Letter to the Editor Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Sep 26, 2024; 16(9): 846-853
Published online Sep 26, 2024. doi: 10.4252/wjsc.v16.i9.846
Innovative mesenchymal stem cell treatments for fatty liver disease
Fei-Qiong Gao, Department of Endocrinology, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310003, Zhejiang Province, China
Jia-Qi Zhu, Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang Province, China
Xu-Dong Feng, Department of Clinical Laboratory, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo 315000, Zhejiang Province, China
ORCID number: Fei-Qiong Gao (0000-0002-3572-408X); Jia-Qi Zhu (0000-0001-9472-4074); Xu-Dong Feng (0000-0002-4103-7737).
Co-first authors: Fei-Qiong Gao and Jia-Qi Zhu.
Author contributions: Gao FQ and Zhu JQ made equal contributions to this study. Gao FQ and Zhu JQ prepared and wrote the manuscript; Feng XD wrote and revised the manuscript; and all authors have read and approved the final manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Xu-Dong Feng, MD, PhD, Doctor, Department of Clinical Laboratory, Ningbo Medical Center Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, No. 57 Xingning Road, Ningbo 315000, Zhejiang Province, China. xdfeng@zju.edu.cn
Received: July 22, 2024
Revised: August 26, 2024
Accepted: September 9, 2024
Published online: September 26, 2024
Processing time: 63 Days and 22.1 Hours

Abstract

The incidence of non-alcoholic fatty liver disease (NAFLD) and alcohol-associated liver disease (ALD) is increasing year by year due to changes in the contemporary environment and dietary structure, and is an important public health problem worldwide. There is an urgent need to continuously improve the understanding of their disease mechanisms and develop novel therapeutic strategies. Mesenchymal stem cells (MSCs) have shown promise as a potential therapeutic strategy in therapeutic studies of NAFLD and ALD. NAFLD and ALD have different triggers and their specific mechanisms of disease progression are different, but both involve disease processes such as hepatocellular steatosis and potential fibrosis, cirrhosis, and even hepatocellular carcinoma. MSCs have metabolic regulatory, anti-apoptotic, antioxidant, and immunomodulatory effects that together promote liver injury repair and functional recovery, and have demonstrated positive results in preclinical studies. This editorial is a continuum of Jiang et al’s review focusing on the advantages and limitations of MSCs and their derivatives as therapeutics for NAFLD and ALD. They detail how MSCs attenuate the progression of NAFLD by modulating molecular pathways involved in glucolipid metabolism, inflammation, oxidative stress, endoplasmic reticulum stress, and fibrosis. Based on recent advances, we discuss MSCs and their derivatives as therapeutic strategies for NAFLD and ALD, providing useful information for their clinical application.

Key Words: Alcohol-associated liver disease; Non-alcoholic fatty liver disease; Mesenchymal stem cells; Cell therapy; Inflammation

Core Tip: Mesenchymal stem cells (MSCs) and their derivatives are a promising therapeutic approach for non-alcoholic fatty liver disease and alcohol-associated liver disease. MSCs, which come from diverse sources and are of low immunogenicity, can attenuate disease progression by modulating key molecular pathways, such as glycolipid metabolism, inflammation, oxidative stress, and fibrosis. In addition, derivatives of MSCs are also considered as a therapeutic strategy due to their ability to retain some of the beneficial effects of MSCs while reducing the risks inherent in cell therapy. However, further studies are needed to emphasize their important mechanistic role in liver injury repair. Refining protocols for the clinical application of MSCs under the prerequisite of a well-defined mechanistic understanding may allow utilizing the full benefits of MSCs in the treatment of liver disease to enhance liver reparability and provide new hope for the treatment of non-alcoholic fatty liver disease and alcohol-associated liver disease.



