1
|
Li H, Zhang Y, Du S, Shen J, Liu X, Jing J. "Remodeling the intestinal immune microenvironment": immune regulation and tissue regeneration by mesenchymal stem/stromal cells in the repair microenvironment of inflammatory bowel disease. Front Immunol 2025; 16:1543702. [PMID: 40433382 PMCID: PMC12106535 DOI: 10.3389/fimmu.2025.1543702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
The global prevalence of inflammatory bowel disease (IBD) has significantly increased in recent decades. IBD is a long-term, recurring, gastrointestinal inflammatory condition that mainly comprises two primary clinical types: ulcerative colitis and Crohn's disease. The current treatment paradigm for IBD primarily focuses on symptom management. However, this approach does not support mucosal epithelial repair, maintenance of barrier homeostasis, or regulation of biological functions in the gut. Conventional therapies rely on the frequent use of high-dose medications, including antibiotics, nonsteroidal anti-inflammatory drugs, biological agents, and immunomodulators. Recently, mesenchymal stem/stromal cells (MSCs) have gained interest in tissue regeneration owing to their unique ability to differentiate and secrete regulatory factors, including extracellular vesicles (EVs), which play crucial roles in abnormal organization. Various routes of administration have been explored in preclinical and clinical studies to deliver MSCs from diverse tissue sources. The routes include intraperitoneal, intravenous, and local (intracolonic or rectal) delivery. The MSCs employed were obtained from various tissues, including bone marrow, umbilical cord, and adipose tissue. This article reviews the research framework for the application of MSCs and EVs secretion in the treatment of IBD, emphasizing key immunological effects, such as immune microenvironment regulation, intestinal barrier stabilization, and therapeutic approaches targeting intestinal barrier disorders. The discussion primarily focuses on the advantages of MSCs over other biologics, impairment of gut mucosal tissue-resident mesenchymal stem cells in IBD development, immune targets (at the cellular and molecular levels) within the framework of IBD, and the reparative effects of MSCs in the microenvironment of IBD. We aimed to present an overview of the current trends in MSC research and therapy, as well as to identify the challenges and future directions that must be addressed to advance research on MSC-mediated therapeutic strategies for IBD.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Jing
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
2
|
Zhao Q, Shao M, Ma L, Zhou R. Insights into Modeling Inflammatory Bowel Disease from Stem Cell Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10887-8. [PMID: 40299197 DOI: 10.1007/s12015-025-10887-8] [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] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is a multifactorial, immune-mediated condition marked by chronic gastrointestinal inflammation. This condition significantly impairs patients' quality of life and represents a major public health challenge globally. Pathogenesis of IBD arises from complex interplay among genetic predisposition, environmental factors, immune dysregulation, and microbial dysbiosis. Although significant strides have been made in unraveling these mechanisms, existing therapeutic options remain inadequate in addressing the full spectrum of clinical needs, underscoring the urgent demand for innovative strategies. Regenerative medicine has emerged as a promising frontier, offering novel tools for therapeutic development. We briefly consolidated current knowledge on IBD pathogenesis and treatments, emphasized the pivotal potential of human intestinal organoids (including adult stem cell-derived organoids and pluripotent stem cell- derived organoids) as a robust platform for mechanistic studies and treatment exploration. Leveraging this technology, we aim to advance personalized and next-generation therapies for IBD.
Collapse
Affiliation(s)
- Qi Zhao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Miaoli Shao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Lisha Ma
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Renfang Zhou
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China.
| |
Collapse
|
3
|
Zhang L, Wang H, Zhao L, Zhang J, Sun W, Chu J, Zhao H, Yang C, Yan S, Chen X, Xu D. Unraveling the interplay between mesenchymal stem cells, gut microbiota, and systemic sclerosis: therapeutic implications. Microbiol Spectr 2025:e0157624. [PMID: 40272189 DOI: 10.1128/spectrum.01576-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/28/2025] [Indexed: 04/25/2025] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease with progressive fibrotic disorders in multiple organs. Mesenchymal stem cells (MSCs) have shown great potential in treating SSc, but the exact regulatory mechanism is not fully understood. In this study, we used human umbilical cord-derived MSCs (hUC-MSCs) to treat SSc mice induced by bleomycin. The gut microbiota composition and predicted functions were analyzed using 2bRAD sequencing of fecal samples from control, SSc, and MSCs-treated mice. Treatment with MSCs improved the bleomycin-induced SSc mice, characterized by significantly reduced collagen deposition and dermal thickness. The gut microbiota of SSc mice exhibited lower species evenness and was clearly separated from the control mice based on beta diversity. MSC treatment led to a significant reduction of conditionally pathogenic bacteria enriched in SSc, including Akkermansia muciniphila and Parasutterella excrementihominis. Conversely, the relative abundance of butyrate-producing bacteria, such as Roseburia, Butyricicoccus porcorum, and Gemmiger formicilis, was notably increased in MSCs-treated SSc mice. Additionally, the functional analysis revealed that MSCs intervention effectively enhanced sulfur metabolism, tryptophan metabolism, citrate cycle, RNA polymerase, and beta-lactam resistance. In summary, the findings in the present study have suggested the close association between gut microbiota and metabolic dysbiosis in mice with SSc. The administration of MSCs has been shown to regulate the disrupted metabolic pathways in SSc mice, thus restoring the normal function of the gut microbiota. This study provides valuable insights into the specific gut microbiota and metabolic pathways involved in the efficacy of MSC treatment, thereby proposing a novel therapeutic strategy for SSc. IMPORTANCE Human umbilical cord-derived mesenchymal stem cells (HUC‑MSCs) demonstrate efficacy in alleviating skin thickening and collagen deposition in systemic sclerosis (SSc) mice, which also regulate the gut microbiota composition and function. Specifically, MSC intervention leads to a notable increase in butyrate-producing bacteria, a decrease in Akkermansia muciniphila and Parasutterella excrementihominis, and a reversal of the dysregulated microbial function in SSc mice. These findings underscore the potential significance of gut microbiota in the therapeutic effects of MSCs in SSc.
Collapse
Affiliation(s)
- Lili Zhang
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Hui Wang
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Lu Zhao
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Jin Zhang
- Department of Rheumatology and Immunology, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Wenchang Sun
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Jinjin Chu
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Haobin Zhao
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Chunjuan Yang
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
- Department of Rheumatology and Immunology, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery, the Affiliated Hospital, Shandong Second Medical University, Weifang, China
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Xiaohua Chen
- Department of Nuclear Medicine, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Donghua Xu
- Medical Research Center, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
- Department of Rheumatology and Immunology, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
4
|
Ho CT, Kao YC, Shyu YM, Wang IC, Liu QX, Liu SW, Huang SC, Chiu H, Hsu LW, Hsu TS, Hsieh WC, Huang CC. Assembly of MSCs into a spheroid configuration increases poly(I:C)-mediated TLR3 activation and the immunomodulatory potential of MSCs for alleviating murine colitis. Stem Cell Res Ther 2025; 16:172. [PMID: 40221757 PMCID: PMC11993957 DOI: 10.1186/s13287-025-04297-3] [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: 12/10/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is associated with significant clinical challenges due to the limitations of current therapeutic approaches. Mesenchymal stem cell (MSC)-based therapies have shown promise in alleviating IBD owing to their potent immunomodulatory properties. However, the therapeutic efficacy of these cells remains suboptimal, primarily due to the harsh peritoneal microenvironment, which compromises MSC viability and functional capacity after transplantation. METHODS To address these limitations, this study aimed to improve MSC engraftment and functionality by assembling MSCs into three-dimensional (3D) spheroids and priming them with the Toll-like receptor 3 (TLR3) agonist polyinosinic-polycytidylic acid (poly(I:C)). Their potential for treating IBD was evaluated using male C57BL/6 mice with dextran sulfate sodium-induced colitis. RESULTS While 3D spheroid formation alone upregulated TLR3 expression and increased MSC survival under oxidative stress, poly(I:C) priming had a pronounced synergistic effect, significantly increasing MSC-mediated splenocyte modulation and oxidative stress resistance. In a murine colitis model, compared with unprimed spheroids or MSC suspensions, poly(I:C)-primed MSC spheroids administered intraperitoneally exhibited increased survival and therapeutic efficacy, effectively alleviating colitis symptoms, reducing colonic inflammation, and promoting tissue recovery. CONCLUSION Collectively, these findings highlight the synergistic benefits of combining 3D spheroid assembly with TLR3 activation as an innovative strategy to improve the therapeutic efficacy of MSC-based treatments for IBD and other inflammatory diseases by increasing post-engraftment cell survival and immunomodulatory capacity.
Collapse
Affiliation(s)
- Chao-Ting Ho
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ying-Chi Kao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yueh-Ming Shyu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - I-Ching Wang
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Qiao-Xuan Liu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih-Chen Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Han Chiu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Wen Hsu
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu , Taiwan
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Wan-Chen Hsieh
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Choudhery MS, Arif T, Mahmood R, Mushtaq A, Niaz A, Hassan Z, Zahid H, Nayab P, Arshad I, Arif M, Majid M, Harris DT. Induced Mesenchymal Stem Cells: An Emerging Source for Regenerative Medicine Applications. J Clin Med 2025; 14:2053. [PMID: 40142860 PMCID: PMC11943107 DOI: 10.3390/jcm14062053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Regenerative medicine is gaining interest in the medical field due to the limitations of conventional treatments, which often fail to address the underlying cause of disease. In recent years, stem cell-based therapies have evolved as a promising alternative approach to treat those diseases that cannot be cured using conventional medicine. Adult stem cells, particularly the mesenchymal stem cells (MSCs), have attracted a lot of attention due to their ability to regenerate and repair human tissues and organs. MSCs isolated from adult tissues are well characterized and are currently the most common type of cells for use in regenerative medicine. However, their low number in adult donor tissues, donor-age and cell-source related heterogeneity, limited proliferative and differentiation potential, and early senescence in in vitro cultures, negatively affect MSC regenerative potential. These factors restrict MSC use for research as well as for clinical applications. To overcome these problems, MSCs with superior regenerative potential are required. Induced MSCs (iMSCs) are obtained from induced pluripotent stem cells (iPSCs). These cells are patient-specific, readily available, and have relatively superior regenerative potential and, therefore, can overcome the problems associated with the use of primary MSCs. In this review, the authors aim to discuss the characteristics, regenerative potential, and limitations of MSCs for regenerative medicine applications. The main methods to generate iMSCs from iPSCs have been discussed in detail. In addition, the proposed criteria for their molecular characterization, applications of iMSCs for disease modeling and drug discovery, as well as potential use in regenerative medicine have been explored in detail.
Collapse
Affiliation(s)
- Mahmood S. Choudhery
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Taqdees Arif
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Ruhma Mahmood
- Department of Pediatric Surgery, Allama Iqbal Medical College, Jinnah Hospital, Lahore 54700, Pakistan;
| | - Asad Mushtaq
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Ahmad Niaz
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Zaeema Hassan
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Hamda Zahid
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Pakeeza Nayab
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Iqra Arshad
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Mehak Arif
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - Mashaim Majid
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 50161, Pakistan; (M.S.C.); (T.A.); (A.M.); (A.N.); (Z.H.); (H.Z.); (P.N.); (I.A.); (M.A.); (M.M.)
| | - David T. Harris
- Department of Immunobiology, University of Arizona Health Sciences Biorepository, College of Medicine, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
6
|
Jammes M, Tabasi A, Bach T, Ritter T. Healing the cornea: Exploring the therapeutic solutions offered by MSCs and MSC-derived EVs. Prog Retin Eye Res 2025; 105:101325. [PMID: 39709150 DOI: 10.1016/j.preteyeres.2024.101325] [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: 09/18/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Affecting a large proportion of the population worldwide, corneal disorders constitute a concerning health hazard associated to compromised eyesight or blindness for most severe cases. In the last decades, mesenchymal stem/stromal cells (MSCs) demonstrated promising abilities in improving symptoms associated to corneal diseases or alleviating these affections, especially through their anti-inflammatory, immunomodulatory and pro-regenerative properties. More recently, MSC therapeutic potential was shown to be mediated by the molecules they release, and particularly by their extracellular vesicles (EVs; MSC-EVs). Consequently, using MSC-EVs emerged as a pioneering strategy to mitigate the risks related to cell therapy while providing MSC therapeutic benefits. Despite the promises given by MSC- and MSC-EV-based approaches, many improvements are considered to optimize the therapeutic significance of these therapies. This review aspires to provide a comprehensive and detailed overview of current knowledge on corneal therapies involving MSCs and MSC-EVs, the strategies currently under evaluation, and the gaps remaining to be addressed for clinical implementation. From encapsulating MSCs or their EVs into biomaterials to enhance the ocular retention time to loading MSC-EVs with therapeutic drugs, a wide range of ground-breaking strategies are currently contemplated to lead to the safest and most effective treatments. Promising research initiatives also include diverse gene therapies and the targeting of specific cell types through the modification of the EV surface, paving the way for future therapeutic innovations. As one of the most important challenges, MSC-EV large-scale production strategies are extensively investigated and offer a wide array of possibilities to meet the needs of clinical applications.
Collapse
Affiliation(s)
- Manon Jammes
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Abbas Tabasi
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Trung Bach
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway, Ireland; CURAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
7
|
Wang Q, Tang X, Wang Y, Zhang D, Li X, Liu S. The role of extracellular vesicles in non-alcoholic steatohepatitis: Emerging mechanisms, potential therapeutics and biomarkers. J Adv Res 2025; 69:157-168. [PMID: 38494073 PMCID: PMC11954800 DOI: 10.1016/j.jare.2024.03.009] [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: 09/11/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH), an emerging global healthcare problem, has become the leading cause of liver transplantation in recent decades. No effective therapies in the clinic have been proven due to the incomplete understanding of the pathogenesis of NASH, and further studies are expected to continue to delve into the mechanisms of NASH. Extracellular vesicles (EVs), which are small lipid membrane vesicles carrying proteins, microRNAs and other molecules, have been identified to play a vital role in cell-to-cell communication and are involved in the development and progression of various diseases. In recent years, there has been increasing interest in the role of EVs in NASH. Many studies have revealed that EVs mediate important pathological processes in NASH, and the role of EVs in NASH is distinct and variable depending on their origin cells and target cells. This review outlines the emerging mechanisms of EVs in the development of NASH and the preclinical evidence related to stem cell-derived EVs as a potential therapeutic strategy for NASH. Moreover, possible strategies involving EVs as clinical diagnostic, staging and prognostic biomarkers for NASH are summarized.
