Human umbilical cord-derived mesenchymal stem cells improve the function of liver in rats with acute-on-chronic liver failure via downregulating Notch and Stat1/Stat3 signaling

Yulin He, Xingrong Guo, Tingyu Lan, Jianbo Xia, Jinsong Wang, Bei Li, Chunyan Peng, Yue Chen, Xiang Hu, Zhongji Meng, Yulin He, Xingrong Guo, Tingyu Lan, Jianbo Xia, Jinsong Wang, Bei Li, Chunyan Peng, Yue Chen, Xiang Hu, Zhongji Meng

Abstract

Background: Effective treatments for acute-on-chronic liver failure (ACLF) are lacking. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) have been applied in tissue regeneration and repair, acting through paracrine effects, cell fusion, and actual transdifferentiation. The present study was designed to investigate the therapeutic potential of hUC-MSCs in acute-on-chronic liver injury (ACLI) and ACLF rat models.

Methods: Wistar rats aged 6 weeks were intraperitoneally administered porcine serum (PS) at a dose of 0.5 mL twice per week for 11 weeks to generate an immune liver fibrosis model. After 11 weeks, rats with immune liver fibrosis were injected intravenously with lipopolysaccharide (LPS) to induce an ACLI model or combined LPS and D-galactosamine (D-GalN) to induce an ACLF model. The rats with ACLI or ACLF were injected intravenously with 2×106 hUC-MSCs, 4×106 hUC-MSCs, or 0.9% sodium chloride as a control. The rats were sacrificed at 1, 2, 4, and 6 weeks (ACLI rats) or 4, 12, and 24 h (ACLF rats). The blood and liver tissues were collected for biochemical and histological investigation.

Results: The application of hUC-MSCs in rats with ACLI and ACLF led to a significant decrease in the serum levels of ALT, AST, TBil, DBil, ALP, ammonia, and PT, with ALB gradually returned to normal levels. Inflammatory cell infiltration and collagen fiber deposition in liver tissues were significantly attenuated in ACLI rats that received hUC-MSCs. Inflammatory cell infiltration and apoptosis in liver tissues of ACLF rats that received hUC-MSCs were significantly attenuated. Compared with those in the rats that received 0.9% sodium chloride, a significant reduction in proinflammatory cytokine levels and elevated serum levels of hepatocyte growth factor (HGF) were found in ACLF rats that received hUC-MSCs. Furthermore, Notch, IFN-γ/Stat1, and IL-6/Stat3 signaling were inhibited in ACLI/ACLF rats that received hUC-MSCs.

Conclusions: hUC-MSC transplantation can improve liver function, the degree of fibrosis, and liver damage and promote liver repair in rats with ACLI or ACLF, mediated most likely by inhibiting Notch signaling and reversing the imbalance of the Stat1/Stat3 pathway.