TO THE EDITOR

Liver disease is responsible for 2 million deaths per year, about 4% of all deaths (1 in 25 deaths worldwide), and deaths are mainly attributed to complications of cirrhosis and hepatocellular carcinoma, with cirrhosis most commonly associated with viral hepatitis, and alcoholic and non-alcoholic fatty liver disease (NAFLD)[1]. The epidemiology and burden of NAFLD and alcohol-associated liver disease (ALD) are changing due to the rising prevalence of obesity and increased alcohol consumption. Age-standardized death rates for NAFLD-associated cirrhosis increased between 2012 and 2017, while age-standardized death rates for cirrhosis of other etiologies declined[2]. Although NAFLD and ALD share fatty liver/steatosis, steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma, the two diseases are different in many ways, including risk factors, mechanisms of lipotoxicity, gut microbiome, mitochondrial dysfunction, and other pathogenic pathways[3]. Both NAFLD and ALD, as common chronic liver diseases, may eventually progress to end-stage liver disease, for which liver transplantation remains the only effective treatment but is limited by donor organ shortages, lifelong immunosuppression, and expensive treatment. Currently, cellular therapies have been investigated as a novel alternative treatment for chronic steatosis and inflammatory and fibrotic liver diseases.

Mesenchymal stem cells (MSCs) can be derived from a wide range of biological tissues, including bone marrow, adipose, umbilical cord, and placenta, and have a wide range of proliferative capacity and pluripotent biological properties[4]. They are promising for therapeutic applications due to their easy accessibility and proliferation in culture, genetic stability, low immunogenicity, and therapeutic properties in tissue repair and immunomodulation. MSCs treatment for NAFLD and ALD provides hepatoprotection, modulates inflammatory processes and angiogenesis, and may benefit liver disease by restoring liver function and reducing inflammation and fibrosis[5]. Furthermore, numerous preclinical studies have demonstrated that extracellular vesicles released from MSCs (MSC-EVs) have considerable potential in the treatment of liver diseases, and optimization of MSC-EVs culture conditions or modification of MSC-EVs further facilitates their development and clinical application in the treatment of liver diseases[6]. The minireview by Jiang et al[7] provides valuable insights into the advances in the study of MSCs therapy for the alleviation of NAFLD. Our editorial examines the in-depth mechanisms of MSCs and their derivatives in the treatment of NAFLD and ALD, with the aim of helping to understand and refine the protocols for the clinical application of MSCs and providing new hope for the treatment of NAFLD and ALD.

MSCs-based therapy and NAFLD

Numerous preclinical studies have demonstrated that MSCs from different sources have a role in attenuating obesity, glucose metabolism, hepatic steatosis, inflammation, and fibrosis, which is becoming increasingly prominent in cell therapy for NAFLD[8-10] (Table 1). This may also be realized by their effects on gut microecology[11]. Immunomodulation is an important factor influencing MSC transplantation, and it has been suggested that MSCs may show clinical value in the treatment of NAFLD/non-alcoholic steatohepatitis (NASH) through their ability to inhibit CD4+ T cell activation[12,13]. MSCs can also play a therapeutic role in ameliorating hepatic steatotoxicity and metabolic disturbances in the context of NAFLD by regulating endoplasmic reticulum stress and calcium homeostasis through sarco(endo)plasmic reticulum Ca(2+)-ATPase[14]. Hepatocyte growth factor was identified as a key functional cytokine secreted by menstrual-derived endometrial stem cells, which reduces hepatic Rnf186 expression and regulates glycolipid metabolism through the AMP-activated protein kinase/mechanistic target of rapamycin pathway to alleviate NAFLD[15]. Not only through the action of secreted factors, MSCs can also rescue dysfunctional mitochondria by transferring mitochondria, thus alleviating steatosis, liver function, and glucolipid metabolism disorders in NAFLD[16,17]. Due to the pluripotency of MSCs, their differentiated hepatocyte-like cells may serve as a source of replacement cells for primary hepatocytes, which, together with their anti-inflammatory and regeneration-promoting properties, could contribute to the recovery of NASH-injured liver after transplantation[18]. MSCs with different special pretreatment protocols may play similar beneficial roles in the treatment of different kinds of diseases. For example, antioxidant-up-regulated modified MSCs play an important role in reducing oxidative stress, improving glucose tolerance, reducing systemic inflammation, and ameliorating fatty liver disease[19]. MSC exosomes also showed similar enhancement effects after pretreatment, e.g., curcumin[20] pretreatment and pan-peroxisome proliferator-activated receptor agonist induction[21].