Collapse
Affiliation(s)
- Qianrong Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xiangning Tang
- Department of endocrinology, the Second Affiliated Hospital of University of South China, 421001 Hunan Province, China
| | - Yu Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Danyi Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
8
|
Sheu JJ, Yeh JN, Chen YC, Chiang JY, Sung PH, Huang CR, Li YC, Yip HK. Shock wave-pretreated ADMSCs of cell-sheet scaffold (CSS) patched on the left ventricular wall (LVW) inhibited LVW remodeling in mini-pig MI: role of CSS on counteracting Laplace's Law of LVW stress - experimental study. Int J Surg 2024; 110:7546-7562. [PMID: 39497545 PMCID: PMC11634124 DOI: 10.1097/js9.0000000000002119] [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: 04/08/2024] [Accepted: 10/05/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND We investigated whether shock wave (SW)-pretreated autologous adipocyte-derived mesenchymal stem cells (ADMSCs) seeded in the cell-sheet scaffold (CSS) could inhibit left ventricular (LV) remodeling and improve LV ejection fraction (LVEF) in old myocardial infarction (MI). METHODS Mini-pigs ( n =20) were divided into group 1 (sham-operated control), group 2 (old MI), group 3 (old MI + autologous ADMSCs/1.0×10 7 in CSS on LV myocardium), and group 4 [old MI + SW (0.12 mJ/mm 2 for total 140 shots)-pretreated ADMSCs in CSS on LV myocardium]. Treatments started on day 28 after MI induction. In-vivo and in-vitro studies were conducted. RESULTS Cell viability/relative mitochondria DNA expression/mitochondrial cytochrome C/adenosine triphosphate concentration in ADMCSs and protein expressions of angiogenesis factors [vascular endothelial growth factor (VEGF)/stromal cell-derived factor-1 (SDF-1)/mitochondrial respiratory chain complexes I-IV/oxygen consumption rate] were higher in group 4 than in group 3 ( P <0.001). By day 180, LVEF and small vessel numbers in the peri-infarct or infarct area were highest in group 1, lowest in group 2, and significantly lower in group 3 than in group 4. In contrast, the LV dimension was opposite to the pattern of change in LVEF in all groups ( P <0.0001). The basal/middle/apical infarct and fibrotic areas were inversely related to LVEF in all groups (all P <0.0001). Protein levels of angiogenetic markers (SDF-1α/C-X-C chemokine receptor type 4/VEGF/angiopoietin-1) were significantly and persistently increased from groups 1 to 4. In contrast, protein levels of endothelial cell markers (von Willebrand factor or endothelial nitric oxide synthase) showed an identical pattern to LVEF in all groups (all P <0.0001). CONCLUSION SW pretreatment of ADMSCs seeded in CSS offered significant benefits in preserving LV performance and ameliorating LV remodeling in mini-pigs with old MI.
Collapse
Affiliation(s)
- Jiunn-Jye Sheu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan, ROC
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan, ROC
| | - Jui-Ning Yeh
- Department of Cardiology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yin-Chia Chen
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - John Y. Chiang
- Department of Computer Science and Engineering, National Sun Yat-Sen University, Kaohsiung, Taiwan, ROC
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Pei-Hsun Sung
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan, ROC
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan, ROC
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Chi-Ruei Huang
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan, ROC
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Yi-Chen Li
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | - Hon-Kan Yip
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan, ROC
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan, ROC
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| |
Collapse
|
9
|
Xiong Z, Li X, Xie M, Guo J, Yin S, Huang D, Jin L, Wang C, Zhang F, Mao C, Chen H, Luo D, Tang H, Chen X, Lian L. Small extracellular vesicles derived from adipose mesenchymal stem cells alleviate intestinal fibrosis by inhibiting the FAK/Akt signaling pathway via MFGE8. J Gastroenterol 2024; 59:1092-1106. [PMID: 39305336 DOI: 10.1007/s00535-024-02152-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/13/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND Intestinal fibrosis is one of the most frequent and severe complications of Crohn's disease. Accumulating studies have reported that adipose mesenchymal stem cell-derived small extracellular vesicles (AMSC-sEVs) could alleviate renal fibrosis, hepatic fibrosis, etc., while their potential for treating intestinal fibrosis remains uncertain. Therefore, this study aims to determine the therapeutic effects of AMSC-sEVs on intestinal fibrosis and identify the mechanisms underlying these effects. METHODS AMSC-sEVs were characterized using transmission electron microscopy, nanoparticle tracking analysis, and western blot. Whether AMSC-sEVs exert antifibrotic effects was investigated in two different murine models of intestinal fibrosis. Besides, AMSC-sEVs were co-cultured with primary human fibroblasts and CCD18co during transforming growth factor (TGF)-β1 stimulation. Label-free proteomics and rescue experiments were performed to identify candidate molecules in AMSC-sEVs. Transcriptome sequencing revealed changes in mRNA levels among different groups. Lastly, proteins related to relevant signaling pathways were identified by western blotting, and their expression and activation status were assessed. RESULTS AMSC-sEVs positively expressed CD63 and Alix and presented a classical "rim of a cup" and granule shape with approximately 43-100 nm diameter. AMSCs significantly alleviated intestinal fibrosis through secreted sEVs in vitro and in vivo. The milk fat globule-EGF factor 8 (MFGE8) was stably enriched in AMSC-sEVs and was an active compound contributing to the treatment of intestinal fibrosis by AMSCs. Mechanistically, AMSC-sEV-based therapies attenuated intestinal fibrosis by inhibiting the FAK/Akt signaling pathway. CONCLUSIONS MFGE8-containing AMSC-sEVs attenuate intestinal fibrosis, partly through FAK/Akt pathway inhibition.
Collapse
Affiliation(s)
- Zhizhong Xiong
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xianzhe Li
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Medical Faculty Heidelberg, Heidelberg University, 69120, Heidelberg, Germany
| | - Minghao Xie
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jianping Guo
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shi Yin
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dayin Huang
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Longyang Jin
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Caiqin Wang
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fengxiang Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaobin Mao
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huaxian Chen
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dandong Luo
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Haijie Tang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xijie Chen
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lei Lian
- Department of Gastrointestinal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, 26 Yuancun Er Heng Rd. Guangzhou, Guangzhou, China.
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
10
|
Jo CH, Lee SY, Son YB, Lee WJ, Choe YH, Lee HJ, Oh SJ, Kim TS, Hong CY, Lee SL, Rho GJ. Regulation of Colonic Inflammation and Macrophage Homeostasis of IFN-γ-Primed Canine AMSCs in Experimental Colitis in Mice. Animals (Basel) 2024; 14:3283. [PMID: 39595338 PMCID: PMC11591378 DOI: 10.3390/ani14223283] [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: 09/17/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have shown potential in treating immune-mediated diseases due to their immunomodulatory properties, which can be enhanced by priming with inflammatory cytokines like interferon-gamma (IFN-γ). This study evaluates the therapeutic effects of IFN-γ-primed canine adipose tissue-derived MSCs (AMSCs) in a mouse model of inflammatory bowel disease (IBD). Canine AMSCs were primed with 50 ng/mL recombinant canine IFN-γ for 48 h, and the effects were compared to those seen in naïve (unprimed) AMSCs. IBD was induced in mice using dextran sodium sulfate (DSS), and AMSCs were injected intraperitoneally on days 1 and 3. The mice treated with IFN-γ-primed AMSCs showed improved clinical outcomes, including a reduced disease activity index (DAI), less body weight loss, and longer colon length compared to the mice treated with naïve AMSCs. A histological analysis revealed less damage to the intestinal structures and reduced inflammatory cell infiltration. IFN-γ priming led to a shift in the immune cell balance in the gut, decreasing pro-inflammatory macrophages (Ly6Chi) and increasing anti-inflammatory macrophages (Ly6Clo/MHC-IIhi). This was associated with the reduced expression of inflammatory cytokine genes (Il-1β, Il-6, and Il-18) and increased expression of the intestinal stem cell marker Lgr5. These findings suggest that IFN-γ-primed AMSCs offer enhanced therapeutic potential for treating CE in veterinary medicine.
Collapse
Affiliation(s)
- Chan-Hee Jo
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
| | - Sang-Yun Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
| | - Young-Bum Son
- Department of Obstetrics, College of Veterinary Medicine, Chonnam National University, 300 Yonbongdong, Buk-gu, Gwangju 500-757, Republic of Korea;
| | - Won-Jae Lee
- College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Yong-Ho Choe
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
| | - Hyeon-Jeong Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
| | - Seong-Ju Oh
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
| | - Tae-Seok Kim
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
| | - Chae-Yeon Hong
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
| | - Sung-Lim Lee
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
- Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Gyu-Jin Rho
- College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea; (C.-H.J.); (S.-Y.L.); (Y.-H.C.); (H.-J.L.); (S.-J.O.); (T.-S.K.); (C.-Y.H.)
- Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
11
|
Wu Z, Su Y, Li J, Liu X, Liu Y, Zhao L, Li L, Zhang L. Induced pluripotent stem cell-derived mesenchymal stem cells: whether they can become new stars of cell therapy. Stem Cell Res Ther 2024; 15:367. [PMID: 39415276 PMCID: PMC11484330 DOI: 10.1186/s13287-024-03968-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/29/2024] [Indexed: 10/18/2024] Open
Abstract
Stem cell therapy constitutes a pivotal subject in contemporary discourse, with donor stem cells having been employed in research and clinical treatments for several decades. Primary cell transplantation encompasses diverse stem cell types, including ectomesenchymal stem cells, hematopoietic stem cells, and various stem cell derivatives such as vesicles and extracellular vesicles. Nevertheless, the emergence of cell engineering techniques has heralded a new epoch in stem cell therapy, markedly broadening their therapeutic potential. Induced pluripotent stem cells (iPSCs) epitomize a significant milestone in modern medical biology. This groundbreaking discovery offers significant potential in disciplines such as biology, pathophysiology, and cellular regenerative medicine. As a result, iPSCs derived differentiated cells have become a pioneering avenue for cell therapy research. Induced mesenchymal stem cells (iMSCs), derived from iPSCs, represent a novel frontier in MSCs related research. Empirical evidence suggests that iMSCs demonstrate enhanced proliferative capacities compared to natural MSCs, with diminished age-related variability and heterogeneity. Numerous clinical trials have highlighted the prospective superiority of iMSCs. This article synthesizes current basic research and clinical trials pertaining to iMSCs, aiming to provide a reference point for future research endeavors.
Collapse
Affiliation(s)
- Zewen Wu
- Shanxi Bethune Hospital, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, 030032, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, 030032, China
| | - Yazhen Su
- Shanxi Bethune Hospital, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Jingxuan Li
- Shanxi Bethune Hospital, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Xinling Liu
- Shanxi Bethune Hospital, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Yang Liu
- Shanxi Bethune Hospital, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Li Zhao
- Shanxi Medical University, Taiyuan, 030000, China
| | - Linxin Li
- Shanxi Bethune Hospital, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China
| | - Liyun Zhang
- Shanxi Bethune Hospital, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan, 030032, China.
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, 030032, China.
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic Diseases), Taiyuan, 030032, China.
| |
Collapse
|
12
|
Shi X, Zhang K, Yu F, Qi Q, Cai X, Zhang Y. Advancements and Innovative Strategies in Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cell Therapy: A Comprehensive Review. Stem Cells Int 2024; 2024:4073485. [PMID: 39377039 PMCID: PMC11458320 DOI: 10.1155/2024/4073485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/24/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
The effectiveness and safety of mesenchymal stem cell (MSC) therapy have been substantiated across various diseases. Nevertheless, challenges such as the restricted in vitro expansion capacity of tissue-derived MSCs and the clinical instability due to the high heterogeneity of isolated cells require urgent resolution. The induced pluripotent stem cell-derived MSCs (iPSC-MSCs), which is differentiated from iPSCs via specific experimental pathways, holds considerable potential as a substitute for tissue derived MSCs. Multiple studies have demonstrated that iPSCs can be differentiated into iPSC-MSCs through diverse differentiation strategies. Research suggests that iPSC-MSCs, when compared to tissue derived MSCs, exhibit superior characteristics in terms of proliferation ability, immune modulation capacity, and biological efficiency. In this review, we meticulously described and summarized the experimental methods of iPSC differentiation into iPSC-MSCs, the application of iPSC-MSCs in various disease models, the latest advancements in clinically relevant iPSC-derived cell products, and the development strategies for the next generation of iPSC-derived therapy products (not only cell products but also their derivatives).
Collapse
Affiliation(s)
- Xiaoyu Shi
- State Industrial Base for Stem Cell Engineering Products, Tianjin 300384, China
| | - Kun Zhang
- State Industrial Base for Stem Cell Engineering Products, Tianjin 300384, China
| | - Fengshi Yu
- State Industrial Base for Stem Cell Engineering Products, Tianjin 300384, China
| | - Qi Qi
- State Industrial Base for Stem Cell Engineering Products, Tianjin 300384, China
| | - Xiaoyu Cai
- State Industrial Base for Stem Cell Engineering Products, Tianjin 300384, China
| | - Yu Zhang
- VCANBIO Cell and Gene Engineering Corp. Ltd., Tianjin, China
| |
Collapse
|
13
|
Wang P, Zhang Y, Li Z, Zhou S, Tang Q, Wang Z, Xiao R, Feng M, Wu L, Liang D. Mesenchymal Stem Cells Derived from Human Urine-Derived iPSCs Exhibit Low Immunogenicity and Reduced Immunomodulatory Profile. Int J Mol Sci 2024; 25:10394. [PMID: 39408724 PMCID: PMC11476417 DOI: 10.3390/ijms251910394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Human-induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) represent a promising and renewable cell source for therapeutic applications. A systematic evaluation of the immunological properties and engraftment potential of iMSCs generated from urine-derived iPSCs is lacking, which has impeded their broader application. In this study, we differentiated urine-derived iPSCs into iMSCs and assessed their fundamental MSC characteristics, immunogenicity, immunomodulatory capacity and in vivo engraftment. Compared to umbilical cord-derived MSCs (UCMSCs), iMSCs demonstrated an enhanced proliferative capacity, a higher level of regenerative gene expression, and lower immunogenicity, strengthening resistance to apoptosis induced by allogeneic peripheral blood mononuclear cells (PBMCs) and the NK-92 cell line. In addition, iMSCs exhibited a diminished ability to inhibit T cell proliferation and activation compared with UCMSCs. Transcriptomic analyses further revealed the decreased expression of immune regulatory factors in iMSCs. After transfusion into mouse models, iMSCs engrafted in the lungs, liver, and spleen and exhibited the ability to migrate to tumor tissues. Our results indicated that iMSCs generated from urine-derived iPSCs have a significant replicative capacity, low immunogenicity and unique immunomodulatory properties, and hence offer obvious advantages in immune privilege and allogenic therapeutic application prospects.