Keywords: ACLF; Liver fibrosis; Liver injury; Notch; Paracrine; Stat1; Stat3; hUC-MSCs.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Schedules of the experiments in rats. a Schematic design of hUC-MSC treatment for rats with ACLI. b Schematic design of hUC-MSC treatment for rats with ACLF. c–e Liver fibrosis/cirrhosis was verified by ELISA for serum HA and PIIINP (c), HE staining (d), and Masson staining (e) of liver specimens from rats that received PS treatment. PS injection. Sacrifice. LPS infusion. D-GalN injection. hUC-MSCs infusion
Fig. 2
Fig. 2
The effect of hUC-MSC transplantation in improving liver function in rats with ACLI. ACLI rats were transplanted with hUC-MSCs or 0.9% sodium chloride as controls, and serum levels of ALT (a), AST (b), TBil (c), DBil (d), ALP (e), and ALB (f) were detected with an automatic biochemical analyzer (n = 3/group). Data are presented as the mean ± SD. *P < 0.05
Fig. 3
Fig. 3
The effects of hUC-MSC transplantation on improving liver function and coagulation function in ACLF rats. ACLF rats were transplanted with hUC-MSCs or 0.9% sodium chloride as a control. Serum levels of ALT (a) and AST (b), plasma levels of ammonia (c), and serum levels of ALB (d), TBil (e), and DBil (f) were detected with an automatic biochemical analyzer, and plasma PT (g) and INR (h) were detected with a coagulation analyzer (4h, n = 3/group; 12h and 24h, n = 4/group). Data are presented as the mean ± SD. *P < 0.05, **P < 0.01
Fig. 4
Fig. 4
Histopathological recovery of liver tissues from rats with ACLI transplanted with hUC-MSCs. ACLI rats were transplanted with hUC-MSCs or 0.9% sodium chloride as a control, and liver sections were used for histological investigation with HE staining (a) or Masson staining (b) and microscopic examination (n = 3/group). Representative photographs are shown for the liver histological presentations of rats with ACLI transplanted with hUC-MSCs or 0.9% sodium chloride as a control at weeks 0, 1, 2, 4, and 6
Fig. 5
Fig. 5
Histopathological recovery of liver tissues from ACLF rats transplanted with hUC-MSCs. ACLF rats were transplanted with hUC-MSCs or 0.9% sodium chloride as a control, and liver sections were used for histological investigation with HE staining and microscopic examination (4h, n = 3/group; 12h and 24h, n = 4/group). Representative photographs are shown for the liver histological presentations of ACLF rats transplanted with hUC-MSCs or 0.9% sodium chloride as a control at hours 4, 12, and 24
Fig. 6
Fig. 6
The effects of hUC-MSC transplantation on liver fibrosis in rats with ACLI. ACLI rats were transplanted with hUC-MSCs or 0.9% sodium chloride as a control, and liver sections from ACLI rats 6 weeks post-hUC-MSC transplantation were used for immunohistochemical staining of α-SMA, desmin, MMP9, and TIMP1 and microscopic examination (n = 3/group). Representative photographs are shown (a). Positive staining was quantified and is presented as the mean ± SD (b). *P < 0.05, **P < 0.01
Fig. 7
Fig. 7
The effects of hUC-MSC transplantation on hepatocyte regeneration in ACLF rats. Liver sections from ACLF rats 12 and 24 h post-hUC-MSC transplantation or 0.9% sodium chloride injection as a control were used for immunohistochemical staining of AFP, CK18, HGF, and PCNA and microscopic examination (n = 4/group). Representative photographs are shown (a). Positive staining was quantified and is presented as the mean ± SD (b, c). d Serum levels of HGF were detected by ELISA (4h, n = 3/group; 12h and 24h, n = 4/group). Data are presented as the mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 8
Fig. 8
The effects of hUC-MSC transplantation on the production of proinflammatory cytokines and antiinflammatory cytokines in rats with ACLI. ACLI rats were transplanted with hUC-MSCs or 0.9% sodium chloride as a control. The serum levels of the cytokines TNF-α (a), IFN-γ (b), IL-6 (c), IL-1β (d), TGF-β1 (e), IL-4 (f), and IL-10 (g) were detected by ELISA at weeks 1, 2, 4, and 6 (n = 3/group). Data are presented as the mean ± SD. *P < 0.05
Fig. 9
Fig. 9
The effects of hUC-MSC transplantation on the production of proinflammatory cytokines in ACLF rats. ACLF rats were transplanted with hUC-MSCs or 0.9% sodium chloride as a control. The serum levels of the cytokines TNF-α (a), IFN-γ (b), IL-6 (c), and IL-1β (d) were detected by ELISA at hours 4, 12, and 24 (4h, n = 3/group; 12h and 24h, n = 4/group). Data are presented as the mean ± SD. *P < 0.05, **P < 0.01
Fig. 10
Fig. 10
The effects of hUC-MSC transplantation on Notch signaling pathway-related genes and Stat1/Stat3 signaling molecules in ACLF rats. Total RNA and protein were extracted from the liver tissues from rats with ACLF transplanted with hUC-MSCs or 0.9% sodium chloride as a control(n = 4/group). The mRNA expression levels of Notch1, Hes1, and P21 were detected by quantitative PCR (a). Total protein was subjected to western blotting analysis. Relative protein expression of Stat1, pStat1, Stat3, pStat3, c-Myc, Bcl2, and CyclinD1 were quantitated (b, c). The mRNA expression levels of c-Myc, Bcl2, and CyclinD1 were detected by quantitative PCR (d). Liver sections were used for immunohistochemically staining of pStat1 and pStat3. Representative photographs and the quantitative analysis were shown (e). Data were presented as the mean ± SD. *P < 0.05

