1
|
Costela-Ruiz VJ, González-Vigil E, Espinosa-Ibáñez O, Alcázar – Caballero RM, Melguizo-Rodríguez L, Fernández-López O, Arias-Santiago S. Application of allogeneic adult mesenchymal stem cells in the treatment of venous ulcers: A phase I/II randomized controlled trial protocol. PLoS One 2025; 20:e0323173. [PMID: 40373055 PMCID: PMC12080757 DOI: 10.1371/journal.pone.0323173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/26/2025] [Indexed: 05/17/2025] Open
Abstract
OBJECTIVE To evaluate the feasibility, safety and efficacy of the cutaneous application of Bioengineered Artificial Mesenchymal Sheet (BAMS) in venous leg ulcers (VLUs) versus conventional treatment. METHODS This protocol is based on the design of a Phase I/II, multicenter, randomized, controlled, open-label clinical trial investigating the application of a biological dressing supplemented with mesenchymal stem cells (NCT05962931). The clinical trial is being conducted in 2 primary care units within the Granada Metropolitan Health District. A total of 20 patients with VLUs are being randomized (1:1) into 2 intervention arms: a control group and a treatment group. The intervention in the treatment group consists of the local application of 4 doses of BAMS, administered once per week, while the control group receives conventional therapy. Feasibility will be assessed based on the ability to complete the administration of 4 doses in at least 80% of the patients in the treatment group. Safety will be evaluated by analyzing the incidence of adverse effects and serious adverse effects. Efficacy will be assessed in terms of the percentage of wound closure (measured by wound area reduction), macroscopic assessment of the lesion (visual macroscopic analysis and RESVECH 2.0 scale), analysis of growth factors and inflammatory cytokines (ELISA test), pain levels (VAS scale) and quality of life (CIVIQ 20). RESULTS If confirmed, BAMS-based therapy may provide an effective treatment for VLUs, potentially reducing wound closure time and associated complications. This therapy could significantly enhance patients' quality of life due to the regenerative and analgesic properties of the biological dressing. DISCUSSION Given the biological activity of mesenchymal stem cells, an accelerated healing effect is expected in the treatment group. This could lead to shorter healing times for chronic wounds, resulting in significant benefits for patients, healthcare professionals, and overall healthcare costs. TRIAL REGISTRATION NCT05962931.
Collapse
Affiliation(s)
- Víctor J. Costela-Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
- Instituto Investigación Biosanitaria, ibs. Granada, Spain
| | - Encarnación González-Vigil
- Andalusian Health Service, Granada Metropolitan Health District, Primary Care Unit of Atarfe (Granada), Granada, Spain
| | - Olga Espinosa-Ibáñez
- Tissue Engineering and Cell Production Unit, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | - Lucía Melguizo-Rodríguez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
- Instituto Investigación Biosanitaria, ibs. Granada, Spain
| | - Olga Fernández-López
- Andalusian Network for the Design and Translation of Advanced Therapies, Junta de Andalucía, Seville, Spain
| | - Salvador Arias-Santiago
- Instituto Investigación Biosanitaria, ibs. Granada, Spain
- Tissue Engineering and Cell Production Unit, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Dermatology Department, Hospital Universitario Virgen de las Nieves, Granada. Spain
- Dermatology Department, School of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
2
|
Wang Y, Wu B, Tao Y, Wang M, Wu D, Chen E, Tang H. Therapeutic effect of hUC-MSCs from different transplantation routes on acute liver failure in rats. Front Med (Lausanne) 2025; 12:1525719. [PMID: 40417693 PMCID: PMC12098624 DOI: 10.3389/fmed.2025.1525719] [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: 11/10/2024] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
Objective Acute liver failure (ALF) is a rare yet serious clinical syndrome. Recent studies have indicated that stem cells can effectively treat this condition. However, the optimal route for stem cell transplantation in the treatment of ALF remains unclear. This study aims to investigate the most effective transplantation route for stem cell therapy in ALF. Methods Human umbilical cord mesenchymal stem cells (hUC-MSCs) expressing both luciferase and green fluorescent protein were generated using a lentiviral vector. The hUC-MSCs were transplanted via the tail vein, portal vein, and abdominal cavity. The survival and distribution of the transplanted hUC-MSCs in rats were assessed through in vivo imaging and immunofluorescence. Furthermore, the therapeutic effects of hUC-MSCs transplanted via different routes on ALF were compared. Results The survival time of hUC-MSCs transplanted via the tail vein and portal vein was shorter compared to those transplanted intraperitoneally. The distribution of hUC-MSCs varied by transplantation route: those injected via the tail vein and portal vein were primarily found in the lungs and liver, respectively, while intraperitoneally transplanted hUC-MSCs predominantly localized in the abdominal cavity. In ALF rats, hUC-MSCs transplanted via the tail vein and portal vein improved survival rates, enhanced liver pathology, and reduced levels of inflammatory cytokines in liver tissue. In contrast, abdominal transplantation of hUC-MSCs showed no significant therapeutic effect. Conclusion hUC-MSCs transplanted via the tail vein and portal vein exhibited similar therapeutic effects on ALF; however, abdominal transplantation of hUC-MSCs showed no significant effect.
Collapse
Affiliation(s)
- Yonghong Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Bei Wu
- Department of Hepatology, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Yachao Tao
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Menglan Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Aydın M, Akyüz S, Yanik H, Yildirim E, Başak AM, Güven HE, Gülap Y, Yilmaz KB. Autologous adipose-derived tissue stromal vascular fraction and intralesional epidermal growth factor combined application in patients with diabetic foot. J Wound Care 2025; 34:xxx-xxxviii. [PMID: 40056382 DOI: 10.12968/jowc.2024.0107] [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] [Indexed: 03/10/2025]
Abstract
OBJECTIVE The aim of this study was to evaluate the effect on wound healing of intralesional epidermal growth factor (iEGF) (Heberprot-p; Hasbiotech, Cuba) and autologous adipose-derived tissue stromal vascular fraction (AD-tSVF) applied in the closure of tissue defects. METHOD The patients included in the study were separated into three approximately equal groups: Group 1 with iEGF+AD-tSVF applied; Group 2 with iEGF only applied; and Group 3 with conventional wound care products applied. Granulation tissue was taken from the wound bed before the application of iEGF and AD-tSVF and at intervals thereafter for flow cytometry analysis. RESULTS Group 1 included 11 patients; Group 2 included 10 patients; and Group 3 included 10 patients. The time to re-epithelialisation was determined as 187.60±68.78 days in Group 3 patients compared with Group 1 (72.27±10.33 days) and Group 2 (70.50±18.02 days) (p<0.001). Following the application of iEGF to the wound bed, an increase was observed in M2 macrophage (CD209+), and M1 macrophage (CD38+) levels. The (CD34+) stem cells obtained from the granulation tissue after the application of AD-tSVF were determined to still be statistically significantly increased in the wound bed on the 21st day. CONCLUSION The results of this study demonstrated that the application of iEGF and iEGF+ AD-tSVF significantly shortened the wound healing period compared with conventional methods. AD-tSVF stands as an effective option, especially in the patient group with halted or delayed wound healing despite the application of iEGF. Moreover, the significant increase (p<0.001) in the level of M2 macrophages (CD209+), M1 macrophages (CD38+) and stem cells (CD34+) provided by this treatment modality showed that it contributed to wound healing at the cellular level.
Collapse
Affiliation(s)
- Mustafa Aydın
- MD, Orthopaedic Surgeon, Department of Orthopedics and Traumatology, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Simay Akyüz
- PhD, RN, Assistant Professor, University of Health Sciences, Gulhane Faculty of Nursing, Ankara, Turkey
| | - Hamdullah Yanik
- PhD, Molecular Biology and Genetics Specialist, Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
| | - Eda Yildirim
- MD, Orthopaedic Surgeon, Department of Orthopedics and Traumatology, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Ali Murat Başak
- MD, Orthopaedic Surgeon, Department of Orthopedics and Traumatology, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Hikmet Erhan Güven
- MD, Associate Professor, General Surgeon, Department of General Surgery, Etlik City Hospital, Ankara, Turkey
| | - Yasin Gülap
- MD, General Surgeon, Department of General Surgery, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
| | - Kerim Bora Yilmaz
- PhD, Molecular Biology and Genetics Specialist, Department of Basic Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
- MD, General Surgeon, Department of General Surgery, University of Health Sciences Gulhane Training and Research Hospital, Ankara, Turkey
- MD, Professor, General Surgeon, Department of Medical and Surgical Research, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| |
Collapse
|
4
|
Chen S, Jiang Y, Chai X, Chen Z, Tian H, Liu M, Zhu T, ShangGuan W, Wu X. Uterine-derived exosomes induce the M2 polarization of macrophages via miR-210-3p/ATP5D to promote endometriosis progression. Life Sci 2025; 363:123383. [PMID: 39798647 DOI: 10.1016/j.lfs.2025.123383] [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: 10/22/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
AIMS Endometriosis development is associated with peritoneal immune microenvironment abnormality; however, the specific mechanism is uncertain. We aimed to investigate the effects and underlying mechanisms of uterine cavity-derived exosomes on macrophage polarization and endometriosis progression. MATERIALS AND METHODS Uterine cavity-derived exosomes, miR-210-3p inhibitor or siATP5D were used to treat macrophages. Then evaluated the polarization of macrophages. By co-culturing of treated macrophages with endometrial stromal cells in vitro and an endometriosis C57BL6 mouse model to assess the role of uterine-derived exosomes and macrophages in the development of endometriosis. KEY FINDINGS Uterine cavity-derived exosomes could increase miR-210-3p expression and induce M2 macrophage polarization. Mechanistically, miR-210-3p can restrict ATP5D expression in macrophages, which leads to M2 polarization. In vivo experiments confirmed that macrophages lentivirally transduced with miR-210-3p can significantly decrease the growth and implantation of mouse endometriosis. SIGNIFICANCE In summary, our findings suggest that exosomes derived from the uterine cavity may drive macrophages towards M2 and promote endometriosis progression via miR-210-3p/ATP5D.
