the score of interstitial fibrosis.
10. CHBP and CsA can reduce renal cell apoptosis, ISEL detected apoptotic cells mainly located in renal tubule and interstitial region, and some cells have polymorphonuclear (FIG. 4A, C), and the glomerular region has few apoptotic cells. From the total number of apoptotic cells in the small tract, interstitial and luminal areas, IR significantly increased the total apoptotic cell number, CsA decreased the total apoptotic cell number at 2 and 8 weeks, and CHBP decreased only at 8 weeks (fig. 4B).
11. CHBP changes Caspase-3 mRNA and protein, and qPCR and Western blot are respectively used for detecting the expression of Caspase-3 mRNA and protein. Caspase-3 mRNA levels at both time points increased with the IR group, further increased in the CsA group at 2 weeks, and decreased under CHBP treatment at 8 weeks (FIGS. 5A, E).
At 8 weeks, expression of the 32kD caspase-3 precursor was reduced by CsA (FIGS. 5B, C, and F, G). Levels of 17kD active caspase-3 were elevated in the IR group at 2 weeks and continued to be elevated in the CsA group at 8 weeks, but were all reversed by CHBP treatment (FIG. 5D, H).
12. The CHBP reduces HMGB1 protein, and expression of HMGB1 is detected by Western blotting (FIG. 6A). CsA was elevated only at 8 weeks compared to the IR group, and expression of HMGB1 in the 2-week IR group and the 8-week CsA-treated group was reduced after CHBP treatment (fig. 6B, C).
13. The different proteins differed between week 2 and week 8, and the protein chip was used to simultaneously detect 18 proteins in the kidney (fig. 7A, B). At week 2, p 70S 6 kinase, mTOR, and caspase-3 were reduced in expression compared to the IR group in CsA group with or without CHBP treatment (FIG. 7C-E), whereas in CsA treated group CHBP increased p53 expression (FIG. 7F).
At 8 weeks, CsA expression of IR group S6ribosomal protein, GSK-3 β and caspase-3 was downregulated by the presence/absence of CHBP, and the IR group caspase-3 was also decreased by CHBP. However, CHBP increased the expression of S6ribosomal protein in the CsA-treated group (FIGS. 7G-I).
14. CHBP reversed the increase in caspase-3 expression and apoptosis in TCMK-1 cells caused by CsA, and caspase-3 mRNA expression levels increased with CsA concentration (2.5-20. mu.g/ml) (FIG. 8A). At 20, 40 μ g/ml, the percentage of early apoptotic cells increased significantly (fig. 8B, C), but the proportion of early apoptosis decreased significantly after treatment with CHBP, with a minimum at 20ng/ml (fig. 8D, E).
15. In the TCMK-1 cell model, caspase-3 siRNA reduced caspase-3 mRNA and apoptosis, and caspase-3 mRNA expression was reduced from 10-30nM in the CASP-3siRNA treated group compared to the control with or without NCsiRNA using 20. mu.g/ml CsA, with maximal inhibition at 30nM/ml, reduced to 64.85% (FIG. 9A). Caspase-3 mRNA expression was significantly increased under CsA but was reversed by CHBP with/without CASP-3siRNA (FIG. 9B). Similarly, CsA significantly increased early apoptotic cells, but was reversible by CHBP with or without CASP-3siRNA or NCsiRNA (FIGS. 9C, D). More interestingly, combination treatment with CHBP and CASP-3siRNA could further reduce early apoptotic cells compared to the CsA + CHBP + NCsiRNA group.
16. The primary protection of CHBP for IR injury was at 2 weeks and protection for CsA nephrotoxicity was at 8 weeks, urinary albumin/creatinine as a better biomarker than SCr. The potential signaling pathway for renal protection by CHBP is associated with different intercellular proteins, such as mTOR at 2 weeks and GSK-3 β at 8 weeks, but caspase-3 is associated at both time points. The CHBP combined with CASP-3siRNA has a profound protective effect on CsA-induced injury in TECs.
Drawings
FIG. 1 is a graph of urine albumin/creatinine and SCR analysis according to example 1 of the present application;
FIG. 2 is a TID semi-quantitative score chart of H & E stained sections in example 2 of the present application.