Table 1 Mesenchymal stem cell treatments for non-alcoholic fatty liver disease/non-alcoholic steatohepatitis and alcohol associated liver disease.
Ref.
Cell source
Secretome
Model
Disease
Function and mechanism
Du et al[15]MenSCsHGFMouseNAFLDRnf186 regulated glucose and lipid metabolism through the AMPK/mTOR pathway
HGF decreased the expression of hepatic Rnf186
Wang et al[13]Mouse BM-MSCs-MouseNAFLDSuppressed the activation of CD4+ T cells
Hu et al[9]Human UC-MSCs-MouseNASHAlleviated obesity, glucose metabolism, hepatic steatosis, inflammation, and fibrosis
Regulated lipid metabolism and the PPAR signaling pathway
Yang et al[11]Human UC-MSCs-MouseNASHAlleviated hepatic steatosis, inflammation, and fibrosis
Reversed the microbiome and metabolome disorders
Li et al[14]Rat BM-MSCs-Mouse/HepG2 cellsNAFLDRegulation of ER stress and the calcium homeostasis via SERCA
Bi et al[16]BM-MSCsMitochondriaMouse/hepatocytesNAFLDMitochondrial transfer from BMSCs rescued dysfunction mitochondria
Nickel et al[17]Human BM-MSCsMitochondriaMouseNASHResolution of NASH in mouse livers involved the donation of human mitochondria to the mouse hepatocytes
Domingues et al[19]Antioxidant-upregulated human AD-MSCs-MouseDiet-induced obeseReduced oxidative stress post-antioxidant-upregulated MSC delivery, intraperitoneally, and reduced systemic inflammation and fat accumulation in the liver
Winkler et al[18]Human BM-MSCs-MouseNASHTransplantation of MSC-derived human hepatocyte-like cells corrects NASH in mice by restoring triglyceride depositions, reducing inflammation and augmenting the regenerative capacity of the liver
Cai et al[32]BM-MSCs-MouseChronic alcoholic hepatitisThrough the PI3K/NF-κB and PI3K/mTOR pathways
Modulation of natural killer B cells and follicular helper T cells
Huai et al[37]Human UC-MSCsFGF21MouseALDEnabled macrophages to exhibit anti-inflammatory inclination
Li et al[38]Lysophosphatidic acid receptors and sphingosine-1-phosphate receptors-co-treated human AD-MSCs-MouseALDAmeliorated histological damage, oxidative stress, inflammation, fibrosis, and lipid metabolism dysfunction, and enhanced alcohol metabolizing enzyme activity
Ge et al[39]BM-MSCs/BM-MSCs pre-activated with TLR3-MouseChronic-binge alcoholProtection against alcohol-induced intestinal and hepatic injury and immune dysfunction
Hernandez et al[40]Human UC-MSCs-MouseAlcohol binge drinkingActivated stem cells resulted in marked improvement in survival and in recovery of hepatic chemistries
Chung et al[34]Sk-MSCsHGFMouse/human colonic Caco-2/tc7 cellsAlcoholic liver damageReduced inflammatory responses in the liver and gut
Wan et al[35]BM-MSCsTSG-6MouseAlcoholic hepatitisSecreted TSG-6 to inhibit STAT3 activation and to reduce liver injury
Wan et al[33]BM-MSCs-MouseAlcoholic hepatitisInhibited hepatic neutrophil and macrophage infiltration, and alleviated oxidative stress

Small extracellular vesicles derived from MSCs have shown promise in animal models for the treatment of NAFLD, which has led to consideration of their clinical translation[22,23] (Table 2). Kang et al[24] demonstrated that human umbilical cord MSCs exosomes (hUC-MSC-Exos) attenuated hepatocellular steatosis, reduced inflammatory macrophages as well as tumor necrosis factor-α and interleukin-6 to inhibit hepatic inflammatory response, and suppressed oxidative stress to alleviate NASH through the nuclear factor erythroid 2-related factor 2/NAD(P)H quinone oxidoreductase-1 antioxidant signaling pathway. In addition, hUC-MSC-Exos can prevent NAFLD by modulating lipid homeostasis through transferring calcium/calmodulin-dependent protein kinase 1 to improve lipid accumulation, inhibit fatty acid synthesis, and enhance fatty acid oxidation[25]. In addition, MSC-Exos are rich in nucleic acids that play important roles in inhibiting lipid accumulation, reactive oxygen species generation, inflammation, and liver fibrosis, which provide potential therapeutic value for NAFLD treatment, such as miR-24-3p[26], miR-223-3p[27], miR-627-5p[28], and miR-96-5p[29]. MSCs not only promote mitochondrial homeostasis in hepatocytes through mitochondrial transfer. Furthermore, it was shown that RNF31 transport via MSCs-derived small extracellular vesicles has a substantial impact on the regulation of hepatocyte mitochondrial homeostasis, hepatic steatosis, and recovery of liver function[30]. MSCs release a large number of molecules in conditioned medium, which may have similar beneficial effects in ameliorating NAFLD without pitfalls such as cellular obstruction, and have the potential for clinical application[31]. Clinical trials regarding MSCs therapy for NAFLD are also being conducted progressively and are expected to provide new therapeutic options for clinical application in patients[8].