Collapse
Affiliation(s)
- Shengnan Chen
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Ying Jiang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Xiaoshan Chai
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhaoying Chen
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hao Tian
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Min Liu
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Tianyu Zhu
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wanwan ShangGuan
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Xianqing Wu
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital of Central South University, Changsha 410011, China.
| |
Collapse
|
5
|
Yarahmadi A, Dorri Giv M, Hosseininejad R, Rezaie A, Mohammadi N, Afkhami H, Farokhi A. Mesenchymal stem cells and their extracellular vesicle therapy for neurological disorders: traumatic brain injury and beyond. Front Neurol 2025; 16:1472679. [PMID: 39974358 PMCID: PMC11835705 DOI: 10.3389/fneur.2025.1472679] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025] Open
Abstract
Traumatic brain injury (TBI) is a complex condition involving mechanisms that lead to brain dysfunction and nerve damage, resulting in significant morbidity and mortality globally. Affecting ~50 million people annually, TBI's impact includes a high death rate, exceeding that of heart disease and cancer. Complications arising from TBI encompass concussion, cerebral hemorrhage, tumors, encephalitis, delayed apoptosis, and necrosis. Current treatment methods, such as pharmacotherapy with dihydropyridines, high-pressure oxygen therapy, behavioral therapy, and non-invasive brain stimulation, have shown limited efficacy. A comprehensive understanding of vascular components is essential for developing new treatments to improve blood vessel-related brain damage. Recently, mesenchymal stem cells (MSCs) have shown promising results in repairing and mitigating brain damage. Studies indicate that MSCs can promote neurogenesis and angiogenesis through various mechanisms, including releasing bioactive molecules and extracellular vesicles (EVs), which help reduce neuroinflammation. In research, the distinctive characteristics of MSCs have positioned them as highly desirable cell sources. Extensive investigations have been conducted on the regulatory properties of MSCs and their manipulation, tagging, and transportation techniques for brain-related applications. This review explores the progress and prospects of MSC therapy in TBI, focusing on mechanisms of action, therapeutic benefits, and the challenges and potential limitations of using MSCs in treating neurological disorders.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Masoumeh Dorri Giv
- Nuclear Medicine Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Hosseininejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Azin Rezaie
- Department of Microbiology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Narges Mohammadi
- Department of Molecular Cell Biology and Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Arastoo Farokhi
- Department of Anesthesiology, Kermanshah University of Medical Sciences, Imam Reza Hospital, Kermanshah, Iran
| |
Collapse
|
6
|
Tasdemir NK, Kilicarslan B, Imren G, Karaosmanoglu B, Taskiran EZ, Bayram C. Hierarchical TiO 2 nanotube arrays enhance mesenchymal stem cell adhesion and regenerative potential through surface nanotopography. J R Soc Interface 2025; 22:20240642. [PMID: 39999880 PMCID: PMC11858746 DOI: 10.1098/rsif.2024.0642] [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: 09/15/2024] [Revised: 11/20/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
The concept of preconditioning mesenchymal stem cells (MSCs) under different stress conditions or with bioactive molecules is introduced to optimize their therapeutic potential. This study investigates the physicochemical effect of hierarchical TiO2 nanotube arrays, a versatile and easy-to-prepare nanosurface, on MSC behaviour. By precisely controlling the nanotopography through anodization, we demonstrate the significant influence of surface properties on MSC adhesion, proliferation and differentiation. Electrostatic interactions between surface charge and proteins play a crucial role in these cellular responses. In addition, preconditioning MSCs under specific conditions enhances their therapeutic potential by optimizing paracrine signalling and homing properties. Higher surface charges and increasing spiky character of surface roughness of titania samples after anodization at 60 V significantly upregulated chemokine receptor type 4 (CXCR4) and vascular endothelial growth factor A (VEGFA), indicating the enhanced migratory and angiogenic potential of MSCs. The study reveals the mechanotransductive effects of nanotopography on MSC differentiation, suggesting that tailored surface features can direct cellular fate. These findings highlight the potential of hierarchical TiO2 nanotube arrays as a promising platform for regenerative medicine, offering a novel approach to improve tissue engineering and therapeutic outcomes.
Collapse
Affiliation(s)
- Nur Kubra Tasdemir
- Department of Medical and Surgical Research, Hacettepe University, Institute of Health Sciences, Ankara, Turkey
| | - Bogac Kilicarslan
- Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
- Advanced Technologies Application and Research Centre, Hacettepe University, Ankara, Turkey
| | - Gozde Imren
- Department of Medical and Surgical Research, Hacettepe University, Institute of Health Sciences, Ankara, Turkey
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Beren Karaosmanoglu
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ekim Z. Taskiran
- Department of Medical and Surgical Research, Hacettepe University, Institute of Health Sciences, Ankara, Turkey
- Department of Medical Genetics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Cem Bayram
- Department of Nanotechnology and Nanomedicine, Graduate School of Science and Engineering, Hacettepe University, Ankara, Turkey
- Advanced Technologies Application and Research Centre, Hacettepe University, Ankara, Turkey
| |
Collapse
|
7
|
Meenakshi Sundaram RS, Rupert S, Srinivasan P, Sathyanesan J, Govarthanan K, Jeyaraman N, Ramasubramanian S, Jeyaraman M, Chung HY, Gangadaran P, Ahn BC. Decoding Cytokine Dynamics: Wharton's Jelly Stromal Cells and Chondro-Differentiates in PHA-Stimulated Co-Culture. Cells 2025; 14:174. [PMID: 39936966 PMCID: PMC11817647 DOI: 10.3390/cells14030174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION Articular cartilage damage presents a significant clinical challenge, with limited options for effective regeneration. Mesenchymal stromal cells (MSCs) derived from Wharton's jelly (WJ) are a promising cell source for cartilage repair due to their regenerative and immunomodulatory properties. While undifferentiated MSCs have demonstrated potent immunoregulatory effects, the immunomodulatory potential of chondrocytes derived from WJ-MSCs remains underexplored, particularly under inflammatory conditions. This study investigates the differential cytokine expression profiles of WJ-MSC-derived chondrocytes and undifferentiated MSCs under inflammatory stimulation with phytohemagglutinin (PHA) to understand their immunomodulatory capacities. MATERIALS AND METHODS WJ-MSCs were differentiated into chondrocytes using a micromass culture system. Differentiated chondrocytes were then co-cultured with immune cells under PHA-induced inflammatory conditions. Control groups included co-cultured cells without PHA activation and chondrocytes activated with PHA in the absence of immune cell interaction. Cytokine expression profiles were analyzed using the RT2 Customized Gene Array to evaluate pro- and anti-inflammatory markers. Morphological changes were assessed microscopically. The immunomodulatory responses of chondrocytes were compared to those of undifferentiated MSCs under the same experimental conditions. RESULTS Chondrocytes co-cultured with immune cells under PHA activation exhibited downregulation of IDO, HLA-G, PDGF, IL-10, TNF-α, IL-6, and IFN-γ compared to undifferentiated MSCs in similar conditions. In non-PHA co-cultured conditions, chondrocytes showed increased expression of IL-6, IFN-γ, IL-4, VEGF, iNOS, PDGF, PTGS-2 and TGF-β, while TNF-α, IL-10, IDO and HLA-G were decreased. In contrast, chondrocytes activated with PHA without immune cell interaction displayed reduced expression of HLA-G and TNF-α, with no significant changes in IL-6, IFN-γ, IL-4, IL-10, VEGF, PDGF, PTGS-2, TGF-β, IDO, and iNOS compared to PHA-stimulated undifferentiated MSCs. CONCLUSION This study demonstrates that chondrocytes derived from WJ-MSCs exhibit limited immunomodulatory potential compared to undifferentiated MSCs, particularly under PHA-induced inflammatory conditions. Undifferentiated MSCs showed superior regulation of key cytokines associated with immune modulation. These findings suggest that maintaining MSCs in an undifferentiated state may be advantageous for therapeutic applications targeting inflammatory conditions, such as osteoarthritis. Future research should explore strategies to enhance the immunomodulatory efficacy of chondrocytes, potentially through genetic modification or adjunctive therapies.
Collapse
Affiliation(s)
- Raja Sundari Meenakshi Sundaram
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Secunda Rupert
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Prasanna Srinivasan
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Jeswanth Sathyanesan
- Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India; (R.S.M.S.); (S.R.); (P.S.)
| | - Kavitha Govarthanan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India;
| | - Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600017, Tamil Nadu, India; (N.J.); (M.J.)
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Swaminathan Ramasubramanian
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr. MGR Educational and Research Institute, Chennai 600017, Tamil Nadu, India; (N.J.); (M.J.)
- Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India;
| | - Ho Yun Chung
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Republic of Korea;
| | - Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
8
|
Wang Y, Zhang Y, Ma M, Zhuang X, Lu Y, Miao L, Lu X, Cui Y, Cui W. Mechanisms underlying the involvement of peritoneal macrophages in the pathogenesis and novel therapeutic strategies for dialysis-induced peritoneal fibrosis. Front Immunol 2024; 15:1507265. [PMID: 39749340 PMCID: PMC11693514 DOI: 10.3389/fimmu.2024.1507265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Long-term exposure of the peritoneum to peritoneal dialysate results in pathophysiological changes in the anatomical organization of the peritoneum and progressive development of peritoneal fibrosis. This leads to a decline in peritoneal function and ultrafiltration failure, ultimately necessitating the discontinuation of peritoneal dialysis, severely limiting the potential for long-term maintenance. Additionally, encapsulating peritoneal sclerosis, a serious consequence of peritoneal fibrosis, resulting in patients discontinuing PD and significant mortality. The causes and mechanisms underlying peritoneal fibrosis in patients undergoing peritoneal dialysis remain unknown, with no definitive treatment available. However, abnormal activation of the immune system appears to be involved in altering the structure of the peritoneum and promoting fibrotic changes. Macrophage infiltration and polarization are key contributors to pathological injury within the peritoneum, showing a strong correlation with the epithelial-to-mesenchymal transition of mesothelial cells and driving the process of fibrosis. This article discusses the role and mechanisms underlying macrophage activation-induced peritoneal fibrosis resulting from PD by analyzing relevant literature from the past decade and provides an overview of recent therapeutic approaches targeting macrophages to treat this condition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yingchun Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University,
Changchun, China
| |
Collapse
|
9
|
Yu Q, Zhang L, Wang Z, Wang Q, Sun X, Deng W, Cao X, Yu J, Xu X. Anti-inflammatory oligosaccharide licensed mesenchymal stem cells allow prolonged survival of septic rats via the promotion of glutathione synthesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156173. [PMID: 39471736 DOI: 10.1016/j.phymed.2024.156173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/16/2024] [Accepted: 10/20/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) possess the capability to mitigate multiorgan failure (MOF) and reduce mortality rates in sepsis. However, their survival is significantly limited due to oxidative stress responses triggered by excessive sepsis inflammation. Previous studies have demonstrated that the paracrine effect of MSCs can be enhanced by cytokine stimuli such as IL-1β, TNF-α, and IFN-γ, a process known as inflammatory licensing. This enhancement, however, may potentially lead to the apoptosis of MSCs. PURPOSE To investigate the therapeutic effects of Fructus Lycii oligosaccharide (FLO)-nasal mucosa-derived ectodermal MSCs (EMSCs) on septic rats and the underlying mechanisms. STUDY DESIGN AND METHODS FLO was screened from 21 distinct saccharides derived from traditional Chinese medicine (TCM), utilizing macrophage lipid raft chromatography prepared by our laboratory as the primary screening method.. The comparison of EMSCs primed with/without FLO was assessed through RNA-seq. Cecal ligation and puncture (CLP) surgery was performed in the CLP, EMSCs, and FLO-EMSCs groups (n = 10). The NC group underwent cecal ligation without puncture. The therapeutic effects of EMSCs and FLO-EMSCs on septic rats were evaluated through multiple tests including RT-PCR, western blot, histochemical staining, etc. RESULTS: FLO promoted M2 polarization of macrophages and enhanced the paracrine effect of EMSCs, without inducing apoptosis. Furthermore, FLO promoted GSH synthesis in EMSCs, aiding in the removal of reactive oxygen species (ROS) within these cells. The FLO-treated EMSCs demonstrated enhanced protection against pyroptosis in macrophages, thereby preventing immune paralysis during sepsis. CONCLUSION This study presents an innovative approach for enhancing the anti-inflammatory properties of MSCs using a TCM-derived oligosaccharide, thereby improving their therapeutic efficacy in sepsis models.
Collapse
Affiliation(s)
- Qingtong Yu
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China
| | - Linzhi Zhang
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China
| | - Zhe Wang
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China
| | - Qilong Wang
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China
| | - Xuan Sun
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China
| | - Wenwen Deng
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China
| | - Xia Cao
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China.
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, 212013, ZhenJiang, PR China.
| |
Collapse
|
10
|
Tsai SC, Wan BH, Tsai FJ, Yang JS. Artificial intelligence (AI)-powered bibliometric analysis of global trends in mesenchymal stem cells (MSCs)-derived exosome research: 2014-2023. Biomedicine (Taipei) 2024; 14:61-77. [PMID: 39777112 PMCID: PMC11703396 DOI: 10.37796/2211-8039.1470] [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: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction In recent years, significant progress has been made in regenerative medicine, specifically in using mesenchymal stem cells (MSCs) due to their regenerative and differentiating abilities. An exciting development in this area is the utilization of exosomes derived from MSCs, which have shown promise in tissue restoration, immune system modulation, and cancer treatment. Objectives This study aims to analyze global research trends and the academic impact of MSCs-derived exosomes from 2014 to 2023, providing a comprehensive overview of this emerging field. Materials and methods The Web of Science database selected 948 relevant publications from 2014 to 2023. Artificial intelligence (AI)-bibliometric tools, including Bibliometrix, CiteSpace, and VOSviewer, were employed to analyze and visualize the data. The focus was on publication quantity, research nations, institutional partnerships, keywords, and research focal points. Results The study revealed that China, Japan, Taiwan, and the United States are the leaders in publication volume and impact in MSCs-derived exosome research. China has the highest number of publications, while the United States and Iran excel in research quality and influence. Primary research themes were identified through keyword and clustering analyses, including tissue repair, immune modulation, bone regeneration, and cancer treatment. The study also emphasized the importance of international collaboration, with China and the United States demonstrating the most robust cooperation. Conclusion MSCs-derived exosome research rapidly expands worldwide, showing promising prospects in regenerative medicine and cell therapy. With continued research and international collaboration, MSCs-derived exosomes are expected to play a vital role in future therapeutic application.
Collapse
Affiliation(s)
- Shih-Chang Tsai
- Department of Biological Science and Technology, China Medical University, Taichung,
Taiwan
| | - Bing-Han Wan
- Department of Biological Science and Technology, China Medical University, Taichung,
Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung,
Taiwan
- China Medical University Children’s Hospital, Taichung,
Taiwan
- Department of Medical Genetics, China Medical University Hospital, Taichung,
Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung,
Taiwan
| |
Collapse
|
11
|
Fang WH, Vangsness CT. Orthobiologic Products: Preservation Options for Orthopedic Research and Clinical Applications. J Clin Med 2024; 13:6577. [PMID: 39518716 PMCID: PMC11546119 DOI: 10.3390/jcm13216577] [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: 09/11/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The biological products used in orthopedics include musculoskeletal allografts-such as bones, tendons, ligaments, and cartilage-as well as biological therapies. Musculoskeletal allografts support the body's healing process by utilizing preserved and sterilized donor tissue. These allografts are becoming increasingly common in surgical practice, allowing patients to avoid more invasive procedures and the risks associated with donor site morbidity. Bone grafting is one of the most frequently used procedures in orthopedics and traumatology. Biologic approaches aim to improve clinical outcomes by enhancing the body's natural healing capacity and reducing inflammation. They serve as an alternative to surgical interventions. While preliminary results from animal studies and small-scale clinical trials have been promising, the field of biologics still lacks robust clinical evidence supporting their efficacy. Biological therapies include PRP (platelet-rich plasma), mesenchymal stem cells (MSCs)/stromal cells/progenitor cells, bone marrow stem/stromal cells (BMSCs), adipose stem/stromal cells/progenitor cells (ASCs), cord blood (CB), and extracellular vesicles (EVs), including exosomes. The proper preservation and storage of these cellular therapies are essential for future use. Preservation techniques include cryopreservation, vitrification, lyophilization, and the use of cryoprotective agents (CPAs). The most commonly used CPA is DMSO (dimethyl sulfoxide). The highest success rates and post-thaw viability have been achieved by preserving PRP with a rate-controlled freezer using 6% DMSO and storing other cellular treatments using a rate-controlled freezer with 5% or 10% DMSO as the CPA. Extracellular vesicles (EVs) have shown the best results when lyophilized with 50 mM or 4% trehalose to prevent aggregation and stored at room temperature.