FIG. 3 is a graph showing the scores of Masson trichrome stained sections in example 2 of the present application.
FIG. 4 is a graph of apoptotic cell staining in example 3 of the present application.
FIG. 5 is a graph showing the expression of caspase-3 mRNA detected by qPCR and the expression of caspase-3 protein detected by western blot in example 5 of the present application.
FIG. 6 is an expression diagram of HMGB1 protein detected by western blot in example 4 of the present application.
FIG. 7 is a diagram of detection of 18 proteins on the protein chip in example 6 of the present application.
FIG. 8 is a graph showing the effect of CsA and CHBP on TCMK-1 cell caspase-3 mRNA expression and apoptosis in example 7 of the present application.
FIG. 9 is a graph showing the effect of CASP-3siRNA on TCMK-1 cell caspase-3 mRNA expression and apoptosis in example 8 of the present application.
Detailed Description
The following examples illustrate specific steps of the present invention, but are not intended to limit the invention.
The terms used in the present invention have meanings generally understood by those of ordinary skill in the art unless otherwise specified.
The invention is described in further detail below with reference to specific examples and with reference to data. It should be understood that these examples are intended to illustrate the present invention, and are not intended to limit the scope of the present invention in any way.
In the following examples, various procedures and methods not described in detail are conventional methods well known in the art.
25-30g of male BALB/c mice were purchased from the university of Nantong laboratory animal center. CHBP was purchased from Shanghai institute of medicine, Chinese academy of sciences, and protein chip kit was purchased from Chemiesce Readout, Denfoss, USA. All animal handling was performed according to the requirements of the university of southwest university committee on animal care and use and the ethical committee on animal care in Jiangsu province.
Example 1
The application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection is to establish a mouse kidney injury model: male BALB/c mice, 25-30g, were randomly divided into 5 groups of 6 mice each, i.e. n-6, with control groups: the abdominal cavity and renal pedicles were exposed and anesthetized with 0.01ml/g of 1% pentobarbital; IR group: separating two renal pedicles, clamping them for 30min with vascular clamps to discolor the kidneys, and then releasing the vascular clamps to allow reperfusion of the blood for 2 or 8 weeks; IR + CsA group: dissolving 2mg CsA in 1ml olive oil, and intragastrically administering to IR mice per day according to 35mg/kg body weight standard; IR + CHBP group: dissolving 0.11mg CHBP in 32ml 0.9% saline, and injecting into IR mouse abdominal cavity once every 3 days according to 24nmol/kg body weight standard; IR + CsA + CHBP group: IR mice were treated with CsA and CHBP simultaneously by intraabdominal injection of 2mg CsA in 1ml olive oil, intragastric administration of 35mg/kg body weight standard to IR mice per day, intraabdominal injection of 0.11mg CHBP in 32ml 0.9% saline every 3 days, and intraabdominal injection of 24nmol/kg body weight standard to IR mice.
Urine samples were collected at weeks 2, 4, 6 and 8 using a metabolism cage, animals were sacrificed at weeks 2 or 8, blood samples and kidney samples were collected, kidney samples were fixed with 10% (w/v) neutral buffered formalin, stored at-80 ℃ for later use after snap freezing in liquid nitrogen, urine samples were measured for urinary albumin/creatinine using an automated biochemical analyzer, and blood samples were measured for serum creatinine (SCr) using an automated biochemical analyzer.
As shown in FIG. 1A, B, at weeks 2 and 4, the CsA-treated group increased the urinary albumin/creatinine ratio (mg/. mu.mol) compared to the IR group, and at weeks 6 and 8, as shown in FIG. 1C, D, the IR group increased the urinary albumin/creatinine ratio compared to the control group, and at all time points with/without CsA treatment, as shown in FIGS. 1A-D, the CHBP treatment in the IR group significantly decreased the ratio.
As shown in fig. 1E, the dynamic distribution of urinary albumin/creatinine was higher in the IR group than in the control and CHBP-treated groups at all time points of the subsequent 2-8 weeks, while CsA further increased the urinary albumin/creatinine ratio, but the difference between the control and IR groups appeared to increase with time, while CHBP combined CsA/CsA-free treatment diminished the difference between them. The trends for the two CsA groups with/without CHBP were unexpectedly similar, both peaking at week 4, declining at week 6, and then settling at week 8, despite the remote distance between the two.
