1
|
Zhang L, Deng Y, Bai X, Wei X, Ren Y, Chen S, Deng H. Cell therapy for end-stage liver disease: Current state and clinical challenge. Chin Med J (Engl) 2024; 137:2808-2820. [PMID: 39602326 DOI: 10.1097/cm9.0000000000003332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Indexed: 11/29/2024] Open
Abstract
ABSTRACT Liver disease involves a complex interplay of pathological processes, including inflammation, hepatocyte necrosis, and fibrosis. End-stage liver disease (ESLD), such as liver failure and decompensated cirrhosis, has a high mortality rate, and liver transplantation is the only effective treatment. However, to overcome problems such as the shortage of donor livers and complications related to immunosuppression, there is an urgent need for new treatment strategies that need to be developed for patients with ESLD. For instance, hepatocytes derived from donor livers or stem cells can be engrafted and multiplied in the liver, substituting the host hepatocytes and rebuilding the liver parenchyma. Stem cell therapy, especially mesenchymal stem cell therapy, has been widely proved to restore liver function and alleviate liver injury in patients with severe liver disease, which has contributed to the clinical application of cell therapy. In this review, we discussed the types of cells used to treat ESLD and their therapeutic mechanisms. We also summarized the progress of clinical trials around the world and provided a perspective on cell therapy.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | |
Collapse
|
2
|
Zhang Y, He X, Ge Z, Wang B, Ni M, Cai G. Investigating the differential therapeutic efficacy and mechanisms of human umbilical cord mesenchymal stem cells at various passages in osteoarthritis treatment. Tissue Cell 2024; 90:102499. [PMID: 39126832 DOI: 10.1016/j.tice.2024.102499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024]
Abstract
This study aimed to assess the clinical efficacy of umbilical cord mesenchymal stem cells (hUC-MSCs) from different passages (P3, P8, and P13) in the treatment of knee osteoarthritis (OA) and explore the underlying mechanisms. The hUC-MSCs from each passage were characterized and evaluated for their stemness, migration, proliferation, and marker expression. Rats with OA were treated with hUC-MSCs from each passage, and the therapeutic effects were assessed based on knee swelling, discomfort, and pathological examination of the knee joint. Co-culture experiments were conducted to examine the ability of hUC-MSCs to stimulate type II collagen synthesis and inhibit MMP13 expression in chondrocytes. Telomere length and telomerase activity of hUC-MSCs from each passage were measured to investigate the reasons for the observed differences in clinical efficacy. The results revealed that P3 and P8 hUC-MSCs exhibited superior osteogenic and chondrogenic differentiation potential compared to P13, while P13 demonstrated stronger adipogenic differentiation. The wound healing rate was significantly higher in the P3 and P8 groups compared to P13. All hUC-MSC groups expressed high levels of CD90 and CD105, indicating their mesenchymal stem cell characteristics, while CD31 and CD45 were not expressed. CD105 expression was significantly reduced in the P13 group. In the treatment of rat osteoarthritis, there were no significant differences in knee swelling, discomfort, Mankin scores, and pathological findings between P3 and P8 hUC-MSC treatments. However, there was a significant difference between the 8th and 13th passages. Co-culture experiments showed that hUC-MSCs from P3 and P8 enhanced type II collagen synthesis and reduced MMP13 expression in chondrocytes. Although no significant difference was observed between the P3 and P8 groups, a significant difference was found between the P13 and P8 groups. Telomere length analysis revealed that P13 samples had significantly shorter telomeres compared to both P3 and P8. The telomerase activity was positive in P3 and P8 hUC-MSCs, indicating no significant difference between these passages, while it was negative in P13 hUC-MSCs. In conclusion, P3 and P8 hUC-MSCs exhibited superior therapeutic potential for knee osteoarthritis compared to P13, possibly due to their enhanced differentiation capacity and telomerase activity.
Collapse
Affiliation(s)
- Yingkai Zhang
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China; Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University, Shanghai City 200032, PR China
| | - Xianwei He
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Zhe Ge
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Bingnan Wang
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Miaozhong Ni
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Guoping Cai
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China.
| |
Collapse
|
3
|
Choi YJ, Kim JH, Lee Y, Pyeon HJ, Yoo IK, Yoo JH. Anti-fibrogenic effect of umbilical cord-derived mesenchymal stem cell-conditioned media in human esophageal fibroblasts. Sci Rep 2024; 14:22233. [PMID: 39333200 PMCID: PMC11437107 DOI: 10.1038/s41598-024-73091-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024] Open
Abstract
Esophageal fibrosis can develop due to caustic or radiation injuries. Umbilical cord-derived mesenchymal stem cells (UC-MSCs) are known to mitigate fibrosis in various organs. However, the potential effects of UC-MSCs on human esophageal fibrosis remain underexplored. This study investigated the anti-fibrogenic properties and mechanisms of UC-MSC-derived conditioned media (UC-MSC-CM) on human esophageal fibroblasts (HEFs). HEFs were treated with TGF-β1 and then cultured with UC-MSC-CM, and the expression levels of extracellular matrix (ECM) components, RhoA, myocardin related transcription factor A (MRTF-A), serum response factor (SRF), Yes-associated protein (YAP), and transcriptional coactivator with PDZ-binding motif (TAZ) were measured. UC-MSC-CM suppressed TGF-β1-induced fibrogenic activation in HEFs, as evidenced by the downregulation of ECM. UC-MSC-CM diminished the expression of RhoA, MRTF-A, and SRF triggered by TGF-β1. In TGF-β1-stimulated HEFs, UC-MSC-CM decreased the nuclear localization of MRTF-A and YAP. Additionally, UC-MSC-CM diminished the TGF-β1-induced nuclear expressions of YAP and TAZ, while concurrently enhancing the cytoplasmic presence of phosphorylated YAP. Furthermore, UC-MSC-CM reduced TGF-β1-induced phosphorylation of Smad2. These findings suggest that UC-MSC-CM may inhibit TGF-β1-induced fibrogenic activation in HEFs by targeting the Rho-mediated MRTF/SRF and YAP/TAZ pathways, as well as the Smad2 pathway. This indicates its potential as a stem cell therapy for esophageal fibrosis.
Collapse
Affiliation(s)
- Yoon Jeong Choi
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea
- Institute of Basic Medical Sciences, CHA University School of Medicine, Seongnam, 13496, South Korea
| | - Jee Hyun Kim
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea
| | - Yeonju Lee
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea
| | - Hee Jang Pyeon
- R&D Division, CHA Biotech Co., Ltd, Seongnam, 13488, South Korea
| | - In Kyung Yoo
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea.
| | - Jun Hwan Yoo
- Department of Gastroenterology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, South Korea.
- Institute of Basic Medical Sciences, CHA University School of Medicine, Seongnam, 13496, South Korea.
| |
Collapse
|
4
|
Zeng Y, Wu Z, Chen G, Liu G, Zhang B, Zhou Y, Chen M, Yao R, Shi Y. Peripheral Injection of hUC-MSCs in the Treatment of Acute Liver Failure: A Pre-Clinical Cohort Study in Rhesus Monkeys. Stem Cells Int 2024; 2024:4654912. [PMID: 39045027 PMCID: PMC11265939 DOI: 10.1155/2024/4654912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/25/2024] Open
Abstract
Background Using a toxin-induced lethal acute liver failure (ALF) monkey model, we have recently shown that early peripheral infusion of human umbilical cord mesenchymal stem cells (hUC-MSCs) can alleviate liver damage and improve animal survival by suppressing the activation of circulating monocytes and the subsequent cytokine storm. Here, we explored whether the administration of hUC-MSCs could still improve ALF when the cytokine storm is fully developed. Method We treated ALF monkeys with peripheral delivery of hUC-MSCs at 48 hr after toxin challenge. Liver indices, histology, imaging, and animal survival were recorded and analyzed. Results In our cohort study, we conducted and demonstrated that the infusion of hUC-MSCs significantly improved liver histology, effectively controlled inflammatory cytokine storms, and increased survival rates. Additionally, the administration of a higher dose of hUC-MSCs (2 × 107/monkey) yielded superior outcomes compared to a lower dose (1 × 107/monkey). Conclusion Treatment of hUC-MSCs can significantly improve the pathological and survival outcomes of ALF even when the cytokine storm has been fully developed, indicating a promising clinical solution for ALF.