Collapse
Affiliation(s)
- Peiyun Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Ying Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Zhixing Li
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Shenglan Zhou
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Qiyu Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Zujia Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Rou Xiao
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Mai Feng
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410078, China;
| | - Lingqian Wu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| | - Desheng Liang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China; (P.W.); (Y.Z.); (Z.L.); (S.Z.); (Q.T.); (Z.W.); (R.X.); (L.W.)
| |
Collapse
|
14
|
Xu J, Li P, Li Z, Liu S, Guo H, Lesser CF, Ke J, Zhao W, Mou X. Gut bacterial type III secretion systems aggravate colitis in mice and serve as biomarkers of Crohn's disease. EBioMedicine 2024; 107:105296. [PMID: 39216231 PMCID: PMC11402190 DOI: 10.1016/j.ebiom.2024.105296] [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: 02/18/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Mesenteric adipose tissue (mAT) hyperplasia, known as creeping fat, is a pathologic characteristic of Crohn's disease (CD). In our previously reported cohort, we observed that Achromobacter pulmonis was the most abundant and prevalent bacteria cultivated from creeping fat. METHODS A whole genomic sequencing and identification of T3SS orthologs of mAT-derived A. pulmonis were used. A functional type III secretion system (T3SS) mediated the pathogenic potential of A. pulmonis in vitro and in mouse colitis model. Furthermore, a T3SS Finder pipeline was introduced to evaluate gut bacterial T3SS orthologs in the feces of CD patients, ulcerative colitis and colorectal cancer patients. FINDINGS Here, we reveal that mAT-derived A. pulmonis possesses a functional T3SS, aggravates colitis in mice via T3SS, and exhibits T3SS-dependent cytotoxicity via a caspase-independent mechanism in macrophages and epithelial cells, which demonstrated the pathogenic potential of the T3SS-harboring A. pulmonis. Metagenomic analyses demonstrate an increased abundance of Achromobacter in the fecal of Crohn's disease patients compared to healthy controls. A comprehensive comparison of total microbial vT3SS abundance in various intestine diseases demonstrated that the specific enrichment of vT3SS genes was shown in fecal samples of CD, neither ulcerative colitis nor colorectal cancer patients, and ten T3SS gene-based biomarkers for CD were discovered and validated in a newly recruited CD cohort. Furthermore, treatment with exclusive enteral nutrition (EEN), an intervention that improves CD patient symptomatology, was found associated with a significant reduction in the prevalence of T3SS genes in fecal samples. INTERPRETATION These findings highlight the pathogenic significance of T3SSs in the context of CD and identify specific T3SS genes that could potentially function as biomarkers for diagnosing and monitoring the clinical status of CD patients. FUNDING This work is supported by the National Key Research and Development Program of China (2020YFA0907800), the China Postdoctoral Science Foundation (2023M744089), the National Natural Science Foundation of China (32000096), the Shenzhen Science and Technology Programs (KQTD20200820145822023, RCIC20231211085944057, and ZDSYS20220606100803007), National Key Clinical Discipline, Guangdong Provincial Clinical Research Center for Digestive Diseases (2020B1111170004), Qingfeng Scientific Research Fund of the China Crohn's & Colitis Foundation (CCCF) (CCCF-QF-2022B71-1), and the Sixth Affiliated Hospital, Sun Yat-sen University Clinical Research 1010 Program 1010CG(2023)-08. These funding provided well support for this research work, which involved data collection, analysis, interpretation, patient recruitment and so on.
Collapse
Affiliation(s)
- Jun Xu
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Peijie Li
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhenye Li
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Sheng Liu
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Huating Guo
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Cammie F Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA; Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jia Ke
- Department of General Surgery (Intestinal Surgery), The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| | - Wenjing Zhao
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China.
| | - Xiangyu Mou
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong 518107, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, China.
| |
Collapse
|
15
|
Zhou X, Liu J, Wu F, Mao J, Wang Y, Zhu J, Hong K, Xie H, Li B, Qiu X, Xiao X, Wen C. The application potential of iMSCs and iMSC-EVs in diseases. Front Bioeng Biotechnol 2024; 12:1434465. [PMID: 39135947 PMCID: PMC11317264 DOI: 10.3389/fbioe.2024.1434465] [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: 05/17/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
The immune system, functioning as the body's "defense army", plays a role in surveillance, defense. Any disruptions in immune system can lead to the development of immune-related diseases. Extensive researches have demonstrated the crucial immunoregulatory role of mesenchymal stem cells (MSCs) in these diseases. Of particular interest is the ability to induce somatic cells under specific conditions, generating a new cell type with stem cell characteristics known as induced pluripotent stem cell (iPSC). The differentiation of iPSCs into MSCs, specifically induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs), hold promise as a potential solution to the challenges of MSCs, potentially serving as an alternative to traditional drug therapies. Moreover, the products of iMSCs, termed induced pluripotent stem cell-derived mesenchymal stem cell-derived extracellular vesicles (iMSC-EVs), may exhibit functions similar to iMSCs. With the biological advantages of EVs, they have become the focus of "cell-free therapy". Here, we provided a comprehensive summary of the biological impact of iMSCs on immune cells, explored the applications of iMSCs and iMSC-EVs in diseases, and briefly discussed the fundamental characteristics of EVs. Finally, we overviewed the current advantages and challenges associated with iMSCs and iMSC-EVs. It is our hope that this review related to iMSCs and iMSC-EVs will contribute to the development of new approaches for the treatment of diseases.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jinyu Liu
- Department of Obstetrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiangbin Xiao
- Department of Cardiovascular, People’s Hospital of Jianyang, Jianyang, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
16
|
Winston T, Song Y, Shi H, Yang J, Alsudais M, Kontaridis MI, Wu Y, Gaborski TR, Meng Q, Cooney RN, Ma Z. Lineage-Specific Mesenchymal Stromal Cells Derived from Human iPSCs Showed Distinct Patterns in Transcriptomic Profile and Extracellular Vesicle Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308975. [PMID: 38757640 PMCID: PMC11267277 DOI: 10.1002/advs.202308975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/16/2024] [Indexed: 05/18/2024]
Abstract
Over the past decades, mesenchymal stromal cells (MSCs) have been extensively investigated as a potential therapeutic cell source for the treatment of various disorders. Differentiation of MSCs from human induced pluripotent stem cells (iMSCs) has provided a scalable approach for the biomanufacturing of MSCs and related biological products. Although iMSCs shared typical MSC markers and functions as primary MSCs (pMSCs), there is a lack of lineage specificity in many iMSC differentiation protocols. Here, a stepwise hiPSC-to-iMSC differentiation method is employed via intermediate cell stages of neural crest and cytotrophoblast to generate lineage-specific MSCs with varying differentiation efficiencies and gene expression. Through a comprehensive comparison between early developmental cell types (hiPSCs, neural crest, and cytotrophoblast), two lineage-specific iMSCs, and six source-specific pMSCs, are able to not only distinguish the transcriptomic differences between MSCs and early developmental cells, but also determine the transcriptomic similarities of iMSC subtypes to postnatal or perinatal pMSCs. Additionally, it is demonstrated that different iMSC subtypes and priming conditions affected EV production, exosomal protein expression, and cytokine cargo.
Collapse
Affiliation(s)
- Tackla Winston
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Yuanhui Song
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Huaiyu Shi
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Junhui Yang
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
| | - Munther Alsudais
- Departments of Biomedical and Chemical EngineeringRochester Institute of TechnologyOne Lomb Memorial DriveRochesterNY14623USA
| | - Maria I. Kontaridis
- Department of Biomedical Research and Translational MedicineMasonic Medical Research Institute2150 Bleecker StreetUticaNY13501USA
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical CenterHarvard Medical School330 Brookline AveBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBuilding C, 240 Longwood AveBostonMA02115USA
| | - Yaoying Wu
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
- Department of Microbiology & ImmunologySUNY Upstate Medical University766 Irving AvenueSyracuseNY13210USA
| | - Thomas R. Gaborski
- Departments of Biomedical and Chemical EngineeringRochester Institute of TechnologyOne Lomb Memorial DriveRochesterNY14623USA
| | - Qinghe Meng
- Department of SurgeryState University of New York Upstate Medical University750 East Adams StreetSyracuseNY13210USA
- Sepsis Interdisciplinary Research CenterState University of New York Upstate Medical University766 Irving AvenueSyracuseNY13210USA
| | - Robert N. Cooney
- Department of SurgeryState University of New York Upstate Medical University750 East Adams StreetSyracuseNY13210USA
- Sepsis Interdisciplinary Research CenterState University of New York Upstate Medical University766 Irving AvenueSyracuseNY13210USA
| | - Zhen Ma
- Department of Biomedical & Chemical EngineeringSyracuse University329 Link HallSyracuseNY13244USA
- BioInspired Institute for Materials and Living SystemsSyracuse University318 Bowne HallSyracuseNY13244USA
- Department of BiologySyracuse University107 College PlSyracuseNY13210USA
| |
Collapse
|
17
|
Soufihasanabad S, Mahmoudi M, Taghavi-Farahabadi M, Mirsanei Z, Mahmoudi Lamouki R, Mirza Abdalla JK, Babaei E, Hashemi SM. In vivo polarization of M2 macrophages by mesenchymal stem cell-derived extracellular vesicles: A novel approach to macrophage polarization and its potential in treating inflammatory diseases. Med Hypotheses 2024; 187:111353. [DOI: 10.1016/j.mehy.2024.111353] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
18
|
Wei S, Li M, Wang Q, Zhao Y, Du F, Chen Y, Deng S, Shen J, Wu K, Yang J, Sun Y, Gu L, Li X, Li W, Chen M, Ling X, Yu L, Xiao Z, Dong L, Wu X. Mesenchymal Stromal Cells: New Generation Treatment of Inflammatory Bowel Disease. J Inflamm Res 2024; 17:3307-3334. [PMID: 38800593 PMCID: PMC11128225 DOI: 10.2147/jir.s458103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract, which has a high recurrence rate and is incurable due to a lack of effective treatment. Mesenchymal stromal cells (MSCs) are a class of pluripotent stem cells that have recently received a lot of attention due to their strong self-renewal ability and immunomodulatory effects, and a large number of experimental and clinical models have confirmed the positive therapeutic effect of MSCs on IBD. In preclinical studies, MSC treatment for IBD relies on MSCs paracrine effects, cell-to-cell contact, and its mediated mitochondrial transfer for immune regulation. It also plays a therapeutic role in restoring the intestinal mucosal barrier through the homing effect, regulation of the intestinal microbiome, and repair of intestinal epithelial cells. In the latest clinical trials, the safety and efficacy of MSCs in the treatment of IBD have been confirmed by transfusion of autologous or allogeneic bone marrow, umbilical cord, and adipose MSCs, as well as their derived extracellular vesicles. However, regarding the stable and effective clinical use of MSCs, several concerns emerge, including the cell sources, clinical management (dose, route and frequency of administration, and pretreatment of MSCs) and adverse reactions. This article comprehensively summarizes the effects and mechanisms of MSCs in the treatment of IBD and its advantages over conventional drugs, as well as the latest clinical trial progress of MSCs in the treatment of IBD. The current challenges and future directions are also discussed. This review would add knowledge into the understanding of IBD treatment by applying MSCs.
Collapse
Affiliation(s)
- Shulin Wei
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Mingxing Li
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Qin Wang
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Yueshui Zhao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Fukuan Du
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Yu Chen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Shuai Deng
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Jing Shen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Ke Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Jiayue Yang
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Yuhong Sun
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Li Gu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Xiaobing Li
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Wanping Li
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Meijuan Chen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Xiao Ling
- Department of Obstetrics, Luzhou Maternal & Child Health Hospital (Luzhou Second People’s Hospital), Luzhou, Sichuan, 646100, People’s Republic of China
| | - Lei Yu
- Department of Obstetrics, Luzhou Maternal & Child Health Hospital (Luzhou Second People’s Hospital), Luzhou, Sichuan, 646100, People’s Republic of China
| | - Zhangang Xiao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| | - Lishu Dong
- Department of Obstetrics, Luzhou Maternal & Child Health Hospital (Luzhou Second People’s Hospital), Luzhou, Sichuan, 646100, People’s Republic of China
| | - Xu Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646100, People’s Republic of China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, 646100, People’s Republic of China
| |
Collapse
|
19
|
Qiao Y, Tang X, Liu Z, Ocansey DKW, Zhou M, Shang A, Mao F. Therapeutic Prospects of Mesenchymal Stem Cell and Their Derived Exosomes in the Regulation of the Gut Microbiota in Inflammatory Bowel Disease. Pharmaceuticals (Basel) 2024; 17:607. [PMID: 38794176 PMCID: PMC11124012 DOI: 10.3390/ph17050607] [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: 04/04/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have shown great potential in the treatment of several inflammatory diseases due to their immunomodulatory ability, which is mediated by exosomes secreted by MSCs (MSC-Exs). The incidence of inflammatory bowel disease (IBD) is increasing globally, but there is currently no long-term effective treatment. As an emerging therapy, MSC-Exs have proven to be effective in alleviating IBD experimentally, and the specific mechanism continues to be explored. The gut microbiota plays an important role in the occurrence and development of IBD, and MSCs and MSC-Exs can effectively regulate gut microbiota in animal models of IBD, but the mechanism involved and whether the outcome can relieve the characteristic dysbiosis necessary to alleviate IBD still needs to be studied. This review provides current evidence on the effective modulation of the gut microbiota by MSC-Exs, offering a basis for further research on the pathogenic mechanism of IBD and MSC-Ex treatments through the improvement of gut microbiota.