As shown in FIG. 1F, G, there was no significant difference in the SCr levels between all groups at week 2 and week 8.
Example 2
Histological evaluation:
kidney specimens were paraffin-embedded sections, stained with hematoxylin and eosin (H & E), and semi-quantitative assessment of the degree of tubulointerstitial damage (TID) was scored from 0 to 4 points: the percentage of the damaged area is less than 5 percent, and 0 is obtained; 5% -25% to obtain 1 point; 25% -50% to obtain 2 points; 50% -75% to obtain 3 points; 4 points are obtained when the content exceeds 75 percent; the parameters evaluated include renal tubule dilation and vacuole in the renal tubular lumen, interstitial dilation i.e. edema or fibrosis, inflammatory cell infiltration, protein casts, intraluminal cells or cell debris; each section was scored by randomly selecting 12 cortical fields at 200x magnification.
As shown in fig. 2A-D, semi-quantitative analysis of TID in H & E stained sections, TID scores were significantly higher for the IR group at weeks 2 and 8 than for the control group, CsA was further higher at week 8 than for the IR group, TID was improved by CHBP treatment only at week 2 for the IR group, and TID was improved by the CsA group at both time points.
Masson trichrome staining, with tubulointerstitial fibrosis detected, 20 cortical fields were randomly selected at 400x magnification per section and scanned using Image-Pro Plus software to assess collagen deposition.
As shown in FIGS. 3A-D, trichrome staining by Masson showed a significant increase in interstitial fibrosis in the IR group compared to the control group, but was improved by CHBP at both time points, with the CsA treated group having CHBP that significantly reduced the score of interstitial fibrosis at week 8.
Example 3
In situ end-labeling of apoptotic cells:
sections the fragmented dna (isel) was labeled in situ with terminal deoxynucleotidyl transferase (TdT) using the ApopTag peroxidase kit, sections were digested with 40 μ g/mL proteinase K for 15 minutes at 37 ℃, then incubated with TdT and digoxigenin-dUTP for 60 minutes at 37 ℃, after adding digoxigenin-peroxidase complex for 30 minutes, sections were visualized with 3' -amino-9-ethylcarbazole (AEC) substrate and 20 fields of view were selected for detection of apoptotic cells in tubules, luminal and interstitial regions in 200x fields.
As shown in fig. 4A-C, the apoptotic cells detected by ISEL were mainly localized in the tubular and interstitial regions, some cells had polymorphonuclear nuclei, and apoptotic cells were rarely found in the glomerular region, and the total apoptotic cell count was greatly increased by IR, decreased by CsA at 2 and 8 weeks, and decreased by CHBP only at 8 weeks, as seen in fig. 4C.
Example 4
Western blot analysis:
preparation of 25 microgram of kidney protein was isolated on 12-15% polyacrylamide denaturing gel and transferred to PVDF membrane, blocked with 5% milk prior to incubation with caspase-3 antibody, HMGB1 antibody, internal reference beta-actin, followed by incubation with peroxidase-coupled secondary antibody, scanning for visualization using molecular imager Chemi Doc XRS + system, and semi-quantitative analysis by Image Lab software.
As shown in fig. 6A, expression of HMGB1 was detected by Western blotting. As shown in fig. 6B, C, CsA was elevated only at 8 weeks compared to the IR group, and expression of HMGB1 in the 2-week IR group and the 8-week CsA-treated group was reduced after CHBP treatment.
Example 5
Real-time quantitative PCR:
detection of Caspase-3 mRNA, Caspase-3 and housekeeping gene GAPDH probes in kidney and TCMK-1 cells in the StepOne Plus real-time PCR System by Reverse Transcription (RT) real-time quantitative polymerase chain reaction (qPCR) were labeled with 6-carboxyfluorescein (FAM), 2. mu.g of total RNA extracted with Trizol reagent was used for reverse transcription to cDNA, 2. mu.l of cDNA was amplified in qPCR reaction buffer with Taq polymerase, where the reaction system containing 900nM forward, reverse primers and 250nM probe was performed at 95 ℃ for 10 min followed by 40 cycles at 95 ℃ for 15 sec and 60 ℃ for 1 min, with normal as control and GAPDH as correction, using 2-ΔΔCtThe method calculates caspase-3 mRNA expression.