Collapse
Affiliation(s)
- William H. Fang
- Department of Orthopedic Surgery, Valley Health Systems, 620 Shadow Lane, Las Vegas, NV 89106, USA
| | - C. Thomas Vangsness
- Department of Orthopedic Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| |
Collapse
|
12
|
Pal D, Das P, Roy S, Mukherjee P, Halder S, Ghosh D, Nandi SK. Recent trends of stem cell therapies in the management of orthopedic surgical challenges. Int J Surg 2024; 110:6330-6344. [PMID: 38716973 PMCID: PMC11487011 DOI: 10.1097/js9.0000000000001524] [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: 12/29/2023] [Accepted: 04/14/2024] [Indexed: 10/20/2024]
Abstract
Emerged health-related problems especially with increasing population and with the wider occurrence of these issues have always put the utmost concern and led medicine to outgrow its usual mode of treatment, to achieve better outcomes. Orthopedic interventions are one of the most concerning hitches, requiring advancement in several issues, that show complications with conventional approaches. Advanced studies have been undertaken to address the issue, among which stem cell therapy emerged as a better area of growth. The capacity of the stem cells to renovate themselves and adapt into different cell types made it possible to implement its use as a regenerative slant. Harvesting the stem cells, particularly mesenchymal stem cells (MSCs) is easier and can be further grown in vitro . In this review, we have discussed orthopedic-related issues including bone defects and fractures, nonunions, ligament and tendon injuries, degenerative changes, and associated conditions, which require further approaches to execute better outcomes, and the advanced strategies that can be tagged along with various ways of application of MSCs. It aims to objectify the idea of stem cells, with a major focus on the application of MSCs from different sources in various orthopedic interventions. It also discusses the limitations, and future scopes for further approaches in the field of regenerative medicine. The involvement of MSCs may transition the procedures in orthopedic interventions from predominantly surgical substitution and reconstruction to bio-regeneration and prevention. Nevertheless, additional improvements and evaluations are required to explore the effectiveness and safety of mesenchymal stem cell treatment in orthopedic regenerative medicine.
Collapse
Affiliation(s)
| | - Pratik Das
- Department of Veterinary Surgery and Radiology
| | - Subhasis Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal
| | - Prasenjit Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal
| | | | | | | |
Collapse
|
13
|
Sirpilla O, Kenderian SS. Bioengineering MSCs for untapped therapeutic potential. Immunotherapy 2024; 16:907-911. [PMID: 39268677 PMCID: PMC11486229 DOI: 10.1080/1750743x.2024.2390819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/07/2024] [Indexed: 09/17/2024] Open
Affiliation(s)
- Olivia Sirpilla
- T Cell Engineering, Mayo Clinic, Rochester, MN55905, USA
- Division of Hematology, Mayo Clinic, Rochester, MN55905, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN55905, USA
- Division of Hematology, Mayo Clinic, Rochester, MN55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN55905, USA
| |
Collapse
|
14
|
Beauregard MA, Bedford GC, Brenner DA, Sanchez Solis LD, Nishiguchi T, Abhimanyu, Longlax SC, Mahata B, Veiseh O, Wenzel PL, DiNardo AR, Hilton IB, Diehl MR. Persistent tailoring of MSC activation through genetic priming. Mol Ther Methods Clin Dev 2024; 32:101316. [PMID: 39282077 PMCID: PMC11396059 DOI: 10.1016/j.omtm.2024.101316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/05/2024] [Indexed: 09/18/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) are an attractive platform for cell therapy due to their safety profile and unique ability to secrete broad arrays of immunomodulatory and regenerative molecules. Yet, MSCs are well known to require preconditioning or priming to boost their therapeutic efficacy. Current priming methods offer limited control over MSC activation, yield transient effects, and often induce the expression of pro-inflammatory effectors that can potentiate immunogenicity. Here, we describe a genetic priming method that can both selectively and sustainably boost MSC potency via the controlled expression of the inflammatory-stimulus-responsive transcription factor interferon response factor 1 (IRF1). MSCs engineered to hyper-express IRF1 recapitulate many core responses that are accessed by biochemical priming using the proinflammatory cytokine interferon-γ (IFN-γ). This includes the upregulation of anti-inflammatory effector molecules and the potentiation of MSC capacities to suppress T cell activation. However, we show that IRF1-mediated genetic priming is much more persistent than biochemical priming and can circumvent IFN-γ-dependent expression of immunogenic MHC class II molecules. Together, the ability to sustainably activate and selectively tailor MSC priming responses creates the possibility of programming MSC activation more comprehensively for therapeutic applications.
Collapse
Affiliation(s)
| | - Guy C. Bedford
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | - Tomoki Nishiguchi
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Abhimanyu
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Santiago Carrero Longlax
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Barun Mahata
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Pamela L. Wenzel
- Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Andrew R. DiNardo
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Isaac B. Hilton
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Michael R. Diehl
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Chemistry, Rice University, Houston, TX, USA
| |
Collapse
|
15
|
Przywara D, Petniak A, Gil-Kulik P. Optimizing Mesenchymal Stem Cells for Regenerative Medicine: Influence of Diabetes, Obesity, Autoimmune, and Inflammatory Conditions on Therapeutic Efficacy: A Review. Med Sci Monit 2024; 30:e945331. [PMID: 39154207 PMCID: PMC11340262 DOI: 10.12659/msm.945331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 08/19/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising tool that may be used in regenerative medicine. Thanks to their ability to differentiate and paracrine signaling, they can be used in the treatment of many diseases. Undifferentiated MSCs can support the regeneration of surrounding tissues through secreted substances and exosomes. This is possible thanks to the production of growth factors. These factors stimulate the growth of neighboring cells, have an anti-apoptotic effect, and support angiogenesis, and MSCs also have an immunomodulatory effect. The level of secreted factors may vary depending on many factors. Apart from the donor's health condition, it is also influenced by the source of MSCs, methods of harvesting, and even the banking of cells. This work is a review of research on how the patient's health condition affects the properties of obtained MSCs. The review discusses the impact of the patient's diabetes, obesity, autoimmune diseases, and inflammation, as well as the impact of the source of MSCs and methods of harvesting and banking cells on the phenotype, differentiation capacity, anti-inflammatory, angiogenic effects, and proliferation potential of MSCs. Knowledge about specific clinical factors allows for better use of the potential of stem cells and more appropriate targeting of procedures for collecting, multiplying, and banking these cells, as well as for their subsequent use. This article aims to review the characteristics, harvesting, banking, and paracrine signaling of MSCs and their role in diabetes, obesity, autoimmune and inflammatory diseases, and potential role in regenerative medicine.
Collapse
|
16
|
Morita S, Iwatake M, Suga S, Takahashi K, Sato K, Miyagi-Shiohira C, Noguchi H, Baba Y, Yukawa H. Establishment of a stem cell administration imaging method in bleomycin-induced pulmonary fibrosis mouse models. Sci Rep 2024; 14:18905. [PMID: 39143270 PMCID: PMC11325036 DOI: 10.1038/s41598-024-67586-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 07/12/2024] [Indexed: 08/16/2024] Open
Abstract
Pulmonary fibrosis is a progressive disease caused by interstitial inflammation. Treatments are extremely scarce; therapeutic drugs and transplantation therapies are not widely available due to cost and a lack of donors, respectively. Recently, there has been a high interest in regenerative medicine and exponential advancements in stem cell-based therapies have occurred. However, a sensitive imaging technique for investigating the in vivo dynamics of transplanted stem cells has not yet been established and the mechanisms of stem cell-based therapy remain largely unexplored. In this study, we administered mouse adipose tissue-derived mesenchymal stem cells (mASCs) labeled with quantum dots (QDs; 8.0 nM) to a mouse model of bleomycin-induced pulmonary fibrosis in an effort to clarify the relationship between in vivo dynamics and therapeutic efficacy. These QD-labeled mASCs were injected into the trachea of C57BL/6 mice seven days after bleomycin administration to induce fibrosis in the lungs. The therapeutic effects and efficacy were evaluated via in vivo/ex vivo imaging, CT imaging, and H&E staining of lung sections. The QD-labeled mASCs remained in the lungs longer and suppressed fibrosis. The 3D imaging results showed that the transplanted cells accumulated in the peripheral and fibrotic regions of the lungs. These results indicate that mASCs may prevent fibrosis. Thus, QD labeling could be a suitable and sensitive imaging technique for evaluating in vivo kinetics in correlation with the efficacy of cell therapy.
Collapse
Affiliation(s)
- Saho Morita
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Mayumi Iwatake
- Institutes of Innovation for Future Society, Institute of Nano-Life-Systems, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan.
| | - Sakura Suga
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Kazuomi Takahashi
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit Frontier, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya, 466-8550, Japan
| | - Kazuhide Sato
- Institutes of Innovation for Future Society, Institute of Nano-Life-Systems, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
- Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit Frontier, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya, 466-8550, Japan
- Nagoya University Graduate School of Medicine, 65 Tsuruma, Showa-ku, Nagoya, 466-8550, Japan
- FOREST-Souhatsu, JST, Tokyo, Japan
- Development of Quantum-Nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya, 466-8550, Japan
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, 903-0215, Japan
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, 903-0215, Japan
| | - Yoshinobu Baba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
- Institutes of Innovation for Future Society, Institute of Nano-Life-Systems, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
- Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba, 263-8555, Japan
| | - Hiroshi Yukawa
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan.
- Institutes of Innovation for Future Society, Institute of Nano-Life-Systems, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan.
- Nagoya University Graduate School of Medicine, 65 Tsuruma, Showa-ku, Nagoya, 466-8550, Japan.
- Development of Quantum-Nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya, 466-8550, Japan.
- Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba, 263-8555, Japan.