Collapse
Affiliation(s)
- Yaru Qiao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China; (Y.Q.); (Z.L.); (D.K.W.O.); (M.Z.)
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang 222006, China;
| | - Xiaohua Tang
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang 212300, China;
| | - Ziyue Liu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China; (Y.Q.); (Z.L.); (D.K.W.O.); (M.Z.)
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China; (Y.Q.); (Z.L.); (D.K.W.O.); (M.Z.)
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast CC0959347, Ghana
| | - Mengjiao Zhou
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China; (Y.Q.); (Z.L.); (D.K.W.O.); (M.Z.)
| | - Anquan Shang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang 222006, China;
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, China; (Y.Q.); (Z.L.); (D.K.W.O.); (M.Z.)
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang 222006, China;
| |
Collapse
|
20
|
Zhang S, Lee Y, Liu Y, Yu Y, Han I. Stem Cell and Regenerative Therapies for the Treatment of Osteoporotic Vertebral Compression Fractures. Int J Mol Sci 2024; 25:4979. [PMID: 38732198 PMCID: PMC11084822 DOI: 10.3390/ijms25094979] [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: 04/09/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Osteoporotic vertebral compression fractures (OVCFs) significantly increase morbidity and mortality, presenting a formidable challenge in healthcare. Traditional interventions such as vertebroplasty and kyphoplasty, despite their widespread use, are limited in addressing the secondary effects of vertebral fractures in adjacent areas and do not facilitate bone regeneration. This review paper explores the emerging domain of regenerative therapies, spotlighting stem cell therapy's transformative potential in OVCF treatment. It thoroughly describes the therapeutic possibilities and mechanisms of action of mesenchymal stem cells against OVCFs, relying on recent clinical trials and preclinical studies for efficacy assessment. Our findings reveal that stem cell therapy, particularly in combination with scaffolding materials, holds substantial promise for bone regeneration, spinal stability improvement, and pain mitigation. This integration of stem cell-based methods with conventional treatments may herald a new era in OVCF management, potentially improving patient outcomes. This review advocates for accelerated research and collaborative efforts to translate laboratory breakthroughs into clinical practice, emphasizing the revolutionary impact of regenerative therapies on OVCF management. In summary, this paper positions stem cell therapy at the forefront of innovation for OVCF treatment, stressing the importance of ongoing research and cross-disciplinary collaboration to unlock its full clinical potential.
Collapse
Affiliation(s)
- Songzi Zhang
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| | - Yunhwan Lee
- Department of Medicine, School of Medicine, CHA University, Seongnam-si 13496, Republic of Korea;
| | - Yanting Liu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| | - Yerin Yu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (S.Z.); (Y.L.); (Y.Y.)
| |
Collapse
|
21
|
Ren R, Jiang J, Li X, Zhang G. Research progress of autoimmune diseases based on induced pluripotent stem cells. Front Immunol 2024; 15:1349138. [PMID: 38720903 PMCID: PMC11076788 DOI: 10.3389/fimmu.2024.1349138] [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: 12/04/2023] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Autoimmune diseases can damage specific or multiple organs and tissues, influence the quality of life, and even cause disability and death. A 'disease in a dish' can be developed based on patients-derived induced pluripotent stem cells (iPSCs) and iPSCs-derived disease-relevant cell types to provide a platform for pathogenesis research, phenotypical assays, cell therapy, and drug discovery. With rapid progress in molecular biology research methods including genome-sequencing technology, epigenetic analysis, '-omics' analysis and organoid technology, large amount of data represents an opportunity to help in gaining an in-depth understanding of pathological mechanisms and developing novel therapeutic strategies for these diseases. This paper aimed to review the iPSCs-based research on phenotype confirmation, mechanism exploration, drug discovery, and cell therapy for autoimmune diseases, especially multiple sclerosis, inflammatory bowel disease, and type 1 diabetes using iPSCs and iPSCs-derived cells.
Collapse
Affiliation(s)
| | | | | | - Guirong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| |
Collapse
|
22
|
Jeong SY, Park BW, Kim J, Lee S, You H, Lee J, Lee S, Park JH, Kim J, Sim W, Ban K, Park J, Park HJ, Kim S. Hyaluronic acid stimulation of stem cells for cardiac repair: a cell-free strategy for myocardial infarct. J Nanobiotechnology 2024; 22:149. [PMID: 38570846 PMCID: PMC10993512 DOI: 10.1186/s12951-024-02410-x] [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: 10/26/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI), a representative form of ischemic heart disease, remains a huge burden worldwide. This study aimed to explore whether extracellular vesicles (EVs) secreted from hyaluronic acid (HA)-primed induced mesenchymal stem cells (HA-iMSC-EVs) could enhance the cardiac repair after MI. RESULTS HA-iMSC-EVs showed typical characteristics for EVs such as morphology, size, and marker proteins expression. Compared with iMSC-EVs, HA-iMSC-EVs showed enhanced tube formation and survival against oxidative stress in endothelial cells, while reduced reactive oxygen species (ROS) generation in cardiomyocytes. In THP-1 macrophages, both types of EVs markedly reduced the expression of pro-inflammatory signaling players, whereas HA-iMSC-EVs were more potent in augmenting anti-inflammatory markers. A significant decrease of inflammasome proteins was observed in HA-iMSC-EV-treated THP-1. Further, phospho-SMAD2 as well as fibrosis markers in TGF-β1-stimulated cardiomyocytes were reduced in HA-iMSC-EVs treatment. Proteomic data showed that HA-iMSC-EVs were enriched with multiple pathways including immunity, extracellular matrix organization, angiogenesis, and cell cycle. The localization of HA-iMSC-EVs in myocardium was confirmed after delivery by either intravenous or intramyocardial route, with the latter increased intensity. Echocardiography revealed that intramyocardial HA-iMSC-EVs injections improved cardiac function and reduced adverse cardiac remodeling and necrotic size in MI heart. Histologically, MI hearts receiving HA-iMSC-EVs had increased capillary density and viable myocardium, while showed reduced fibrosis. CONCLUSIONS Our results suggest that HA-iMSC-EVs improve cardiac function by augmenting vessel growth, while reducing ROS generation, inflammation, and fibrosis in MI heart.
Collapse
Affiliation(s)
- Seon-Yeong Jeong
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Bong-Woo Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
- Catholic High-Performance Cell Therapy Center and Department of Medical Life Science, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Jimin Kim
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Seulki Lee
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Haedeun You
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Joohyun Lee
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea
| | - Susie Lee
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Jinju Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Woosup Sim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Joonghoon Park
- Graduate School of International Agricultural Technology, Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do, 25354, South Korea
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seoho-gu, Seoul, 06591, Republic of Korea.
- Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Songpa‑gu, Seoul, 05855, South Korea.
| |
Collapse
|
23
|
DeFrates KG, Tong E, Cheng J, Heber‐Katz E, Messersmith PB. A Pro-Regenerative Supramolecular Prodrug Protects Against and Repairs Colon Damage in Experimental Colitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304716. [PMID: 38247203 PMCID: PMC10987129 DOI: 10.1002/advs.202304716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/13/2023] [Indexed: 01/23/2024]
Abstract
Structural repair of the intestinal epithelium is strongly correlated with disease remission in inflammatory bowel disease (IBD); however, ulcer healing is not addressed by existing therapies. To address this need, this study reports the use of a small molecule prolyl hydroxylase (PHD) inhibitor (DPCA) to upregulate hypoxia-inducible factor one-alpha (HIF-1α) and induce mammalian regeneration. Sustained delivery of DPCA is achieved through subcutaneous injections of a supramolecular hydrogel, formed through the self-assembly of PEG-DPCA conjugates. Pre-treatment of mice with PEG-DPCA is shown to protect mice from epithelial erosion and symptoms of dextran sodium sulfate (DSS)-induced colitis. Surprisingly, a single subcutaneous dose of PEG-DPCA, administered after disease onset, leads to accelerated weight gain and complete restoration of healthy tissue architecture in colitic mice. Rapid DPCA-induced restoration of the intestinal barrier is likely orchestrated by increased expression of HIF-1α and associated targets leading to an epithelial-to-mesenchymal transition. Further investigation of DPCA as a potential adjunctive or stand-alone restorative treatment to combat active IBD is warranted.
Collapse
Affiliation(s)
- Kelsey G. DeFrates
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
| | - Elaine Tong
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
| | - Jing Cheng
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
| | | | - Phillip B. Messersmith
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
- Department of Materials Science and EngineeringUniversity of California, BerkeleyBerkeleyCA94720USA
- Materials Sciences DivisionLawrence Berkeley National LaboratoryBerkeleyCA94720USA
| |
Collapse
|
24
|
Manda V, Pavelka J, Lau E. Proteomics applications in next generation induced pluripotent stem cell models. Expert Rev Proteomics 2024; 21:217-228. [PMID: 38511670 PMCID: PMC11065590 DOI: 10.1080/14789450.2024.2334033] [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] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Induced pluripotent stem (iPS) cell technology has transformed biomedical research. New opportunities now exist to create new organoids, microtissues, and body-on-a-chip systems for basic biology investigations and clinical translations. AREAS COVERED We discuss the utility of proteomics for attaining an unbiased view into protein expression changes during iPS cell differentiation, cell maturation, and tissue generation. The ability to discover cell-type specific protein markers during the differentiation and maturation of iPS-derived cells has led to new strategies to improve cell production yield and fidelity. In parallel, proteomic characterization of iPS-derived organoids is helping to realize the goal of bridging in vitro and in vivo systems. EXPERT OPINIONS We discuss some current challenges of proteomics in iPS cell research and future directions, including the integration of proteomic and transcriptomic data for systems-level analysis.
Collapse
Affiliation(s)
- Vyshnavi Manda
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jay Pavelka
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Edward Lau
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
25
|
Teshima T. Heterogeneity of mesenchymal stem cells as a limiting factor in their clinical application to inflammatory bowel disease in dogs and cats. Vet J 2024; 304:106090. [PMID: 38417670 DOI: 10.1016/j.tvjl.2024.106090] [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: 10/28/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
Inflammatory bowel disease (IBD) is a major subtype of chronic enteropathies in dogs and cats. Conventional drugs such as immunomodulatory medicines as glucocorticoids and/or other anti-inflammatory are mainly applied for treatment. However, these drugs are not always effective to maintain remission from IBD and are limited by unacceptable side effects. Hence, more effective and safe therapeutic options need to be developed. Mesenchymal stem cells (MSCs) are multipotent stem cells with a self-renewal capacity, and have immunomodulatory, anti-inflammatory, anti-fibrotic, and tissue repair properties. Therefore, the application of MSCs as an alternative therapy for IBD has great potential in veterinary medicine. The efficacy of adipose tissue-derived MSC (ADSC) therapy for IBD in dogs and cats has been reported, including numerous studies in animal models. However, treatment outcomes in clinical trials of human IBD patients have not been consistent with preclinical studies. MSC-based therapy for various diseases has received widespread attention, but various problems in such therapy remain, among which no consensus has been reached on the preparation and treatment procedures for MSCs, and cellular heterogeneity of MSCs may be an issue. This review describes the current status of ADSC therapy for canine and feline IBD and summarizes the cellular heterogeneity of canine ADSCs, to highlight the necessity for further reduction or elimination of MSCs heterogeneity and standardization of MSC-based therapies.
Collapse
Affiliation(s)
- Takahiro Teshima
- Laboratory of Veterinary Internal Medicine, School of Veterinary Medicine, Faculty of Veterinary Science, Japan; Research Center for Animal Life Science, Nippon Veterinary and Life Science University, Musashino, Tokyo 180-8602, Japan.
| |
Collapse
|
26
|
Thandar M, Yang X, Zhu Y, Zhang X, Chen Z, Huang S, Chi P. Dysbiosis of gut microbiota and metabolites is associated with radiation-induced colorectal fibrosis and is restored by adipose-derived mesenchymal stem cell therapy. Life Sci 2024; 341:122502. [PMID: 38350495 DOI: 10.1016/j.lfs.2024.122502] [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: 11/22/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/15/2024]
Abstract
AIMS This study aimed to investigate the effects of adipose-derived mesenchymal stem cells (ADSCs) on radiation-induced colorectal fibrosis (RICF) along with the associated dysbiosis of gut microbiota and metabolites. MAIN METHODS Fecal microbiota were assessed through 16S rRNA gene sequencing, and the fecal metabolome was characterized using liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry. The correlation between microbiota and metabolome data was explored. KEY FINDINGS ADSC injection demonstrated a significant restoration of radiation-induced intestinal damage in vivo. At the phylum level, irradiated rats exhibited an increase in Bacteroidota and Campilobacterota, and a decrease in Firmicutes and Desulfobacterota, contrasting with the ADSC treatment group. Metabolomic analysis revealed 72 differently expressed metabolites (DEMs) from gas chromatography-mass spectrometry and 284 DEMs from liquid chromatography-mass spectrometry in the radiation group compared to the blank group. In the ADSC treatment group versus the radiation group, 36 DEMs from gas chromatography-mass spectrometry and 341 DEMs from liquid chromatography-mass spectrometry were identified. KEGG enrichment analysis implicated pathways such as steroid hormone biosynthesis, gap junction, primary bile acid biosynthesis, citrate cycle, cAMP signaling pathway, and alanine, aspartate, and glutamate metabolism during RICF progression and after treated with ADSCs. Correlation analysis highlighted the role of ADSCs in modulating the metabolic process of Camelledionol in fecal Bacteroides. SIGNIFICANCE These findings underscore the potential of ADSCs in reversing dysbiosis and restoring normal colonic flora in the context of RICF, offering valuable insights for therapeutic interventions targeting radiation-induced complications.