Collapse
Affiliation(s)
- Yuting Zeng
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| | - Zhenru Wu
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| | - Gen Chen
- Development and Application of Human Major Disease Monkey Model Key Laboratory of SichuanSichuan Yibin Horizontal and Vertical Biotechnology Co., Ltd., Yibin 644601, China
| | - Guoqiang Liu
- Development and Application of Human Major Disease Monkey Model Key Laboratory of SichuanSichuan Yibin Horizontal and Vertical Biotechnology Co., Ltd., Yibin 644601, China
| | - Bo Zhang
- Sichuan Stem Cell Bank and Sichuan Neo-Life Stem Cell Biotech Inc., Chengdu 610037, China
| | - Yongjie Zhou
- Laboratory of Liver TransplantationWest China HospitalSichuan University, Chengdu 610041, China
| | - Menglin Chen
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| | - Rong Yao
- Department of Emergency MedicineEmergency Medical LaboratoryWest China HospitalSichuan University, Chengdu, Sichuan, China
| | - Yujun Shi
- Liver Transplant CenterTransplant Center and Key Laboratory of Transplant Engineering and ImmunologyNHCWest China HospitalSichuan University, Chengdu, China
- Institute of clinical PathologyWest China HospitalSichuan University, Chengdu, China
| |
Collapse
|
5
|
Jin X, Zhang J, Zhang Y, He J, Wang M, Hei Y, Guo S, Xu X, Liu Y. Different origin-derived exosomes and their clinical advantages in cancer therapy. Front Immunol 2024; 15:1401852. [PMID: 38994350 PMCID: PMC11236555 DOI: 10.3389/fimmu.2024.1401852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Exosomes, as a class of small extracellular vesicles closely related to the biological behavior of various types of tumors, are currently attracting research attention in cancer diagnosis and treatment. Regarding cancer diagnosis, the stability of their membrane structure and their wide distribution in body fluids render exosomes promising biomarkers. It is expected that exosome-based liquid biopsy will become an important tool for tumor diagnosis in the future. For cancer treatment, exosomes, as the "golden communicators" between cells, can be designed to deliver different drugs, aiming to achieve low-toxicity and low-immunogenicity targeted delivery. Signaling pathways related to exosome contents can also be used for safer and more effective immunotherapy against tumors. Exosomes are derived from a wide range of sources, and exhibit different biological characteristics as well as clinical application advantages in different cancer therapies. In this review, we analyzed the main sources of exosomes that have great potential and broad prospects in cancer diagnosis and therapy. Moreover, we compared their therapeutic advantages, providing new ideas for the clinical application of exosomes.
Collapse
Affiliation(s)
- Xiaoyan Jin
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jing Zhang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
- The Second Affiliated Hospital of Xi‘an Medical University, Xi’an, Shaanxi, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Jing He
- Laboratory of Obstetrics and Gynecology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Mingming Wang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yu Hei
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Shutong Guo
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Xiangrong Xu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yusi Liu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
6
|
Rofaani E, Mardani MW, Yutiana PN, Amanda O, Darmawan N. Differentiation of mesenchymal stem cells into vascular endothelial cells in 3D culture: a mini review. Mol Biol Rep 2024; 51:781. [PMID: 38913199 DOI: 10.1007/s11033-024-09743-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Mesenchymal Stem Cells, mesodermal origin and multipotent stem cells, have ability to differentiate into vascular endothelial cells. The cells are squamous in morphology, inlining, and protecting blood vessel tissue, as well as maintaining homeostatic conditions. ECs are essential in vascularization and blood vessels formation. The differentiation process, generally carried out in 2D culture systems, were relied on growth factors induction. Therefore, an artificial extracellular matrix with relevant mechanical properties is essential to build 3D culture models. Various 3D fabrication techniques, such as hydrogel-based and fibrous scaffolds, scaffold-free, and co-culture to endothelial cells were reviewed and summarized to gain insights. The obtained MSCs-derived ECs are shown by the expression of endothelial gene markers and tubule-like structure. In order to mimicking relevant vascular tissue, 3D-bioprinting facilitates to form more complex microstructures. In addition, a microfluidic chip with adequate flow rate allows medium perfusion, providing mechanical cues like shear stress to the artificial vascular vessels.
Collapse
Affiliation(s)
- E Rofaani
- Group Research of Theranostics, Research Center for Vaccine and Drug, Research Organization of Health, National Research and Innovation Agency, LAPTIAB Building No 611 PUSPIPTEK or KST BJ Habibie, Tangerang Selatan, Banten, 15315, Indonesia.
| | - M W Mardani
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sebelas Maret University, Ir. Sutami Street No. 36A, Jebres District, Surakarta, Central Java, 57126, Indonesia
| | - P N Yutiana
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sebelas Maret University, Ir. Sutami Street No. 36A, Jebres District, Surakarta, Central Java, 57126, Indonesia
| | - O Amanda
- Department of Technique of Biomedis, Faculty of Technique of Industry, Institut Teknologi Sumatera, Jalan Terusan Ryacudu, Way Huwi, Jati Agung, Lampung Selatan, Lampung, 35365, Indonesia
| | - N Darmawan
- Laboratory of Inorganic Chemistry, Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Kampus IPB Dramaga, Bogor, West Java, 16880, Indonesia
| |
Collapse
|
7
|
Kao YH, Chang CY, Lin YC, Chen PH, Lee PH, Chang HR, Chang WY, Chang YC, Wun SF, Sun CK. Mesenchymal Stem Cell-Derived Exosomes Mitigate Acute Murine Liver Injury via Ets-1 and Heme Oxygenase-1 Up-regulation. Curr Stem Cell Res Ther 2024; 19:906-918. [PMID: 37723631 DOI: 10.2174/1574888x19666230918102826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/21/2023] [Accepted: 08/11/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs)-derived exosomes have been previously demonstrated to promote tissue regeneration in various animal disease models. This study investigated the protective effect of exosome treatment in carbon tetrachloride (CCl4)-induced acute liver injury and delineated possible underlying mechanism. METHODS Exosomes collected from conditioned media of previously characterized human umbilical cord-derived MSCs were intravenously administered into male CD-1 mice with CCl4-induced acute liver injury. Biochemical, histological and molecular parameters were used to evaluate the severity of liver injury. A rat hepatocyte cell line, Clone-9, was used to validate the molecular changes by exosome treatment. RESULTS Exosome treatment significantly suppressed plasma levels of AST, ALT, and pro-inflammatory cytokines, including IL-6 and TNF-α, in the mice with CCl4-induced acute liver injury. Histological morphometry revealed a significant reduction in the necropoptic area in the injured livers following exosome therapy. Consistently, western blot analysis indicated marked elevations in hepatic expression of PCNA, c-Met, Ets-1, and HO-1 proteins after exosome treatment. Besides, the phosphorylation level of signaling mediator JNK was significantly increased, and that of p38 was restored by exosome therapy. Immunohistochemistry double staining confirmed nuclear Ets-1 expression and cytoplasmic localization of c-Met and HO-1 proteins. In vitro studies demonstrated that exosome treatment increased the proliferation of Clone-9 hepatocytes and protected them from CCl4-induced cytotoxicity. Kinase inhibition experiment indicated that the exosome-driven hepatoprotection might be mediated through the JNK pathway. CONCLUSION Exosome therapy activates the JNK signaling activation pathway as well as up-regulates Ets-1 and HO-1 expression, thereby protecting hepatocytes against hepatotoxin-induced cell death.
Collapse
Affiliation(s)
- Ying-Hsien Kao
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Chih-Yang Chang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Yu-Chun Lin
- Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, 52445, Taiwan
| | - Po-Han Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Po-Huang Lee
- Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, 52445, Taiwan
- Committee for Integration and Promotion of Advanced Medicine and Biotechnology, E-Da Healthcare Group, Kaohsiung, 82445, Taiwan
| | - Huoy-Rou Chang
- Departments of Biomedical Engineering, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Wen-Yu Chang
- Department of Dermatology, EDa Cancer Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
- The School of Medicine for International Students, College of Medicine, IShou University, Kaohsiung, 82445, Taiwan
| | - Yo-Chen Chang
- Department of Ophthalmology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Shen-Fa Wun
- Departments of Biomedical Engineering, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Cheuk-Kwan Sun
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
- The School of Medicine for International Students, College of Medicine, IShou University, Kaohsiung, 82445, Taiwan
| |
Collapse
|
8
|
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. [PMID: 38155924 PMCID: PMC10753838 DOI: 10.3389/fbioe.2023.1322514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Liver disease has emerged as a significant worldwide health challenge due to its diverse causative factors and therapeutic complexities. The majority of liver diseases ultimately progress to end-stage liver disease and liver transplantation remains the only effective therapy with the limitations of donor organ shortage, lifelong immunosuppressants and expensive treatment costs. Numerous pre-clinical studies have revealed that extracellular vesicles released by mesenchymal stem cells (MSC-EV) exhibited considerable potential in treating liver diseases. Although natural MSC-EV has many potential advantages, some characteristics of MSC-EV, such as heterogeneity, uneven therapeutic effect, and rapid clearance in vivo constrain its clinical translation. In recent years, researchers have explored plenty of ways to improve the therapeutic efficacy and rotation rate of MSC-EV in the treatment of liver disease. In this review, we summarized current strategies to enhance the therapeutic potency of MSC-EV, mainly including optimization culture conditions in MSC or modifications of MSC-EV, aiming to facilitate the development and clinical application of MSC-EV in treating liver disease.