Table 2 Mesenchymal stem cell derivatives for treatment of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis.
Ref.
Cell source
Secretome
Model
Disease
Function and mechanism
Kang et al[24]Human UC-MSC-EV-Mouse/HepG2 and AML12 cellsNASHNrf2/NQO-1 antioxidant signaling pathway
Yang et al[25]Human UC-MSC-EVCAMKK1Mouse/hepatic cellsNAFLDCAMKK1-mediated lipid homoeostasis regulation
Chen et al[30]MSC-sEVRNF31MouseNAFLDRegulation of mitochondrial homeostasis
Du et al[26]Human UC-MSC-ExoMiRNA-24-3pMouse/hepatocytesNAFLDMiR-24-3p directly targeted Kelch-like ECH-associated protein 1, and suppressed its expression
Restrained lipid accumulation, ROS generation, and inflammation
Niu et al[27]AD-MSC-EVMiRNA-223-3pMouse/hepatocytesNAFLDMiR-223-3p displayed suppressive effects on lipid accumulation and liver fibrosis through E2F1 inhibition
Cheng et al[28]Human UC-MSC-ExoMiRNA-627-5pRat/L-02 cellsNAFLDMiR-627-5p improved glucose and lipid metabolism and alleviated liver damage by repressing FTO expression
El-Derany et al[29]BM-MSC-ExoMiRNA-96-5pMouseNASHCaspase-2 signaling inhibition
Tawfeek and Kasem[20]Curcumin-preconditioned MSC-Exo-Mouse/HepG2 cellsNASHRegulated inflammatory, oxidative stress, and mitochondrial-dependent apoptosis-associated ASK-JNK-BAX genes
Kim et al[21]Pan-peroxisome proliferator-activated receptor agonist-primed induced MSC-EV-MouseNASHReduced steatotic changes and ameliorated ER stress and mitochondiral oxidative stress induced by inflammation
Zhang et al[23]MSC-sEV-MouseNASHPolarized pro-fibrotic M2 macrophages without exacerbating liver fibrosis
Yang et al[31]MSCs conditional medium-Mouse/L-02 cellsNAFLDImproved mitochondrial function and alleviated inflammation and apoptosis by regulating SIRT1
MSCs-based therapy and ALD

Despite numerous studies dedicated to demonstrating the therapeutic potential of MSCs in the management of acute and chronic liver diseases, the field of utilizing MSCs in the treatment of ALD has not yet been sufficiently reported in the scientific literature (Table 1). Cai et al[32] previously reported that the combination of bone marrow-derived MSCs (BM-MSCs) and Lactobacillus plumosus culture supernatant may ameliorate the symptoms of alcoholic hepatitis through modulation of the phosphoinositide 3-kinase/nuclear factor kappa B and phosphoinositide 3-kinase/mechanistic target of rapamycin pathways, as well as modulation of natural killer B cells and follicular helper T cells. MSCs have been used to treat many inflammatory diseases, and studies have shown that MSCs are effective in treating alcoholic hepatitis through their ability to inhibit hepatic neutrophil and macrophage infiltration and attenuate oxidative stress[33]. Skeletal muscle satellite cell-derived MSCs exert anti-inflammatory effects through the secretion of prostaglandin E2 and hepatocyte growth factor, which may attenuate alcoholic liver injury by decreasing the inflammatory response in the liver and intestine[34]. Tumor necrosis factor-stimulated gene-6 secreted by BM-MSCs is able to inhibit signal transducer and activator of transcription 3 activation, thereby reducing liver injury and treating alcoholic hepatitis[35]. Application of tumor necrosis factor-stimulated gene-6 alone attenuates alcohol-induced liver injury and fibrosis by blocking the cleavage of CD44 to form the CD44 intracellular domain[36]. Pretreated MSCs likewise had enhanced therapeutic effects on ALD, and fibroblast growth factor 21-overexpressing MSCs promoted the immunomodulatory function of MSCs on macrophages through metabolic regulation of oxidative phosphorylation, causing macrophages to exhibit anti-inflammatory tendencies and thus alleviating ALD[37]. Lysophosphatidic acid receptor and sphingosine-1-phosphate receptor-co-treated human adipose-derived MSCs exhibit significant therapeutic efficacy that could enhance their efficacy in the future treatment of ALD[38]. Toll-like receptor 3-preactivated BM-MSCs can be used to protect against alcohol-induced intestinal and hepatic injury[39]. Activated hUC-MSCs significantly improved hepatocyte survival and recovery of hepatocyte chemical function compared with non-activated stem cells, which has important clinical applications[40].

