1
|
Goyal A, Afzal M, Khan NH, Goyal K, Srinivasamurthy SK, Gupta G, Benod Kumar K, Ali H, Rana M, Wong LS, Kumarasamy V, Subramaniyan V. Targeting p53-p21 signaling to enhance mesenchymal stem cell regenerative potential. Regen Ther 2025; 29:352-363. [PMID: 40248767 PMCID: PMC12004386 DOI: 10.1016/j.reth.2025.03.007] [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: 01/26/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are properties of self-renewal and differentiation potentials and thus are very appealing to regenerative medicine. Nevertheless, their therapeutic potential is frequently constrained by senescence, limited proliferation, and stress-induced apoptosis. The key role of the p53-p21 biology in MSC biology resides in safeguarding genomic stability while promoting senescence and limiting regenerative capacity upon over-activation demonstrated. This pathway is a key point for improving MSC function and exploiting the inherent limitations. Recent advances indicate that senescence can be delayed by targeting the p53-p21 signaling and improved MSC proliferation and differentiation capacity. PFT-α pharmacological agents transiently inhibit p53 from increasing proliferation and lineage-specific differentiation, while antioxidants such as hydrogen-rich saline and epigallocatechin 3 gallate (EGCG) suppress oxidative stress and attenuate p53 p21 signaling. Genetic tools like CRISPR-Cas9 and RNA interference also precisely modulate TP53 and CDKN1A expression to optimize MSC functionality. The interplay of p53-p21 with pathways like Wnt/β-catenin and MAPK further highlights opportunities for combinatorial therapies to enhance MSC resilience and regenerative outcomes. This review aims to offer a holistic view of how p53-p21 targeting can further the regenerative potential of MSCs, resolving senescence, proliferation, and stress resilience towards advanced therapeutics built on MSCs.
Collapse
Affiliation(s)
- Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical & Health Sciences University, P.O. Box 11172, Ras Al Khaimah, United Arab Emirates
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - K. Benod Kumar
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
2
|
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
|
3
|
Ding Y, Wang J, Zheng X, Chen Y, Zhu F, Lin F, Ma K, Liang X, Han S. Mixed lymphocyte reaction-conditioned MSC-derived extracellular vesicles enhance graft survival via miR-638-mediated immunoregulation. Stem Cells Transl Med 2025; 14:szaf009. [PMID: 40261200 PMCID: PMC12012893 DOI: 10.1093/stcltm/szaf009] [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: 11/26/2024] [Accepted: 02/08/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) require priming by proinflammatory stimuli for optimal immunosuppressive effects. Our previous work identified mixed lymphocyte reaction-conditioned medium (MLR-CdM) as a potent enhancer of MSC immunosuppressive properties. This study evaluates the immunomodulatory potential of MSC-derived extracellular vesicles preconditioned with MLR-CdM (MSC-EVMLR) compared to IFN-γ (MSC-EVIFN), focusing on key miRNAs and mechanisms involved. METHODS We assessed the ability of MSC-EVMLR and MSC-EVIFN to modulate lymphocyte proliferation and cytokine expression in vitro. To identify potential effector molecules within MSC-EVMLR, we performed miRNA array analysis combined with dose-response experiments using MLR-CdM under varying stimulation conditions. We used a murine allogeneic heterotopic heart transplantation model to investigate the impact of MSC-EVMLR on graft survival and its immunomodulatory effects. RESULTS MSC-EVMLR outperformed MSC-EVIFN in suppressing lymphocyte proliferation and steering cytokine expression toward an anti-inflammatory profile in vitro. Through miRNA array analysis and dose-response experiments with MLR-CdM, miR-638 was identified as a potential effector molecule in MSC-EVMLR. In vivo study demonstrated that MSC-EVMLR significantly prolonged graft survival, which was associated with a marked decreased proinflammatory cytokines IL6 and IFN-γ and increase in regulatory T cells (Tregs) and within the transplanted heart tissue. These effect was significantly reduced upon miR-638 knockdown. Additionally, the miR-638/Fosb axis was identified as a key pathway that promoted Treg differentiation and induced immune tolerance. CONCLUSIONS Preconditioning MSCs with MLR-CdM, a blend of inflammatory stimuli, potentiates the immunoregulatory capacity of MSC-EV beyond the effects of IFN-γ stimulation alone. This study advances the understanding of MSC-EV-based therapies in transplantation.
Collapse
Affiliation(s)
- Yue Ding
- Department of Organ Transplantation, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), 200003 Shanghai, People’s Republic of China
| | - Jiyuan Wang
- Department of Organ Transplantation, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), 200003 Shanghai, People’s Republic of China
| | - Xueyang Zheng
- Department of Organ Transplantation, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), 200003 Shanghai, People’s Republic of China
| | - Yu Chen
- Department of Organ Transplantation, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), 200003 Shanghai, People’s Republic of China
| | - Fanyuan Zhu
- Department of Organ Transplantation, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), 200003 Shanghai, People’s Republic of China
| | - Fang Lin
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, People’s Republic of China
| | - Kexin Ma
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, People’s Republic of China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, People’s Republic of China
- Shanghai East Hospital, Jinzhou Medical University, 121001 Jinzhou, People’s Republic of China
| | - Xiaoting Liang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, People’s Republic of China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, People’s Republic of China
| | - Shu Han
- Department of Organ Transplantation, Shanghai Changzheng Hospital (Second Affiliated Hospital of Naval Medical University), 200003 Shanghai, People’s Republic of China
| |
Collapse
|
4
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
5
|
Liang Z, Xie H, Wu D. Immune mediated inflammatory diseases: moving from targeted biologic therapy, stem cell therapy to targeted cell therapy. Front Immunol 2025; 16:1520063. [PMID: 40260258 PMCID: PMC12009864 DOI: 10.3389/fimmu.2025.1520063] [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: 10/30/2024] [Accepted: 03/13/2025] [Indexed: 04/23/2025] Open
Abstract
Despite the advancements in targeted biologic therapy for immune-mediated inflammatory diseases (IMIDs), significant challenges persist, including challenges in drug maintenance, primary and secondary non-responses, and adverse effects. Recent data have strengthened the evidence supporting stem cell therapy as an experimental salvage therapy into a standard treatment option. Recent preclinical and clinical studies suggested that chimeric antigen receptor T cell (CAR-T) therapy, which depleting tissue and bone marrow B cells, may lead to improvement, even inducing long-lasting remissions for patients with IMIDs. In this review, we address the unmet needs of targeted biologic therapy, delineate the critical differences between stem cell transplantation and CAR-T therapy, evaluate the current status of CAR-T therapy for IMIDs and explore its potential and existing limitations.