References

    1. Moon AM, Singal AG, Tapper EB. Contemporary epidemiology of chronic liver disease and cirrhosis. Clin Gastroenterol Hepatol. 2019;18(12):2650–66.
    1. Sarin SK, Choudhury A, Sharma MK, Maiwall R, Al Mahtab M, Rahman S, et al. Acute-on-chronic liver failure: consensus recommendations of the Asian Pacific association for the study of the liver (APASL): an update. Hepatol Int. 2019;13(4):353–390. doi: 10.1007/s12072-019-09946-3.
    1. Sarin SK, Choudhury A. Acute-on-chronic liver failure: terminology, mechanisms and management. Nat Rev Gastroenterol Hepatol. 2016;13(3):131–149. doi: 10.1038/nrgastro.2015.219.
    1. Nevens F, Laleman W. Artificial liver support devices as treatment option for liver failure. Best Pract Res Clin Gastroenterol. 2012;26(1):17–26. doi: 10.1016/j.bpg.2012.01.002.
    1. Larsen FS. Artificial liver support in acute and acute-on-chronic liver failure. Curr Opin Crit Care. 2019;25(2):187–191. doi: 10.1097/MCC.0000000000000584.
    1. Murray KF, Carithers RL., Jr AASLD practice guidelines: evaluation of the patient for liver transplantation. Hepatology. 2005;41(6):1407–1432. doi: 10.1002/hep.20704.
    1. Finkenstedt A, Nachbaur K, Zoller H, Joannidis M, Pratschke J, Graziadei IW, Vogel W. Acute-on-chronic liver failure: excellent outcomes after liver transplantation but high mortality on the wait list. Liver Transpl. 2013;19(8):879–886. doi: 10.1002/lt.23678.
    1. Michelena J, Altamirano J, Abraldes JG, Affo S, Morales-Ibanez O, Sancho-Bru P, et al. Systemic inflammatory response and serum lipopolysaccharide levels predict multiple organ failure and death in alcoholic hepatitis. Hepatology. 2015;62(3):762–772. doi: 10.1002/hep.27779.
    1. Meng Z, Zhang Y, Wei Z, Liu P, Kang J, Ma D, et al. High serum resistin associates with intrahepatic inflammation and necrosis: an index of disease severity for patients with chronic HBV infection. BMC Gastroenterol. 2017;17(1):6. doi: 10.1186/s12876-016-0558-5.
    1. Bernardi M, Moreau R, Angeli P, Schnabl B, Arroyo V. Mechanisms of decompensation and organ failure in cirrhosis: from peripheral arterial vasodilation to systemic inflammation hypothesis. J Hepatol. 2015;63(5):1272–1284. doi: 10.1016/j.jhep.2015.07.004.
    1. Zhao RH, Shi Y, Zhao H, Wu W, Sheng JF. Acute-on-chronic liver failure in chronic hepatitis B: an update. Expert Rev Gastroenterol Hepatol. 2018;12(4):341–350. doi: 10.1080/17474124.2018.1426459.
    1. Li H, Xia Q, Zeng B, Li ST, Liu H, Li Q, Li J, Yang SY, Dong XJ, Gao T, Munker S, Liu Y, Liebe R, Xue F, Li QG, Chen XS, Liu Q, Zeng H, Wang JY, Xie Q, Meng QH, Wang JF, Mertens PR, Lammert F, Singer MV, Dooley S, Ebert MPA, Qiu DK, Wang TL, Weng HL. Submassive hepatic necrosis distinguishes HBV-associated acute on chronic liver failure from cirrhotic patients with acute decompensation. J Hepatol. 2015;63(1):50–59. doi: 10.1016/j.jhep.2015.01.029.
    1. Claria J, Stauber RE, Coenraad MJ, Moreau R, Jalan R, Pavesi M, et al. Systemic inflammation in decompensated cirrhosis: characterization and role in acute-on-chronic liver failure. Hepatology. 2016;64(4):1249–1264. doi: 10.1002/hep.28740.
    1. Wasmuth HE, Kunz D, Yagmur E, Timmer-Stranghoner A, Vidacek D, Siewert E, et al. Patients with acute on chronic liver failure display "sepsis-like" immune paralysis. J Hepatol. 2005;42(2):195–201. doi: 10.1016/j.jhep.2004.10.019.