- Department of Quantum Life Science, Graduate School of Science, Chiba University, Chiba, 265-8522, Japan.
| |
Collapse
|
17
|
Lyu Z, Xin M, Oyston DR, Xue T, Kang H, Wang X, Wang Z, Li Q. Cause and consequence of heterogeneity in human mesenchymal stem cells: Challenges in clinical application. Pathol Res Pract 2024; 260:155354. [PMID: 38870711 DOI: 10.1016/j.prp.2024.155354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
Human mesenchymal stem cells (hMSCs) are mesoderm-derived adult stem cells with self-proliferation capacity, pluripotent differentiation potency, and excellent histocompatibility. These advantages make hMSCs a promising tool in clinical application. However, the majority of clinical trials using hMSC therapy for diverse human diseases do not achieve expectations, despite the prospective pre-clinical outcomes in animal models. This is partly attributable to the intrinsic heterogeneity of hMSCs. In this review, the cause of heterogeneity in hMSCs is systematically discussed at multiple levels, including isolation methods, cultural conditions, donor-to-donor variation, tissue sources, intra-tissue subpopulations, etc. Additionally, the effect of hMSCs heterogeneity on the contrary role in tumor progression and immunomodulation is also discussed. The attempts to understand the cellular heterogeneity of hMSCs and its consequences are important in supporting and improving therapeutic strategies for hMSCs.
Collapse
Affiliation(s)
- Zhao Lyu
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Miaomiao Xin
- Assisted Reproductive Center, Women's & Children's Hospital of Northwest, Xi'an, Shaanxi, China; University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, Vodnany, Czech Republic
| | - Dale Reece Oyston
- Department of Evolution, Ecology and Behaviour, University of Liverpool, Liverpool, UK
| | - Tingyu Xue
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Hong Kang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Xiangling Wang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Zheng Wang
- Medical Center of Hematology, the Second Affiliated Hospital, Army Medical University, Chongqing, Sichuan, China.
| | - Qian Li
- Changsha Medical University, Changsha, Hunan, China.
| |
Collapse
|
18
|
Zhang Z, Wu W, Li M, Du L, Li J, Yin X, Zhang W. Mesenchymal stem cell–derived extracellular vesicles: A novel nanoimmunoregulatory tool in musculoskeletal diseases. NANO TODAY 2024; 57:102343. [DOI: 10.1016/j.nantod.2024.102343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Yang Q, Huang J, Liu Y, Mai Q, Zhou Y, Zhou L, Zeng L, Deng K. Human Umbilical Cord Mesenchymal Stem Cells Promote Anti-Inflammation and Angiogenesis by Targeting Macrophages in a Rat Uterine Scar Model. Stem Cell Rev Rep 2024; 20:1555-1568. [PMID: 38703310 DOI: 10.1007/s12015-024-10730-6] [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] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) have demonstrated efficacy in repairing uterine scars, although the underlying mechanisms remain unclear. METHODS Uterine injury was surgically induced in a rat model, followed by immediate transplantation of 5 × 10 ^ 5 hUC-MSCs to each side of the uterus. Uterine morphology was evaluated at days 14 and 30 using HE and Masson staining. Immunohistochemistry assessed macrophage polarization, angiogenesis and endometrial receptivity in the endometrium. Additionally, the regulatory effects of hUC-MSCs on macrophage polarization were explored through coculture. qRT-PCR quantified the expression of anti-inflammatory (IL10 and Arg1) and pro-inflammatory (iNOS and TNF-α) factors. Western blotting evaluated CD163 expression. RESULTS Transplantation of hUC-MSCs promoted the healing of uterine injuries and tissue regeneration while inhibiting tissue fibrosis. Immunohistochemistry at days 14 and 30 post-transplantation demonstrated the polarization of macrophages toward the M2 phenotype in the uterine injury area in the presence of hUC-MSCs. Furthermore, hUC-MSC transplantation improved angiogenesis and endometrial receptivity in the uterine injury rat model, associated with increased IL10 expression. hUC-MSC-induced angiogenesis can be resisted by depleted macrophages. In vitro coculture experiments further demonstrated that hUC-MSCs promoted IL10 expression in macrophages while suppressing TNF-α and iNOS expression. Western blotting showed enhanced CD163 expression in macrophages following hUC-MSC treatment. CONCLUSIONS hUC-MSCs contribute to the healing of uterine injuries by targeting macrophages to promote angiogenesis and the expression of anti-inflammatory factors.
Collapse
Affiliation(s)
- Qian Yang
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China
| | - Jinfa Huang
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China
| | - Yixuan Liu
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China
| | - Qiqing Mai
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China
| | - Yuan Zhou
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China
| | - Lei Zhou
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China
| | - Lingling Zeng
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China
| | - Kaixian Deng
- Gynecology Department, Shunde Hospital, Southern Medical University, Foshan, 528308, China.
| |
Collapse
|
20
|
Hacimoto SYS, Cressoni ACL, da Silva LECM, Padovan CC, Ferriani RA, Rosa-e-Silva JC, Meola J. Selection of reference miRNAs for RT-qPCR assays in endometriosis menstrual blood-derived mesenchymal stem cells. PLoS One 2024; 19:e0306657. [PMID: 39078824 PMCID: PMC11288454 DOI: 10.1371/journal.pone.0306657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 06/20/2024] [Indexed: 08/02/2024] Open
Abstract
Choosing appropriate reference genes or internal controls to normalize RT-qPCR data is mandatory for the interexperimental reproducibility of gene expression data obtained by RT-qPCR in most studies, including those on endometriosis. Particularly for miRNAs, the choice for reference genes is challenging because of their physicochemical and biological characteristics. Moreover, the retrograde menstruation theory, mesenchymal stem cells in menstrual blood (MenSCs), and changes in post-transcriptional regulatory processes through miRNAs have gained prominence in the scientific community as important players in endometriosis. Therefore, we originally explored the stability of 10 miRNAs expressions as internal control candidates in conditions involving the two-dimensional culture of MenSCs from healthy women and patients with endometriosis. Here, we applied multiple algorithms (geNorm, NormFinder, Bestkeeper, and delta Ct) to screen reference genes and assessed the comprehensive stability classification of miRNAs using RefFinder. Pairwise variation calculated using geNorm identified three miRNAs as a sufficient number of reference genes for accurate normalization. MiR-191-5p, miR-24-3p, and miR-103a-3p were the best combination for suitable gene expression normalization. This study will benefit similar research, but is also attractive for regenerative medicine and clinics that use MenSCs, miRNA expression, and RT-qPCR.
Collapse
Affiliation(s)
- Sabrina Yukari Santos Hacimoto
- Department of Gynecology and Obstetrics of Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana Clara Lagazzi Cressoni
- Department of Gynecology and Obstetrics of Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Cristiana Carolina Padovan
- Department of Gynecology and Obstetrics of Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rui Alberto Ferriani
- Department of Gynecology and Obstetrics of Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- National Institute of Hormones and Women’s Health (Hormona), CNPq, Porto Alegre, Rio Grande do Sul, Brazil
| | - Júlio César Rosa-e-Silva
- Department of Gynecology and Obstetrics of Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Juliana Meola
- Department of Gynecology and Obstetrics of Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- National Institute of Hormones and Women’s Health (Hormona), CNPq, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
21
|
Wang X, He W, Huang H, Han J, Wang R, Li H, Long Y, Wang G, Han X. Recent Advances in Hydrogel Technology in Delivering Mesenchymal Stem Cell for Osteoarthritis Therapy. Biomolecules 2024; 14:858. [PMID: 39062572 PMCID: PMC11274544 DOI: 10.3390/biom14070858] [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: 05/28/2024] [Revised: 07/06/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Osteoarthritis (OA), a chronic joint disease affecting over 500 million individuals globally, is characterized by the destruction of articular cartilage and joint inflammation. Conventional treatments are insufficient for repairing damaged joint tissue, necessitating novel therapeutic approaches. Mesenchymal stem cells (MSCs), with their potential for differentiation and self-renewal, hold great promise as a treatment for OA. However, challenges such as MSC viability and apoptosis in the ischemic joint environment hinder their therapeutic effectiveness. Hydrogels with biocompatibility and degradability offer a three-dimensional scaffold that support cell viability and differentiation, making them ideal for MSC delivery in OA treatment. This review discusses the pathological features of OA, the properties of MSCs, the challenges associated with MSC therapy, and methods for hydrogel preparation and functionalization. Furthermore, it highlights the advantages of hydrogel-based MSC delivery systems while providing insights into future research directions and the clinical potential of this approach.
Collapse
Affiliation(s)
- Xiangjiang Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Wentao He
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Hao Huang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Collage of Physics and Optoelectronics Engineering, Shenzhen University, Shenzhen 518060, China;
| | - Jiali Han
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Ruren Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Hongyi Li
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Ying Long
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Guiqing Wang
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| | - Xianjing Han
- The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan 511518, China; (X.W.); (W.H.); (J.H.); (R.W.); (H.L.); (Y.L.)
| |
Collapse
|
22
|
Szeliga A, Grymowicz M, Kostrzak A, Smolarczyk R, Bala G, Smolarczyk K, Meczekalski B, Suchta K. Bone: A Neglected Endocrine Organ? J Clin Med 2024; 13:3889. [PMID: 38999458 PMCID: PMC11242793 DOI: 10.3390/jcm13133889] [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: 05/25/2024] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/14/2024] Open
Abstract
Bone has traditionally been viewed in the context of its structural contribution to the human body. Foremost providing necessary support for mobility, its roles in supporting calcium homeostasis and blood cell production are often afterthoughts. Recent research has further shed light on the ever-multifaceted role of bone and its importance not only for structure, but also as a complex endocrine organ producing hormones responsible for the autoregulation of bone metabolism. Osteocalcin is one of the most important substances produced in bone tissue. Osteocalcin in circulation increases insulin secretion and sensitivity, lowers blood glucose, and decreases visceral adipose tissue. In males, it has also been shown to enhance testosterone production by the testes. Neuropeptide Y is produced by various cell types including osteocytes and osteoblasts, and there is evidence suggesting that peripheral NPY is important for regulation of bone formation. Hormonal disorders are often associated with abnormal levels of bone turnover markers. These include commonly used bone formation markers (bone alkaline phosphatase, osteocalcin, and procollagen I N-propeptide) and commonly used resorption markers (serum C-telopeptides of type I collagen, urinary N-telopeptides of type I collagen, and tartrate-resistant acid phosphatase type 5b). Bone, however, is not exclusively comprised of osseous tissue. Bone marrow adipose tissue, an endocrine organ often compared to visceral adipose tissue, is found between trabecula in the bone cortex. It secretes a diverse range of hormones, lipid species, cytokines, and other factors to exert diverse local and systemic effects.