Collapse
Affiliation(s)
- Lijuan Zheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Jing Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Linna Guo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Zhuofan Zhai
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shuang Xia
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Zhiyu Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Jing Chang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yizhu Jiang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xinran Huang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jingyi Ge
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Changsha, China
| |
Collapse
|
9
|
Yuan M, Yao L, Chen P, Wang Z, Liu P, Xiong Z, Hu X, Li L, Jiang Y. Human umbilical cord mesenchymal stem cells inhibit liver fibrosis via the microRNA-148a-5p/SLIT3 axis. Int Immunopharmacol 2023; 125:111134. [PMID: 37918086 DOI: 10.1016/j.intimp.2023.111134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/08/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) have garnered considerable attention as prospective modalities of treatment for liver fibrosis (LF). The inhibition of hepatic stellate cell (HSC) activation underlies the anti-fibrotic effects of hUC-MSCs. However, the precise mechanism by which hUC-MSCs impede HSC activation remains unclarified. We aimed to elucidate the intrinsic mechanisms underlying the therapeutic effects of hUC-MSCs in LF patients. METHODS Mice with liver cirrhosis induced by carbon tetrachloride (CCl4) were used as experimental models and administered hUC-MSCs via tail-vein injection. The alterations in inflammation and fibrosis were evaluated through histopathological examinations. RNA sequencing (RNA-seq) and bioinformatics analysis were then conducted to investigate the therapeutic mechanism of hUC-MSCs. Finally, an in-vitro experiment involving the co-cultivation of hUC-MSCs or hUC-MSC-derived exosomes (MSC-Exos) with LX2 cells was performed to validate the potential mechanism underlying the hepatoprotective effects of hUC-MSCs in LF patients. RESULTS hUC-MSC therapy significantly improved liver function and alleviated LF in CCl4-induced mice. High-throughput RNA-Seq analysis identified 1142 differentially expressed genes that were potentially involved in mediating the therapeutic effects of hUC-MSCs. These genes play an important role in regulating the extracellular matrix. miRNA expression data (GSE151098) indicated that the miR-148a-5p level was downregulated in LF samples, but restored following hUC-MSC treatment. miR-148a-5p was delivered to LX2 cells by hUC-MSCs via the exosome pathway, and the upregulated expression of miR-148a-5p significantly suppressed the expression of the activated phenotype of LX2 cells. SLIT3 was identified within the pool of potential target genes regulated by miR-148a-5p. Furthermore, hUC-MSC administration upregulated the expression of miR-148a-5p, which played a crucial role in suppressing the expression of SLIT3, thereby palliating fibrosis. CONCLUSIONS hUC-MSCs inhibit the activation of HSCs through the miR-148a-5p/SLIT3 pathway and are thus capable of alleviating LF.
Collapse
Affiliation(s)
- Mengqin Yuan
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lichao Yao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ping Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zheng Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Pingji Liu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiyu Xiong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xue Hu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lanjuan Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310053, China.
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
10
|
Xie Y, Li Y, Yao J, Song X, Wang H, Zhang J, Li X. Protein Lactylation Modification and Proteomics Features in Cirrhosis Patients after UC-MSC Treatment. Curr Issues Mol Biol 2023; 45:8444-8460. [PMID: 37886975 PMCID: PMC10605309 DOI: 10.3390/cimb45100532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Umbilical cord mesenchymal stem cell (UC-MSC) therapy improves liver function in liver cirrhosis patients. This study aimed to elucidate the therapeutic mechanism underlying cell therapy by analyzing changes in the modification and expression of proteins 1 month post-treatment with UC-MSCs. This prospective study included 11 cirrhosis patients who received MSC injection. The laboratory indexes before and after treatment were collected to evaluate the clinical treatment effect of UC-MSCs, and the protein expression and lactylation modification in the liver were comprehensively revealed. Meanwhile, weighted gene co-expression network analysis was used to analyze the co-expression protein modules and their relationship with clinical features. The patients with liver cirrhosis showed an improvement trend after receiving UC-MSC treatment; specifically, the liver protein synthesis function was significantly improved and the coagulation function was also significantly improved. Proteomics combined with lactic acid proteomics revealed 160 lysine lactylation (Kla) sites of 119 proteins. Functional analysis showed that the lactylation-modified proteins were enriched in the pathway of glucose and other substances' metabolism, and many key enzymes of glycolysis and gluconeogenesis were lactated. UC-MSC therapy has a certain clinical effect in the treatment of liver cirrhosis and may act by regulating material metabolism, because the lactylation protein points to energy metabolism.
Collapse
Affiliation(s)
- Ye Xie
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Ying Li
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jia Yao
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China
| | - Xiaojing Song
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Haiping Wang
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China
| | - Jianjun Zhang
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China
| | - Xun Li
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- General Surgery Department, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China
- Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, China
| |
Collapse
|
11
|
Shao J, Xia L, Ye Z, Yang Q, Zhang C, Shi Y, Zhang L, Gu L, Xu C, Chen Y, Chen Y, Pan X, Wu F, Pan R, Liang J, Zhang L. A repeat-dose toxicity study of human umbilical cord mesenchymal stem cells in NOG mice by intravenous injection. Expert Opin Drug Metab Toxicol 2023; 19:857-866. [PMID: 37921457 DOI: 10.1080/17425255.2023.2279243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Stem cell-based therapies have demonstrated great potential in several clinical trials. However, safety data on stem cell application remain inadequate. This study evaluated the toxicity of human umbilical cord mesenchymal stem cells (hUC-MSCs) in NOD/Shi-scid/IL-2 Rγnull (NOG) mice. RESEARCH DESIGN AND METHODS Mice were administered hUC-MSCs intravenously at doses of 3.5 × 106 cells/kg and 3.5 × 107 cells/kg. Toxicity was assessed by clinical observation, behavioral evaluation, pathology, organ weight, and histopathology. We determined the distribution of hUC-MSCs using a validated qPCR method and colonization using immunohistochemistry. RESULTS No significant abnormal effects on clinical responses, body weight, or food intake were observed in the mice, except for two in the high-dose group that died during the last administration. Mouse activity in the high-dose group decreased 6 h after the first administration. Terminal examination revealed dose-dependent changes in hematology. The mice in the high-dose group displayed pulmonary artery wall plaques and mild alveolar wall microthrombi. hUC-MSCs colonized primarily the lung tissues and were largely distributed there 24 h after the final administration. CONCLUSIONS The no observed adverse effect level for intravenous administration of hUC-MSCs in NOG mice over a period of 3 w was 3.5 × 106 cells/kg.
Collapse
Affiliation(s)
- Jinjin Shao
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Lijuan Xia
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Zhichao Ye
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Qian Yang
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Chengda Zhang
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Yuhua Shi
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Lili Zhang
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Liqiang Gu
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Cong Xu
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Ying Chen
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Yunxiang Chen
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Xin Pan
- Zhejiang Key Laboratory of Cell-Based Drug and Applied Technology Development, S-Evans Biosciences Co., Ltd, Hangzhou, China
| | - Feifei Wu
- Zhejiang Key Laboratory of Cell-Based Drug and Applied Technology Development, S-Evans Biosciences Co., Ltd, Hangzhou, China
| | - Ruolang Pan
- Zhejiang Key Laboratory of Cell-Based Drug and Applied Technology Development, S-Evans Biosciences Co., Ltd, Hangzhou, China
| | - Jinfeng Liang
- Zhejiang Center for Drugs and Cosmetics Evaluation, Zhejiang Province Food and Drug Administration, Hangzhou, China
| | - Lijiang Zhang
- Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| |
Collapse
|
12
|
Xue G, Zhang J, Wu L, Sun S, Wu H, Hou Y, Wang J. Differentiation of umbilical cord mesenchymal stem cells into hepatocytes with CYP450 metabolic enzyme activity induced by a liver injury microenvironment. Biochem Biophys Res Commun 2023; 647:47-54. [PMID: 36716645 DOI: 10.1016/j.bbrc.2023.01.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/15/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
The aim of this study was to observe the effect of a simulated liver tissue injury microenvironment on the directed differentiation of umbilical cord mesenchymal stem cells into hepatocytes with CYP450 metabolic activity in vitro, and to explore the mechanisms underlying this directed differentiation. Normal and damaged liver tissue homogenate supernatants (LHS and CCl4-LHS, respectively) were used as induction fluids. After induction for different durations, Western blot and RT-PCR were used to measure the protein and gene expression of the hepatocellular proteins AFP, CK18, ALB, and the CYP450 family. Simultaneously, the metabolic activity of CYP450 in hepatocytes was determined. Compared with the LHS and CCl4-LHS controls, the LHS and CCl4-LHS induction groups showed a significantly elevated protein and gene expression of AFP, CK18, ALB, CYP1A1/2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 (P < 0.05). The metabolic activity of CYP450 in hepatocytes was increased (P < 0.05). In addition, compared with the LHS group, the CCl4-LHS group induced cell differentiation more rapidly and with a higher efficiency. The results suggested that a liver injury microenvironment is conducive for the directed differentiation of umbilical cord mesenchymal stem cells into hepatocytes with metabolic enzyme activity.
Collapse
Affiliation(s)
- Gai Xue
- Department of Sports Human Science, Shijiazhuang University, Shijiazhuang, 050035, Hebei province, China
| | - JinHu Zhang
- Hebei Academy of Chinese Medicine Sciences, Hebei University of Chinese Medicine, Shijiazhuang, 050030, Hebei Province, China
| | - LiYing Wu
- Department of Pharmacy, The 980th Hospital of the People's Liberation Army Joint Logistics Support Force, Shijiazhuang, 050082, Hebei Province, China
| | - Shuang Sun
- Department of Sports Human Science, Shijiazhuang University, Shijiazhuang, 050035, Hebei province, China
| | - HongHai Wu
- Department of Pharmacy, The 980th Hospital of the People's Liberation Army Joint Logistics Support Force, Shijiazhuang, 050082, Hebei Province, China
| | - YanNing Hou
- Department of Pharmacy, The 980th Hospital of the People's Liberation Army Joint Logistics Support Force, Shijiazhuang, 050082, Hebei Province, China
| | - JinXuan Wang
- Department of Sports Human Science, Shijiazhuang University, Shijiazhuang, 050035, Hebei province, China.
| |
Collapse
|
13
|
Liu Y, Amissah OB, Huangfang X, Wang L, Dieu Habimana JD, Lv L, Ding X, Li J, Chen M, Zhu J, Mukama O, Sun Y, Li Z, Huang R. Large-scale expansion of human umbilical cord-derived mesenchymal stem cells using PLGA@PLL scaffold. BIORESOUR BIOPROCESS 2023; 10:18. [PMID: 36915643 PMCID: PMC9994782 DOI: 10.1186/s40643-023-00635-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/31/2023] [Indexed: 03/16/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are highly important in biomedicine and hold great potential in clinical treatment for various diseases. In recent years, the capabilities of MSCs have been under extensive investigation for practical application. Regarding therapy, the efficacy usually depends on the amount of MSCs. Nevertheless, the yield of MSCs is still limited due to the traditional cultural methods. Herein, we proposed a three-dimensional (3D) scaffold prepared using poly lactic-co-glycolic acid (PLGA) nanofiber with polylysine (PLL) grafting, to promote the growth and proliferation of MSCs derived from the human umbilical cord (hUC-MSCs). We found that the inoculated hUC-MSCs adhered efficiently to the PLGA scaffold with good affinity, fast growth rate, and good multipotency. The harvested cells were ideally distributed on the scaffold and we were able to gain a larger yield than the traditional culturing methods under the same condition. Thus, our cell seeding with a 3D scaffold could serve as a promising strategy for cell proliferation in the large-scale production of MSCs. Moreover, the simplicity and low preparation cost allow this 3D scaffold to extend its potential application beyond cell culture. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1186/s40643-023-00635-6.