Collapse
Affiliation(s)
- Zhenguo Liang
- Department of Rheumatology and Immunology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Hui Xie
- Department of Clinical Research and Development, Antengene Corporation, Shanghai, China
| | - Dongze Wu
- Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Grignano MA, Pisani S, Gregorini M, Rainaudo G, Avanzini MA, Croce S, Valsecchi C, Ceccarelli G, Islami T, Margiotta E, Portalupi V, De Mauri A, Stea ED, Pattonieri EF, Iadarola P, Viglio S, Conti B, Rampino T. Engineered ATP-Loaded Extracellular Vesicles Derived from Mesenchymal Stromal Cells: A Novel Strategy to Counteract Cell ATP Depletion in an In Vitro Model. Int J Mol Sci 2025; 26:3424. [PMID: 40244293 PMCID: PMC11990007 DOI: 10.3390/ijms26073424] [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: 03/03/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
The use of adenosine triphosphate (ATP) has shown promising effects in alleviating ischemic damage across various tissues. However, the penetration of ATP into kidney tubular cells presents a challenge due to their unique anatomical and physiological properties. In this study, we introduce a novel bioinspired drug delivery system utilizing extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) and engineered to carry ATP. ATP-loaded liposomes (ATP-LPs) and ATP-loaded EVs (ATP-EVs) were prepared using microfluidic technology, followed by characterization of their morphology (DLS, NTA, SEM, TEM), ATP content, and release rate at 37 °C (pH 7.4). Additionally, the delivery efficacy of ATP-LPs and ATP-EVs was evaluated in vitro on renal cells (HK2 cells) under chemically induced ischemia. The results indicated successful ATP enrichment in EVs, with ATP-EVs showing no significant changes in morphology or size compared to naïve EVs. Notably, ATP-EVs demonstrated superior ATP retention compared to ATP-LPs, protecting the ATP from degradation in the extracellular environment. In an ATP-depleted HK2 cell model, only ATP-EVs effectively restored ATP levels, preserving cell viability and reducing apoptotic gene expression (BCL2-BAX). This study is the first to successfully demonstrate the direct delivery of ATP into renal tubular cells in vitro using EVs as carriers.
Collapse
Affiliation(s)
- Maria Antonietta Grignano
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Silvia Pisani
- Department of Drug Sciences, University of Pavia, Viale Torquato Taramelli 12, 27100 Pavia, Italy;
| | - Marilena Gregorini
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| | - Giorgia Rainaudo
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Maria Antonietta Avanzini
- Pediatric Haematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.A.); (C.V.)
- Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Stefania Croce
- Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Chiara Valsecchi
- Pediatric Haematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.A.); (C.V.)
- Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Gabriele Ceccarelli
- Human Anatomy Unit, Department of Public Health, Experimental Medicine and Forensic, University of Pavia, 27100 Pavia, Italy;
- Centre for Health Technologies (CHT), University of Pavia, 27100 Pavia, Italy
| | - Tefik Islami
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Elisabetta Margiotta
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Valentina Portalupi
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Andreana De Mauri
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Emma Diletta Stea
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Eleonora Francesca Pattonieri
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
| | - Paolo Iadarola
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Simona Viglio
- Lung Transplantation Unit, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Viale Torquato Taramelli 12, 27100 Pavia, Italy;
| | - Teresa Rampino
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.A.G.); (M.G.); (G.R.); (T.I.); (E.M.); (V.P.); (A.D.M.); (E.D.S.); (E.F.P.); (T.R.)
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
7
|
Zhao L, Li N, Shi X, Zhang J, Gao M, Wei Y, Li X, Du B, Sun D, Nian H, Wei R. Enhanced Therapeutic Effect of IL-10-ADSCs on Rabbit Autoimmune Dacryoadenitis By Suppressing T Follicular Helper Cell Responses Via miR-142-5p/RC3H1 Axis. Invest Ophthalmol Vis Sci 2025; 66:66. [PMID: 40261659 DOI: 10.1167/iovs.66.4.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025] Open
Abstract
Purpose Mesenchymal stem cells (MSCs) represent a promising therapeutic strategy in clinical research for dry eye, and their immunomodulatory effects can be enhanced through genetic modification. In this study, we constructed interleukin-10 (IL-10) gene-modified adipose-derived MSCs (IL-10-ADSCs) and investigated their protective effects and underlying mechanisms on rabbit autoimmune dacryoadenitis, an animal model of autoimmune dry eye. Methods ADSCs were isolated from rabbit adipose tissue and transduced with IL-10 overexpressing lentivirus. Then the preventive and therapeutic effects of IL-10-ADSCs on rabbit autoimmune dacryoadenitis were evaluated. Flow cytometry and Western blot were performed to assess the immunomodulatory effects of IL-10-ADSCs on T follicular helper (Tfh) cells. Bioinformatic analyses and functional gain and loss assays were used to determine the molecular mechanism underlying the effects of IL-10-ADSCs on Tfh responses. Results We demonstrated that IL-10-ADSCs maintain the cell surface phenotype and multi-differentiation potentials of MSCs. Intravenous injection of IL-10-ADSCs markedly attenuated autoimmune dacryoadenitis, yielding significantly superior clinical and pathological improvements compared to ADSCs. Further investigation revealed that IL-10-ADSCs administration significantly suppressed Tfh cell responses in vivo and in vitro, contributing to reduced inflammation and improved tissue damage. Mechanistically, IL-10-ADSCs exert their suppressive function on Tfh cells partially through the miR-142-5p/RC3H1 axis. Notably, IL-10-ADSCs subconjunctivally administered after disease onset efficiently ameliorated the severity of autoimmune dacryoadenitis. Conclusions IL-10-ADSCs ameliorate autoimmune dacryoadenitis by suppressing Tfh cell responses via suppressing the miR-142-5p/RC3H1 axis. The enhanced therapeutic effects of IL-10-ADSCs could be of significant value in improving the effectiveness of stem cell therapy in autoimmune dry eye.