Collapse
Affiliation(s)
- Mya Thandar
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China
| | - Xiaojie Yang
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China; Department of Thoracic Surgery, Third Affiliated Hospital of Chongqing Medical University, Chongqing 401100, China
| | - Yuanchang Zhu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China
| | - Xueying Zhang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China
| | - Zhifen Chen
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Training Center of Minimally Invasive Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China.
| | - Shenghui Huang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Training Center of Minimally Invasive Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China.
| | - Pan Chi
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China; Training Center of Minimally Invasive Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China.
| |
Collapse
|
27
|
Cai X, Li Y, Gao F, Muhammad B, Yang H. Therapeutic effect and study of human umbilical cord blood mononuclear cells in patients with ischaemic bowel disease. Sci Rep 2024; 14:6121. [PMID: 38480861 PMCID: PMC10937724 DOI: 10.1038/s41598-024-56720-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 03/17/2024] Open
Abstract
Ischaemic bowel disease (ICBD) is a group of intestinal ischaemia syndromes caused by various aetiologies of reduced intestinal blood flow or vascular occlusion. ICBD can present as abdominal pain, bloody stool, and diarrhoea. This disease often occurs in middle-aged and elderly individuals with cardiovascular and cerebrovascular diseases. The incidence of ischaemic bowel disease has been increasing for decades, and it is difficult to diagnose, resulting in rapid disease progression and a high mortality rate. Therefore, fully understanding this disease, improving the diagnosis rate of this disease, and finding appropriate treatment methods are urgently needed to improve the condition and prognosis of patients. Umbilical cord blood stem cells are accessible, have weak immunogenicity, and have various biological functions, such as angiogenesis, inflammation and immune regulation. Many studies have confirmed that cord blood stem cells can relieve ischaemia, and these cells have attracted tremendous amounts of attention in regenerative medicine in recent years. In this paper, we discuss the clinical characteristics of ICBD, analyse the characteristics of human umbilical cord blood mononuclear cells (HUCB-MNCs), and use its to treat ischaemic bowel disease. Additionally, we compare the clinical manifestations and related indicators before and after treatment to evaluate the efficacy and safety of these methods.
Collapse
Affiliation(s)
- Xiaoxiao Cai
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238 Jingshi East Road, Jinan, Shandong, China
- Graduate Department of Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, Shandong, China
| | - Yonghao Li
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238 Jingshi East Road, Jinan, Shandong, China
- Graduate Department of Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, Shandong, China
| | - Fengyu Gao
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238 Jingshi East Road, Jinan, Shandong, China
| | - Bilal Muhammad
- Graduate Department of Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, Shandong, China
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766 Jingshi Road, Jinan, Shandong, China
| | - Hongli Yang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766 Jingshi Road, Jinan, Shandong, China.
| |
Collapse
|
28
|
Saadh MJ, Mohamed AH, Almoyad MAA, Allela OQB, Amin AH, Malquisto AA, Jin WT, Sârbu I, AlShamsi F, Elsaid FG, Akhavan-Sigari R. Dual role of mesenchymal stem/stromal cells and their cell-free extracellular vesicles in colorectal cancer. Cell Biochem Funct 2024; 42:e3962. [PMID: 38491792 DOI: 10.1002/cbf.3962] [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: 12/30/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 03/18/2024]
Abstract
Colorectal cancer (CRC) is one of the main causes of cancer-related deaths. However, the surgical control of the CRC progression is difficult, and in most cases, the metastasis leads to cancer-related mortality. Mesenchymal stem/stromal cells (MSCs) with potential translational applications in regenerative medicine have been widely researched for several years. MSCs could affect tumor development through secreting exosomes. The beneficial properties of stem cells are attributed to their cell-cell interactions as well as the secretion of paracrine factors in the tissue microenvironment. For several years, exosomes have been used as a cell-free therapy to regulate the fate of tumor cells in a tumor microenvironment. This review discusses the recent advances and current understanding of assessing MSC-derived exosomes for possible cell-free therapy in CRC.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, Jordan
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Asma'a H Mohamed
- Biomedical Engineering Department, College of Engineering and Technologies, Al-Mustaqbal University, Babil, Hilla, Iraq
| | - Muhammad Ali Abdullah Almoyad
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Mushait, Saudi Arabia
| | | | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - April Ann Malquisto
- Abuyog Community College, Abuyog Leyte, Philippines
- ESL Science Teacher, Tacloban City, Tacloban, Philippines
- Department of Art Sciences and Education, Tacloban City, Philippines
| | - Wong Tze Jin
- Department of Science and Technology, Faculty of Humanities, Management and Science, Universiti Putra Malaysia Bintulu Campus, Sarawak, Malaysia
- Institute for Mathematical Research, Universiti Putra Malaysia, Selangor, Malaysia
| | - Ioan Sârbu
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, Romania
| | - Faisal AlShamsi
- Dubai Health Authority, Primary Health Care Department, Dubai, United Arab Emirates
| | - Fahmy Gad Elsaid
- Biology Department, College of Science, King Khalid University, Asir, Abha, Al-Faraa, Saudi Arabia
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
29
|
Bu X, Gao Y, Pan W, Liu L, Wang J, Yin Z, Ping B. Human Amniotic Membrane-Derived Mesenchymal Stem Cells Prevent Acute Graft-Versus-Host Disease in an Intestinal Microbiome-Dependent Manner. Transplant Cell Ther 2024; 30:189.e1-189.e13. [PMID: 37939900 DOI: 10.1016/j.jtct.2023.11.005] [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: 08/13/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Acute graft-versus-host disease (aGVHD) represents a fatal severe complication after allogeneic hematopoietic stem cell transplantation. As a promising cell therapeutic strategy of aGVHD, the mechanism of mesenchymal stem cells (MSC) to ameliorate aGVHD has not been fully clarified, especially in the field of intestinal homeostasis including the intestinal microbiome involved in the pathogenesis of aGVHD. The present study aimed to explore the effect of MSC on intestinal homeostasis including the intestinal barrier and intestinal microbiome and its metabolites, as well as the role of intestinal microbiome in the preventive process of hAMSCs ameliorating aGVHD. The preventive effects of human amniotic membrane-derived MSC (hAMSCs) was assessed in humanized aGVHD mouse models. Immunohistochemistry and RT-qPCR were used to evaluate intestinal barrier function. The 16S rRNA sequencing and targeted metabolomics assay were performed to observe the alternation of intestinal microbiome and the amounts of medium-chain fatty acids (MCFAs) and short-chain fatty acids (SCFAs), respectively. Flow cytometry was performed to analyze the frequencies of T immune cells. Through animal experiments, we found that hAMSCs had the potential to prevent aGVHD. HAMSCs could repair the damage of intestinal barrier structure and function, as well as improve the dysbiosis of intestinal microbiome induced by aGVHD, and meanwhile, upregulate the concentration of metabolites SCFAs, so as to reshape intestinal homeostasis. Gut microbiota depletion and fecal microbial transplantation confirmed the involvement of intestinal microbiome in the preventive process of hAMSCs on aGVHD. Our findings showed that hAMSCs prevented aGVHD in an intestinal microbiome-dependent manner, which might shed light on a new mechanism of hAMSCs inhibiting aGVHD and promote the development of new prophylaxis regimes for aGVHD prevention.
Collapse
Affiliation(s)
- Xiaoyin Bu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Hematology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ya Gao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weifeng Pan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhao Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Baohong Ping
- Department of Hematology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
30
|
Zhao M, Liu S, Wang Y, Lv K, Lou P, Zhou P, Zhu J, Li L, Cheng J, Lu Y, Liu J. The mitochondria‒paraspeckle axis regulates the survival of transplanted stem cells under oxidative stress conditions. Theranostics 2024; 14:1517-1533. [PMID: 38389853 PMCID: PMC10879866 DOI: 10.7150/thno.88764] [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: 08/02/2023] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Rationale: Stem cell-based therapies have emerged as promising tools for tissue engineering and regenerative medicine, but their therapeutic efficacy is largely limited by the oxidative stress-induced loss of transplanted cells at injured tissue sites. To address this issue, we aimed to explore the underlying mechanism and protective strategy of ROS-induced MSC loss. Methods: Changes in TFAM (mitochondrial transcription factor A) signaling, mitochondrial function, DNA damage, apoptosis and senescence in MSCs under oxidative stress conditions were assessed using real-time PCR, western blotting and RNA sequencing, etc. The impact of TFAM or lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) knockdown or overexpression on mitochondrial function, DNA damage repair, apoptosis and senescence in MSCs was also analyzed. The effect of mitochondrion-targeted antioxidant (Mito-TEMPO) on the survival of transplanted MSCs was evaluated in a mouse model of renal ischemia/reperfusion (I/R) injury. Results: Mitochondrial ROS (mtROS) bursts caused defects in TFAM signaling and overall mitochondrial function, which further impaired NEAT1 expression and its mediated paraspeckle formation and DNA repair pathways in MSCs, thereby jointly promoting MSC senescence and death under oxidative stress. In contrast, targeted inhibition of the mtROS bursts is a sufficient strategy for attenuating early transplanted MSC loss at injured tissue sites, and coadministration of Mito-TEMPO improved the local retention of transplanted MSCs and reduced oxidative injury in ischemic kidneys. Conclusions: This study identified the critical role of the mitochondria‒paraspeckle axis in regulating cell survival and may provide insights into developing advanced stem cell therapies for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Meng Zhao
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Shuyun Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yizhuo Wang
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Lv
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peng Lou
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pingya Zhou
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaying Zhu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Lan Li
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingqiu Cheng
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanrong Lu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Department of General Surgery and NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
31
|
Huang P, Sun R, Xu C, Jiang Z, Zuo M, Li Y, Liu R, Gong P, Han Y, Fang J, Li P, Shao C, Shi Y. Glucocorticoid activates STAT3 and NF-κB synergistically with inflammatory cytokines to enhance the anti-inflammatory factor TSG6 expression in mesenchymal stem/stromal cells. Cell Death Dis 2024; 15:70. [PMID: 38238297 PMCID: PMC10796730 DOI: 10.1038/s41419-024-06430-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024]
Abstract
Glucocorticoid (GC) is essential for maintaining immune homeostasis. While GC is known to regulate the expression of genes related to inflammation in immune cells, the effects of GC, especially in the presence of inflammation, on non-immune cells remain largely unexplored. In particular, the impact of GC on inflammatory cytokine-induced immune modulatory responses of tissue stromal cells is unknown, though it has been widely used to modulate tissue injuries. Here we found that GC could enhance the expression of TSG6, a vital tissue repair effector molecule, in IFNγ and TNFα treated human umbilical cord (UC)-MSCs. NF-κB activation was found to be required for GC-augmented TSG6 upregulation. STAT3, but not STAT1, was also found to be required for the TSG6 upregulation in MSCs exposed to IFNγ, TNFα and GC. Moreover, the phosphorylation (activation) of STAT3 was attenuated when NF-κB was knocked down. Importantly, human UC-MSCs pretreated with a cocktail containing GC, IFNγ, and TNFα could significantly enhance the therapeutic effect of human UC-MSCs in an acute lung injury mouse model, as reflected by reduced infiltration of immune cells and down-regulation of iNOS in macrophages in the lung. Together, the findings reveal a novel link between GR, NF-κB and STAT3 in regulating the immunomodulatory and regenerative properties of MSCs, providing novel information for the understanding and treatment of inflammatory conditions.
Collapse
Affiliation(s)
- Peiqing Huang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Rongrong Sun
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Chenchang Xu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Zixuan Jiang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Muqiu Zuo
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Yinghong Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Pixia Gong
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Yuyi Han
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Peishan Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China.
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China.
| |
Collapse
|
32
|
Kim H, Lee SK, Hong S, Park TS, Kim J, Kim S, Kim TM. Pan PPAR agonist stimulation of induced MSCs produces extracellular vesicles with enhanced renoprotective effect for acute kidney injury. Stem Cell Res Ther 2024; 15:9. [PMID: 38167146 PMCID: PMC10763307 DOI: 10.1186/s13287-023-03577-0] [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: 06/01/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) has a complex pathophysiology and imposes serious health concerns worldwide. Extracellular vesicles (EVs) derived from induced mesenchymal stem cells (iMSCs) have been recognized as novel cell-free therapeutics for various inflammatory and degenerative disorders. In this study, we investigated whether iMSCs stimulated with a pan-peroxisome proliferator-activated receptor (PPAR) agonist could enhance the therapeutic efficacy of EVs against AKI. METHODS Human iMSCs were primed with or without lanifibranor, a PPAR agonist for 24 h, and EVs were collected after an additional 24 h. The basic characteristics of EVs were evaluated using cryo-transmission electron microscopy imaging, immunoblot detection of EV markers, nanoparticle tracking analysis, and localization in AKI kidneys. In vitro, the potential of the EVs to promote the growth and survival of HK-2 cells undergoing cisplatin-induced apoptosis and anti-inflammatory effects in M1-polarized THP-1 was compared. Subsequently, AKI was induced in BALB/c mice using cisplatin. After 8 and 24 h of cisplatin treatment, iMSC-EVs or pan-PPAR-iMSC-EVs were injected intravascularly. At 96 h after cisplatin administration, the renoprotective effects of iMSC-EVs or pan-PPAR-iMSC-EVs in inhibiting inflammation and apoptosis were compared using serum biochemistry, histology, immunohistochemistry, and gene expression analysis by qPCR. RESULTS Both EV types expressed EV markers and had typical EV morphology, and their localization in the renal tissue was confirmed. The proliferation and survival of HK-2 cells were higher in pan-PPAR-iMSC-EVs than those in iMSC-EVs. In M1-polarized THP-1 cells, the reduction in the mRNA expression of inflammatory cytokines was more significant in pan-PPAR-iMSC-EVs than that in iMSC-EVs. In the mouse model of cisplatin-induced AKI, pan-PPAR-iMSC-EVs markedly enhanced renoprotective effects compared to iMSC-EVs. Specifically, pan-PPAR-iMSC-EVs reduced tissue inflammation, immune cell infiltration, and apoptosis. Pan-PPAR-iMSC-EVs also increased renal capillary density. CONCLUSION Priming iMSCs with a PPAR agonist significantly improved the therapeutic potential of EVs by reducing inflammation and apoptosis. The reported strategy may contribute to the development of a novel cell-free option for AKI treatment. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Hongduk Kim
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do, 25354, South Korea
| | - Seul Ki Lee
- Brexogen Research Center, Brexogen Inc., Songpa-gu, Seoul, 05855, South Korea
| | - Sungok Hong
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do, 25354, South Korea
| | - Tae Sub Park
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do, 25354, South Korea
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang, Gangwon-do, 25354, South Korea
| | - Jimin Kim
- Brexogen Research Center, Brexogen Inc., Songpa-gu, Seoul, 05855, South Korea
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Songpa-gu, Seoul, 05855, South Korea
| | - Tae Min Kim
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do, 25354, South Korea.