As shown in FIG. 5A, E, Caspase-3 mRNA and protein expression was measured by qPCR and Western blot, and Caspase-3 mRNA levels at two time points were increased in the IR group, further increased in the CsA group at 2 weeks, and decreased under CHBP treatment at 8 weeks.
As shown in FIG. 5B, C, F, G, at 8 weeks, expression of the 32kD caspase-3 precursor was reduced by CsA, 17kD active caspase-3 levels were elevated in the IR group at 2 weeks and continued to be elevated in the CsA group at 8 weeks, as shown in FIG. 5D, H, but all were reversible by CHBP treatment.
Example 6
Protein chip analysis:
protein chip kit (Chemiescence Readout, denver, usa) which can simultaneously detect 18 phosphorylated or cleaved signal molecules using 50 μ g protein, acquire images by briefly exposing slides to a molecular imaging system according to the chip instructions, and perform semi-quantitative analysis by scanning the bulk density using Alpha View software 3.3.
As shown in FIG. 7A, B, the protein chip was used to detect 18 proteins in kidney simultaneously, and at 2 weeks, p 70S 6 kinase was present in CsA group with or without CHBP treatment, as shown in FIG. 7C-E, and both mTOR and caspase-3 expression were reduced compared to IR group, while in CsA treated group, CHBP increased p53 expression as shown in FIG. 7F.
As shown in FIGS. 7G-I, at 8 weeks, expression of IR group S6ribosomal protein, GSK-3 β and caspase-3 was downregulated by CsA with/without CHBP, and the IR group caspase-3 was also decreased by CHBP. However, in the CsA-treated group, CHBP increased the expression of S6ribosomal protein.
Example 7
TCMK-1 cell culture:
TCMK-1 cells (mouse renal epithelial cell line, CCL 139) were cultured in DMEM/F12 medium containing 10% fetal bovine serum, 100 units/ml penicillin G and 100. mu.g/ml streptomycinTM) Caspase-3 siRNAs (CASP-3 siRNAs, 10, 20, 30, and 40nM) were transfected into TCMK-1 cells using Lipofectamine @ RNAiMAX for 24 hours at cell length fusions of 60-70% with/without CHBP (2.5, 5, 10, 20, and 40 μ g/ml), the sequence of the double-stranded CASP-3siRNA being: sense strand 5'-GCUUCUUCAGAGGCGACUAtt-3' and antisense strand 5'-UAGUCGCCUCUGAAGAAGCta-3', in which the negative control siRNA (NCsiRNA, #4390843, Thermo Fisher Scientific) did not target any known mammalian genes, siRNA transfected cells were cultured for 4-6 hours and CsA was added to cells with/without CHBP treatment.
As shown in FIG. 8A, the expression level of caspase-3 mRNA increased with increasing CsA concentration (2.5-20. mu.g/ml), and at 20, 40. mu.g/ml, the percentage of early apoptotic cells was significantly increased as shown in FIG. 8B, C, but the proportion of early apoptosis after treatment with CHBP was significantly decreased, as shown in FIG. 8D, E, which was lowest at 20 ng/ml.
Example 8
Flow cytometry for apoptosis:
TCMK-1 cells were harvested, Cell pellets resuspended in buffer and incubated with annexin-V and PI away from light for 15 minutes, and samples were analyzed by BD FACS Calibur flow cytometer using Cell Quest research software, with annexin-V or PI alone and no dye set as controls, counting 10,000 cells, with quadrant dot plots showing: viable cells (Annexin V-/PI-), early apoptotic cells (Annexin V +/PI-), late apoptotic cells (Annexin V +/PI +) or necrotic cells (Annexin V-/PI +), the number of each type of cell being expressed as a percentage of total gated cells.
As shown in FIG. 9A, the caspase-3 mRNA expression in the CASP-3siRNA treated group was reduced from 10-30nM, with the maximum inhibition at 30nM being 64.85% compared to the control with or without NCsiRNA using 20. mu.g/ml CsA, and the caspase-3 mRNA expression was significantly increased by CsA, as shown in FIG. 9B, but was reversed by the presence/absence of CASP-3siRNA, and similarly, CsA significantly increased early apoptotic cells, but with or without CASP-3siRNA or NCsiRNA, as shown in FIG. 9C, D, the combined treatment with CHBP and CASP-3siRNA further reduced early apoptotic cells compared to the CsA + CHBP + NCsiRNA group.