Collapse
Affiliation(s)
- Yujie Liu
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,Guangzhou Junyuankang Biotechnology Co., Ltd., Guangzhou, 510530 China
| | - Obed Boadi Amissah
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | | | - Ling Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China
| | - Jean de Dieu Habimana
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Linshuang Lv
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Xuanyan Ding
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Junyi Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ming Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China
| | - Jinmin Zhu
- GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436 China
| | - Omar Mukama
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Yirong Sun
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| | - Zhiyuan Li
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,University of Chinese Academy of Sciences, Beijing, 100049 China.,School of Life Sciences, University of Science and Technology of China, Hefei, 230027 China.,GZMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436 China.,GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013 China
| | - Rongqi Huang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China.,GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530 China
| |
Collapse
|
14
|
Liu P, Mao Y, Xie Y, Wei J, Yao J. Stem cells for treatment of liver fibrosis/cirrhosis: clinical progress and therapeutic potential. Stem Cell Res Ther 2022; 13:356. [PMID: 35883127 PMCID: PMC9327386 DOI: 10.1186/s13287-022-03041-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
Cost-effective treatment strategies for liver fibrosis or cirrhosis are limited. Many clinical trials of stem cells for liver disease shown that stem cells might be a potential therapeutic approach. This review will summarize the published clinical trials of stem cells for the treatment of liver fibrosis/cirrhosis and provide the latest overview of various cell sources, cell doses, and delivery methods. We also describe the limitations and strengths of various stem cells in clinical applications. Furthermore, to clarify how stem cells play a therapeutic role in liver fibrosis, we discuss the molecular mechanisms of stem cells for treatment of liver fibrosis, including liver regeneration, immunoregulation, resistance to injury, myofibroblast repression, and extracellular matrix degradation. We provide a perspective for the prospects of future clinical implementation of stem cells.
Collapse
Affiliation(s)
- Pinyan Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yongcui Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ye Xie
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jiayun Wei
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jia Yao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China. .,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China.
| |
Collapse
|
15
|
Yao L, Hu X, Dai K, Yuan M, Liu P, Zhang Q, Jiang Y. Mesenchymal stromal cells: promising treatment for liver cirrhosis. Stem Cell Res Ther 2022; 13:308. [PMID: 35841079 PMCID: PMC9284869 DOI: 10.1186/s13287-022-03001-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/13/2022] [Indexed: 11/11/2022] Open
Abstract
Liver fibrosis is a wound-healing process that occurs in response to severe injuries and is hallmarked by the excessive accumulation of extracellular matrix or scar tissues within the liver. Liver fibrosis can be either acute or chronic and is induced by a variety of hepatotoxic causes, including lipid deposition, drugs, viruses, and autoimmune reactions. In advanced fibrosis, liver cirrhosis develops, a condition for which there is no successful therapy other than liver transplantation. Although liver transplantation is still a viable option, numerous limitations limit its application, including a lack of donor organs, immune rejection, and postoperative complications. As a result, there is an immediate need for a different kind of therapeutic approach. Recent research has shown that the administration of mesenchymal stromal cells (MSCs) is an attractive treatment modality for repairing liver injury and enhancing liver regeneration. This is accomplished through the cell migration into liver sites, immunoregulation, hepatogenic differentiation, as well as paracrine mechanisms. MSCs can also release a huge variety of molecules into the extracellular environment. These molecules, which include extracellular vesicles, lipids, free nucleic acids, and soluble proteins, exert crucial roles in repairing damaged tissue. In this review, we summarize the characteristics of MSCs, representative clinical study data, and the potential mechanisms of MSCs-based strategies for attenuating liver cirrhosis. Additionally, we examine the processes that are involved in the MSCs-dependent modulation of the immune milieu in liver cirrhosis. As a result, our findings lend credence to the concept of developing a cell therapy treatment for liver cirrhosis that is premised on MSCs. MSCs can be used as a candidate therapeutic agent to lengthen the survival duration of patients with liver cirrhosis or possibly reverse the condition in the near future.
Collapse
Affiliation(s)
- Lichao Yao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Xue Hu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Kai Dai
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Mengqin Yuan
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Pingji Liu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Qiuling Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
16
|
Liu X, Li X, Wu G, Qi P, Zhang Y, Liu Z, Li X, Yu Y, Ye X, Li Y, Yang D, Teng Y, Shi C, Jin X, Qi S, Liu Y, Wang S, Liu Y, Cao F, Kong Q, Wang Z, Zhang H. Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Deliver miR-21 to Promote Corneal Epithelial Wound Healing through PTEN/PI3K/Akt Pathway. Stem Cells Int 2022; 2022:1252557. [PMID: 35873535 PMCID: PMC9303509 DOI: 10.1155/2022/1252557] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/19/2022] [Accepted: 06/23/2022] [Indexed: 12/30/2022] Open
Abstract
Objective Rapid restoration of corneal epithelium integrity after injury is particularly important for preserving corneal transparency and vision. Mesenchymal stem cells (MSCs) can be taken into account as the promising regenerative therapeutics for improvement of wound healing processes based on the variety of the effective components. The extracellular vesicles form MSCs, especially exosomes, have been considered as important paracrine mediators though transferring microRNAs into recipient cell. This study investigated the mechanism of human umbilical cord MSC-derived small extracellular vesicles (HUMSC-sEVs) on corneal epithelial wound healing. Methods HUMSC-sEVs were identified by transmission electron microscopy, nanoparticle tracking analysis, and Western blot. Corneal fluorescein staining and histological staining were evaluated in a corneal mechanical wound model. Changes in HCEC proliferation after HUMSC-sEVs or miR-21 mimic treatment were evaluated by CCK-8 and EdU assays, while migration was assessed by in vitro scratch wound assay. Full-length transcriptome sequencing was performed to identify the differentially expressed genes associated with HUMSC-sEVs treatment, followed by validation via real-time PCR and Western blot. Results The sEVs derived from HUMSCs can significantly promote corneal epithelial cell proliferation, migration in vitro, and corneal epithelial wound healing in vivo. Similar effects were obtained after miR-21 transfection, while the beneficial effects of HUMSC-sEVs were partially negated by miR-21 knockdown. Results also show that the benefits are associated with decreased PTEN level and activated the PI3K/Akt signaling pathway in HCECs. Conclusion HUMSC-sEVs could enhance the recovery of corneal epithelial wounds though restraining PTEN by transferring miR-21 and may represent a promising novel therapeutic agent for corneal wound repair.