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang Daero 1447, Pyeongchang, Gangwon-do, 25354, South Korea.
| |
Collapse
|
33
|
Liu A, Li C, Wang C, Liang X, Zhang X. Impact of Mesenchymal Stem Cells on the Gut Microbiota and Microbiota Associated Functions in Inflammatory Bowel Disease: A Systematic Review of Preclinical Evidence on Animal Models. Curr Stem Cell Res Ther 2024; 19:981-992. [PMID: 37817517 DOI: 10.2174/011574888x250413230920051715] [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: 03/07/2023] [Revised: 07/25/2023] [Accepted: 08/11/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a global health problem in which gut microbiota dysbiosis plays a pivotal pathogenic role. Mesenchymal stem cells (MSCs) therapy has shown promising application prospects for its powerful immune regulation and tissue repair ability. Recent experimental data suggest that MSCs also regulate the composition of gut microbiota. The current review analyzed, for the first time, the research data linking MSCs and gut microbiota modulation in IBD models aiming at assessing the role of gut microbiota in MSCs repair of IBD. METHODS A comprehensive and structured literature search was performed up to January 2023 on the PubMed, Web of Science, and Scopus databases. The quality and risk of bias assessment followed the PRISMA guidelines and SYRCLE's tool. RESULTS A total of nine pre-clinical studies on animal models were included. Although the dose and route of MSCs applied were quite heterogeneous, results showed that MSCs displayed protective effects on intestinal inflammation, including mice general assessment, immunoregulation, and intestinal barrier integrity. Meanwhile, studies showed positive effects on the composition of gut flora with MSCs administration, which had been characterized by restoration of Firmicutes/ Bacteroides balance and reduction of Proteobacteria. The beneficial bacteria Akkermansia, Bifidobacterium, and Lactobacillus were also distinctly enriched, and the pathogenic bacteria Escherichia-Shigella was conversely decreased. The alpha and beta diversity were also regulated to resemble those of healthy mice. Microbial metabolic functions, such as biosynthesis of secondary bile acid and sphingolipid metabolism, and some biological behaviors related to cell regeneration were also up-regulated, while cancer function and poorly characterized cellular function were down-regulated. CONCLUSION Current data support the remodeling effect on gut microbiota with MSC administration, which provides a potential therapeutic mechanism for MSCs in the treatment of IBD. Additional studies in humans and animal models are warranted to further confirm the role of gut microflora in MSCs repairing IBD.
Collapse
Affiliation(s)
- Airu Liu
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Chenyang Li
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Chen Wang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Xiaonan Liang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Xiaolan Zhang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| |
Collapse
|
34
|
Jingyi L, Lin W, Yuan C, Lingling Z, Qianqian J, Anlong X, Yansong G. Intravenous transplantation of bone marrow-derived mesenchymal stem cells improved behavioral deficits and altered fecal microbiota composition of BTBR mice. Life Sci 2024; 336:122330. [PMID: 38065352 DOI: 10.1016/j.lfs.2023.122330] [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: 10/01/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
AIMS It is recognized that autism spectrum disorder (ASD) is a highly complex neurodevelopmental disorder with communication deficits as well as multiple social barriers. The core symptoms of ASD are not treatable with current therapeutics. Therefore, finding new treatment strategies for ASD is urgently needed. Mesenchymal stem cells (MSC) have been shown to be a promising therapeutic approach in previous studies. However, the underlying mechanisms of MSC treatment for ASD through gut microbiota remain unclear and require further investigation. MAIN METHODS BTBR mice were used as ASD model and then randomly assigned to the human bone marrow-derived mesenchymal stem cell (hBMMSC) intravenous treatment group or vehicle treatment group. C57BL/6J (C57) mice served as control. Multiple social behavioral tests were performed during the 6-week period and fecal samples were collected at different time points for 16 s rRNA sequencing analysis. KEY FINDINGS The administration of hBMMSC improved social deficits of BTBR mice in the open field test (OFT), light-dark box test (LBT), novel object recognition (NOR), and free social test (FST), while also significantly reducing stereotypic behaviors. Additionally, hBMMSC administration notably reversed the alterations of microbiota abundance in BTBR mice, particularly the Firmicutes/Bacteroidetes ratio. Several specific differential taxa were further selected and showed a correlation with the prognosis and behavioral scores of ASD. SIGNIFICANCE Overall, intravenous treatment with hBMMSC had a beneficial impact on ASD by ameliorating social deficits and modifying microbiota compositions. This outcome indicates that hBMMSC intravenous transplantation could be a promising therapeutic strategy for enhancing ASD symptoms improvements.
Collapse
Affiliation(s)
- Li Jingyi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Wang Lin
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Chen Yuan
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Zhang Lingling
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Jiang Qianqian
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China
| | - Xu Anlong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.
| | - Gao Yansong
- Jiuzhitang Maker (Beijing) Cell Technology Co., LTD., Beijing 102600, China; Hunan Provincial Key Laboratory of Critical Quality Attribute of Cell Therapy Products, Changsha, Hunan Province, China.
| |
Collapse
|
35
|
He T, Zhou B, Sun G, Yan Q, Lin S, Ma G, Yao Q, Wu X, Zhong Y, Gan D, Huo S, Jin W, Chen D, Bai X, Cheng T, Cao H, Xiao G. The bone-liver interaction modulates immune and hematopoietic function through Pinch-Cxcl12-Mbl2 pathway. Cell Death Differ 2024; 31:90-105. [PMID: 38062244 PMCID: PMC10781991 DOI: 10.1038/s41418-023-01243-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 01/12/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are used to treat infectious and immune diseases and disorders; however, its mechanism(s) remain incompletely defined. Here we find that bone marrow stromal cells (BMSCs) lacking Pinch1/2 proteins display dramatically reduced ability to suppress lipopolysaccharide (LPS)-induced acute lung injury and dextran sulfate sodium (DSS)-induced inflammatory bowel disease in mice. Prx1-Cre; Pinch1f/f; Pinch2-/- transgenic mice have severe defects in both immune and hematopoietic functions, resulting in premature death, which can be restored by intravenous injection of wild-type BMSCs. Single cell sequencing analyses reveal dramatic alterations in subpopulations of the BMSCs in Pinch mutant mice. Pinch loss in Prx1+ cells blocks differentiation and maturation of hematopoietic cells in the bone marrow and increases production of pro-inflammatory cytokines TNF-α and IL-1β in monocytes. We find that Pinch is critical for expression of Cxcl12 in BMSCs; reduced production of Cxcl12 protein from Pinch-deficient BMSCs reduces expression of the Mbl2 complement in hepatocytes, thus impairing the innate immunity and thereby contributing to infection and death. Administration of recombinant Mbl2 protein restores the lethality induced by Pinch loss in mice. Collectively, we demonstrate that the novel Pinch-Cxcl12-Mbl2 signaling pathway promotes the interactions between bone and liver to modulate immunity and hematopoiesis and may provide a useful therapeutic target for immune and infectious diseases.
Collapse
Affiliation(s)
- Tailin He
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Bo Zhou
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Guohuan Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin, China
| | - Qinnan Yan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Sixiong Lin
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Donghao Gan
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Shaochuan Huo
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Wenfei Jin
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin, China.
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China.
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China.
| |
Collapse
|
36
|
Fu Y, Zhang C, Xie H, Wu Z, Tao Y, Wang Z, Gu M, Wei P, Lin S, Li R, He Y, Sheng J, Xu J, Wang J, Pan Y. Human umbilical cord mesenchymal stem cells alleviated TNBS-induced colitis in mice by restoring the balance of intestinal microbes and immunoregulation. Life Sci 2023; 334:122189. [PMID: 37865178 DOI: 10.1016/j.lfs.2023.122189] [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: 05/01/2023] [Revised: 09/07/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
AIMS Human umbilical cord mesenchymal stem cells (HUMSCs) have been documented to be effective for several immune disorders including inflammatory bowel diseases (IBD). However, it remains unclear how HUMSCs function in regulating immune responses and intestinal flora in the trinitrobenzene sulfonic acid (TNBS)-induced IBD model. MATERIALS AND METHODS We assessed the regulatory effects of HUMSCs on the gut microbiota, T lymphocyte subpopulations and related immune cytokines in the TNBS-induced IBD model. The mice were divided into the normal, TNBS, and HUMSC-treated groups. The effect of HUMSCs was evaluated by Hematoxylin and Eosin (H&E) staining, fluorescence-activated cell sorting (FACS), and enzyme-linked immunosorbent assay (ELISA) analyses. Metagenomics Illumina sequencing was conducted for fecal samples. KEY FINDINGS We demonstrated that the disease symptoms and pathological changes in the colon tissues of TNBS-induced colitis mice were dramatically ameliorated by HUMSCs, which improved the gut microbiota and rebalanced the immune system, increasing the abundance of healthy bacteria (such as Lactobacillus murinus and Lactobacillus johnsonii), the Firmicutes/Bacteroidetes ratio, and the proportion of Tregs; the Th1/Th17 ratio was decreased. Consistently, the expression levels of IFN-γ and IL-17 were significantly decreased, and transforming growth factor-β1 (TGF-β1) levels were significantly increased in the plasma of colitis mice HUMSC injection. SIGNIFICANCE Our experiment revealed that HUMSCs mitigate acute colitis by regulating the rebalance of Th1/Th17/Treg cells and related cytokines and remodeling the gut microbiota, providing potential future therapeutic targets in IBD.
Collapse
Affiliation(s)
- Yanxia Fu
- Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, China
| | - Chen Zhang
- Chinese PLA General Hospital and Medical School, Beijing 100853, China; Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Hui Xie
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Zisheng Wu
- Chinese PLA General Hospital and Medical School, Beijing 100853, China
| | - Yurong Tao
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Ziyu Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Meng Gu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Panjian Wei
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Shuye Lin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Ruoran Li
- Chinese PLA General Hospital and Medical School, Beijing 100853, China
| | - Yuqi He
- Department of Gastroenterology, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Jianqiu Sheng
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Junfeng Xu
- Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China.
| |
Collapse
|
37
|
Tsai ET, Peng SY, Wu YR, Lin TC, Chen CY, Liu YH, Tseng YH, Hsiao YJ, Tseng HC, Lai WY, Lin YY, Yang YP, Chiou SH, Chen SP, Chien Y. HLA-Homozygous iPSC-Derived Mesenchymal Stem Cells Rescue Rotenone-Induced Experimental Leber's Hereditary Optic Neuropathy-like Models In Vitro and In Vivo. Cells 2023; 12:2617. [PMID: 37998352 PMCID: PMC10670753 DOI: 10.3390/cells12222617] [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: 09/23/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) hold promise for cell-based therapy, yet the sourcing, quality, and invasive methods of MSCs impede their mass production and quality control. Induced pluripotent stem cell (iPSC)-derived MSCs (iMSCs) can be infinitely expanded, providing advantages over conventional MSCs in terms of meeting unmet clinical demands. METHODS The potential of MSC therapy for Leber's hereditary optic neuropathy (LHON) remains uncertain. In this study, we used HLA-homozygous induced pluripotent stem cells to generate iMSCs using a defined protocol, and we examined their therapeutic potential in rotenone-induced LHON-like models in vitro and in vivo. RESULTS The iMSCs did not cause any tumorigenic incidence or inflammation-related lesions after intravitreal transplantation, and they remained viable for at least nine days in the mouse recipient's eyes. In addition, iMSCs exhibited significant efficacy in safeguarding retinal ganglion cells (RGCs) from rotenone-induced cytotoxicity in vitro, and they ameliorated CGL+IPL layer thinning and RGC loss in vivo. Optical coherence tomography (OCT) and an electroretinogram demonstrated that iMSCs not only prevented RGC loss and impairments to the retinal architecture, but they also improved retinal electrophysiology performance. CONCLUSION The generation of iMSCs via the HLA homozygosity of iPSCs offers a compelling avenue for overcoming the current limitations of MSC-based therapies. The results underscore the potential of iMSCs when addressing retinal disorders, and they highlight their clinical significance, offering renewed hope for individuals affected by LHON and other inherited retinal conditions.
Collapse
Affiliation(s)
- En-Tung Tsai
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan; (E.-T.T.)
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Shih-Yuan Peng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - You-Ren Wu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Tai-Chi Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chih-Ying Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Yu-Hao Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Yu-Hsin Tseng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Yu-Jer Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Huan-Chin Tseng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Yi-Ying Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan; (E.-T.T.)
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Genomic Research Center, Academia Sinica, Taipei 115024, Taiwan
| | - Shih-Pin Chen
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan; (E.-T.T.)