Example 9
Statistical analysis:
statistical analysis was performed using GraphPad Prism 6.0 software, data expressed as mean ± Standard Error of Mean (SEM), analysis of variance (ANOVA) was used to compare results between three or more groups, two-sided t-test was used to compare results between two groups, P ≦ 0.05 was considered statistically significant, and all data from cell culture studies represent at least three independent experiments.
Sequence listing
<110> university of southeast Tong
AFFILIATED HOSPITAL OF NANTONG University
<120> application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection
<160> 2
<170> SIPOSequenceListing 1.0
<210> 1
<211> 21
<212> DNA/RNA
<213> Artificial sequence (2 Ambystoma latex x Ambystoma jeffersonia)
<400> 1
gcuucuucag aggcgacuat t 21
<210> 2
<211> 21
<212> DNA/RNA
<213> Artificial sequence (2 Ambystoma latex x Ambystoma jeffersonia)
<400> 2
uagucgccuc ugaagaagct a 21
Claims (10)
1. An application of cyclic erythropoietin-derived peptide in protecting kidney injury and cyclosporin A injury.
2. The use of a cyclic erythropoietin-derived peptide according to claim 1 for kidney injury and cyclosporin a injury protection, characterized by the steps of:
the first step is as follows: establishing a mouse kidney injury model: male BALB/c mice, 25-30g, were randomly divided into 5 groups of 6 mice each, i.e. n =6, with control groups: the abdominal cavity and renal pedicles were exposed and anesthetized with 0.01ml/g of 1% pentobarbital; IR group: separating two renal pedicles, clamping them for 30min with vascular clamps to discolor the kidneys, and then releasing the vascular clamps to allow reperfusion of the blood for 2 or 8 weeks; IR + CsA group: 2mg CsA was dissolved in 1ml olive oil and the IR group mice were gavaged daily at 35mg/kg body weight; IR + CHBP group: dissolving 0.11mg CHBP in 32ml 0.9% physiological saline, and injecting into IR group mice abdominal cavity according to 24nmol/kg body weight standard once every 3 days; IR + CsA + CHBP group: the IR group mice were treated simultaneously with CsA gavage and CHBP once every 3 days;
the second step is that: collecting urine samples at weeks 2, 4, 6 and 8 using a metabolism cage, at week 2 or 8, sacrificing the animals, collecting blood samples and kidney samples, fixing the kidney samples with 10% (w/v) neutral formalin buffer solution, quick freezing in liquid nitrogen and storing at-80 ℃ for later use, measuring urine albumin/creatinine using an automated biochemical analyzer for urine samples, and measuring serum creatinine (SCr) using an automated biochemical analyzer for blood samples;
the third step: kidney specimens were paraffin-embedded sections, stained with hematoxylin and eosin (H & E), and semi-quantitative assessment of the degree of tubulointerstitial damage (TID) was scored from 0 to 4 points: the percentage of the damaged area is less than 5 percent, and 0 is obtained; 5% -25% to obtain 1 point; 25% -50% to obtain 2 points; 50% -75% to obtain 3 points; 4 points are obtained when the content exceeds 75 percent; the parameters evaluated include tubular dilation and vacuoles within the tubular lumen, interstitial dilation, inflammatory cell infiltration, protein casts, intraluminal cells or cell debris; each section was scored by randomly selecting 12 cortical fields at 200x magnification;
the fourth step: masson trichrome staining, detecting tubulointerstitial fibrosis, randomly selecting 20 cortical fields for each section under 400x magnification, and scanning using Image-Pro Plus software to assess collagen deposition;
fifth, the sections were labeled in situ with terminal deoxynucleotidyl transferase (TdT) fragmented dna (isel) using the ApopTag peroxidase kit: digesting the section with 40 mug/mL proteinase K for 15 minutes at 37 ℃, then incubating the section with TdT and digoxigenin-dUTP for 60 minutes at 37 ℃, adding digoxigenin-peroxidase complex for 30 minutes, developing the section with 3' -amino-9-ethylcarbazole (AEC) substrate, and selecting 20 fields in 200x fields to detect apoptotic cells of tubules, lumen and interstitial region;