Collapse
Affiliation(s)
- Lu Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Na Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Xinrui Shi
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Jiachen Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Min Gao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Yankai Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Xuejia Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Bei Du
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
| | - Deming Sun
- Doheny Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, United States
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
- https://orcid.org/0000-0002-9983-3410
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin, China
- https://orcid.org/0000-0002-9708-0355
| |
Collapse
|
8
|
Wiewiórska-Krata N, Foroncewicz B, Mucha K, Zagożdżon R. Cell therapies for immune-mediated disorders. Front Med (Lausanne) 2025; 12:1550527. [PMID: 40206475 PMCID: PMC11980423 DOI: 10.3389/fmed.2025.1550527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 04/11/2025] Open
Abstract
Immune-mediated disorders are a broad range of diseases, arising as consequence of immune defects, exaggerated/misguided immune response or a mixture of both conditions. Their frequency is on a rise in the developed societies and they pose a significant challenge for diagnosis and treatment. Traditional pharmacological, monoclonal antibody-based or polyclonal antibody replacement-based therapies aiming at modulation of the immune responses give very often dissatisfactory results and/or are burdened with unacceptable adverse effects. In recent years, a new group of treatment modalities has emerged, utilizing cells as living drugs, especially with the use of the up-to-date genetic engineering. These modern cellular therapies are designed to offer a high potential for more targeted, safe, durable, and personalized treatment options. This work briefly reviews the latest advances in the treatment of immune-mediated disorders, mainly those related to exaggeration of the immune response, with such cellular therapies as hematopoietic stem cells (HSCs), mesenchymal stromal cells (MSCs), regulatory T cells (Tregs), chimeric antigen receptor (CAR) T cells and others. We highlight the main features of these therapies as new treatment options for taming the dysregulated immune system. Undoubtfully, in near future such therapies can provide lasting remissions in a range of immune-mediated disorders with reduced treatment burden and improved quality of life for the patients.
Collapse
Affiliation(s)
- Natalia Wiewiórska-Krata
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- ProMix Center (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Foroncewicz
- ProMix Center (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Mucha
- ProMix Center (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Radosław Zagożdżon
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
9
|
Xie Y, Chai M, Xing Y, Zhou P, Wei P, Hua H. miRNA let-7f-5p-encapsulated labial gland MSC-derived EVs ameliorate experimental Sjögren's syndrome by suppressing Th17 cells via targeting RORC/IL-17A signaling axis. J Nanobiotechnology 2025; 23:228. [PMID: 40114173 PMCID: PMC11927278 DOI: 10.1186/s12951-025-03308-y] [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: 03/08/2025] [Indexed: 03/22/2025] Open
Abstract
Sjögren's syndrome (SS) is an autoimmune disease primarily affecting salivary glands, with xerostomia as a distinct clinical manifestation. This disease also poses a significantly increased risk of lymphoma, severely impacting patients' quality of life. The imbalance between Th17 and Treg cells plays a critical role in SS progression, driving severe immune dysregulation, chronic inflammation, and escalating tissue dysfunction. However, current clinical treatments for SS still remain limited, and it continues to be recognized as a refractory disease. Therefore, the development of novel and effective therapeutic strategies is a pressing demand in clinical research. In recent years, extracellular vesicle (EV) therapy has emerged as a promising approach for autoimmune disease treatment, showing encouraging outcomes in modulating immune balance and alleviating symptoms. EVs carry diverse cargo, among which microRNAs (miRNAs) are highly abundant and play critical roles. These small RNAs are essential for EV-mediated functions, particularly in regulating gene expression and modulating the immune microenvironment. Our research team first isolated labial gland mesenchymal stem cells (LGMSCs) and their derived EVs (LGMSC-EVs), which offer potential therapeutic advantages in SS due to their salivary gland origin. Then we screened and identified the highly enriched miRNA let-7f-5p as a key regulator through miRNA profiling analysis. To achieve better therapeutic outcomes, we transfected exogenous miRNA let-7f-5p into LGMSC-EVs to upregulate its expression, thereby constructing let-7f-5p-encapsulated LGMSC-EVs. These modified EVs were subsequently tested in an experimental SS mouse model to evaluate their therapeutic potential. The upregulation of miRNA let-7f-5p in LGMSC-EVs significantly enhanced their therapeutic effects, resulting in clinical improvements such as increased salivary flow and reduced lymphocytic infiltration. Mechanistically, let-7f-5p-encapsulated LGMSC-EVs suppressed Th17 cells by directly targeting the 3'-untranslated region (3'UTR) of RORC, inhibiting the RORC/IL-17A signaling axis, and reducing IL-17A production, thereby restoring Th17/Treg balance and promoting an anti-inflammatory profile. Collectively, this let-7f-5p-encapsulated LGMSC-EV therapy offers a promising target-driven approach for the treatment of SS, achieving improved clinical outcomes and immune rebalance after modification with miRNA let-7f-5p, which presents new potential for the clinical treatment of SS.
Collapse
Affiliation(s)
- Yufei Xie
- Department of Oral Medicine, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Maosheng Chai
- Department of Oral Medicine, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yixiao Xing
- Department of Oral Medicine, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- Department of Stomatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peiru Zhou
- Department of Oral Medicine, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Pan Wei
- Department of Oral Medicine, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Hong Hua
- Department of Oral Medicine, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| |
Collapse
|
10
|
Zhao B, Zhang Z, Guo X, Liu X, Lei M, Guo S, Yao Q, Zhang F, Peng T, Liu A, Jiang B, Zhu D. Mesenchymal stem cell-derived exosomes in renal ischemia-reperfusion injury: a new therapeutic strategy. Int Urol Nephrol 2025; 57:875-884. [PMID: 39520637 DOI: 10.1007/s11255-024-04258-4] [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: 08/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Renal ischemia-reperfusion injury (RIRI) is a serious kidney condition that causes significant damage due to lack of blood flow. This injury leads to oxidative stress and inflammation, which can cause acute tubular necrosis and kidney failure. Stem cell-derived exosomes, small vesicles released by stem cells, have shown promise in treating RIRI. Mesenchymal stem cells (MSCs) have been used to mitigate RIRI, and their exosomes have been found to play a crucial role in repairing damaged tissues. This review explores the key roles of exosomes from different sources of MSCs in RIRI, the potential of MSC-derived exosomes in treating this disease, and future research directions.
Collapse
Affiliation(s)
- Bo Zhao
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
- Xianning Central Hospital, First Affiliated Hospital of Hubei University of Science and Technology , 228 Jingui Road, Xian'an District, 437000, Xianning, Hubei Province, People's Republic of China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437000, Xianning, Hubei Province, People's Republic of China
| | - Zhenwang Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Xiying Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Xiufen Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Min Lei
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Qing Yao
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Feixue Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Tie Peng
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Aimei Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China.
| | - Botao Jiang
- Xianning Central Hospital, First Affiliated Hospital of Hubei University of Science and Technology , 228 Jingui Road, Xian'an District, 437000, Xianning, Hubei Province, People's Republic of China.
| | - Dan Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China.