BM-MSCs transplantation has been recognized as an effective treatment for liver cirrhosis, and there are clinical trials investigating the efficacy and safety of autologous BM-MSCs transplantation for alcoholic cirrhosis[41,42]. Chronic high intake of ethanol can lead to a variety of metabolic changes in the body and increased levels of ethanol and its metabolites in the body’s microenvironment. It has been shown that ethanol inhibits BM-MSCs-mediated hepatocyte renewal in rats, which may affect the clinical application of MSCs for the treatment of ALD[43], and more research should be invested in exploring the in-depth mechanisms and solutions.

CONCLUSION

NAFLD and ALD are common chronic diseases with a poor prognosis. Finding treatments for these diseases can improve the prognosis and quality of life of patients. Stem cell transplantation is an emerging therapy for the treatment of acute and chronic liver diseases. Both MSCs and their derivatives are attractive therapeutic tools that have been widely explored in preclinical and clinical studies. The incidence of NAFLD and ALD is increasing every year and constantly threatening human health. MSCs secrete molecules and EVs that are considered to have potential as therapeutic agents for NFALD and ALD. However, data from preclinical studies of MSC-derived products for the treatment of animal models of ALD are limited. In addition, a large amount of research data is needed to support the current development of clinical applications of MSCs for NAFLD and ALD, including the optimal therapeutic dose, standardized pretreatment modalities, and therapeutic infusion routes. According to the review by Jiang et al[7], MSCs are a promising therapeutic candidate for NAFLD. However, further studies are needed to investigate the relationship between the pathological events that occur during the development of NAFLD and ALD and the cellular and molecular mechanisms associated with the therapeutic effects of MSCs. In conclusion, MSCs are clearly a promising and attractive resource for the development of liver disease therapies, and further comprehensive studies to develop safer and more effective strategies for the treatment of NAFLD and ALD would be beneficial to realize their therapeutic potential.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Ntikoudi A S-Editor: Wang JJ L-Editor: Wang TQ P-Editor: Zhang XD