sixth step, western blot analysis: preparing 25 microgram of kidney protein to separate on 12-15% polyacrylamide denaturing gel and transferring it to PVDF membrane, blocking with 5% milk before incubation with caspase-3 antibody, HMGB1 antibody, internal reference beta-actin, after incubation with peroxidase-coupled secondary antibody, scanning and developing using molecular imager Chemi Doc XRS + system, and semi-quantitative analysis by Image Lab software;
seventh step, real-time quantitative PCR: detection of Caspase-3 mRNA, Caspase-3 and housekeeping gene GAPDH probes in kidney and TCMK-1 cells in StepOne Plus real-time PCR System by Reverse Transcription (RT) real-time quantitative polymerase chain reaction (qPCR) were labeled with 6-carboxyfluorescein (FAM), 2. mu.g of total RNA extracted with Trizol reagent was used for reverse transcription into cDNA, 2. mu.l of cDNA was amplified in qPCR reaction buffer with Taq polymerase, wherein the reaction system containing 900nM forward, reverse primer and 250nM probe was in a StepOne Plus real-time PCR system10 min at 95 ℃ followed by 40 cycles of 15 sec at 95 ℃ and 1 min at 60 ℃ with normal group as control and GAPDH as correction, 2-ΔΔCtThe method calculates the expression of caspase-3 mRNA;
eighth step, protein chip analysis: the protein chip in the protein chip kit simultaneously detects 18 phosphorylated or cracked signal molecules by using 50 mug protein, obtains an image by briefly exposing a glass slide to a molecular imaging system according to a chip use instruction, and performs semi-quantitative analysis by scanning volume density by using Alpha View software 3.3;
ninth step, TCMK-1 cell culture: TCMK-1 cells were cultured in DMEM/F12 medium containing 10% fetal bovine serum, 100 units/ml penicillin G and 100. mu.g/ml streptomycin, the cells were treated with CsA for 24 hours with/without CHBP at 60-70% cell length fusion, Caspase-3 siRNA was transfected into TCMK-1 cells using Lipofectamine @ RNAImax, the sequence of double stranded CASP-3siRNA was: 5'-GCUUCUUCAGAGGCGACUAtt-3' and 5'-UAGUCGCCUCUGAAGAAGCta-3', wherein the negative control siRNA does not target any known mammalian genes, the siRNA transfected cells are cultured for 4-6 hours and CsA is added to the cells with/without CHBP treatment;
step ten, determining apoptosis by a flow cytometer: TCMK-1 cells attached to the wall were collected, the Cell pellet resuspended in buffer and incubated with annexin-V and PI away from light for 15 minutes, and the samples were analyzed by BD FACS Calibur flow cytometer using Cell Quest research software, with annexin-V or PI incubated alone and no dye set as controls, and 10000 cells counted, with the results shown in a quadrant dot plot: viable cells (Annexin V-/PI-), early apoptotic cells (Annexin V +/PI-), late apoptotic cells (Annexin V +/PI +) or necrotic cells (Annexin V-/PI +), the number of each type of cell being expressed as a percentage of total gated cells;
eleventh, statistical analysis: statistical analysis was performed using GraphPad Prism 6.0 software, data expressed as mean ± Standard Error of Mean (SEM), analysis of variance (ANOVA) was used to compare results between three or more groups, two-sided t-test was used to compare results between two groups, P ≦ 0.05 was considered statistically significant, and all data from cell culture studies represent at least three independent experiments.
3. The use of a cyclic erythropoietin-derived peptide according to claim 2 for kidney injury and cyclosporin a injury protection, wherein: in the first step, 2mg of CsA is dissolved in 1ml of olive oil, the mice in the IR group are subjected to intragastric administration according to the standard of 35mg/kg of body weight every day, and 0.11mg of CHBP is dissolved in 32ml of 0.9% physiological saline every 3 days and is subjected to intraperitoneal injection once according to the standard of 24nmol/kg of body weight.