Collapse
Affiliation(s)
- Xiaolong Liu
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xuran Li
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Guangyuan Wu
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Pengfei Qi
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Yanyan Zhang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhiyu Liu
- Department of Laboratory Diagnostics, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xinyue Li
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yu Yu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiangmei Ye
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Yang Li
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Dongguang Yang
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Yueqiu Teng
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Ce Shi
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Xin Jin
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Sen Qi
- Department of Hematology, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuting Liu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Shudan Wang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Liu
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Fenglin Cao
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Qingran Kong
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhenkun Wang
- Central Laboratory, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| | - Hong Zhang
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Barakat M, Hussein AM, Salama MF, Awadalla A, Barakat N, Serria M, El-Shafey M, El-Sherbiny M, El Adl MA. Possible Underlying Mechanisms for the Renoprotective Effect of Retinoic Acid-Pretreated Wharton's Jelly Mesenchymal Stem Cells against Renal Ischemia/Reperfusion Injury. Cells 2022; 11:cells11131997. [PMID: 35805083 PMCID: PMC9266019 DOI: 10.3390/cells11131997] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Objectives: The current work investigated the effect of Wharton jelly mesenchymal stem cells (WJ-MSCs) pretreated with all-trans-retinoic acid (ATRA) on renal ischemia in rats and the possible role of oxidative stress, apoptotic and Wnt/β-Catenin signaling pathways, and inflammatory cytokines in their effects. Methods: The study included 90 male Sprague Dawley rats that were allocated to five groups (n = 18 rats): (I) Sham-operated group (right nephrectomy was performed); (II) Ischemia/reperfusion injury (IRI) group, a sham group with 45-min renal ischemia on the left kidney; (III) ATRA group, an ischemic group with an intravenous (i.v.) administration of ATRA 10 µM, 10 min post-surgery); (IV) WJ-MSCs group, an IRI group with an i.v. administration of 150 µL containing 7 × 106 WJ-MSCs, 10 min post-surgery; (V) WJ-MSCs + ATRA group, an IRI group with an i.v. administration of 150 µL of 7 × 106 WJ-MSCs pretreated with 10 µM ATRA. At the end of the experiments, serum creatinine, BUN micro-albuminuria (MAU), urinary protein, markers of redox state in the left kidney (MDA, CAT, SOD, and GSH), and the expression of Bax, IL-6, HIF-1α, Wnt7B, and β-catenin genes at the level of mRNA as well as for immunohistochemistry for NFkB and β-Catenin markers were analyzed. Results: The current study found that 45-min of renal ischemia resulted in significant impairment of kidney function (evidenced by the increase in serum creatinine, BUN, and urinary proteins) and deterioration of the kidney morphology, which was associated with a significant increase in redox state (evidenced by an increase in MDA and a decrease in GSH, SOD, and CAT), and a significant increase in inflammatory and apoptotic processes (evidenced by an increase in Bax and IL-6, NFkB, Wnt7B, β-catenin and HIF-1α) in kidney tissues (p < 0.05). On the other hand, treatment with ATRA, WJ-MSCs, or a combination of both, caused significant improvement in kidney function and morphology, which was associated with significant attenuation of oxidative stress, apoptotic markers, and inflammatory cytokines (IL6 and NFkB) with the upregulation of HIF-1α and β-catenin in kidney tissues (p < 0.05). Moreover, the renoprotective effect of WJ-MSCs pretreated with ATRA was more potent than WJ-MSCs alone. Conclusions: It is concluded that preconditioning of WJ-MSCs with ATRA may enhance their renoprotective effect. This effect could be due to the upregulation of the beta-catenin/Wnt pathway and attenuation of apoptosis, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Mai Barakat
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
- Institute of Global Public Health and Human Ecology, School of Science and Engineering, American University, Cairo 11835, Egypt
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
- Correspondence: ; Tel.: +20-10-0242-1140
| | - Mohamed F. Salama
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
| | - Amira Awadalla
- Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt; (A.A.); (N.B.)
| | - Nashwa Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt; (A.A.); (N.B.)
| | - Mohamed Serria
- Department of Biochemistry, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed El-Shafey
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
- Physiological Sciences Department, Fakeeh College for Medical Sciences, Jeddah 21461, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 71666, Saudi Arabia;
| | - Mohamed A. El Adl
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (M.B.); (M.F.S.); (M.A.E.A.)
| |
Collapse
|
18
|
Fu M, Yin W, Zhang W, Zhu Y, Ni H, Gong L. MicroRNA-15a inhibits hepatic stellate cell activation and proliferation via targeting SRY-box transcription factor 9. Bioengineered 2022; 13:13011-13020. [PMID: 35611752 PMCID: PMC9276033 DOI: 10.1080/21655979.2022.2068895] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Accumulating research have indicated that microRNAs are associated with the progression of hepatic fibrosis (HF). Nevertheless, the biological role and function of microRNA (miR)-15a in HF are still unknown. Our data revealed that miR-15a expression was decreased in TGF-β1-treated LX-2 cells and CCl4-induced mouse model. Additionally, miR-15a could directly target the 3’‑untranslated region of SRY-box transcription factor 9 (SOX9) to inhibit its expression. miR-15a overexpression attenuated the viability and invasion, but enhanced apoptosis in LX-2 cells. However, miR-15a knockdown had the opposite effects. Interestingly, SOX9 overexpression reversed the changes in cell viability, invasion and apoptosis mediated by miR-15a overexpression. Moreover, the miR-15a overexpression-mediated collagen I and alpha smooth muscle actin (a-SMA) downregulation were reversed by SOX9 overexpression. Overall, miR-15a could inhibit LX-2 cell viability and HF pathogenesis by targeting SOX9 in vitro and in vivo.
Collapse
Affiliation(s)
- Maoying Fu
- Department of Infectious Diseases, The First People's Hospital of Kunshan, China
| | - Weihua Yin
- Department of Infectious Diseases, The First People's Hospital of Kunshan, China
| | - Wei Zhang
- Department of Infectious Diseases, The First People's Hospital of Kunshan, China
| | - Yanfang Zhu
- Department of Infectious Diseases, The First People's Hospital of Kunshan, China
| | - Huihui Ni
- Department of Infectious Diseases, The First People's Hospital of Kunshan, China
| | - Li Gong
- Department of Infectious Diseases, The First People's Hospital of Kunshan, China
| |
Collapse
|
19
|
Hazrati A, Malekpour K, Soudi S, Hashemi SM. Mesenchymal Stromal/Stem Cells and Their Extracellular Vesicles Application in Acute and Chronic Inflammatory Liver Diseases: Emphasizing on the Anti-Fibrotic and Immunomodulatory Mechanisms. Front Immunol 2022; 13:865888. [PMID: 35464407 PMCID: PMC9021384 DOI: 10.3389/fimmu.2022.865888] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
Various factors, including viral and bacterial infections, autoimmune responses, diabetes, drugs, alcohol abuse, and fat deposition, can damage liver tissue and impair its function. These factors affect the liver tissue and lead to acute and chronic liver damage, and if left untreated, can eventually lead to cirrhosis, fibrosis, and liver carcinoma. The main treatment for these disorders is liver transplantation. Still, given the few tissue donors, problems with tissue rejection, immunosuppression caused by medications taken while receiving tissue, and the high cost of transplantation, liver transplantation have been limited. Therefore, finding alternative treatments that do not have the mentioned problems is significant. Cell therapy is one of the treatments that has received a lot of attention today. Hepatocytes and mesenchymal stromal/stem cells (MSCs) are used in many patients to treat liver-related diseases. In the meantime, the use of mesenchymal stem cells has been studied more than other cells due to their favourable characteristics and has reduced the need for liver transplantation. These cells increase the regeneration and repair of liver tissue through various mechanisms, including migration to the site of liver injury, differentiation into liver cells, production of extracellular vesicles (EVs), secretion of various growth factors, and regulation of the immune system. Notably, cell therapy is not entirely excellent and has problems such as cell rejection, undesirable differentiation, accumulation in unwanted locations, and potential tumorigenesis. Therefore, the application of MSCs derived EVs, including exosomes, can help treat liver disease and prevent its progression. Exosomes can prevent apoptosis and induce proliferation by transferring different cargos to the target cell. In addition, these vesicles have been shown to transport hepatocyte growth factor (HGF) and can promote the hepatocytes'(one of the most important cells in the liver parenchyma) growths.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Dai Y, Cui X, Zhang G, Mohsin A, Xu H, Zhuang Y, Guo M. Development of a novel feeding regime for large scale production of human umbilical cord mesenchymal stem/stromal cells. Cytotechnology 2022; 74:351-369. [DOI: 10.1007/s10616-022-00523-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 01/23/2022] [Indexed: 12/21/2022] Open
|
21
|
Yang H, Xie Y, Li T, Liu S, Zeng S, Wang B. A novel minimally invasive OFM technique with orthotopic transplantation of hUC-MSCs and in vivo monitoring of liver metabolic microenvironment in liver fibrosis treatment. Stem Cell Res Ther 2021; 12:534. [PMID: 34627378 PMCID: PMC8502355 DOI: 10.1186/s13287-021-02599-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) transplantation showed promising therapeutic results in liver fibrosis. However, efficient cell delivery method is urgently needed and the therapeutic mechanism remains unclear. This study focused on developing a minimally invasive open-flow microperfusion (OFM) technique, which combined orthotopic transplantation of human umbilical cord-derived (hUC)-MSCs to liver and in vivo monitoring of liver microenvironment in mice with CCl4-induced liver fibrosis. Methods The therapeutic potential of OFM route was evaluated by comparing OFM with intravenous (IV) injection route in terms of hUC-MSCs engraftment at the fibrosis liver, liver histopathological features, liver function and fibrotic markers expression after hUC-MSCs administration. OFM was also applied to sample liver interstitial fluid in vivo, and subsequent metabolomic analysis was performed to investigate metabolic changes in liver microenvironment. Results Compared with IV route, OFM route caused more hUC-MSCs accumulation in the liver and was more effective in improving the remodeling of liver structure and reducing collagen deposition in fibrotic liver. OFM transplantation of hUC-MSCs reduced blood ALT, AST, ALP and TBIL levels and increased ALB levels, to a greater extent than IV route. And OFM route appeared to have a more pronounced effect on ameliorating the CCl4-induced up-regulation of the fibrotic markers, such as α-SMA, collagen I and TGF-β. In vivo monitoring of liver microenvironment demonstrated the metabolic perturbations induced by pathological condition and treatment intervention. Two metabolites and eight metabolic pathways, which were most likely to be associated with the liver fibrosis progression, were regulated by hUC-MSCs administration. Conclusion The results demonstrated that the novel OFM technique would be useful for hUC-MSCs transplantation in liver fibrosis treatment and for monitoring of the liver metabolic microenvironment to explore the underlying therapeutic mechanisms. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02599-w.