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437000, Xianning, Hubei Province, People's Republic of China.
| |
Collapse
|
11
|
Ruan MF, Yin YH, Shao XD, Qi XS. Bone marrow mesenchymal stem cell transplantation for treatment of liver cirrhosis: Recent advances. Shijie Huaren Xiaohua Zazhi 2025; 33:106-113. [DOI: 10.11569/wcjd.v33.i2.106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025] Open
Abstract
The incidence and mortality of cirrhosis have been increasing year by year, making it become an important factor threatening human health. Cirrhosis in the early stage often lacks obvious symptoms, and liver transplantation has become the only effective treatment for end-stage cirrhosis. However, due to the high cost of liver transplantation, the high incidence of complications, and limited donor resources, the application of liver transplantation has been limited. Currently, stem cell therapy has been considered an alternative treatment for cirrhosis. The sources and types of stem cells are diverse, and autologous bone marrow mesenchymal stem cells (BMSCs) have become one of the important cell types involved in tissue regeneration due to their great differentiation potential, abundant content, and ease of obtaining and preparation. This review summarizes the mechanisms, transplantation routes, safety, effectiveness, cell origin, advantages, and limitations of BMSCs in the treatment of cirrhosis by reviewing the relevant literature.
Collapse
Affiliation(s)
- Meng-Fan Ruan
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Jinzhou Medical University), Shenyang 110840, Liaoning Province, China
- Postgraduate College, Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
| | - Yu-Hang Yin
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Jinzhou Medical University), Shenyang 110840, Liaoning Province, China
- Postgraduate College, China Medical University, Shenyang 110122, Liaoning Province, China
| | - Xiao-Dong Shao
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Jinzhou Medical University), Shenyang 110840, Liaoning Province, China
| | - Xing-Shun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Jinzhou Medical University), Shenyang 110840, Liaoning Province, China
- Postgraduate College, Jinzhou Medical University, Jinzhou 121001, Liaoning Province, China
- Postgraduate College, China Medical University, Shenyang 110122, Liaoning Province, China
| |
Collapse
|
12
|
Shestakova VA, Smirnova EI, Atiakshin DA, Kisel AA, Koryakin SN, Litun EV, Saburov VO, Demyashkin GA, Lagoda TS, Yakimova AO, Kabakov AE, Ignatyuk MA, Yatsenko EM, Kudlay DA, Ivanov SA, Shegay PV, Kaprin AD, Baranovskii DS, Komarova LN, Klabukov ID. Impact of Minimally Manipulated Cell Therapy on Immune Responses in Radiation-Induced Skin Wound Healing. Int J Mol Sci 2025; 26:1994. [PMID: 40076619 PMCID: PMC11900442 DOI: 10.3390/ijms26051994] [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: 12/30/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
The current treatment of radiation-induced skin wounds utilizes mainly conventional therapies, including topical steroids, creams, ointments, and hydrogel dressings, which do not take into account the immunologic changes that occur in the skin after radiation exposure. Therefore, it is relevant to consider alternative therapies and their impact on changes in the immune landscape of the skin. The aim of this study was to investigate the effect of allogeneic minimally manipulated keratinocytes and fibroblasts on rat skin repair and the development of immune responses. We found that the use of cell therapy compared to treatment with syntazone ointment and no treatment resulted in faster healing and a reduction in the size of radiation-induced skin wounds, area of inflammation, and edema. Additionally, in the group receiving the cell therapy application, there was an observed increase in the number of mast cells (MCs), activation of MC interaction with M2 macrophages, a reduction in the direct contact of MCs with the vascular bed, an increase in the content of collagen fibers due to the intensification of collagen fibrillogenesis, and a restoration of their histotopographic organization. Thus, the positive effect of cell therapy based on allogeneic minimally manipulated keratinocytes and fibroblasts on skin regeneration indicated that it can be used in clinical practice to improve the effectiveness of rehabilitation after radiation therapy.
Collapse
Affiliation(s)
- Victoria A. Shestakova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Ekaterina I. Smirnova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Dmitrii A. Atiakshin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anastas A. Kisel
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Sergey N. Koryakin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Evgeniy V. Litun
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Vyacheslav O. Saburov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Grigory A. Demyashkin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Tatyana S. Lagoda
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Anna O. Yakimova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Alexander E. Kabakov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Michael A. Ignatyuk
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Elena M. Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Dmitry A. Kudlay
- Immunology Department, Institute of Immunology FMBA of Russia, 115552 Moscow, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Sergey A. Ivanov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Peter V. Shegay
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Denis S. Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- University Hospital Basel, Basel University, 4001 Basel, Switzerland
- Research and Educational Resource Center for Cellular Technologies, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Lyudmila N. Komarova
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Ilya D. Klabukov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| |
Collapse
|
13
|
Tew VK, Barathan M, Nordin F, Law JX, Ng MH. Emerging Role of Mesenchymal Stromal Cell and Exosome Therapies in Treating Cognitive Impairment. Pharmaceutics 2025; 17:284. [PMID: 40142948 PMCID: PMC11945939 DOI: 10.3390/pharmaceutics17030284] [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: 11/18/2024] [Revised: 12/27/2024] [Accepted: 01/09/2025] [Indexed: 03/28/2025] Open
Abstract
Cognitive aging, characterized by the gradual decline in cognitive functions such as memory, attention, and problem-solving, significantly impacts daily life. This decline is often accelerated by neurodegenerative diseases, particularly Alzheimer's Disease (AD) and Parkinson's Disease (PD). AD is marked by the accumulation of amyloid-beta plaques and tau tangles, whereas PD involves the degeneration of dopaminergic neurons. Both conditions lead to severe cognitive impairment, greatly diminishing the quality of life for affected individuals. Recent advancements in regenerative medicine have highlighted mesenchymal stromal cells (MSCs) and their derived exosomes as promising therapeutic options. MSCs possess regenerative, neuroprotective, and immunomodulatory properties, which can promote neurogenesis, reduce inflammation, and support neuronal health. Exosomes, nanosized vesicles derived from MSCs, provide an efficient means for delivering bioactive molecules across the blood-brain barrier, targeting the underlying pathologies of AD and PD. While these therapies hold great promise, challenges such as variability in MSC sources, optimal dosing, and effective delivery methods need to be addressed for clinical application. The development of robust protocols, along with rigorous clinical trials, is crucial for validating the safety and efficacy of MSC and exosome therapies. Future research should focus on overcoming these barriers, optimizing treatment strategies, and exploring the integration of MSC and exosome therapies with lifestyle interventions. By addressing these challenges, MSC- and exosome-based therapies could offer transformative solutions for improving outcomes and enhancing the quality of life for individuals affected by cognitive aging and neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia (F.N.); (J.X.L.)