4. The use of a cyclic erythropoietin-derived peptide according to claim 3 for kidney injury and cyclosporin A injury protection, wherein: the third step is interstitial expansion as edema or fibrosis.
5. The use of a cyclic erythropoietin-derived peptide according to claim 1 for kidney injury and cyclosporin a injury protection, wherein: the sixth step prepares 25 micrograms of kidney protein: taking out the tissue from-80 ℃, adding 1ml of protein lysate (prepared by mixing protease inhibitor PMSF and protein lysate RIPA according to the ratio of 1: 100) into every 100mg of tissue after thawing, homogenizing on an ice homogenizer, mixing in an ice bath for 30min, then transferring into an EP tube, centrifuging for 15min at 4 ℃ and 12000g, taking the supernatant, measuring the protein concentration by using a protein quantification kit BCA method, taking a certain amount of protein solution, adding 6 times of protein loading and diluting to 1 x, boiling for 5min with boiling water to denature the protein, and storing at 4 ℃.
6. The use of a cyclic erythropoietin-derived peptide according to claim 1 for kidney injury and cyclosporin a injury protection, wherein: in the ninth step, TCMK-1 cells are mouse renal epithelial cell line CCL139 cells.
7. The use of a cyclic erythropoietin-derived peptide according to claim 1 for kidney injury and cyclosporin a injury protection, wherein: the CsA treatment in the ninth step was continued for 24 hours with CsA concentrations of 2.5, 5, 10, 20 and 40. mu.g/ml, respectively.
8. The use of a cyclic erythropoietin-derived peptide according to claim 1 for kidney injury and cyclosporin a injury protection, wherein: caspase-3 siRNA was transfected into TCMK-1 cells in the ninth step, where CASP-3siRNA was 10, 20, 30 and 40 nM.
9. The use of a cyclic erythropoietin-derived peptide according to claim 1 for kidney injury and cyclosporin a injury protection, wherein: the sequence of the double-stranded CASP-3siRNA in the ninth step is as follows: sense strand 5'-GCUUCUUCAGAGGCGACUAtt-3' and antisense strand 5'-UAGUCGCCUCUGAAGAAGCta-3'.
10. The use of a cyclic erythropoietin-derived peptide according to claim 1 for kidney injury and cyclosporin a injury protection, wherein: the negative control siRNA in the ninth step is NCsiRNA, #4390843, Thermo Fisher Scientific.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202010740606.4A CN112076309A (en) | 2020-07-27 | 2020-07-27 | Application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202010740606.4A CN112076309A (en) | 2020-07-27 | 2020-07-27 | Application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection |
Publications (1)
Publication Number | Publication Date |
---|---|
CN112076309A true CN112076309A (en) | 2020-12-15 |
Family
ID=73735196
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202010740606.4A Pending CN112076309A (en) | 2020-07-27 | 2020-07-27 | Application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN112076309A (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN112972653A (en) * | 2021-02-23 | 2021-06-18 | 温州医科大学附属第二医院(温州医科大学附属育英儿童医院) | Application of CHBP in promotion of ultra-long random flap survival |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040219603A1 (en) * | 2003-03-27 | 2004-11-04 | Prasad Devarajan | Method and kit for detecting the early onset of renal tubular cell injury |
CN109908331A (en) * | 2019-04-30 | 2019-06-21 | 青岛大学附属医院 | Application of the Hexarelin in preparation protection ischemia-reperfusion injury of kidney drug/pharmaceutical composition |
-
2020
- 2020-07-27 CN CN202010740606.4A patent/CN112076309A/en active Pending
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20040219603A1 (en) * | 2003-03-27 | 2004-11-04 | Prasad Devarajan | Method and kit for detecting the early onset of renal tubular cell injury |
CN102183656A (en) * | 2003-03-27 | 2011-09-14 | 儿童医院医疗中心 | A method and kit for detecting the early onset of renal tubular cell injury |
CN109908331A (en) * | 2019-04-30 | 2019-06-21 | 青岛大学附属医院 | Application of the Hexarelin in preparation protection ischemia-reperfusion injury of kidney drug/pharmaceutical composition |
Non-Patent Citations (3)
Title |
---|