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (S.-Y.P.); (Y.-R.W.); (Y.-H.L.); (Y.-J.H.); (Y.-Y.L.); (Y.-P.Y.)
| |
Collapse
|
38
|
Li C, Rui Q, Dong X, Ning S, Zhou J, Wu H, Jiang C, Cui Y, Liu J, Jiang J, Qin L. Human amnion-derived mesenchymal stem cells improve subclinical hypothyroidism by immunocompetence mediating apoptosis inhibition on thyroid cells in aged mice. Cell Tissue Res 2023; 394:309-323. [PMID: 37572164 PMCID: PMC10638193 DOI: 10.1007/s00441-023-03822-1] [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: 11/30/2022] [Accepted: 07/27/2023] [Indexed: 08/14/2023]
Abstract
Subclinical hypothyroidism (SCH) affects 10% of the global population, which is most prevalent in women and the elderly. However, it remains debatable whether the elderly with subclinical hypothyroidism needs thyroxine supplement. Human amnion-derived mesenchymal stem cells (hAMSCs) could play important roles in autoimmune diseases, suggesting that hAMSC be a candidate to regulate the thyroid function of female age-related subclinical hypothyroidism. Herein, we established the model of SCH in the aged female mice. This study was designed to investigate whether human amnion-derived mesenchymal stem cells (hAMSC) could effect on immune regulation, apoptosis inhibition of thyroid cells, thyroid function, blood lipid levels, and heart function. In addition, qualified hAMSCs were intravenously injected into aged female SCH mice via the tail vein on day 0 and day 10. The levels of thyroid hormone and blood lipids as well as cardiac function, serum immunological indexes, and apoptosis of thyroid cells were then analyzed on day 5, 10, 15, and 20; meanwhile, the quantity of Th1, Th2, Th17, and Treg immune cells in peripheral blood was evaluated before and on day 20 post-injection. Our study demonstrated that after hAMSC transplantation, the thyroid functions, blood lipid levels, and heart function indexes of age-related SCH (AR-SCH) mice were significantly improved. Consistent with this, Th1 and Treg cells increased significantly, while Th2 and Th17 cells decreased in peripheral blood. Apoptosis was also suppressed in the thyroid cells. In summary, hAMSC delivery can potentially be a safe and effective therapy for treating SCH in the elderly, improving related complications by immunomodulatory and apoptosis inhibition.
Collapse
Affiliation(s)
- Chuyu Li
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Qiang Rui
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210000, China
| | - Xiaohan Dong
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Song Ning
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Jing Zhou
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Huimin Wu
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Chunyan Jiang
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Jun Jiang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
| | - Lianju Qin
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
| |
Collapse
|
39
|
Kim J, Lee SK, Jeong SY, You H, Han SD, Park S, Kim S, Kim TM. Multifaceted action of stem cell-derived extracellular vesicles for nonalcoholic steatohepatitis. J Control Release 2023; 364:S0168-3659(23)00706-X. [PMID: 39491172 DOI: 10.1016/j.jconrel.2023.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) is a chronic liver disease associated with metabolic syndrome. Extracellular vesicles (EVs) are essential signaling mediators containing functional biomolecules. EVs are secreted from various cell types, and recent studies have shown that mesenchymal stem cell-derived EVs have therapeutic potential against immune and metabolic diseases. In this study, we investigated whether EVs from induced mesenchymal stem cells (iMSC-EVs) regulate AMPK signaling and lipid metabolism using cell-based studies and two different mouse models of NASH (methionine/choline-deficient diet-induced and ob/ob mice). Protein analysis revealed that iMSC-EVs carry cargo proteins with the potential to regulate lipid metabolism. iMSC-EVs inhibited free fatty acid release from adipose tissues by downregulating the activity of lipolytic genes in NASH. In addition, iMSC-EVs improved hepatic steatosis by modulating AMPK signaling, which plays essential role in metabolic homeostasis in the liver. Moreover, iMSC-EVs reduced CD36 expression, contributing to the blockade of free fatty acid transport to the liver of NASH mice. Finally, iMSC-EVs reduced inflammation, endoplasmic reticulum stress, and apoptosis while promoting hepatic regeneration of the NASH liver. In conclusion, iMSC-EVs can potentially serve as cell-free therapeutics for NASH owing to their multifaceted modality.
Collapse
Affiliation(s)
- Jimin Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Seul Ki Lee
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Seon-Yeong Jeong
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Haedeun You
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Sang-Deok Han
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Somi Park
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Songpa-Gu, Seoul 05855, South Korea
| | - Tae Min Kim
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea; Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-do 25354, South Korea.
| |
Collapse
|
40
|
Jiang H, Tang Q, Zheng D, Gu Y, Man C. Parathyroid hormone enhances the therapeutic effect of mesenchymal stem cells on temporomandibular joint osteoarthritis in rats. AMERICAN JOURNAL OF STEM CELLS 2023; 12:73-82. [PMID: 38021454 PMCID: PMC10658131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/12/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVES Temporomandibular joint osteoarthritis (TMJOA) is a degenerative disease affecting the joint, which is characterized by injury to the articular cartilage, as well as changes in the synovial and subchondral bone. TMJOA has a high incidence rate, without any effective treatment. Despite the therapeutic potential of mesenchymal stem cells (MSCs) in various diseases, their efficacy in treating TMJOA is constrained by the local hypoxic conditions and elevated reactive oxygen species (ROS) environment within the damaged temporomandibular joint. In recent years, many studies have reported that parathyroid hormone (PTH) can effectively treat TMJOA, and has an important impact on MSC differentiation. Therefore, we hypothesized that PTH may influence the potential of MSCs, thereby improving their therapeutic effect on TMJOA. METHODS First, we isolated and cultured rat bone marrow MSCs, and evaluated their proliferation and differentiation after adding PTH. Next, the in vitro environment of hypoxia and high ROS was established by hypoxia condition and H2O2 treatment, and the resistance of PTH-treated MSCs to hypoxia and ROS was subsequently investigated. Finally, PTH-treated MSCs were used to treat TMJOA in a rat model to evaluate the efficacy of PTH. RESULTS PTH enhanced the proliferation ability of MSCs, promoted the osteogenic differentiation of MSCs, and improved the tolerance of MSCs to hypoxia and ROS. Finally, the therapeutic effect of PTH-treated MSCs on TMJOA was significantly improved. CONCLUSION PTH enhances the therapeutic effect of MSCs on TMJOA in rats.
Collapse
Affiliation(s)
- Haitao Jiang
- Department of Oral and Maxillofacial Trauma and Orthognathic Surgery, Stomatological Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Qiuyu Tang
- Honghuagang District Stomatological Hospital of Zunyi CityZunyi, Guizhou, China
| | - Dexin Zheng
- Department of Oral and Maxillofacial Trauma and Orthognathic Surgery, Stomatological Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Yunkai Gu
- Department of Stomatology, Affiliated Hospital of Jiangnan UniversityWuxi, Jiangsu, China
| | - Cheng Man
- Department of Oral and Maxillofacial Trauma and Orthognathic Surgery, Stomatological Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| |
Collapse
|
41
|
O’Reilly C, Mills S, Rea MC, Lavelle A, Ghosh S, Hill C, Ross RP. Interplay between inflammatory bowel disease therapeutics and the gut microbiome reveals opportunities for novel treatment approaches. MICROBIOME RESEARCH REPORTS 2023; 2:35. [PMID: 37849974 PMCID: PMC7615213 DOI: 10.20517/mrr.2023.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023]
Abstract
Inflammatory bowel disease (IBD) is a complex heterogeneous disorder defined by recurring chronic inflammation of the gastrointestinal tract, attributed to a combination of factors including genetic susceptibility, altered immune response, a shift in microbial composition/microbial insults (infection/exposure), and environmental influences. Therapeutics generally used to treat IBD mainly focus on the immune response and include non-specific anti-inflammatory and immunosuppressive therapeutics and targeted therapeutics aimed at specific components of the immune system. Other therapies include exclusive enteral nutrition and emerging stem cell therapies. However, in recent years, scientists have begun to examine the interplay between these therapeutics and the gut microbiome, and we present this information here. Many of these therapeutics are associated with alterations to gut microbiome composition and functionality, often driving it toward a "healthier profile" and preclinical studies have revealed that such alterations can play an important role in therapeutic efficacy. The gut microbiome can also improve or hinder IBD therapeutic efficacy or generate undesirable metabolites. For certain IBD therapeutics, the microbiome composition, particularly before treatment, may serve as a biomarker of therapeutic efficacy. Utilising this information and manipulating the interactions between the gut microbiome and IBD therapeutics may enhance treatment outcomes in the future and bring about new opportunities for personalised, precision medicine.
Collapse
Affiliation(s)
- Catherine O’Reilly
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61C996, Ireland
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
- Authors contributed equally
| | - Susan Mills
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
- Authors contributed equally
| | - Mary C. Rea
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61C996, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Subrata Ghosh
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Colin Hill
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - R. Paul Ross
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| |
Collapse
|
42
|
Liu A, Liang X, Wang W, Wang C, Song J, Guo J, Sun D, Wang D, Song M, Qian J, Zhang X. Human umbilical cord mesenchymal stem cells ameliorate colon inflammation via modulation of gut microbiota-SCFAs-immune axis. Stem Cell Res Ther 2023; 14:271. [PMID: 37749611 PMCID: PMC10521524 DOI: 10.1186/s13287-023-03471-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a global health problem in which gut microbiota dysbiosis plays a pivotal pathogenic role. Mesenchymal stem cells (MSCs) therapy has emerged as a prospective novel tool for managing IBD, and which can also regulate the composition of gut microbiota. However, the functional significance of MSCs-induced changes in gut microbiome is poorly understood. METHODS Here, we investigated for the first time the role of gut microbiota in mediating the protective effect of human umbilical cord MSCs (HUMSCs) on DSS-induced colitis. Gut microbiota alteration and short-chain fatty acids (SCFAs) production were analyzed through 16S rRNA sequencing and targeted metabolomics. Spectrum antibiotic cocktail (ABX), fecal microbiota transplantation (FMT) and sterile fecal filtrate (SFF) were employed to evaluate the protective effect of intestinal flora and its metabolites. Cytokine microarray, Enzyme-linked immunosorbent assay (ELISA), and flow cytometry were conducted to assess the effect on CD4+T homeostasis. RESULTS Here, we investigated for the first time the role of gut microbiota in mediating the protective effect of MSCs on DSS-induced colitis. By performing gut microbiota depletion and fecal microbiota transplantation (FMT) experiments, we revealed that MSCs derived from human umbilical cord ameliorated colon inflammation and reshaped T-cells immune homeostasis via remodeling the composition and diversity of gut flora, especially up-regulated SCFAs-producing bacterial abundance, such as Akkermansia, Faecalibaculum, and Clostridia_UCG_014. Consistently, targeted metabolomics manifested the increased SCFAs production with MSCs administration, and there was also a significant positive correlation between differential bacteria and SCFAs. Meanwhile, combined with sterile fecal filtrate (SFF) gavage experiments, the underlying protective mechanism was further associated with the improved Treg/Th2/Th17 balance in intestinal mucosa mediated via the increased microbiota-derived SCFAs production. CONCLUSION The present study advances understanding of MSCs in the protective effects on colitis, providing evidence for the new role of the microbiome-metabolite-immune axis in the recovery of colitis by MSCs.
Collapse
Affiliation(s)
- Airu Liu
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Xiaonan Liang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Wenxin Wang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Chen Wang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Jia Song
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Jinbo Guo
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Donglei Sun
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Dong Wang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Mei Song
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Jiaming Qian
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China.
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing City, China.
| | - Xiaolan Zhang
- Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang City, China.
| |
Collapse
|
43
|
Arakawa M, Sakamoto Y, Miyagawa Y, Nito C, Takahashi S, Nitahara-Kasahara Y, Suda S, Yamazaki Y, Sakai M, Kimura K, Okada T. iPSC-derived mesenchymal stem cells attenuate cerebral ischemia-reperfusion injury by inhibiting inflammatory signaling and oxidative stress. Mol Ther Methods Clin Dev 2023; 30:333-349. [PMID: 37637385 PMCID: PMC10448333 DOI: 10.1016/j.omtm.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/11/2023] [Indexed: 08/29/2023]
Abstract
Induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs) hold great promise as a cell source for transplantation into injured tissues to alleviate inflammation. However, the therapeutic efficacy of iMSC transplantation for ischemic stroke remains unknown. In this study, we evaluated the therapeutic effects of iMSC transplantation on brain injury after ischemia-reperfusion using a rat transient middle cerebral artery occlusion model and compared its therapeutic efficacy with that of bone marrow mesenchymal stem cells (BMMSCs). We showed that iMSCs and BMMSCs reduced infarct volumes after reperfusion and significantly improved motor function on days 3, 7, 14, 28, and 56 and cognitive function on days 28 and 56 after reperfusion compared with the vehicle group. Furthermore, immunological analyses revealed that transplantation of iMSCs and BMMSCs inhibited microglial activation and expression of proinflammatory cytokines and suppressed oxidative stress and neuronal cell death in the cerebral cortex at the ischemic border zone. No difference in therapeutic effect was observed between the iMSC and BMMSC groups. Taken together, our results demonstrate that iMSC therapy can be a practical alternative as a cell source for attenuation of brain injury and improvement of neurological function because of the unlimited supply of uniform therapeutic cells.
Collapse
Affiliation(s)
- Masafumi Arakawa
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yuki Sakamoto
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yoshitaka Miyagawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Laboratory for Clinical Research, Collaborative Research Center, Nippon Medical School, Tokyo, Japan
| | - Shiro Takahashi
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yuko Nitahara-Kasahara
- Division of Molecular and Medical Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yoshiyuki Yamazaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Mashito Sakai
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kazumi Kimura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
44
|
Yang J, Li D, Zhang M, Lin G, Hu S, Xu H. From the updated landscape of the emerging biologics for IBDs treatment to the new delivery systems. J Control Release 2023; 361:568-591. [PMID: 37572962 DOI: 10.1016/j.jconrel.2023.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/14/2023]
Abstract
Inflammatory bowel diseases (IBDs) treatments have shifted from small-molecular therapeutics to the oncoming biologics. The first-line biologics against the moderate-to-severe IBDs are mainly involved in antibodies against integrins, cytokines and cell adhesion molecules. Besides, other biologics including growth factors, antioxidative enzyme, anti-inflammatory peptides, nucleic acids, stem cells and probiotics have also been explored at preclinical or clinical studies. Biologics with variety of origins have their unique potentials in attenuating immune inflammation or gut mucosa healing. Great advances in use of biologics for IBDs treatments have been archived in recent years. But delivering issues for biologic have also been confronted due to their liable nature. In this review, we will focus on biologics for IBDs treatments in the recent publications; summarize the current landscapes of biologics and their promise to control disease progress. Alternatively, the confronted challenges for delivering biologics will also be analyzed. To combat these drawbacks, some new delivering strategies are provided: firstly, designing the functional materials with high affinity toward biologics; secondly, the delivering vehicle systems to encapsulate the liable biologics; thirdly, the topical adhering delivery systems as enema. To our knowledge, this review is the first study to summarize the updated usage of the oncoming biologics for IBDs, their confronted challenges in term of delivery and the potential combating strategies.