Collapse
Affiliation(s)
- Anna Szeliga
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Monika Grymowicz
- Department of Gynecological Endocrinology, Warsaw Medical University, 00-315 Warsaw, Poland
| | - Anna Kostrzak
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Roman Smolarczyk
- Department of Gynecological Endocrinology, Warsaw Medical University, 00-315 Warsaw, Poland
| | - Gregory Bala
- UCD School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | | | - Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Katarzyna Suchta
- Department of Gynecological Endocrinology, Warsaw Medical University, 00-315 Warsaw, Poland
| |
Collapse
|
23
|
Sousa AR, Gonçalves DC, Neves BG, Santos‐Coquillat A, Oliveira MB, Mano JF. Encapsulated Mesenchymal Stromal Cells as Cyclic Providers of Immunomodulatory Secretomes: A Living on-Demand Delivery System. Adv Healthc Mater 2024; 13:e2304012. [PMID: 38545848 PMCID: PMC11468815 DOI: 10.1002/adhm.202304012] [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: 11/15/2023] [Revised: 02/28/2024] [Indexed: 04/09/2024]
Abstract
The stimulation of mesenchymal stromal cells (MSCs) with inflammatory molecules is often used to boost their therapeutic effect. Prolonged exposure to inflammatory molecules has been explored to improve their action because MSCs therapies seem to be improved transiently with such stimuli. However, the possibility of cyclically stimulating MSCs to recover their optimized therapeutic potential is still to be elucidated, although the efficacy of cell-based therapies may be dependent on the ability to readapt to the relapse pathological conditions. Here, the response of MSCs, encapsulated in alginate hydrogels and cultured for 22 d, is explored using three different regimes: single, continuous, and intermittent stimulation with IFNγ. Exposure to IFNγ leads to a decrease in the secretion of IL-10, which is cyclically countered by IFNγ weaning. Conditioned media collected at different stages of pulsatile stimulation show an immunomodulatory potential toward macrophages, which directly correlates with IL-10 concentration in media. To understand whether the correlation between cyclic stimulation of MSCs and other biological actions can be observed, the effect on endothelial cells is studied, showcasing an overall modest influence on tube formation. Overall, the results describe the response of encapsulated MSCs to unusual pulsatile simulation regimens, exploring encapsulated MSCs as a living on-demand release system of tailored secretomes with recoverable immunomodulatory action.
Collapse
Affiliation(s)
- Ana Rita Sousa
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of AveiroAveiro3810‐193Portugal
| | - Diana C. Gonçalves
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of AveiroAveiro3810‐193Portugal
| | - Beatriz Guapo Neves
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of AveiroAveiro3810‐193Portugal
| | - Ana Santos‐Coquillat
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of AveiroAveiro3810‐193Portugal
| | - Mariana B. Oliveira
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of AveiroAveiro3810‐193Portugal
| | - João F. Mano
- Department of ChemistryCICECO – Aveiro Institute of MaterialsUniversity of AveiroAveiro3810‐193Portugal
| |
Collapse
|
24
|
Waqar MA, Zaman M, Khan R, Shafeeq Ur Rahman M, Majeed I. Navigating the tumor microenvironment: mesenchymal stem cell-mediated delivery of anticancer agents. J Drug Target 2024; 32:624-634. [PMID: 38652480 DOI: 10.1080/1061186x.2024.2347356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/21/2024] [Indexed: 04/25/2024]
Abstract
Scientific knowledge of cancer has advanced greatly throughout the years, with most recent studies findings includes many hallmarks that capture disease's multifaceted character. One of the novel approach utilised for the delivery of anti-cancer agents includes mesenchymal stem cell mediated drug delivery. Mesenchymal stem cells (MSCs) are non-haematopoietic progenitor cells that may be extracted from bone marrow, tooth pulp, adipose tissue and placenta/umbilical cord blood dealing with adult stem cells. MSCs are mostly involved in regeneration of tissue, they have also been shown to preferentially migrate to location of several types of tumour in-vivo. Usage of MSCs ought to improve both effectiveness and safety of anti-cancer drugs by enhancing delivery efficiency of anti-cancer therapies to tumour site. Numerous researches has demonstrated that various drugs, when delivered via mesenchymal stem cell mediated delivery can elicit anti-tumour effect of cells in cancers of breast cells and thyroid cells. MSCs have minimal immunogenicity because to lack of co-stimulatory molecule expression, which means there is no requirement for immunosuppression after allogenic transplantation. This current review elaborates recent advancements of mesenchyma stem cell mediated drug delivery of anti-cancer agents along with its mechanism and previously reported studies of drugs manufactured via this drug delivery system.
Collapse
Affiliation(s)
- Muhammad Ahsan Waqar
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | - Muhammad Zaman
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Rabeel Khan
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | | | - Imtiaz Majeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
25
|
Zhao E, Chen J, Qiu D, Liang R, Lu D, Tan W, Qin Y, Liu Q. STING-deficiency in lung resident mesenchymal stromal cells contributes to the alleviation of LPS-induced lung injury. Biochem Biophys Res Commun 2024; 714:149973. [PMID: 38657444 DOI: 10.1016/j.bbrc.2024.149973] [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: 04/02/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by acute diffuse inflammatory lung injury with a high mortality rate. Mesenchymal stromal cells (MSC) are pluripotent adult cells that can be extracted from a variety of tissues, including the lung. Lung-resident MSC (LR-MSC) located around vascular vessels and act as important regulators of lung homeostasis, regulating the balance between lung injury and repair processes. LR-MSC support the integrity of lung tissue by modulating immune responses and releasing trophic factors. Studies have reported that the STING pathway is involved in the progression of lung injury inflammation, but the specific mechanism is unclear. In this study, we found that STING deficiency could ameliorate lipopolysaccharides (LPS)-induced acute lung injury, STING knockout (STING KO) LR-MSC had an enhanced treatment effect on acute lung injury. STING depletion protected LR-MSC from LPS-induced apoptosis. RNA-sequencing and Western blot results showed that STING KO LR-MSC expressed higher levels of MSC immunoregulatory molecules, such as Igfbp4, Icam1, Hgf and Cox2, than WT LR-MSC. This study highlights that LR-MSC have a therapeutic role in acute lung injury, and we demonstrate that STING deficiency can enhance the immunomodulatory function of LR-MSC in controlling lung inflammation. Thus, STING can be used as an intervention target to enhance the therapeutic effect of MSC.
Collapse
Affiliation(s)
- Erming Zhao
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiawen Chen
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Dongbo Qiu
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Rukang Liang
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Di Lu
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Weikeng Tan
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Yunfei Qin
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Qiuli Liu
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
26
|
Gao G, Li L, Li C, Liu D, Wang Y, Li C. Mesenchymal stem cells: Guardians of women's health. Regen Ther 2024; 26:1087-1098. [PMID: 39582803 PMCID: PMC11585475 DOI: 10.1016/j.reth.2024.10.011] [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: 09/05/2024] [Revised: 10/13/2024] [Accepted: 10/23/2024] [Indexed: 11/26/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted more and more attention because of their multidirectional differentiation potential, immune regulatory abilities and self-renewal capacity. In recent years, their use has become prominent in the domains of regenerative medicine and tissue engineering. MSCs have shown promise in therapeutic studies for a variety of diseases and have become a new source of innovative solutions for the treatment of some obstetric and gynecological diseases. This review systematically presents the latest research on the use of MSCs in the treatment of obstetrics- and gynecology-related diseases. Specifically, this review encompasses the latest findings related to the role of MSCs in premature ovarian failure, polycystic ovary syndrome, ovarian cancer, fallopian tube-related diseases, uterine adhesions, endometriosis, cesarean scar defects, postmenopausal osteoporosis, and pelvic floor dysfunction. The shortcomings and challenges of the future use of MSCs in disease treatment are also discussed, with the intent to motivate improvements in MSC applications in clinical therapy. It is believed that with further research, MSCs will play a more important role in the treatment of obstetrics- and gynecology-related diseases.