YUANYUAN WU 等: "Protective Effects of HBSP on Ischemia Reperfusion and Cyclosporine A Induced Renal Injury", CLINICAL AND DEVELOPMENTAL IMMUNOLOGY * |
YUANYUAN WU 等: "Transcriptional Profiles in Ischemia/Reperfusion Injured Murine Kidneys Synergistically Protected by Erythropoietin Derived Peptide CHBP and Caspase-3 siRNA", PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS * |
YUFANG ZHANG 等: "Long-Term Protection of CHBP Against Combinational Renal Injury Induced by Both Ischemia– Reperfusion and Cyclosporine A in Mice", FRONT. IMMUNOL * |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN112972653A (en) * | 2021-02-23 | 2021-06-18 | 温州医科大学附属第二医院(温州医科大学附属育英儿童医院) | Application of CHBP in promotion of ultra-long random flap survival |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Ling et al. | Therapeutic role of TGF-β–neutralizing antibody in mouse cyclosporin A nephropathy: morphologic improvement associated with functional preservation | |
Asai et al. | Magnesium supplementation prevents experimental chronic cyclosporine a nephrotoxicity via renin-angiotensin system independent mechanism. | |
Shu et al. | Catalpol ameliorates endothelial dysfunction and inflammation in diabetic nephropathy via suppression of RAGE/RhoA/ROCK signaling pathway | |
Nespoux et al. | Gene knockout of the Na+-glucose cotransporter SGLT2 in a murine model of acute kidney injury induced by ischemia-reperfusion | |
Kodama et al. | A human serum albumin–thioredoxin fusion protein prevents experimental contrast-induced nephropathy | |
Lindblom et al. | Delineating a role for the mitochondrial permeability transition pore in diabetic kidney disease by targeting cyclophilin D | |
Gouyon et al. | Long-term effects of in utero exposure to cyclosporin A on renal function in the rabbit | |
Lin et al. | Metformin attenuates cyclosporine A-induced renal fibrosis in rats | |
Yang et al. | Influence of the renin-angiotensin system on epidermal growth factor expression in normal and cyclosporine-treated rat kidney | |
Deaciuc et al. | Inhibition of caspases in vivo protects the rat liver against alcohol‐induced sensitization to bacterial lipopolysaccharide | |
Lim et al. | Long-term treatment with cyclosporine decreases aquaporins and urea transporters in the rat kidney | |
Srichai et al. | Apoptosis of the thick ascending limb results in acute kidney injury | |
CN112076309A (en) | Application of cyclic erythropoietin-derived peptide in kidney injury and cyclosporin A injury protection | |
Nakao et al. | Low-dose carbon monoxide inhibits progressive chronic allograft nephropathy and restores renal allograft function | |
Li et al. | Klotho regulates epithelial-to-mesenchymal transition in vitro via Wnt/β-catenin pathway and attenuates chronic allograft dysfunction in a rat renal transplant model | |
Harb et al. | Nicorandil prevents the nephrotoxic effect of cyclosporine-A in albino rats through modulation of HIF-1α/VEGF/eNOS signaling | |
Homma et al. | Activation of renal angiotensin type 1 receptor contributes to the pathogenesis of progressive renal injury in a rat model of chronic cardiorenal syndrome | |
Carlessi et al. | Exendin‐4 attenuates brain death–induced liver damage in the rat | |
Hu et al. | Role of (pro) renin receptor in cyclosporin A-induced nephropathy | |
Kimura et al. | Protective effects of C-type natriuretic peptide on cisplatin-induced nephrotoxicity in mice | |
Han et al. | The role of NF-kB in the downregulation of organic cation transporter 2 expression and renal cation secretion in kidney disease | |
CN112795637A (en) | Use of inhibitors targeting IL-17C for the treatment of inflammation-related chronic kidney disease | |
Sun et al. | Blockade of angiotensin II with losartan attenuates transforming growth factor-β1 inducible gene-h3 (βig-h3) expression in a model of chronic cyclosporine nephrotoxicity | |
WO2022105870A1 (en) | Methods of treating systemic lupus erythematosus using btk inhibitors | |
AU2023217319A1 (en) | Neutralization of acyl-coa binding protein confers autophagy-dependent organ protection |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination | ||
RJ01 | Rejection of invention patent application after publication | ||
RJ01 | Rejection of invention patent application after publication |
Application publication date: 20201215 |