Collapse
Affiliation(s)
- Hui Yang
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Yuanyuan Xie
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Tuo Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Shuo Liu
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Sheng Zeng
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Bin Wang
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
22
|
Sungkar T, Putra A, Lindarto D, Sembiring RJ. Intravenous Umbilical Cord-derived Mesenchymal Stem Cells Transplantation Regulates Hyaluronic Acid and Interleukin-10 Secretion Producing Low-grade Liver Fibrosis in Experimental Rat. Med Arch 2021; 74:177-182. [PMID: 32801431 PMCID: PMC7405996 DOI: 10.5455/medarh.2020.74.177-182] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Introduction: Immunomodulation properties of mesenchymal stem cells have attracted tremendous attention that eventually could regress liver fibrosis process. Aim: The study aims to demonstrate the immunomodulation activities of Umbilical cord-derived Mesenchymal stem cells (UC-MSCs) affecting interleukin-10 (IL-10) and hyaluronic acid (HA) secretion post intraperitoneal injection of CCl4, potent hepatotoxin, induced liver fibrosis among experimental rats. Methods: There were 18 Sprague-Dawley (SD) rats divided into three treatment groups (G1 sham group, G2 untreated liver fibrosis group, and G3 UC-MSCs treated-group) and isolated in Stem Cell and Cancer Research Facility, Semarang, Indonesia. Blood examination was conducted after 3 and 14 days of UC-MSCs transplantation using sandwich based ELISA followed by the histopathological analysis of rat liver tissue. ANOVA and posthoc LSD tests were determined the significance against all groups based on their quantitative measurement. Results: UC-MSCs have been successfully extracted and isolated as well as positive with osteogenic differentiation (Alizarin dye). In further analysis, there were significant mean differences among all groups through the ANOVA test, both IL-10 and HA secretion, concurrent with low-grade liver fibrosis in G3. IL-10 elevates during the early phase of UC-MSCs transplantation, and HA significantly reduced on the 14th day of transplantation, it characterizes the liver fibrosis that has been attenuated. Conclusion: The transplantation of UC-MSCs has given an opportunity for the treatment of a wide range of chronic liver diseases through the immunomodulation properties via its paracrine effects that regulate specific cytokine to suppress fibrosis development.
Collapse
Affiliation(s)
- Taufik Sungkar
- Divison of Gastroenterol-hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Agung Putra
- Stem Cell And Cancer Research (SCCR), Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia.,Department of Pathology, Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia
| | - Dharma Lindarto
- Division of Endocrine Metabolic, Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Rosita Juwita Sembiring
- Department of Clinical Pathology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
23
|
Yao Y, Xia Z, Cheng F, Jang Q, He J, Pan C, Zhang L, Ye Y, Wang Y, Chen S, Su D, Su X, Cheng L, Shi G, Dai L, Deng H. Human placental mesenchymal stem cells ameliorate liver fibrosis in mice by upregulation of Caveolin1 in hepatic stellate cells. Stem Cell Res Ther 2021; 12:294. [PMID: 34016164 PMCID: PMC8139101 DOI: 10.1186/s13287-021-02358-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/26/2021] [Indexed: 02/08/2023] Open
Abstract
Background Liver fibrosis (LF) is a common pathological process characterized by the activation of hepatic stellate cells (HSCs) and accumulation of extracellular matrix. Severe LF causes cirrhosis and even liver failure, a major cause of morbidity and mortality worldwide. Transplantation of human placental mesenchymal stem cells (hPMSCs) has been considered as an alternative therapy. However, the underlying mechanisms and the appropriate time window for hPMSC transplantation are not well understood. Methods We established mouse models of CCl4-injured LF and administered hPMSCs at different stages of LF once a week for 2 weeks. The therapeutic effect of hPMSCs on LF was investigated, according to histopathological and blood biochemical analyses. In vitro, the effect of hPMSCs and the secretomes of hPMSCs on the inhibition of activated HSCs was assessed. RNA sequencing (RNA-seq) analysis, real-time PCR array, and western blot were performed to explore possible signaling pathways involved in treatment of LF with hPMSCs. Results hPMSC treatment notably alleviates experimental hepatic fibrosis, restores liver function, and inhibits inflammation. Furthermore, the therapeutic effect of hPMSCs against mild-to-moderate LF was significantly greater than against severe LF. In vitro, we observed that the hPMSCs as well as the secretomes of hPMSCs were able to decrease the activation of HSCs. Mechanistic dissection studies showed that hPMSC treatment downregulated the expression of fibrosis-related genes, and this was accompanied by the upregulation of Caveolin-1 (Cav1) (p < 0.001). This suggested that the amelioration of LF occurred partly due to the restoration of Cav1 expression in activated HSCs. Upregulation of Cav1 can inhibit the TGF-/Smad signaling pathway, mainly by reducing Smad2 phosphorylation, resulting in the inhibition of activated HSCs, whereas this effect could be abated if Cav1 was silenced in advance by siRNAs. Conclusions Our findings suggest that hPMSCs could provide multifaceted therapeutic benefits for the treatment of LF, and the TGF-/Cav1 pathway might act as a therapeutic target for hPMSCs in the treatment of LF. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02358-x.
Collapse
Affiliation(s)
- Yunqi Yao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Zhemin Xia
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Fuyi Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Qingyuan Jang
- Department of Obstetrics, Sichuan Provincial Hospital for Women and Children, Chengdu, P.R. China
| | - Jiao He
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Cheng Pan
- Department of Plastic and Burn Surgery, West China Hospital, Chengdu, P.R. China
| | - Lin Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Yixin Ye
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Yuan Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Shuang Chen
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Dongsheng Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No. 1, Gao-peng Street, Chengdu, 610041, Sichuan, P.R. China.
| |
Collapse
|
24
|
Lou S, Duan Y, Nie H, Cui X, Du J, Yao Y. Mesenchymal stem cells: Biological characteristics and application in disease therapy. Biochimie 2021; 185:9-21. [PMID: 33711361 DOI: 10.1016/j.biochi.2021.03.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells. In addition to the capacity for self-renewal and multipotential differentiation, MSCs also have the following characteristics. MSCs can exert immunomodulatory functions through interaction with innate or adaptive immune cells, MSCs with poor immunogenicity can be used for allogeneic transplantation, and MSCs can "home" to inflammation and tumour sites. Based on these biological properties, MSCs demonstrate broad clinical application prospects in the treatment of tissue injury, autoimmune diseases, transplantation, cancer and other inflammation-related diseases. In this review we describe the biological characteristics of MSCs and discuss the research advances of MSCs in regenerative medicine, immunomodulation, oncology, and COVID-19, to fully understand the range of diseases in which MSC therapy may be beneficial.
Collapse
Affiliation(s)
- Songyue Lou
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Henan, 450018, China.
| | - Huizong Nie
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Xujie Cui
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Jialing Du
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Yongfang Yao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Henan, 450018, China; School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
25
|
Li W, Shi L, Hu B, Hong Y, Zhang H, Li X, Zhang Y. Mesenchymal Stem Cell-Based Therapy for Stroke: Current Understanding and Challenges. Front Cell Neurosci 2021; 15:628940. [PMID: 33633544 PMCID: PMC7899984 DOI: 10.3389/fncel.2021.628940] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/14/2021] [Indexed: 12/15/2022] Open
Abstract
Stroke, the most prevalent cerebrovascular disease, causes serious loss of neurological function and is the leading cause of morbidity and mortality worldwide. Despite advances in pharmacological and surgical therapy, treatment for functional rehabilitation following stroke is limited with a consequent serious impact on quality of life. Over the past decades, mesenchymal stem cell (MSCs)-based therapy has emerged as a novel strategy for various diseases including stroke due to their unique properties that include easy isolation, multipotent differentiation potential and strong paracrine capacity. Although MSCs have shown promising results in the treatment of stroke, there remain many challenges to overcome prior to their therapeutic application. In this review, we focus on the following issues: the scientific data from preclinical studies and clinical trials of MSCs in the treatment of stroke; the potential mechanisms underlying MSC-based therapy for stroke; the challenges related to the timing and delivery of MSCs and MSC senescence.
Collapse
Affiliation(s)
- Weifeng Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Linli Shi
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Bei Hu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yimei Hong
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hao Zhang
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuelin Zhang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Lin J, Deng H, Zhang Y, Zou L, Fu Z, Dai J. Effect of human umbilical cord-derived mesenchymal stem cells on murine model of bronchiolitis obliterans like injury. Pediatr Pulmonol 2021; 56:129-137. [PMID: 33085211 DOI: 10.1002/ppul.25128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND Bronchiolitis obliterans is a fatal respiratory disease characterized by the obliteration of small airways. Mesenchymal stem cells (MSCs) is a promising candidate for cell-based therapy. OBJECTIVE To evaluate the therapeutic effect of human umbilical cord-derived mesenchymal stem cells (HUC-MSCs) on a murine model of bronchiolitis obliterans like injury (BOLI). METHOD The murine model of BOLI was established by administrating of diacetyl (DA) via intratracheal instillation. Treatment of HUC-MSCs or HUC-MSCs culture medium (HUC-MSCs-CM) was conducted in the BOLI model. RESULTS The pathogenic manifestations, lung function, and the number of neutrophils were similar between the oropharyngeal inhalation DA group (OPI-DA), intratracheal instillation group (ITI-DA); however, less reduction of weight and higher survival rate were observed in ITI-DA groups. Compared with the control groups, the trend of weight loss was significantly reduced (p < .05), and the pulmonary function was significantly improved (p < .05) in HUC-MSCs and HUC-MSCs-CM groups. Masson staining and hematoxylin and eosin staining showed that the deposition of collagen around bronchioles and blood vessels is less and airway epithelial cells and basal cells in lung tissue repaired better in HUC-MSCs and HUC-MSCs-CM groups compared with the control groups. Immunofluorescence shows the expression of E-cadherin and cytokeratin 5 (CK-5) were significantly higher in HUC-MSCs and HUC-MSCs-CM groups compared with control groups, while HUC-MSCs themselves did not express E-cadherin or CK-5. The DiI label showed HUC-MSCs gradually reduced after 2 days in the bronchus and 4 days in bronchiole. CONCLUSION HUC-MSCs could help to repair airway epithelial cells in a murine model of BOLI. It might be related to paracrine factors of HUC-MSCs.