Collapse
Affiliation(s)
- Guanwen Gao
- Peking University Shenzhen Clinical Institute of Shantou University Medical College, Shenzhen, 518036, China
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| | - Li Li
- Department of Internal Medicine, Jinan Central Hospital Affiliated to Shandong University, Ji Nan, 250000, China
| | - Changling Li
- Department of Obstetrics and Gynecology, Pingyi People's Hospital, Linyi City, Shandong Province, 276000, China
| | - Degao Liu
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| | - Yunfei Wang
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| | - Changzhong Li
- Center of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen Key Laboratory on Technology for Early Diagnosis of Major Gynecologic Diseases, Shenzhen, 518036, China
| |
Collapse
|
27
|
Koch DW, Froneberger A, Berglund A, Connard S, Souther A, Schnabel LV. IL-1β + TGF-β2 dual-licensed mesenchymal stem cells have reduced major histocompatibility class I expression and positively modulate tenocyte migration, metabolism, and gene expression. J Am Vet Med Assoc 2024; 262:S61-S72. [PMID: 38547589 PMCID: PMC11187728 DOI: 10.2460/javma.23.12.0708] [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: 12/22/2023] [Accepted: 03/08/2024] [Indexed: 04/24/2024]
Abstract
OBJECTIVE The study objectives were to 1) determine the mesenchymal stem cell (MSC) surface expression of major histocompatibility complex (MHC) class I and transcriptome-wide gene expression changes following IL-1β + TGF-β2 dual licensing and 2) evaluate if IL-1β + TGF-β2 dual-licensed MSCs had a greater ability to positively modulate tenocyte function compared to naive MSCs. SAMPLE Equine bone marrow-derived MSCs from 6 donors and equine superficial digital flexor tenocytes from 3 donors. METHODS Experiments were performed in vitro. Flow cytometry and bulk RNA sequencing were utilized to determine naive and dual-licensed MSC phenotype and transcriptome-wide changes in gene expression. Conditioned media were generated from MSCs and utilized in tenocyte cell culture assays as a method to determine the effect of MSC paracrine factors on tenocyte function. RESULTS Dual-licensed MSCs have a reduced expression of MHC class I and exhibit enrichment in functional pathways associated with the extracellular matrix, cell signaling, and tissue development. Additionally, dual-licensed MSC-conditioned media significantly improved in vitro tenocyte migration and metabolism to a greater degree than naive MSC-conditioned media. In tenocytes exposed to IL-1β, dual-licensed conditioned media also positively modulated tenocyte gene expression. CLINICAL RELEVANCE Our data indicate that conditioned media containing paracrine factors secreted from dual-licensed MSCs significantly modulates in vitro tenocyte function, which may confer benefits in vivo to healing tendons following injury. Additionally, due to reduced MHC class I expression in dual-licensed MSCs, this technique may also provide an avenue to provide an effective "off-the-shelf" allogenic source of MSCs.
Collapse
Affiliation(s)
- Drew W. Koch
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Anna Froneberger
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | - Alix Berglund
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Shannon Connard
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| | - Alexis Souther
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | - Lauren V. Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC
| |
Collapse
|
28
|
Shimizu Y, Ntege EH, Takahara E, Matsuura N, Matsuura R, Kamizato K, Inoue Y, Sowa Y, Sunami H. Adipose-derived stem cell therapy for spinal cord injuries: Advances, challenges, and future directions. Regen Ther 2024; 26:508-519. [PMID: 39161365 PMCID: PMC11331855 DOI: 10.1016/j.reth.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Spinal cord injury (SCI) has limited treatment options for regaining function. Adipose-derived stem cells (ADSCs) show promise owing to their ability to differentiate into multiple cell types, promote nerve cell survival, and modulate inflammation. This review explores ADSC therapy for SCI, focusing on its potential for improving function, preclinical and early clinical trial progress, challenges, and future directions. Preclinical studies have demonstrated ADSC transplantation's effectiveness in promoting functional recovery, reducing cavity formation, and enhancing nerve regrowth and myelin repair. To improve ADSC efficacy, strategies including genetic modification and combination with rehabilitation are being explored. Early clinical trials have shown safety and feasibility, with some suggesting motor and sensory function improvements. Challenges remain for clinical translation, including optimizing cell survival and delivery, determining dosing, addressing tumor formation risks, and establishing standardized protocols. Future research should focus on overcoming these challenges and exploring the potential for combining ADSC therapy with other treatments, including rehabilitation and medication.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Eisaku Takahara
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Rikako Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Kota Kamizato
- Department of Anesthesiology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, 1-98, Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, 329-0498, Tochigi, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| |
Collapse
|
29
|
Chen X, Yang N, Li B, Gao X, Wang Y, Wang Q, Liu X, Zhang Z, Zhang R. Visualization Analysis of Small Extracellular Vesicles in the Application of Bone-Related Diseases. Cells 2024; 13:904. [PMID: 38891036 PMCID: PMC11171653 DOI: 10.3390/cells13110904] [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: 04/11/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Small extracellular vesicles were shown to have similar functional roles to their parent cells without the defect of potential tumorigenicity, which made them a great candidate for regenerative medicine. The last twenty years have witnessed the rapid development of research on small extracellular vesicles. In this paper, we employed a scientometric synthesis method to conduct a retrospective analysis of small extracellular vesicles in the field of bone-related diseases. The overall background analysis consisted the visualization of the countries, institutions, journals, and authors involved in research. The current status of the research direction and future trends were presented through the analysis of references and keywords, which showed that engineering strategies, mesenchymal stem cell derived exosomes, and cartilage damage were the most concerning topics, and scaffold, osteoarthritis, platelet-rich plasma, and senescence were the future trends. We also discussed the current problems and challenges in practical applications, including the in-sight mechanisms, the building of relevant animal models, and the problems in clinical trials. By using CiteSpace, VOSviewer, and Bibliometrix, the presented data avoided subjective selectivity and tendency well, which made the conclusion more reliable and comprehensive. We hope that the findings can provide new perspectives for researchers to understand the evolution of this field over time and to search for novel research directions.
Collapse
Affiliation(s)
- Xinjiani Chen
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ning Yang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
| | - Bailei Li
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xinyu Gao
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
| | - Yayu Wang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qin Wang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaojun Liu
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
- Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 318000, China
| | - Zhen Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
| | - Rongqing Zhang
- Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 314006, China; (X.C.); (N.Y.); (B.L.); (X.G.); (Y.W.); (Q.W.); (X.L.)
- Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, 705 Yatai Road, Jiaxing 314006, China
- Taizhou Innovation Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing 318000, China
| |
Collapse
|
30
|
Saleh RO, Al-Hawary SIS, Hammoud A, Hjazi A, Ayad Abdulrazzaq S, Rajput P, Alawsi T, Alnajar MJ, Alawadi A. The long non-coding RNAs (lncRNA) in the pathogenesis of gastric cancer cells: molecular mechanisms and involvement miRNAs. Mol Biol Rep 2024; 51:615. [PMID: 38704760 DOI: 10.1007/s11033-024-09546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024]
Abstract
A complex sequence of occurrences, including host genetic vulnerability, Helicobacter pylori infection, and other environmental variables, culminate in gastric cancer (GC). The development of several genetic and epigenetic changes in oncogenes and tumor suppressor genes causes dysregulation of several signaling pathways, which upsets the cell cycle and the equilibrium between cell division and apoptosis, leading to GC. Developments in computational biology and RNA-seq technology enable quick detection and characterization of long non-coding RNAs (lncRNAs). Recent studies have shown that long non-coding RNAs (lncRNAs) have multiple roles in the development of gastric cancer. These lncRNAs interact with molecules of protein, RNA, DNA, and/or combinations. This review article explores several gastric cancer-associated lncRNAs, such as ADAMTS9-AS2, UCA1, XBP-1, and LINC00152. These various lncRNAs could change GC cell apoptosis, migration, and invasion features in the tumor microenvironment. This review provides an overview of the most recent research on lncRNAs and GC cell apoptosis, migration, invasion, and drug resistance, focusing on studies conducted in cancer cells and healthy cells during differentiation.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Ahmad Hammoud
- Department of Medical and Technical Information Technology, Bauman Moscow State Technical University, Moscow, Russia.
- Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Mishref Campus, Kuwait City, Kuwait.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences , Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | | | - Pranchal Rajput
- School of Applied and Life Sciences, Divison of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Taif Alawsi
- Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
- Department of Laser and Optoelectronics Engineering, University of Technology, Baghdad, Iraq
| | | | - Ahmed Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al-Qadisiyyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
31
|
Niu X, Xu X, Xu C, Cheuk YC, Rong R. Recent Advances of MSCs in Renal IRI: From Injury to Renal Fibrosis. Bioengineering (Basel) 2024; 11:432. [PMID: 38790298 PMCID: PMC11117619 DOI: 10.3390/bioengineering11050432] [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: 02/29/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Renal fibrosis is a pathological endpoint of maladaptation after ischemia-reperfusion injury (IRI), and despite many attempts, no good treatment has been achieved so far. At the core of renal fibrosis is the differentiation of various types of cells into myofibroblasts. MSCs were once thought to play a protective role after renal IRI. However, growing evidence suggests that MSCs have a two-sided nature. In spite of their protective role, in maladaptive situations, MSCs start to differentiate towards myofibroblasts, increasing the myofibroblast pool and promoting renal fibrosis. Following renal IRI, it has been observed that Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs) and Renal Resident Mesenchymal Stem Cells (RR-MSCs) play important roles. This review presents evidence supporting their involvement, discusses their potential mechanisms of action, and suggests several new targets for future research.