References
1.  Devarbhavi H, Asrani SK, Arab JP, Nartey YA, Pose E, Kamath PS. Global burden of liver disease: 2023 update. J Hepatol. 2023;79:516-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 300]  [Reference Citation Analysis (1)]
2.  Huang DQ, Terrault NA, Tacke F, Gluud LL, Arrese M, Bugianesi E, Loomba R. Global epidemiology of cirrhosis - aetiology, trends and predictions. Nat Rev Gastroenterol Hepatol. 2023;20:388-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 145]  [Article Influence: 145.0]  [Reference Citation Analysis (0)]
3.  Singal AK. Similarities and differences between non-alcoholic fatty liver disease (NAFLD) & alcohol-associated liver disease (ALD). Transl Gastroenterol Hepatol. 2021;6:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
4.  Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci. 2019;76:3323-3348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 295]  [Article Influence: 59.0]  [Reference Citation Analysis (0)]
5.  Korkida F, Stamatopoulou A, Roubelakis MG. Recent Advances in Mesenchymal Stem/Stromal Cell-Based Therapy for Alcohol-Associated Liver Disease and Non-alcoholic Fatty Liver Disease. Stem Cells Transl Med. 2024;13:107-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
6.  Zheng L, Gong H, Zhang J, Guo L, Zhai Z, Xia S, Hu Z, Chang J, Jiang Y, Huang X, Ge J, Zhang B, Yan M. Strategies to improve the therapeutic efficacy of mesenchymal stem cell-derived extracellular vesicle (MSC-EV): a promising cell-free therapy for liver disease. Front Bioeng Biotechnol. 2023;11:1322514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
7.  Jiang Y, Yusoff NM, Du J, Moses EJ, Lin JT. Current perspectives on mesenchymal stem cells as a potential therapeutic strategy for non-alcoholic fatty liver disease. World J Stem Cells. 2024;16:760-772.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (5)]
8.  Sakai Y, Fukunishi S, Takamura M, Kawaguchi K, Inoue O, Usui S, Takashima S, Seki A, Asai A, Tsuchimoto Y, Nasti A, Bich Ho TT, Imai Y, Yoshimura K, Murayama T, Yamashita T, Arai K, Yamashita T, Mizukoshi E, Honda M, Wada T, Harada K, Higuchi K, Kaneko S. Clinical trial of autologous adipose tissue-derived regenerative (stem) cells therapy for exploration of its safety and efficacy. Regen Ther. 2021;18:97-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
9.  Hu J, Li S, Zhong X, Wei Y, Sun Q, Zhong L. Human umbilical cord mesenchymal stem cells attenuate diet-induced obesity and NASH-related fibrosis in mice. Heliyon. 2024;10:e25460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
10.  Watanabe T, Tsuchiya A, Takeuchi S, Nojiri S, Yoshida T, Ogawa M, Itoh M, Takamura M, Suganami T, Ogawa Y, Terai S. Development of a non-alcoholic steatohepatitis model with rapid accumulation of fibrosis, and its treatment using mesenchymal stem cells and their small extracellular vesicles. Regen Ther. 2020;14:252-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 49]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
11.  Yang Z, Xia Q, Lu D, Yue H, Zhang J, Li Y, Zhang B, Li X, Cao M. Human mesenchymal stem cells treatment improved hepatic lesions and reversed gut microbiome disorder in non-alcoholic steatohepatitis. Aging (Albany NY). 2020;12:21660-21673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
12.  Wang H, Zhang H, Huang B, Miao G, Yan X, Gao G, Luo Y, Chen H, Chen W, Yang L. Mesenchymal stem cells reverse highfat dietinduced nonalcoholic fatty liver disease through suppression of CD4+ T lymphocytes in mice. Mol Med Rep. 2018;17:3769-3774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
13.  Wang H, Wang D, Yang L, Wang Y, Jia J, Na D, Chen H, Luo Y, Liu C. Compact bone-derived mesenchymal stem cells attenuate nonalcoholic steatohepatitis in a mouse model by modulation of CD4 cells differentiation. Int Immunopharmacol. 2017;42:67-73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
14.  Li L, Zeng X, Liu Z, Chen X, Li L, Luo R, Liu X, Zhang J, Liu J, Lu Y, Cheng J, Chen Y. Mesenchymal stromal cells protect hepatocytes from lipotoxicity through alleviation of endoplasmic reticulum stress by restoring SERCA activity. J Cell Mol Med. 2021;25:2976-2993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