| |
Collapse
|
14
|
Rayat Pisheh H, Sani M. Mesenchymal stem cells derived exosomes: a new era in cardiac regeneration. Stem Cell Res Ther 2025; 16:16. [PMID: 39849585 PMCID: PMC11756228 DOI: 10.1186/s13287-024-04123-2] [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: 10/31/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
Despite significant strides in medical treatments and surgical procedures for cardiovascular diseases, these conditions continue to be a major global health concern. The persistent need for innovative therapeutic approaches to mend damaged heart tissue highlights the complexity and urgency of this medical challenge. In recent years, stem cells have emerged as a promising tool for tissue regeneration, but challenges such as graft rejection and tumor formation have limited their clinical application. Exosomes, extracellular vesicles containing a diverse array of biomolecules, have garnered significant attention for their potential in regenerative medicine. The cardioprotective and reparative properties of mesenchymal stem cell-derived exosomes hold promise for the treatment of heart diseases. These exosomes can modulate various cellular processes, including angiogenesis, apoptosis, and inflammation, thereby enhancing cardiac function. Despite the growing interest, there remains a lack of comprehensive reviews synthesizing the molecular mechanisms, preclinical, and clinical evidence related to the specific role of MSC-derived exosomes in cardiac therapies. This review aims to fill that gap by exploring the potential of MSC-derived exosomes as a therapeutic strategy for cardiac diseases. This review explores the potential of mesenchymal stem cell-derived exosomes as a therapeutic strategy for cardiac diseases. We discuss the molecular mechanisms underlying their cardioprotective effects, summarize preclinical and clinical studies investigating their efficacy, and address the challenges and future perspectives of exosome-based therapies. The collective evidence suggests that MSC-derived exosomes hold promise as a novel and effective therapeutic approach for cardiac diseases.
Collapse
Affiliation(s)
- Hossein Rayat Pisheh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
Ginting AR, Munir D, Amin MM, Darlan DM, Putra A, Rusda M, Mutiara E, Mayasari E, Rozi MF. Mesenchymal stem cells for immune modulation in systemic lupus erythematosus: From bench research to clinical applications. NARRA J 2024; 4:e994. [PMID: 39816093 PMCID: PMC11731813 DOI: 10.52225/narra.v4i3.994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/15/2024] [Indexed: 01/18/2025]
Abstract
Systemic lupus erythematosus (SLE) is a prevalent autoimmune disease affecting multiple organ systems. Disease progression is inevitable as part of its natural course, necessitating aggressive therapeutic strategies, particularly with the use of immunosuppressants. Long-term use of steroids and other immunosuppressants is associated with significant adverse effects. Mesenchymal stem cells (MSCs) have been shown to modulate the immune response, leading to immunosuppressive effects against self-antigens. MSCs have demonstrated the ability to modulate several immune cell populations, contributing to favorable outcomes in controlling immune and inflammatory conditions. Recent evidence has shown an increase in Treg and Breg cell subsets following MSC administration, along with modulation of other immune cells, including dendritic cells, B cells, and T cells. However, the balance between MSC pro-inflammatory and anti-inflammatory phenotypic activation remains a critical factor in determining therapeutic outcomes. Various covariates also influence the efficacy of MSC therapy. The aim of this study was to provide a comprehensive overview of the utilization of mesenchymal stem cells (MSCs) in SLE treatment, leveraging their immunomodulatory and immunosuppressive capabilities. Understanding the fundamental preclinical effects of MSCs and recent findings from clinical studies may enhance the potential of MSC therapy in the management of SLE patients.
Collapse
Affiliation(s)
- Andi R. Ginting
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Delfitri Munir
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Pusat Unggulan Iptek Tissue Engineering, Universitas Sumatera Utara, Medan, Indonesia
| | - Mustafa M. Amin
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Psychiatry, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Dewi M. Darlan
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research Center, Semarang, Indonesia
- Faculty of Postgraduate Biomedical Science, Universitas Islam Sultan Agung, Semarang, Indonesia
| | - Muhammad Rusda
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Erna Mutiara
- Faculty of Public Health, Universitas Sumatera Utara, Medan, Indonesia
| | - Evita Mayasari
- Department of Microbiology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad F. Rozi
- Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
16
|
Tripathi T, Mohan S, Alfaifi HA, Farasani A, R R, Sharma P, Sharma A, Koul A, Prasad GVS, Rustagi S, Anand J, Sah S, Gaidhane S, Bushi G, Jena D, Khatib MN, Shabil M, Abdelwahab SI, Bhopte K, Pant M, Mehta R, Pandey S, Brar M, Chilakam N, Balaraman AK. Efficacy and safety of stem cell therapy for fistula management: an overview of existing systematic reviews. Int J Surg 2024; 110:7573-7584. [PMID: 39468970 PMCID: PMC11634089 DOI: 10.1097/js9.0000000000002125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Fistulas, abnormal connections between two anatomical structures, significantly impact the quality of life and can result from a variety of causes, including congenital defects, inflammatory conditions, and surgical complications. Stem cell therapy has emerged as a promising alternative due to its potential for regenerative and immunomodulatory effects. This overview of systematic reviews aimed to assess the safety and efficacy of stem cell therapy in managing fistulas, drawing on the evidence available. METHODS This umbrella review was conducted following the Joanna Briggs Institute (JBI) methodology to assess the efficacy and safety of stem cell therapy for treating various types of fistulas. A comprehensive search was performed across multiple electronic databases including PubMed, Embase, Cochrane Register, and Web of Science up to 5 May 2024. Systematic reviews focusing on stem cell therapy for fistulas were included, with data extracted on study design, stem cell types, administration methods, and outcomes. The quality of the reviews was assessed using the AMSTAR 2 tool, and meta-analyses were conducted using R software version 4.3. RESULTS Nineteen systematic reviews were included in our umbrella review. The stem cell therapy demonstrated by significant improvements in clinical remission rates, with a relative risk (RR) of 1.299 (95% CI: 1.192-1.420). Stem cell therapy enhanced fistula closure rates, both short-term (RR=1.481; 95% CI: 1.036-2.116) and long-term (RR=1.422; 95% CI: 1.091-1.854). The safety analysis revealed no significant increase in the risk of adverse events with stem cell therapy, showing a pooled RR of 0.972 (95% CI: 0.739-1.278) for general adverse events and 1.136 (95% CI: 0.821-1.572) for serious adverse events, both of which indicate a safety profile comparable to control treatments. Re-epithelialization rates also improved (RR=1.44; 95% CI: 1.322-1.572). CONCLUSION Stem cell therapy shows promise as an effective and safe treatment for fistulas, particularly in inducing remission and promoting closure of complex fistulas. The findings advocate for further high-quality research to confirm these benefits and potentially incorporate stem cell therapy into standard clinical practice for fistula management. Future studies should focus on long-term outcomes and refining stem cell treatment protocols to optimize therapeutic efficacy.