    1. He B, Zhang Y, Lu MH, Cao YL, Fan YH, Deng JQ, et al. Glucocorticoids can increase the survival rate of patients with severe viral hepatitis B: a meta-analysis. Eur J Gastroenterol Hepatol. 2013;25(8):926–934. doi: 10.1097/MEG.0b013e32835f4cbd.
    1. Fausto N. Liver regeneration and repair: hepatocytes, progenitor cells, and stem cells. Hepatology. 2004;39(6):1477–1487. doi: 10.1002/hep.20214.
    1. Xiang X, Feng D, Hwang S, Ren T, Wang X, Trojnar E, Matyas C, Mo R, Shang D, He Y, Seo W, Shah VH, Pacher P, Xie Q, Gao B. Interleukin-22 ameliorates acute-on-chronic liver failure by reprogramming impaired regeneration pathways in mice. J Hepatol. 2020;72(4):736–745. doi: 10.1016/j.jhep.2019.11.013.
    1. Arab JP, Sehrawat TS, Simonetto DA, Verma VK, Feng D, Tang T, et al. An open-label, dose-escalation study to assess the safety and efficacy of IL-22 agonist F-652 in patients with alcohol-associated hepatitis. Hepatology. 2019;72(2):441–453. doi: 10.1002/hep.31046.
    1. Garg V, Garg H, Khan A, Trehanpati N, Kumar A, Sharma BC, Sakhuja P, Sarin SK. Granulocyte colony-stimulating factor mobilizes CD34(+) cells and improves survival of patients with acute-on-chronic liver failure. Gastroenterology. 2012;142(3):505–512. doi: 10.1053/j.gastro.2011.11.027.
    1. Spees JL, Lee RH, Gregory CA. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res Ther. 2016;7(1):125. doi: 10.1186/s13287-016-0363-7.
    1. Hu C, Zhao L, Wu Z, Li L. Transplantation of mesenchymal stem cells and their derivatives effectively promotes liver regeneration to attenuate acetaminophen-induced liver injury. Stem Cell Res Ther. 2020;11(1):88. doi: 10.1186/s13287-020-01596-9.
    1. Kim JH, Jo CH, Kim HR, Hwang YI. Comparison of immunological characteristics of mesenchymal stem cells from the periodontal ligament, umbilical cord, and adipose tissue. Stem Cells Int. 2018;2018:8429042. doi: 10.1155/2018/8429042.
    1. Yun JW, Ahn JH, Kwon E, Kim SH, Kim H, Jang JJ, Kim WH, Kim JH, Han SY, Kim JT, Kim JH, Kim W, Ku SY, Do BR, Kang BC. Human umbilical cord-derived mesenchymal stem cells in acute liver injury: hepatoprotective efficacy, subchronic toxicity, tumorigenicity, and biodistribution. Regul Toxicol Pharmacol. 2016;81:437–447. doi: 10.1016/j.yrtph.2016.09.029.
    1. Zhang Y, Li Y, Li W, Cai J, Yue M, Jiang L, et al. Therapeutic effect of human umbilical cord mesenchymal stem cells at various passages on acute liver failure in rats. Stem Cells Int. 2018;2018:7159465.
    1. Chai NL, Zhang XB, Chen SW, Fan KX, Linghu EQ. Umbilical cord-derived mesenchymal stem cells alleviate liver fibrosis in rats. World J Gastroenterol. 2016;22(26):6036–6048. doi: 10.3748/wjg.v22.i26.6036.
    1. Yu SJ, Chen LM, Lyu S, Li YY, Yang B, Geng H, Lin H, Wang SY, Xu RN, Wang LF, Shi M, Wang FS. Safety and efficacy of human umbilical cord derived-mesenchymal stem cell transplantation for treating patients with HBV-related decompensated cirrhosis. Zhonghua Gan Zang Bing Za Zhi. 2016;24(1):51–55. doi: 10.3760/cma.j.issn.1007-3418.2016.01.010.
    1. Li YH, Xu Y, Wu HM, Yang J, Yang LH, Yue-Meng W. Umbilical cord-derived mesenchymal stem cell transplantation in hepatitis B virus related acute-on-chronic liver failure treated with plasma exchange and entecavir: a 24-month prospective study. Stem Cell Rev Rep. 2016;12(6):645–653. doi: 10.1007/s12015-016-9683-3.