15.  Du J, Jiang Y, Liu X, Ji X, Xu B, Zhang Y, Liu Y, Zhang T, Lin J. HGF Secreted by Menstrual Blood-Derived Endometrial Stem Cells Ameliorates Non-Alcoholic Fatty Liver Disease Through Downregulation of Hepatic Rnf186. Stem Cells. 2023;41:153-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
16.  Bi Y, Guo X, Zhang M, Zhu K, Shi C, Fan B, Wu Y, Yang Z, Ji G. Bone marrow derived-mesenchymal stem cell improves diabetes-associated fatty liver via mitochondria transformation in mice. Stem Cell Res Ther. 2021;12:602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
17.  Nickel S, Christ M, Schmidt S, Kosacka J, Kühne H, Roderfeld M, Longerich T, Tietze L, Bosse I, Hsu MJ, Stock P, Roeb E, Christ B. Human Mesenchymal Stromal Cells Resolve Lipid Load in High Fat Diet-Induced Non-Alcoholic Steatohepatitis in Mice by Mitochondria Donation. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
18.  Winkler S, Borkham-Kamphorst E, Stock P, Brückner S, Dollinger M, Weiskirchen R, Christ B. Human mesenchymal stem cells towards non-alcoholic steatohepatitis in an immunodeficient mouse model. Exp Cell Res. 2014;326:230-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
19.  Domingues CC, Kundu N, Kropotova Y, Ahmadi N, Sen S. Antioxidant-upregulated mesenchymal stem cells reduce inflammation and improve fatty liver disease in diet-induced obesity. Stem Cell Res Ther. 2019;10:280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 26]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
20.  Tawfeek GA, Kasem HA. Curcumin preconditioned mesenchymal stem cells derived exosomes transplantation ameliorate and protect against non- alcoholic steatohepatitis by regulation the expression of key genes of inflammation and oxidative stress. Transpl Immunol. 2023;78:101837.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
21.  Kim J, Lee SK, Jeong SY, Cho HJ, Park J, Kim TM, Kim S. Cargo proteins in extracellular vesicles: potential for novel therapeutics in non-alcoholic steatohepatitis. J Nanobiotechnology. 2021;19:372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
22.  Shi Y, Yang X, Wang S, Wu Y, Zheng L, Tang Y, Gao Y, Niu J. Human umbilical cord mesenchymal stromal cell-derived exosomes protect against MCD-induced NASH in a mouse model. Stem Cell Res Ther. 2022;13:517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 21]  [Reference Citation Analysis (0)]
23.  Zhang B, Zhang B, Lai RC, Sim WK, Lam KP, Lim SK. MSC-sEV Treatment Polarizes Pro-Fibrotic M2 Macrophages without Exacerbating Liver Fibrosis in NASH. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
24.  Kang Y, Song Y, Luo Y, Song J, Li C, Yang S, Guo J, Yu J, Zhang X. Exosomes derived from human umbilical cord mesenchymal stem cells ameliorate experimental non-alcoholic steatohepatitis via Nrf2/NQO-1 pathway. Free Radic Biol Med. 2022;192:25-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 40]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
25.  Yang F, Wu Y, Chen Y, Xi J, Chu Y, Jin J, Yan Y. Human umbilical cord mesenchymal stem cell-derived exosomes ameliorate liver steatosis by promoting fatty acid oxidation and reducing fatty acid synthesis. JHEP Rep. 2023;5:100746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
26.  Du X, Li H, Han X, Ma W. Mesenchymal stem cells-derived exosomal miR-24-3p ameliorates non-alcohol fatty liver disease by targeting Keap-1. Biochem Biophys Res Commun. 2022;637:331-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Niu Q, Wang T, Wang Z, Wang F, Huang D, Sun H, Liu H. Adipose-derived mesenchymal stem cell-secreted extracellular vesicles alleviate non-alcoholic fatty liver disease via delivering miR-223-3p. Adipocyte. 2022;11:572-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
28.  Cheng L, Yu P, Li F, Jiang X, Jiao X, Shen Y, Lai X. Human umbilical cord-derived mesenchymal stem cell-exosomal miR-627-5p ameliorates non-alcoholic fatty liver disease by repressing FTO expression. Hum Cell. 2021;34:1697-1708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
29.  El-Derany MO, AbdelHamid SG. Upregulation of miR-96-5p by bone marrow mesenchymal stem cells and their exosomes alleviate non-alcoholic steatohepatitis: Emphasis on caspase-2 signaling inhibition. Biochem Pharmacol. 2021;190:114624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
30.  Chen Y, Yang F, Shi Y, Sheng J, Wang Y, Zhang L, Zhou J, Jin Y, Yan Y. RNF31 alleviates liver steatosis by promoting p53/BNIP3-related mitophagy in hepatocytes. Free Radic Biol Med. 2024;219:163-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