Collapse
Affiliation(s)
- Tripti Tripathi
- Department of Physiology, Integral Institute of Medical Sciences and Research, Dashauli, Uttar Pradesh, India
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Hassan A. Alfaifi
- Pharmaceutical Care Administration (Jeddah Second Health Cluster), Ministry of Health, Saudi Arabia
| | - Abdullah Farasani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | | | - Apurva Koul
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, India
| | - G. V. Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Jigisha Anand
- Department of Biotechnology, Graphic Era (Deemed to be University) Clement Town Dehradun, India
- Department of Allied Sciences, Graphic Era Hill University Clement Town Dehradun, India
| | - Sanjit Sah
- Department of Paediatrics, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Shilpa Gaidhane
- One Health Centre, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education, Wardha, India
| | - Ganesh Bushi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Diptismita Jena
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mahalaqua N. Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Muhammed Shabil
- University Center for Research and Development, Chandigarh University, Mohali, Punjab, India
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor, Malaysia
| | | | - Kiran Bhopte
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, India
| | - Manvi Pant
- New Delhi Institute of Management, New Delhi, India
| | - Rachana Mehta
- Clinical Microbiology, RDC, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
- Dr Lal PathLabs - Nepal, Chandol-4, Maharajgunj, Kathmandu, Nepal
| | - Sakshi Pandey
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| | - Manvinder Brar
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, India
| | - Nagavalli Chilakam
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Ashok K. Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor, Malaysia
| |
Collapse
|
17
|
Gelinski Kempe PR, de Castro MV, Coser LDO, Cartarozzi LP, Barraviera B, Ferreira RS, de Oliveira ALR. Combination of Adult Mesenchymal Stem Cell Therapy and Immunomodulation with Dimethyl Fumarate Following Spinal Cord Ventral Root Repair. BIOLOGY 2024; 13:953. [PMID: 39596908 PMCID: PMC11591889 DOI: 10.3390/biology13110953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
Spinal cord injury results in significant motor and sensory loss. In the experimental ventral root avulsion (VRA) model, the ventral (motor) roots are disconnected from the spinal cord surface, disrupting contact between spinal motoneurons and muscle fibers. Axotomized motoneurons typically degenerate within two to three weeks after avulsion, the situation being exacerbated by an increased glial response and chronic inflammation. Nevertheless, root reimplantation has been observed to stimulate regenerative potential in some motoneurons, serving as a model for CNS/PNS regeneration. We hypothesized that a combination of neuroprotective and immunomodulatory therapies is capable of enhancing regenerative responses following nerve root injury and repair. A heterologous fibrin biopolymer (HFB) was used for surgical repair; dimethyl fumarate (DMF) was used for neuroprotection and immunomodulation; and adipose tissue-derived mesenchymal stem cells (AT-MSCs) were used as a source of trophic factors and cytokines that may further enhance neuronal survival. Thus, adult female Lewis rats underwent unilateral VRA of the L4-L6 roots, followed by reimplantation with HFB, AT-MSCs transplantation, and daily DMF treatment for four weeks, with a 12-week postoperative survival period. An evaluation of the results focused on light microscopy, qRT-PCR, and the Catwalk motor function recovery system. Data were analyzed using one-way or two-way ANOVA (p < 0.05). The results indicate that the combined therapy resulted in a reduced glial response and a 70% improvement in behavioral motor recovery. Overall, the data support the potential of combined regenerative approaches after spinal cord root injury.
Collapse
Affiliation(s)
- Paula Regina Gelinski Kempe
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Campinas 13083-865, SP, Brazil; (P.R.G.K.); (M.V.d.C.); (L.d.O.C.); (L.P.C.)
| | - Mateus Vidigal de Castro
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Campinas 13083-865, SP, Brazil; (P.R.G.K.); (M.V.d.C.); (L.d.O.C.); (L.P.C.)
| | - Lilian de Oliveira Coser
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Campinas 13083-865, SP, Brazil; (P.R.G.K.); (M.V.d.C.); (L.d.O.C.); (L.P.C.)
| | - Luciana Politti Cartarozzi
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Campinas 13083-865, SP, Brazil; (P.R.G.K.); (M.V.d.C.); (L.d.O.C.); (L.P.C.)
| | - Benedito Barraviera
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu 01419-901, SP, Brazil; (B.B.); (R.S.F.J.)
- Center for Translational Sciences and Biopharmaceuticals Development, Botucatu 18610-307, SP, Brazil
- Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
| | - Rui Seabra Ferreira
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu 01419-901, SP, Brazil; (B.B.); (R.S.F.J.)
- Center for Translational Sciences and Biopharmaceuticals Development, Botucatu 18610-307, SP, Brazil
| | | |
Collapse
|
18
|
Górska A, Trubalski M, Borowski B, Brachet A, Szymańczyk S, Markiewicz R. Navigating stem cell culture: insights, techniques, challenges, and prospects. Front Cell Dev Biol 2024; 12:1435461. [PMID: 39588275 PMCID: PMC11586186 DOI: 10.3389/fcell.2024.1435461] [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: 06/03/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024] Open
Abstract
Stem cell research holds huge promise for regenerative medicine and disease modeling, making the understanding and optimization of stem cell culture a critical aspect of advancing these therapeutic applications. This comprehensive review provides an in-depth overview of stem cell culture, including general information, contemporary techniques, encountered problems, and future perspectives. The article begins by explaining the fundamental characteristics of various stem cell types, elucidating the importance of proper culture conditions in maintaining pluripotency or lineage commitment. A detailed exploration of established culture techniques sheds light on the evolving landscape of stem cell culture methodologies. Common challenges such as genetic stability, heterogeneity, and differentiation efficiency are thoroughly discussed, with insights into cutting-edge strategies and technologies aimed at addressing these hurdles. Moreover, the article delves into the impact of substrate materials, culture media components, and biophysical cues on stem cell behavior, emphasizing the intricate interplay between the microenvironment and cell fate decisions. As stem cell research advances, ethical considerations and regulatory frameworks become increasingly important, prompting a critical examination of these aspects in the context of culture practices. Lastly, the article explores emerging perspectives, including the integration of artificial intelligence and machine learning in optimizing culture conditions, and the potential applications of stem cell-derived products in personalized medicine. This comprehensive overview aims to serve as a valuable resource for researchers and clinicians, fostering a deeper understanding of stem cell culture and its key role in advancing regenerative medicine and biomedical research.