Collapse
Affiliation(s)
- Jiaojiao Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Dingwei Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Mengjiao Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Gaolong Lin
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
| | - Helin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| |
Collapse
|
45
|
Bu X, Pan W, Wang J, Liu L, Yin Z, Jin H, Liu Q, Zheng L, Sun H, Gao Y, Ping B. Therapeutic Effects of HLA-G5 Overexpressing hAMSCs on aGVHD After Allo-HSCT: Involving in the Gut Microbiota at the Intestinal Barrier. J Inflamm Res 2023; 16:3669-3685. [PMID: 37645691 PMCID: PMC10461746 DOI: 10.2147/jir.s420747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
Background Acute graft-versus-host disease (aGVHD) initiated by intestinal barrier dysfunction and gut microbiota dysbiosis, remains one of the main obstacles for patients undergoing allogenic hematopoietic stem cell transplantation (allo-HSCT) to achieve good prognosis. Studies have suggested that mesenchymal stem cells (MSCs) can suppress immune responses and reduce inflammation, and human leukocyte antigen-G5 (HLA-G5) plays an important role in the immunomodulatory effects of MSCs, but very little is known about the potential mechanisms in aGVHD. Thus, we explored the effect of HLA-G5 on the immunosuppressive properties of human amnion MSCs (hAMSCs) and demonstrated its mechanism related to the gut microbiota at the intestinal barrier in aGVHD. Methods Patients undergoing allo-HSCT were enrolled to detect the levels of plasma-soluble HLA-G (sHLA-G) and regulatory T cells (Tregs). Humanized aGVHD mouse models were established and treated with hAMSCs or HLA-G5 overexpressing hAMSCs (ov-HLA-G5-hAMSCs) to explore the mechanism of HLA-G5 mediated immunosuppressive properties of hAMSCs and the effect of ov-HLA-G5-hAMSCs on the gut microbiota at the intestinal barrier in aGVHD. Results The plasma levels of sHLA-G on day +30 after allo-HSCT in aGVHD patients were lower than those in patients without aGVHD, and the sHLA-G levels were positively correlated with Tregs percentages. ov-HLA-G5-hAMSCs had the potential to inhibit the expansion of CD3+CD4+ T and CD3+CD8+ T cells and promote Tregs differentiation, suppress proinflammatory cytokine secretion but promote anti-inflammatory cytokines release. Besides, ov-HLA-G5-hAMSCs also could reverse the intestinal barrier dysfunction and gut microbiota dysbiosis in aGVHD. Conclusion We demonstrated that HLA-G might work with Tregs to create a regulatory network together to reduce the occurrence of aGVHD. HLA-G5 mediated hAMSCs to exert higher immunosuppressive properties in vivo and reverse the immune imbalance caused by T lymphocytes and cytokines. Furthermore, HLA-G5 overexpressing hAMSCs could restore gut microbiota and intestinal barriers, thereby ameliorating aGVHD.
Collapse
Affiliation(s)
- Xiaoyin Bu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Hematology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Weifeng Pan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Junhui Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Liping Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Zhao Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Hua Jin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Haitao Sun
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| | - Ya Gao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Baohong Ping
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Department of Hematology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| |
Collapse
|
46
|
Yang F, Zheng X, Liang W, Ni B, Lu J, Liu Q, Xu R, He Y, Yee Waye MM, Zhang Q, Chen Y, Zou X, Chen W. Short-Term Clinical Response and Changes in the Fecal Microbiota and Metabolite Levels in Patients with Crohn's Disease After Stem Cell Infusions. Stem Cells Transl Med 2023; 12:497-509. [PMID: 37399531 PMCID: PMC10427961 DOI: 10.1093/stcltm/szad036] [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: 02/09/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023] Open
Abstract
Recent studies have shown a close relationship between the gut microbiota and Crohn's disease (CD). This study aimed to determine whether mesenchymal stem cell (MSC) treatment alters the gut microbiota and fecal metabolite pathways and to establish the relationship between the gut microbiota and fecal metabolites. Patients with refractory CD were enrolled and received 8 intravenous infusions of MSCs at a dose of 1.0 × 106 cells/kg. The MSC efficacy and safety were evaluated. Fecal samples were collected, and their microbiomes were analyzed by 16S rDNA sequencing. The fecal metabolites at baseline and after 4 and 8 MSC infusions were identified by liquid chromatography-mass spectrometry (LC--MS). A bioinformatics analysis was conducted using the sequencing data. No serious adverse effects were observed. The clinical symptoms and signs of patients with CD were substantially relieved after 8 MSC infusions, as revealed by changes in weight, the CD activity index (CDAI) score, C-reactive protein (CRP) level, and erythrocyte sedimentation rate (ESR). Endoscopic improvement was observed in 2 patients. A comparison of the gut microbiome after 8 MSC treatments with that at baseline showed that the genus Cetobacterium was significantly enriched. Linoleic acid was depleted after 8 MSC treatments. A possible link between the altered Cetobacterium abundance and linoleic acid metabolite levels was observed in patients with CD who received MSCs. This study enabled an understanding of both the gut microbiota response and bacterial metabolites to obtain more information about host-gut microbiota metabolic interactions in the short-term response to MSC treatment.
Collapse
Affiliation(s)
- Fan Yang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Infectious Diseases, The First People’s Hospital of Kashi, The Kashi Affiliated Hospital, Sun Yat-Sen University, Kashi, People’s Republic of China
- Postdoctoral Research Station, Xinjiang Medical University, Ürümqi, People’s Republic of China
| | - Xiaofang Zheng
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Weicheng Liang
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Beibei Ni
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Jianxi Lu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Qiuli Liu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ruixuan Xu
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yizhan He
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Mary Miu Yee Waye
- The Nethersole School of Nursing, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Qi Zhang
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Infectious Diseases, The First People’s Hospital of Kashi, The Kashi Affiliated Hospital, Sun Yat-Sen University, Kashi, People’s Republic of China
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yufeng Chen
- Department of Colorectal Surgery & Department of General Surgery & Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xiaoguang Zou
- Department of Infectious Diseases, The First People’s Hospital of Kashi, The Kashi Affiliated Hospital, Sun Yat-Sen University, Kashi, People’s Republic of China
| | - Wenjie Chen
- Biotherapy Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Department of Infectious Diseases, The First People’s Hospital of Kashi, The Kashi Affiliated Hospital, Sun Yat-Sen University, Kashi, People’s Republic of China
- Cell-Gene Therapy Translational Medicine Research Centre, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People’s Republic of China
- NMPA Key Laboratory for Quality Research and Evaluation of Cell Products, Guangzhou, People’s Republic of China
| |
Collapse
|
47
|
Chiou SH, Ong HKA, Chou SJ, Aldoghachi AF, Loh JK, Verusingam ND, Yang YP, Chien Y. Current trends and promising clinical utility of IPSC-derived MSC (iMSC). PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:131-154. [PMID: 37678969 DOI: 10.1016/bs.pmbts.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Mesenchymal stem cells (MSCs) differentiated from human induced pluripotent stem cells (iPSC) or induced MSC (iMSCs) are expected to address issues of scalability and safety as well as the difficulty in producing homogenous clinical grade MSCs as demonstrated by the promising outcomes from preclinical and clinical trials, currently ongoing. The assessment of iMSCs based in vitro and in vivo studies have thus far showed more superior performance as compared to that of the primary or native human MSCs, in terms of cell proliferation, expansion capacity, immunomodulation properties as well as the influence of paracrine signaling and exosomal influence in cell-cell interaction. In this chapter, an overview of current well-established methods in generating a sustainable source of iMSCs involving well defined culture media is discussed followed by the properties of iMSC as compared to that of MSC and its promising prospects for continuous development into potential clinical grade applications.
Collapse
Affiliation(s)
- Shih-Hwa Chiou
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veteran General Hospital, Taipei, Taiwan
| | - Han Kiat Alan Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
| | - Shih-Jie Chou
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veteran General Hospital, Taipei, Taiwan
| | - A F Aldoghachi
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
| | - Jit Kai Loh
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
| | - Nalini Devi Verusingam
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veteran General Hospital, Taipei, Taiwan.
| | - Yueh Chien
- Department of Medical Research, Taipei Veteran General Hospital, Taipei, Taiwan
| |
Collapse
|
48
|
Chen Y, Meng W, Ren G, An N, Zhang J, Liu Z, Wu X, Yin W, Hu X, Liu Z, Feng F, Chen Y. NLRP3 inflammasome inhibition of OP9 cells enhance therapy for inflammatory bowel disease. Heliyon 2023; 9:e18038. [PMID: 37483815 PMCID: PMC10362138 DOI: 10.1016/j.heliyon.2023.e18038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/25/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are becoming more popular in therapy. Therefore, in-depth studies on mesenchymal stem cells in therapy are urgently needed. However, the difficulty in culturing and propagating MSCs in vitro complicates potential studies on MSCs in a murine model. OP9 cells are a stromal cell line from mouse bone marrow, which have similar characteristics and functions to MSCs and can maintain their original characteristics. Because of these properties, OP9 cells have become a suitable substitute for research on MSCs. Previously, we have found that MSCs can cure inflammatory bowel disease in mice. In this study, we aimed to investigate whether OP9 cells can functionally regulate and alleviate inflammatory diseases. We evaluated the therapeutic effect of OP9 cells in the mouse model of inflammatory bowel disease and found OP9 cells were able to ameliorate inflammatory bowel disease. We explored the existence of NLRP3 inflammasome in OP9 cells, and showed better therapeutic effects when the NLRP3 inflammasome was suppressed. Thus, OP9 cell line is similar to MSCs in characteristic and function, and is an ideal substitute for MSCs research. The preliminary exploration of the inflammasome system in OP9 cells lays a theoretical and methodological foundation for further study of MSCs.
Collapse
Affiliation(s)
- Yutong Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Weicheng Meng
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Guangming Ren
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Ning An
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Zhang
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhixin Liu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoshuang Wu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen Yin
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xingbin Hu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zheng Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Fan Feng
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Yaozhen Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
49
|
Tian CM, Zhang Y, Yang MF, Xu HM, Zhu MZ, Yao J, Wang LS, Liang YJ, Li DF. Stem Cell Therapy in Inflammatory Bowel Disease: A Review of Achievements and Challenges. J Inflamm Res 2023; 16:2089-2119. [PMID: 37215379 PMCID: PMC10199681 DOI: 10.2147/jir.s400447] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/03/2023] [Indexed: 05/24/2023] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is a group of chronic inflammatory diseases of the gastrointestinal tract. Repeated inflammation can lead to complications, such as intestinal fistula, obstruction, perforation, and bleeding. Unfortunately, achieving durable remission and mucosal healing (MH) with current treatments is difficult. Stem cells (SCs) have the potential to modulate immunity, suppress inflammation, and have anti-apoptotic and pro-angiogenic effects, making them an ideal therapeutic strategy to target chronic inflammation and intestinal damage in IBD. In recent years, hematopoietic stem cells (HSCs) and adult mesenchymal stem cells (MSCs) have shown efficacy in treating IBD. In addition, numerous clinical trials have evaluated the efficiency of MSCs in treating the disease. This review summarizes the current research progress on the safety and efficacy of SC-based therapy for IBD in both preclinical models and clinical trials. We discuss potential mechanisms of SC therapy, including tissue repair, paracrine effects, and the promotion of angiogenesis, immune regulation, and anti-inflammatory effects. We also summarize current SC engineering strategies aimed at enhancing the immunosuppressive and regenerative capabilities of SCs for treating intestinal diseases. Additionally, we highlight current limitations and future perspectives of SC-related therapy for IBD.
Collapse
Affiliation(s)
- Cheng-Mei Tian
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People’s Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
50
|
Aldoghachi AF, Loh JK, Wang ML, Yang YP, Chien CS, Teh HX, Omar AH, Cheong SK, Yeap SK, Ho WY, Ong AHK. Current developments and therapeutic potentials of exosomes from induced pluripotent stem cells-derived mesenchymal stem cells. J Chin Med Assoc 2023; 86:356-365. [PMID: 36762931 DOI: 10.1097/jcma.0000000000000899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells derived from adult human tissues that have the ability to proliferate in vitro and maintain their multipotency, making them attractive cell sources for regenerative medicine. However, MSCs reportedly show limited proliferative capacity with inconsistent therapeutic outcomes due to their heterogeneous nature. On the other hand, induced pluripotent stem cells (iPSC) have emerged as an alternative source for the production of various specialized cell types via their ability to differentiate from all three primary germ layers, leading to applications in regenerative medicine, disease modeling, and drug therapy. Notably, iPSCs can differentiate into MSCs in monolayer, commonly referred to as induced mesenchymal stem cells (iMSCs). These cells show superior therapeutic qualities compared with adult MSCs as the applications of the latter are restricted by passage number and autoimmune rejection when applied in tissue regeneration trials. Furthermore, increasing evidence shows that the therapeutic properties of stem cells are a consequence of the paracrine effects mediated by their secretome such as from exosomes, a type of extracellular vesicle secreted by most cell types. Several studies that investigated the potential of exosomes in regenerative medicine and therapy have revealed promising results. Therefore, this review focuses on the recent findings of exosomes secreted from iMSCs as a potential noncell-based therapy.
Collapse
Affiliation(s)
- Ahmed Faris Aldoghachi
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
| | - Jit-Kai Loh
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Mong-Lien Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ping Yang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chian-Shiu Chien
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hui Xin Teh
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
| | - Alfaqih Hussain Omar
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, Malaysia
| | - Soon-Keng Cheong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
- National Cancer Council (MAKNA), Kuala Lumpur, Malaysia
| | - Swee Keong Yeap
- Marine Biotechnology, China-ASEAN College of Marine Sciences, Xiamen University Malaysia Campus, Jalan Sunsuria, Bandar Sunsuria, Sepang, Selangor, Malaysia
| | - Wan Yong Ho
- Faculty of Sciences and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Alan Han-Kiat Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Cheras, Malaysia
| |
Collapse
|