Collapse
Affiliation(s)
- Jilei Lin
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Huarong Deng
- Guangzhou Women and Children's Medical Center, Guangdong, China
| | - Yin Zhang
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lin Zou
- Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Center for Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhou Fu
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jihong Dai
- Department of Respiratory Disease, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
27
|
Faruqu FN, Liam‐Or R, Zhou S, Nip R, Al‐Jamal KT. Defined serum-free three-dimensional culture of umbilical cord-derived mesenchymal stem cells yields exosomes that promote fibroblast proliferation and migration in vitro. FASEB J 2021; 35:e21206. [PMID: 33368666 PMCID: PMC7986687 DOI: 10.1096/fj.202001768rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022]
Abstract
Stem cell-derived exosomes are emerging as novel and clinically relevant cell-free therapeutics for regenerative therapy. This work focused on investigating the stimulation of fibroblasts by exosomes derived from umbilical cord-derived mesenchymal stem cells (ucMSC) in a defined serum-free three-dimensional (3D) culture. 3D culture of ucMSC was carried out in medium supplemented with KnockOut serum replacement (KO-medium) using the Aggrewell system. ucMSC in KO-medium formed spheroids with maintained size and integrity throughout culture. This enabled the isolation of vesicles from ucMSC spheroids in KO-medium with sizes that fall within the exosomal size range and were positive for the expression of canonical exosomal markers CD63, CD9, CD81, Alix, and TSG101. The ucMSC-derived exosomes (ExoucMSC ) were shown to significantly increase the migration and proliferation of murine fibroblasts in vitro. To conclude, 3D culture of ucMSC in defined serum-free KO-medium formed viable spheroids which enabled the isolation of ExoucMSC with the potential of accelerating wound healing.
Collapse
Affiliation(s)
- Farid N. Faruqu
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | - Revadee Liam‐Or
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | - Shuai Zhou
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | - Rebecca Nip
- Institute of Pharmaceutical ScienceKing’s College LondonLondonUK
| | | |
Collapse
|
28
|
Na J, Song J, Kim HH, Seok J, Kim JY, Jun JH, Kim GJ. Human placenta-derived mesenchymal stem cells trigger repair system in TAA-injured rat model via antioxidant effect. Aging (Albany NY) 2020; 13:61-76. [PMID: 33406506 PMCID: PMC7835021 DOI: 10.18632/aging.202348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/05/2020] [Indexed: 01/09/2023]
Abstract
Oxidative stress induces damages of various cell types or tissues through a repetitive imbalance between the systemic manifestation of reactive oxygen species (ROS) and detoxification of the reactive intermediates. Thioacetamide (TAA) is well known for causing several degenerative diseases by oxidative stress. However, study of the antioxidant mechanisms of stem cells in TAA-injured rat model is insufficient. Therefore, we investigated the effect of placenta-derived mesenchymal stem cells (PD-MSCs) transplantation on liver and ovary of TAA-injured rat models to study the antioxidant effect in degenerative diseases. In TAA-injured rat model, PD-MSCs engrafted into damaged organ including liver and ovary in PD-MSCs transplanted groups (Tx) compared with non-transplanted groups (NTx) (*p<0.05). Transplanted PD-MSCs reduced inflammatory factors and upregulated oxidative stress factors in Tx compared with NTx (*p<0.05). Also, transplanted PD-MSCs enhanced antioxidants factors and organ functional restoration factors in Tx compared with NTx. These data show that PD-MSC transplantation triggers the regeneration of organ (e.g., liver and ovary) damaged by oxidative stress from TAA treatment via activating antioxidant factors. Therefore, these data suggest the therapeutic potential via antioxidant effect and help understand the therapeutic mechanism of PD-MSCs in damaged tissues such as in liver and reproductive system.
Collapse
Affiliation(s)
- Jeeyoon Na
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph Song
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Hyun Ho Kim
- College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jin Seok
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Jae Yeon Kim
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Ji Hye Jun
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
29
|
Xu C, Zhao J, Li Q, Hou L, Wang Y, Li S, Jiang F, Zhu Z, Tian L. Exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells ameliorate pulmonary fibrosis in a mouse silicosis model. Stem Cell Res Ther 2020; 11:503. [PMID: 33239075 PMCID: PMC7687745 DOI: 10.1186/s13287-020-02023-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background Silicosis is an occupational respiratory disease caused by long-term excessive silica inhalation, which is most commonly encountered in industrial settings. Unfortunately, there is no effective therapy to delay and cure the progress of silicosis. In the recent years, stem cell therapy has emerged as an attractive tool against pulmonary fibrosis (PF) owing to its unique biological characteristics. However, the direct use of stem cells remains limitation by many risk factors for therapeutic purposes. The exclusive utility of exosomes secreted from stem cells, rather than cells, has been considered a promising alternative to overcome the limitations of cell-based therapy while maintaining its advantages. Methods and results In this study, we first employed a three-dimensional (3D) dynamic system to culture human umbilical cord mesenchymal stem cell (hucMSC) spheroids in a microcarrier suspension to yield exosomes from serum-free media. Experimental silicosis was induced in C57BL/6J mice by intratracheal instillation of a silica suspension, with/without exosomes derived from hucMSC (hucMSC-Exos), injection via the tail vein afterwards. The results showed that the gene expression of collagen I (COL1A1) and fibronectin (FN) was upregulated in the silica group as compared to that in the control group; however, this change decreased with hucMSC-Exo treatment. The value of FEV0.1 decreased in the silica group as compared to that in the control group, and this change diminished with hucMSC-Exo treatment. These findings suggested that hucMSC-Exos could inhibit silica-induced PF and regulate pulmonary function. We also performed in vitro experiments to confirm these findings; the results revealed that hucMSC-Exos decreased collagen deposition in NIH-3T3 cells exposed to silica. Conclusions Taken together, these studies support a potential role for hucMSC-Exos in ameliorating pulmonary fibrosis and provide new evidence for improving clinical treatment induced by silica. Supplementary information The online version contains supplementary material available at 10.1186/s13287-020-02023-9.
Collapse
Affiliation(s)
- Chunjie Xu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jing Zhao
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Qiuyue Li
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Lin Hou
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Siling Li
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Fuyang Jiang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
30
|
Bagheri-Mohammadi S. Protective effects of mesenchymal stem cells on ischemic brain injury: therapeutic perspectives of regenerative medicine. Cell Tissue Bank 2020; 22:249-262. [PMID: 33231840 DOI: 10.1007/s10561-020-09885-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022]
Abstract
Cerebral ischemic injury as the main manifestation of stroke can occur in stroke patients (70-80%). Nowadays, the main therapeutic strategy used for ischemic brain injury treatment aims to achieve reperfusion, neuroprotection, and neurorecovery. Also, angiogenesis as a therapeutic approach maybe represents a promising tool to enhance the prognosis of cerebral ischemic stroke. Unfortunately, although many therapeutic approaches as a life-saving gateway for cerebral ischemic injuries like pharmacotherapy and surgical treatments are widely used, they all fail to restore or regenerate damaged neurons in the brain. So, the suitable therapeutic approach would focus on regenerating the lost cells and restore the normal function of the brain. Currently, stem cell-based regenerative medicine introduced a new paradigm approach in cerebral ischemic injuries treatment. Today, in experimental researches, different types of stem cells such as mesenchymal stem cells have been applied. Therefore, stem cell-based regenerative medicine provides the opportunity to inquire and develop a more effective and safer therapeutic approach with the capability to produce and regenerate new neurons in damaged tissues.
Collapse
Affiliation(s)
- Saeid Bagheri-Mohammadi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Departments of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
31
|
Yang J, Ma K, Zhang C, Liu Y, Liang F, Hu W, Bian X, Yang S, Fu X. Burns Impair Blood-Brain Barrier and Mesenchymal Stem Cells Can Reverse the Process in Mice. Front Immunol 2020; 11:578879. [PMID: 33240266 PMCID: PMC7677525 DOI: 10.3389/fimmu.2020.578879] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/07/2020] [Indexed: 12/17/2022] Open
Abstract
Neurological syndromes are observed in numerous patients who suffer burns, which add to the economic burden of societies and families. Recent studies have implied that blood-brain barrier (BBB) dysfunction is the key factor that induces these central nervous system (CNS) syndromes in peripheral traumatic disease, e.g., surgery and burns. However, the effect of burns on BBB and the underlying mechanism remains, largely, to be determined. The present study aimed to investigate the effect of burns on BBB and the potential of umbilical cord-derived mesenchymal stem cells (UC-MSCs), which have strong anti-inflammatory and repairing ability, to protect the integrity of BBB. BBB permeability was evaluated using dextran tracer (immunohistochemistry imaging and spectrophotometric quantification) and western blot, interleukin (IL)-6, and IL-1β levels in blood and brain were measured by enzyme-linked immunosorbent assay. Furthermore, transmission electron microscopy (TEM) was used to detect transcellular vesicular transport (transcytosis) in BBB. We found that burns increased mouse BBB permeability to both 10-kDa and 70-kDa dextran. IL-6 and IL-1β levels increased in peripheral blood and CNS after burns. In addition, burns decreased the level of tight junction proteins (TJs), including claudin-5, occludin, and ZO-1, which indicated increased BBB permeability due to paracellular pathway. Moreover, increased vesicular density after burns suggested increased transcytosis in brain microvascular endothelial cells. Finally, administering UC-MSCs at 1 h after burns effectively reversed these adverse effects and protected the integrity of BBB. These results suggest that burns increase BBB permeability through both paracellular pathway and transcytosis, the potential mechanism of which might be through increasing IL-6 and IL-1β levels and decreasing Mfsd2a level, and appropriate treatment with UC-MSCs can reverse these effects and protect the integrity of BBB after burns.