Collapse
Affiliation(s)
- Aleksandra Górska
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Mateusz Trubalski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Bartosz Borowski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Adam Brachet
- Student Scientific Association, Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| | - Sylwia Szymańczyk
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Renata Markiewicz
- Occupational Therapy Laboratory, Chair of Nursing Development, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
19
|
Harrell CR, Djonov V, Volarevic A, Arsenijevic A, Volarevic V. Molecular Mechanisms Responsible for Mesenchymal Stem Cell-Dependent Attenuation of Tear Hyperosmolarity and Immune Cell-Driven Inflammation in the Eyes of Patients with Dry Eye Disease. Diseases 2024; 12:269. [PMID: 39589943 PMCID: PMC11592843 DOI: 10.3390/diseases12110269] [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: 08/24/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Dry eye disease (DED) is a chronic condition characterized by a decrease in tear production or an increase in tear evaporation, leading to inflammation and damage of the ocular surface. Dysfunction of ion channels, tear hyperosmolarity and immune cell-driven inflammation create a vicious circle responsible for the pathological changes in the eyes of DED patients. Mesenchymal stem cells (MSCs) are adult, rapidly proliferating stem cells that produce a large number of immunoregulatory, angiomodulatory, and growth factors that efficiently reduce tear hyperosmolarity-induced pathological changes, inhibit harmful immune response, and provide trophic support to the injured corneal and conjuctival epithelial cells, goblet cells and acinar cells in lacrimal glands of DED patients. METHODS An extensive research in the literature was implemented in order to elucidate the role of MSCs in the attenuation of tear hyperosmolarity and eye inflammation in patients suffering from DED. RESULTS Findings obtained in preclinical and pilot clinical studies demonstrated that MSCs reduced tear hyperomsolaity-induced pathological changes and suppressed immune cell-driven eye inflammation. Additionally, MSC-based therapy managed to successfully address the most severe DED-related conditions and complications. CONCLUSIONS MSCs should be considered as potentially new therapeutic agents for the treatment of severe DED.
Collapse
Affiliation(s)
- Carl Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N, Palm Harbor, FL 34684, USA;
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland;
| | - Ana Volarevic
- Department of Psychology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia;
| | - Aleksandar Arsenijevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia;
| | - Vladislav Volarevic
- Departments of Genetics, Microbiology and Immunology, Center for Research on Harmful Effects of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34000 Kragujevac, Serbia;
- Faculty of Pharmacy Novi Sad, Trg Mladenaca 5, 21000 Novi Sad, Serbia
| |
Collapse
|
20
|
Wu L, Zhang L, Huang M, Wu Y, Jin S, Zhang Y, Gan X, Yu T, Yu G, Zhang J, Wang X. Mesenchymal Stem Cell-Derived Exosomes: Emerging as a Promising Cell-Free Therapeutic Strategy for Autoimmune Hepatitis. Biomolecules 2024; 14:1353. [PMID: 39595530 PMCID: PMC11592114 DOI: 10.3390/biom14111353] [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: 09/12/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Autoimmune hepatitis (AIH) is an immune-mediated liver disease that currently faces limited treatment options. In its advanced stages, AIH can progress to liver fibrosis and cirrhosis. Recent research has increasingly focused on cell-free therapies, particularly the use of mesenchymal stem cell (MSC)-derived exosomes (Exos), which have shown promise in treating autoimmune diseases, including AIH. MSC-Exos, as microvesicles with low immunogenicity, high safety, and permeability, can deliver RNA, DNA, proteins, lipids, and various drugs for disease treatment, showing promising clinical application prospects. This review provides a comprehensive summary of the current research on MSC-Exos in the treatment of autoimmune hepatitis (AIH) and explores the underlying molecular mechanisms involved. It highlights the significant regulatory effects of MSC-Exos on immune cells and their ability to modify the microenvironment, demonstrating anti-inflammatory and anti-fibrotic properties while promoting liver regeneration. Additionally, this review also discusses potential challenges and future strategies for advancing Exo-based therapies in the treatment of AIH.
Collapse
Affiliation(s)
- Liwen Wu
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Longze Zhang
- Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Minglei Huang
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yan Wu
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Sikan Jin
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yaqi Zhang
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Xinyun Gan
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Ting Yu
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Guang Yu
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi 563003, China
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
21
|
Vakili S, Jafarinia M. Advances in Mesenchymal Stem Cell Research Applications for Female Infertility-Mechanisms, Efficacy Parameters, Challenges and Future Roadmap. Galen Med J 2024; 13:e3632. [PMID: 39483858 PMCID: PMC11525105 DOI: 10.31661/gmj.v13i.3632] [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: 06/08/2024] [Revised: 06/29/2024] [Accepted: 09/24/2024] [Indexed: 11/03/2024] Open
Abstract
Infertility affects approximately 15-20% of couples globally, with female factors contributing to nearly half of cases. Conditions such as polycystic ovary syndrome, endometriosis, tubal damage and premature ovarian failure are leading causes of female infertility. Current treatments like in vitro fertilization (IVF) have limitations and risks. Mesenchymal stem cells (MSCs) have shown therapeutic potential due to their ability to differentiate, secrete trophic factors, and exhibit immunomodulatory and anti-inflammatory properties. They have been demonstrated to repair and regenerate reproductive organs in various preclinical models of infertility related conditions. MSCs have reduced endometriotic lesions, regenerated lost follicles in premature ovarian failure (POF) models, and promoted tubal repair in damage models. Some clinical and preclinical studies have reported improved outcomes with MSC therapy in endometriosis and premature ovarian failure patients. This review discusses the properties and sources of MSCs, their mechanisms of action, preclinical evidence for applications in conditions like POF, polycystic ovary syndrome (PCOS), endometriosis, Asherman syndrome, and preeclampsia, and preliminary clinical data on MSC therapy for female infertility management.
Collapse
Affiliation(s)
- Sina Vakili
- Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz,
Iran
| |
Collapse
|
22
|
Xu S, Zhang Y, Zheng Z, Sun J, Wei Y, Ding G. Mesenchymal stem cells and their extracellular vesicles in bone and joint diseases: targeting the NLRP3 inflammasome. Hum Cell 2024; 37:1276-1289. [PMID: 38985391 DOI: 10.1007/s13577-024-01101-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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024]
Abstract
The nucleotide-binding oligomerization domain-like-receptor family pyrin domain-containing 3 (NLRP3) inflammasome is a cytosolic multi-subunit protein complex, and recent studies have demonstrated the vital role of the NLRP3 inflammasome in the pathological and physiological conditions, which cleaves gasdermin D to induce inflammatory cell death called pyroptosis and mediates the release of interleukin-1 beta and interleukin-18 in response to microbial infection or cellular injury. Over-activation of the NLRP3 inflammasome is associated with the pathogenesis of many disorders affecting bone and joints, including gouty arthritis, osteoarthritis, rheumatoid arthritis, osteoporosis, and periodontitis. Moreover, mesenchymal stem cells (MSCs) have been discovered to facilitate the inhibition of NLRP3 and maybe ideal for treating bone and joint diseases. In this review, we implicate the structure and activation of the NLRP3 inflammasome along with the detail on the involvement of NLRP3 inflammasome in bone and joint diseases pathology. In addition, we focused on MSCs and MSC-extracellular vesicles targeting NLRP3 inflammasomes in bone and joint diseases. Finally, the existing problems and future direction are also discussed.