31.  Yang M, Cui Y, Song J, Cui C, Wang L, Liang K, Wang C, Sha S, He Q, Hu H, Guo X, Zang N, Sun L, Chen L. Mesenchymal stem cell-conditioned medium improved mitochondrial function and alleviated inflammation and apoptosis in non-alcoholic fatty liver disease by regulating SIRT1. Biochem Biophys Res Commun. 2021;546:74-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
32.  Cai C, Chen DZ, Ge LC, Chen WK, Ye SS, Ye WW, Tao Y, Wang R, Li J, Lin Z, Wang XD, Xu LM, Chen YP. Synergistic effects of Lactobacillus rhamnosus culture supernatant and bone marrow mesenchymal stem cells on the development of alcoholic steatohepatitis in mice. Am J Transl Res. 2019;11:5703-5715.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Wan YM, Li ZQ, Liu C, He YF, Wang MJ, Wu XN, Zhang Y, Li YH. Mesenchymal stem cells reduce alcoholic hepatitis in mice via suppression of hepatic neutrophil and macrophage infiltration, and of oxidative stress. PLoS One. 2020;15:e0228889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
34.  Chung JS, Hwang S, Hong JE, Jo M, Rhee KJ, Kim S, Jung PY, Yoon Y, Kang SH, Ryu H, Kim MY, Bae KS, Eom YW. Skeletal muscle satellite cell-derived mesenchymal stem cells ameliorate acute alcohol-induced liver injury. Int J Med Sci. 2022;19:353-363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
35.  Wan YM, Li ZQ, Zhou Q, Liu C, Wang MJ, Wu HX, Mu YZ, He YF, Zhang Y, Wu XN, Li YH, Xu ZY, Wu HM, Xu Y, Yang JH, Wang XF. Mesenchymal stem cells alleviate liver injury induced by chronic-binge ethanol feeding in mice via release of TSG6 and suppression of STAT3 activation. Stem Cell Res Ther. 2020;11:24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
36.  Han J, Lee C, Jeong H, Jeon S, Lee M, Lee H, Choi YH, Jung Y. Tumor necrosis factor-inducible gene 6 protein and its derived peptide ameliorate liver fibrosis by repressing CD44 activation in mice with alcohol-related liver disease. J Biomed Sci. 2024;31:54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
37.  Huai Q, Zhu C, Zhang X, Dai H, Li X, Wang H. Mesenchymal stem/stromal cells armored by FGF21 ameliorate alcohol-induced liver injury through modulating polarization of macrophages. Hepatol Commun. 2024;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
38.  Li M, Lv Y, Chen F, Wang X, Zhu J, Li H, Xiao J. Co-stimulation of LPAR(1) and S1PR(1/3) increases the transplantation efficacy of human mesenchymal stem cells in drug-induced and alcoholic liver diseases. Stem Cell Res Ther. 2018;9:161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
39.  Ge L, Chen D, Chen W, Cai C, Tao Y, Ye S, Lin Z, Wang X, Li J, Xu L, Chen Y. Pre-activation of TLR3 enhances the therapeutic effect of BMMSCs through regulation the intestinal HIF-2α signaling pathway and balance of NKB cells in experimental alcoholic liver injury. Int Immunopharmacol. 2019;70:477-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
40.  Hernandez JC, Yeh DW, Marh J, Choi HY, Kim J, Chopra S, Ding L, Thornton M, Grubbs B, Makowka L, Sher L, Machida K. Activated and nonactivated MSCs increase survival in humanized mice after acute liver injury through alcohol binging. Hepatol Commun. 2022;6:1549-1560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
41.  Suk KT, Yoon JH, Kim MY, Kim CW, Kim JK, Park H, Hwang SG, Kim DJ, Lee BS, Lee SH, Kim HS, Jang JY, Lee CH, Kim BS, Jang YO, Cho MY, Jung ES, Kim YM, Bae SH, Baik SK. Transplantation with autologous bone marrow-derived mesenchymal stem cells for alcoholic cirrhosis: Phase 2 trial. Hepatology. 2016;64:2185-2197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 192]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
42.  Jang YO, Kim YJ, Baik SK, Kim MY, Eom YW, Cho MY, Park HJ, Park SY, Kim BR, Kim JW, Soo Kim H, Kwon SO, Choi EH, Kim YM. Histological improvement following administration of autologous bone marrow-derived mesenchymal stem cells for alcoholic cirrhosis: a pilot study. Liver Int. 2014;34:33-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 126]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
43.  Liu YL, Liu WT, Han MZ, Wang M, Sun CX. [Ethanol-inhibited rat bone marrow mesenchymal stem cell differentiated hepatocytes play a role in liver regeneration]. Zhonghua Gan Zang Bing Za Zhi. 2012;20:55-57.  [PubMed]  [DOI]  [Cited in This Article: ]