Collapse
Affiliation(s)
- Jie Yang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China.,Department of Dermatology, Fourth Medical Center, PLA General Hospital, Beijing, China
| | - Kui Ma
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China
| | - Cuiping Zhang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China
| | - Yufan Liu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China.,Department of Dermatology, Fourth Medical Center, PLA General Hospital, Beijing, China
| | - Feng Liang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China
| | - Wenzhi Hu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China
| | - Xiaowei Bian
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China.,Tianjin Medical University, Tianjin, China
| | - Siming Yang
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China.,Department of Dermatology, Fourth Medical Center, PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, Chinese People's Liberation Army (PLA) General Hospital and PLA Medical College, Beijing, China.,Department of Dermatology, Fourth Medical Center, PLA General Hospital, Beijing, China
| |
Collapse
|
32
|
Yang N, Chen H, Gao Y, Zhang S, Lin Q, Ji X, Li N, Xu W, Liu Y, Jin S. Tanshinone IIA exerts therapeutic effects by acting on endogenous stem cells in rats with liver cirrhosis. Biomed Pharmacother 2020; 132:110815. [PMID: 33113421 DOI: 10.1016/j.biopha.2020.110815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Liver cirrhosis (LC), the major pathway for the progression and development of chronic liver disease, is an advanced stage of liver disease. It is the third most common chronic noncommunicable disease after cardiovascular diseases and malignant tumors. Tanshinone IIA (Tan), an extract of Salvia miltiorrhiza (S. miltiorrhiza), has been proven to promote the proliferation and differentiation of stem cells. Moreover, its protective effect in liver injury has received widespread attention. The present study investigated whether Tan plays a therapeutic role in LC by promoting endogenous stem cell proliferation and differentiation. MATERIALS AND METHODS LC models were established by intraperitoneal injection of an olive oil solution containing 50 % carbon tetrachloride (CCL4) combined with 10 % alcohol in the drinking water. After successful model establishment, the animals were randomly divided into four groups and injected with physiological saline or low-, medium-, or high-dose (10, 20, or 40 mg/kg) Tan for seven consecutive days. The protective effect of Tan on LC was observed by western blotting, serological examination and histopathological staining. Furthermore, immunofluorescence double-labeling of 5-bromo-2-deoxyuridine (BrdU) and the liver cell markers albumin and CK-18 or the liver stem cell markers EPCAM and OV-6 was used to evaluate the proliferation and differentiation of endogenous liver stem cells. RESULTS We confirmed successful establishment of the LC model by observing transaminase levels and hematoxylin-eosin (HE) and Masson staining of liver sections in CCL4-treated and healthy rats. After Tan treatment, HE and Masson staining of paraffin sections of liver tissue showed that Tan treatment significantly improved histological injury to the liver. Serological tests showed that albumin-bilirubin (ALBI) scores and models for end-stage liver disease (MELD) were lower. Immunofluorescence and immunohistochemical staining showed that the newly proliferated cells were colocalized with ALB, OV-6, EPCAM, and CK-18, indicating that new expression of these markers occurred after Tan injection. All results were most significant in the medium-dose treatment group. CONCLUSION Tan can alleviate liver injury induced by CCL4 combined with alcohol in rats and plays a therapeutic role in LC by promoting the proliferation and differentiation of endogenous liver stem cells.
Collapse
Affiliation(s)
- Ningning Yang
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Haoyuan Chen
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Yang Gao
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Sijia Zhang
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Qiuchi Lin
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Xuechun Ji
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Ning Li
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Wanying Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Ying Liu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Street, Nan Gang District, Harbin, Heilongjiang Province, 150081, China.
| |
Collapse
|
33
|
Chen H, Chen X, Wang G. Platelets: A review of their function and effects in liver diseases. LIVER RESEARCH 2020. [DOI: 10.1016/j.livres.2020.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
34
|
Jiang L, Zhang Y, Liu T, Wang X, Wang H, Song H, Wang W. Exosomes derived from TSG-6 modified mesenchymal stromal cells attenuate scar formation during wound healing. Biochimie 2020; 177:40-49. [PMID: 32800897 DOI: 10.1016/j.biochi.2020.08.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/16/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cell (MSC)-derived exosome therapy has emerged as an effective therapy strategy for the pathological scar formation. However, the underlying mechanisms have not been completely understood. In the current study, we investigate the therapeutic effect of TSG-6 modified MSC-derived exosomes on a mouse full-thickness wound model and provide evidence of a possible mechanism for MSC-derived exosomes to prevent from scar formation. Overexpression and knockdown of TSG-6 were conducted by lentivirus infection into hBMSCs. Exosomes were isolated from cell culture and identified by transmission electron microscopy and Western blot. C57BL/6J mice were performed of full-thickness skin wounds and treated with exosomal suspension or TSG-6-neutralizing antibody. H&E staining was subjected to observe the pathological changes of scar tissues. Immunohistochemistry, ELISA, real time-PCR and Western blot were applied to detect the expressions of relevant molecules. The results showed that subcutaneous injection of TSG-6 overexpressed MSC-derived exosomes effectively ameliorated scar pathological injury, decreased inflammatory molecular secretion and attenuated collagen deposition in a mouse skin wound model. Reversely, knockdown of TSG-6 abrogated the therapeutic effect of MSC-derived exosomes on scarring. Moreover, TSG-6-neutralizing antibody counteracted the effect of TSG-6 overexpressed MSC-derived exosomes in preventing scar formation. In conclusion, we demonstrated that exosomes derived from TSG-6 modified MSCs suppressed scar formation via reducing inflammation and inhibiting collagen deposition.
Collapse
Affiliation(s)
- Lan Jiang
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Yanguo Zhang
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China.
| | - Tao Liu
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China.
| | - Xiaoxia Wang
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Huan Wang
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Haifeng Song
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| | - Wenting Wang
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, People's Republic of China
| |
Collapse
|
35
|
Jia Y, Shu X, Yang X, Sun H, Cao H, Cao H, Zhang K, Xu Q, Li G, Yang Y. Enhanced therapeutic effects of umbilical cord mesenchymal stem cells after prolonged treatment for HBV-related liver failure and liver cirrhosis. Stem Cell Res Ther 2020; 11:277. [PMID: 32650827 PMCID: PMC7350639 DOI: 10.1186/s13287-020-01787-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/05/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
Background Umbilical cord mesenchymal stem cells (UCMSCs) have been demonstrated to have good therapeutic effects in the treatment of HBV-related liver diseases. However, the therapeutic effect of UCMSCs on HBV-related liver failure and liver cirrhosis and the variations in the efficacy of UCMSCs after different treatment courses remain poorly understood. Therefore, this study was designed to answer these two questions. Methods This was an observational study that retrospectively considered a 3-year period during which 513 patients who received stem cell infusion and met the criteria of hepatic failure and liver cirrhosis were identified from the databases of the Third Affiliated Hospital of Sun Yat-sen University. The eligible patients were categorized into the liver failure group and liver cirrhosis group. The two groups were divided into different subgroups according to the duration of stem cell therapy. In the liver failure group, group A received more than 4 weeks and group B received less than 4 weeks of stem cell therapy. In the liver cirrhosis group, patients who received more than 4 weeks of stem cell therapy belonged to group C, and the patients in group D received less than 4 weeks of stem cell therapy. The patients were followed up for 24 weeks. The demographics, clinical characteristics, biochemical factors, and model for end-stage liver disease (MELD) scores were recorded and compared among different groups. Results A total of 64 patients met the criteria for liver failure, and 59 patients met the criteria for liver cirrhosis. After UCMSC treatment, the levels of alanine aminotransferase (ALT), glutamic-oxaloacetic transaminase (AST), and total bilirubin (TBIL) at all postbaseline time points were significantly lower than those at baseline in the liver failure group and liver cirrhosis group; the prothrombin activity (PTA) and MELD scores gradually improved in only the liver failure group. Four weeks after UCMSC treatment, patients who received prolonged treatment with UCMSCs had a larger decrease in TBIL levels than patients who terminated treatment with UCMSCs. After more than 4 weeks of UCMSC treatment, there were no statistically significant differences in the changes in ALT, AST, TBIL, and PTA values and MELD scores between patients with liver failure who received prolonged treatment with UCMSCs and patients with liver cirrhosis who received prolonged treatment with UCMSCs at any time point. However, the median decrease and cumulative decrease in the TBIL level of patients with liver failure with a standard 4-week treatment course were larger than those of patients with liver cirrhosis with a standard 4-week treatment course. Conclusion Peripheral infusion of UCMSCs showed good therapeutic effects for HBV-related liver failure and liver cirrhosis. Prolonging the treatment course can increase the curative effect of UCMSCs for end-stage liver disease, especially for patients with cirrhosis.
Collapse
Affiliation(s)
- Yifan Jia
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xin Shu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiaoan Yang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Haixia Sun
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Huijuan Cao
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Hong Cao
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China.
| | - Ka Zhang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China.
| | - Qihuan Xu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Gang Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yang Yang
- Department of Liver Surgery and Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|