Collapse
Affiliation(s)
- Shuangshuang Xu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Ying Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Yanan Wei
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, Shandong Province, China.
| |
Collapse
|
23
|
Yang D, Ge T, Zhou J, Li H, Zhang Y. Aloe-emodin alleviates inflammatory bowel disease in mice by modulating intestinal microbiome homeostasis via the IL-4/IL-13 axis. Heliyon 2024; 10:e34932. [PMID: 39157379 PMCID: PMC11328045 DOI: 10.1016/j.heliyon.2024.e34932] [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: 04/06/2024] [Revised: 07/11/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Inflammatory bowel disease (IBD) is a global health concern. Aloe-emodin (AE) has diverse pharmacological benefits, including anti-inflammatory effects. However, its role in IBD remains unclear, prompting our investigation of its regulatory effects and mechanisms in an IBD mouse model. Methods We studied the therapeutic efficacy of AE in alleviating symptoms and modulating cytokine secretion in a murine model of dextran sulfate sodium (DSS)-induced colitis. BALB/c mice were administered DSS to induce colitis and were subsequently treated with varying doses of AE. Changes in body weight, fecal lipocalin-2 (LCN2) levels, colon tissue histology, and serum cytokine concentrations were evaluated to assess the effects of AE treatment. Additionally, 16 S rRNA sequencing was used to analyze alterations in the composition of the gut microbiota following AE intervention. Finally, the database was used to analyze the signaling pathways associated with IBD in AE and to detect the expression levels of interleukin (IL)-4 pathway using real-time quantitative reverse transcription PCR. Exogenous IL-4 was used in rescue experiments to observe its effects on the disease process of IBD under AE regulation. Results AE treatment resulted in a dose-dependent mitigation of weight loss, reduction in fecal LCN2 levels, and amelioration of histological damage in DSS-induced colitis in mice. The levels of superoxide dismutase and catalase increased, whereas malondialdehyde decreased following AE treatment, indicating a dose-dependent alleviation of colitis symptoms. Furthermore, AE administration attenuated the secretion of pro-inflammatory cytokines, including IL-17, tumor necrosis factor-alpha (TNF-α), and chemokine ligand 1, while promoting the expression of anti-inflammatory cytokines IL-4 and IL-13. Analysis of the gut microbiota revealed that AE effectively suppressed the overgrowth of colitis-associated bacterial species and restored microbial homeostasis. Finally, we found that overexpression of IL-4 was able to reverse the therapeutic effect of AE for DSS-induced IBD. Conclusion AE shows promise in alleviating colitis severity, influencing inflammatory cytokines, and modulating the gut microbiota in an IBD mouse model via the IL-4/IL-13 pathway, suggesting its potential as a natural IBD remedy.
Collapse
Affiliation(s)
| | | | - Jingyi Zhou
- Department of Anorectal Surgery, The First People's Hospital of Lianyungang, NO.6 Zhenhua East Road, Haizhou District, Lianyungang, 222061, Jiangsu, China
| | - Huazhuan Li
- Department of Anorectal Surgery, The First People's Hospital of Lianyungang, NO.6 Zhenhua East Road, Haizhou District, Lianyungang, 222061, Jiangsu, China
| | - Yonggang Zhang
- Department of Anorectal Surgery, The First People's Hospital of Lianyungang, NO.6 Zhenhua East Road, Haizhou District, Lianyungang, 222061, Jiangsu, China
| |
Collapse
|
24
|
Bakinowska E, Bratborska AW, Kiełbowski K, Ćmil M, Biniek WJ, Pawlik A. The Role of Mesenchymal Stromal Cells in the Treatment of Rheumatoid Arthritis. Cells 2024; 13:915. [PMID: 38891047 PMCID: PMC11171813 DOI: 10.3390/cells13110915] [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: 04/20/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory joint disease characterised by the formation of a hyperplastic pannus, as well as cartilage and bone damage. The pathogenesis of RA is complex and involves broad interactions between various cells present in the inflamed synovium, including fibroblast-like synoviocytes (FLSs), macrophages, and T cells, among others. Under inflammatory conditions, these cells are activated, further enhancing inflammatory responses and angiogenesis and promoting bone and cartilage degradation. Novel treatment methods for RA are greatly needed, and mesenchymal stromal cells (MSCs) have been suggested as a promising new regenerative and immunomodulatory treatment. In this paper, we present the interactions between MSCs and RA-FLSs, and macrophages and T cells, and summarise studies examining the use of MSCs in preclinical and clinical RA studies.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (K.K.); (M.Ć.); (W.J.B.)
| | | | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (K.K.); (M.Ć.); (W.J.B.)
| | - Maciej Ćmil
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (K.K.); (M.Ć.); (W.J.B.)
| | - Wojciech Jerzy Biniek
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (K.K.); (M.Ć.); (W.J.B.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (K.K.); (M.Ć.); (W.J.B.)
| |
Collapse
|
25
|
Zhang Q, Lin J, Yang M, Li Z, Zhang M, Bu B. Therapeutic potential of natural killer cells in neuroimmunological diseases. Biomed Pharmacother 2024; 173:116371. [PMID: 38430631 DOI: 10.1016/j.biopha.2024.116371] [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: 12/17/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024] Open
Abstract
Natural killer (NK) cells, a major component of the innate immune system, have prominent immunoregulatory, antitumor proliferation, and antiviral activities. NK cells act as a double-edged sword with therapeutic potential in neurological autoimmunity. Emerging evidence has identified NK cells are involved in the development and progression of neuroimmunological diseases such as multiple sclerosis, neuromyelitis optica spectrum disorders, autoimmune encephalitis, Guillain-Barré Syndrome, chronic inflammatory demyelinating polyneuropathy, myasthenia gravis, and idiopathic inflammatory myopathy. However, the regulatory mechanisms and functional roles of NK cells are highly variable in different clinical states of neuroimmunological diseases and need to be further determined. In this review, we summarize the evidence for the heterogenic involvement of NK cells in the above conditions. Further, we describe cutting-edge NK-cell-based immunotherapy for neuroimmunological diseases in preclinical and clinical development and highlight challenges that must be overcome to fully realize the therapeutic potential of NK cells.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Lin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengge Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhijun Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Bitao Bu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|