    1. Shi M, Zhang Z, Xu R, Lin H, Fu J, Zou Z, Zhang A, Shi J, Chen L, Lv S, He W, Geng H, Jin L, Liu Z, Wang FS. Human mesenchymal stem cell transfusion is safe and improves liver function in acute-on-chronic liver failure patients. Stem Cells Transl Med. 2012;1(10):725–731. doi: 10.5966/sctm.2012-0034.
    1. Xu WX, He HL, Pan SW, Chen YL, Zhang ML, Zhu S, et al. Combination treatments of plasma exchange and umbilical cord-derived mesenchymal stem cell transplantation for patients with hepatitis B virus-related acute-on-chronic liver failure: a clinical trial in China. Stem Cells Int. 2019;2019:4130757.
    1. Gao LR, Zhang NK, Ding QA, Chen HY, Hu X, Jiang S, Li TC, Chen Y, Wang ZG, Ye Y, Zhu ZM. Common expression of stemness molecular markers and early cardiac transcription factors in human Wharton’s jelly-derived mesenchymal stem cells and embryonic stem cells. Cell Transplant. 2013;22(10):1883–1900. doi: 10.3727/096368912X662444.
    1. Li F, Miao L, Sun H, Zhang Y, Bao X, Zhang D. Establishment of a new acute-on-chronic liver failure model. Acta Pharm Sin B. 2017;7(3):326–333. doi: 10.1016/j.apsb.2016.09.003.
    1. Sobani ZA, Paniz GR, Wong M, McCarthy DM. Don't Miss the BoAAT: correctly diagnosing acute-on-chronic liver disease. Dig Dis Sci. 2019;64(10):2780–2783. doi: 10.1007/s10620-019-05816-y.
    1. Guo G, Zhuang X, Xu Q, Wu Z, Zhu Y, Zhou Y, Li Y, Lu Y, Zhang B, Talbot P, Liao J, She J, Bu H, Shi Y. Peripheral infusion of human umbilical cord mesenchymal stem cells rescues acute liver failure lethality in monkeys. Stem Cell Res Ther. 2019;10(1):84. doi: 10.1186/s13287-019-1184-2.
    1. Lee GH. Hepatic encephalopathy in acute-on-chronic liver failure. Hepatol Int. 2015;9(4):520–526. doi: 10.1007/s12072-015-9626-0.
    1. Lin NC, Wu HH, Ho JH, Liu CS, Lee OK. Mesenchymal stem cells prolong survival and prevent lethal complications in a porcine model of fulminant liver failure. Xenotransplantation. 2019;26(6):e12542. doi: 10.1111/xen.12542.
    1. Zhang H, Ming Y, Liu X, Zang C, Chi L, Li D. Gene expression profile changes induced upon umbilical cord mesenchymal cell infusion therapy in a rat model of hepatic cirrhosis. Zhonghua Gan Zang Bing Za Zhi. 2014;22(7):519–524. doi: 10.3760/cma.j.issn.1007-3418.2014.07.009.
    1. Bataller R, Brenner DA. Liver fibrosis. J Clin Invest. 2005;115(2):209–218. doi: 10.1172/JCI24282.
    1. Mederacke I, Hsu CC, Troeger JS, Huebener P, Mu X, Dapito DH, Pradere JP, Schwabe RF. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun. 2013;4(1):2823. doi: 10.1038/ncomms3823.
    1. Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol. 2014;14(3):181–194. doi: 10.1038/nri3623.
    1. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology. 2008;134(6):1655–1669. doi: 10.1053/j.gastro.2008.03.003.
    1. Cai X, Li Z, Zhang Q, Qu Y, Xu M, Wan X, Lu L. CXCL6-EGFR-induced Kupffer cells secrete TGF-beta1 promoting hepatic stellate cell activation via the SMAD2/BRD4/C-MYC/EZH2 pathway in liver fibrosis. J Cell Mol Med. 2018;22(10):5050–5061. doi: 10.1111/jcmm.13787.
    1. Breitkopf K, Godoy P, Ciuclan L, Singer MV, Dooley S. TGF-beta/Smad signaling in the injured liver. Z Gastroenterol. 2006;44(1):57–66. doi: 10.1055/s-2005-858989.
    1. Zhang S, Chen L, Liu T, Zhang B, Xiang D, Wang Z, Wang Y. Human umbilical cord matrix stem cells efficiently rescue acute liver failure through paracrine effects rather than hepatic differentiation. Tissue Eng Part A. 2012;18(13-14):1352–1364. doi: 10.1089/ten.tea.2011.0516.
    1. Xuan J, Feng W, An ZT, Yang J, Xu HB, Li J, Zhao ZF, Wen W. Anti-TGFbeta-1 receptor inhibitor mediates the efficacy of the human umbilical cord mesenchymal stem cells against liver fibrosis through TGFbeta-1/Smad pathway. Mol Cell Biochem. 2017;429(1-2):113–122. doi: 10.1007/s11010-017-2940-1.
    1. Siebel C, Lendahl U. Notch signaling in development, tissue homeostasis, and disease. Physiol Rev. 2017;97(4):1235–1294. doi: 10.1152/physrev.00005.2017.
    1. Adams JM, Jafar-Nejad H. The roles of notch signaling in liver development and disease. Biomolecules. 2019;9(10):608.
    1. Chen Y, Zheng S, Qi D, Guo J, Zhang S, Weng Z. Inhibition of Notch signaling by a gamma-secretase inhibitor attenuates hepatic fibrosis in rats. PLoS One. 2012;7(10):e46512. doi: 10.1371/journal.pone.0046512.
    1. Russell JO, Ko S, Monga SP, Shin D. Notch inhibition promotes differentiation of liver progenitor cells into hepatocytes via sox9b repression in zebrafish. Stem Cells Int. 2019;2019:8451282. doi: 10.1155/2019/8451282.
    1. Tang Y, Li Q, Meng F, Huang X, Li C, Zhou X, et al. Therapeutic potential of HGF-expressing human umbilical cord mesenchymal stem cells in mice with acute liver failure. Int J Hepatol. 2016;2016:5452487. doi: 10.1155/2016/5452487.
    1. Shi D, Zhang J, Zhou Q, Xin J, Jiang J, Jiang L, Wu T, Li J, Ding W, Li J, Sun S, Li J, Zhou N, Zhang L, Jin L, Hao S, Chen P, Cao H, Li M, Li L, Chen X, Li J. Quantitative evaluation of human bone mesenchymal stem cells rescuing fulminant hepatic failure in pigs. Gut. 2017;66(5):955–964. doi: 10.1136/gutjnl-2015-311146.
    1. Trebicka J, Amoros A, Pitarch C, Titos E, Alcaraz-Quiles J, Schierwagen R, Deulofeu C, Fernandez-Gomez J, Piano S, Caraceni P, Oettl K, Sola E, Laleman W, McNaughtan J, Mookerjee RP, Coenraad MJ, Welzel T, Steib C, Garcia R, Gustot T, Rodriguez Gandia MA, Bañares R, Albillos A, Zeuzem S, Vargas V, Saliba F, Nevens F, Alessandria C, de Gottardi A, Zoller H, Ginès P, Sauerbruch T, Gerbes A, Stauber RE, Bernardi M, Angeli P, Pavesi M, Moreau R, Clària J, Jalan R, Arroyo V. Addressing profiles of systemic inflammation across the different clinical phenotypes of acutely decompensated cirrhosis. Front Immunol. 2019;10:476. doi: 10.3389/fimmu.2019.00476.
    1. Schwarzkopf KM, Eberle L, Uschner FE, Klein S, Schierwagen R, Mucke MM, et al. Interleukin-22 in acute-on-chronic liver failure: a matter of ineffective levels, receptor dysregulation or defective signalling? J Hepatol. 2020;73(4):980–982. doi: 10.1016/j.jhep.2020.05.012.
    1. Sharma RR, Pollock K, Hubel A, McKenna D. Mesenchymal stem or stromal cells: a review of clinical applications and manufacturing practices. Transfusion. 2014;54(5):1418–1437. doi: 10.1111/trf.12421.
    1. Eom YW, Shim KY, Baik SK. Mesenchymal stem cell therapy for liver fibrosis. Korean J Intern Med. 2015;30(5):580–589. doi: 10.3904/kjim.2015.30.5.580.

Source: PubMed

3
Abonnere