Collapse
Affiliation(s)
- Xinhao Niu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Xiaoqing Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Cuidi Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| |
Collapse
|
32
|
Moellerberndt J, Niebert S, Fey K, Hagen A, Burk J. Impact of platelet lysate on immunoregulatory characteristics of equine mesenchymal stromal cells. Front Vet Sci 2024; 11:1385395. [PMID: 38725585 PMCID: PMC11079816 DOI: 10.3389/fvets.2024.1385395] [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: 02/12/2024] [Accepted: 03/29/2024] [Indexed: 05/12/2024] Open
Abstract
Multipotent mesenchymal stromal cells (MSC) play an increasing role in the treatment of immune-mediated diseases and inflammatory processes. They regulate immune cells via cell-cell contacts and by secreting various anti-inflammatory molecules but are in turn influenced by many factors such as cytokines. For MSC culture, platelet lysate (PL), which contains a variety of cytokines, is a promising alternative to fetal bovine serum (FBS). We aimed to analyze if PL with its cytokines improves MSC immunoregulatory characteristics, with the perspective that PL could be useful for priming the MSC prior to therapeutic application. MSC, activated peripheral blood mononuclear cells (PBMC) and indirect co-cultures of both were cultivated in media supplemented with either PL, FBS, FBS+INF-γ or FBS+IL-10. After incubation, cytokine concentrations were measured in supernatants and control media. MSC were analyzed regarding their expression of immunoregulatory genes and PBMC regarding their proliferation and percentage of FoxP3+ cells. Cytokines, particularly IFN-γ and IL-10, remained at high levels in PL control medium without cells but decreased in cytokine-supplemented control FBS media without cells during incubation. PBMC released IFN-γ and IL-10 in various culture conditions. MSC alone only released IFN-γ and overall, cytokine levels in media were lowest when MSC were cultured alone. Stimulation of MSC either by PBMC or by PL resulted in an altered expression of immunoregulatory genes. In co-culture with PBMC, the MSC gene expression of COX2, TNFAIP6, IDO1, CXCR4 and MHC2 was upregulated and VCAM1 was downregulated. In the presence of PL, COX2, TNFAIP6, VCAM1, CXCR4 and HIF1A were upregulated. Functionally, while no consistent changes were found regarding the percentage of FoxP3+ cells, MSC decreased PBMC proliferation in all media, with the strongest effect in FBS media supplemented with IL-10 or IFN-γ. This study provides further evidence that PL supports MSC functionality, including their immunoregulatory mechanisms. The results justify to investigate functional effects of MSC cultured in PL-supplemented medium on different types of immune cells in more detail.
Collapse
Affiliation(s)
- Julia Moellerberndt
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - Sabine Niebert
- Institute of Physiology, Pathophysiology, and Biophysics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Kerstin Fey
- Equine Clinic (Internal Medicine), Justus-Liebig-University Giessen, Giessen, Germany
| | - Alina Hagen
- Equine Clinic (Surgery, Orthopedics), Justus-Liebig-University Giessen, Giessen, Germany
| | - Janina Burk
- Institute of Physiology, Pathophysiology, and Biophysics, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
33
|
Li Y, Wang T, Li X, Li W, Lei Y, Shang Q, Zheng Z, Fang J, Cao L, Yu D, Meng Z, Zhang S, Liu R, Liu C, Xu C, Ding Y, Chen Y, Candi E, Melino G, Wang Y, Shi Y, Shao C. SOD2 promotes the immunosuppressive function of mesenchymal stem cells at the expense of adipocyte differentiation. Mol Ther 2024; 32:1144-1157. [PMID: 38310354 PMCID: PMC11163202 DOI: 10.1016/j.ymthe.2024.01.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 11/28/2023] [Accepted: 01/30/2024] [Indexed: 02/05/2024] Open
Abstract
The potent immunomodulatory function of mesenchymal stem/stromal cells (MSCs) elicited by proinflammatory cytokines IFN-γ and TNF-α (IT) is critical to resolve inflammation and promote tissue repair. However, little is known about how the immunomodulatory capability of MSCs is related to their differentiation competency in the inflammatory microenvironment. In this study, we demonstrate that the adipocyte differentiation and immunomodulatory function of human adipose tissue-derived MSCs (MSC(AD)s) are mutually exclusive. Mitochondrial reactive oxygen species (mtROS), which promote adipocyte differentiation, were decreased in MSC(AD)s due to IT-induced upregulation of superoxide dismutase 2 (SOD2). Furthermore, knockdown of SOD2 led to enhanced adipogenic differentiation but reduced immunosuppression capability of MSC(AD)s. Interestingly, the adipogenic differentiation was associated with increased mitochondrial biogenesis and upregulation of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PPARGC1A/PGC-1α) expression. IT inhibited PGC-1α expression and decreased mitochondrial mass but promoted glycolysis in an SOD2-dependent manner. MSC(AD)s lacking SOD2 were compromised in their therapeutic efficacy in DSS-induced colitis in mice. Taken together, these findings indicate that the adipogenic differentiation and immunomodulation of MSC(AD)s may compete for resources in fulfilling the respective biosynthetic needs. Blocking of adipogenic differentiation by mitochondrial antioxidant may represent a novel strategy to enhance the immunosuppressive activity of MSCs in the inflammatory microenvironment.
Collapse
Affiliation(s)
- Yanan Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tingting Wang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Xiaolei Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Wen Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Yan Lei
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Qianwen Shang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Zhiyuan Zheng
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Lijuan Cao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Daojiang Yu
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhenzhen Meng
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Shengchao Zhang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Chunxiao Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Chenchang Xu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Yayun Ding
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Yongjing Chen
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
34
|
Sirpilla O, Sakemura RL, Hefazi M, Huynh TN, Can I, Girsch JH, Tapper EE, Cox MJ, Schick KJ, Manriquez-Roman C, Yun K, Stewart CM, Ogbodo EJ, Kimball BL, Mai LK, Gutierrez-Ruiz OL, Rodriguez ML, Gluscevic M, Larson DP, Abel AM, Wierson WA, Olivier G, Siegler EL, Kenderian SS. Mesenchymal stromal cells with chimaeric antigen receptors for enhanced immunosuppression. Nat Biomed Eng 2024; 8:443-460. [PMID: 38561490 PMCID: PMC12080371 DOI: 10.1038/s41551-024-01195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Allogeneic mesenchymal stromal cells (MSCs) are a safe treatment option for many disorders of the immune system. However, clinical trials using MSCs have shown inconsistent therapeutic efficacy, mostly owing to MSCs providing insufficient immunosuppression in target tissues. Here we show that antigen-specific immunosuppression can be enhanced by genetically modifying MSCs with chimaeric antigen receptors (CARs), as we show for E-cadherin-targeted CAR-MSCs for the treatment of graft-versus-host disease in mice. CAR-MSCs led to superior T-cell suppression and localization to E-cadherin+ colonic cells, ameliorating the animals' symptoms and survival rates. On antigen-specific stimulation, CAR-MSCs upregulated the expression of immunosuppressive genes and receptors for T-cell inhibition as well as the production of immunosuppressive cytokines while maintaining their stem cell phenotype and safety profile in the animal models. CAR-MSCs may represent a widely applicable therapeutic technology for enhancing immunosuppression.
Collapse
Affiliation(s)
- Olivia Sirpilla
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - R Leo Sakemura
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Mehrdad Hefazi
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Truc N Huynh
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Ismail Can
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - James H Girsch
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Erin E Tapper
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Michelle J Cox
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Kendall J Schick
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Claudia Manriquez-Roman
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kun Yun
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Carli M Stewart
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Ekene J Ogbodo
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Brooke L Kimball
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Long K Mai
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Omar L Gutierrez-Ruiz
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Makena L Rodriguez
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Martina Gluscevic
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Daniel P Larson
- Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | - Alex M Abel
- LifEngine Animal Health Laboratories Incorporated, Rochester, MN, USA
| | - Wesley A Wierson
- LifEngine Animal Health Laboratories Incorporated, Rochester, MN, USA
| | - Gloria Olivier
- Department of Business Development, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth L Siegler
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Hetta HF, Elsaghir A, Sijercic VC, Akhtar MS, Gad SA, Moses A, Zeleke MS, Alanazi FE, Ahmed AK, Ramadan YN. Mesenchymal stem cell therapy in diabetic foot ulcer: An updated comprehensive review. Health Sci Rep 2024; 7:e2036. [PMID: 38650719 PMCID: PMC11033295 DOI: 10.1002/hsr2.2036] [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: 11/08/2023] [Revised: 02/06/2024] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
Background Diabetes has evolved into a worldwide public health issue. One of the most serious complications of diabetes is diabetic foot ulcer (DFU), which frequently creates a significant financial strain on patients and lowers their quality of life. Up until now, there has been no curative therapy for DFU, only symptomatic relief or an interruption in the disease's progression. Recent studies have focused attention on mesenchymal stem cells (MSCs), which provide innovative and potential treatment candidates for several illnesses as they can differentiate into various cell types. They are mostly extracted from the placenta, adipose tissue, umbilical cord (UC), and bone marrow (BM). Regardless of their origin, they show comparable features and small deviations. Our goal is to investigate MSCs' therapeutic effects, application obstacles, and patient benefit strategies for DFU therapy. Methodology A comprehensive search was conducted using specific keywords relating to DFU, MSCs, and connected topics in the databases of Medline, Scopus, Web of Science, and PubMed. The main focus of the selection criteria was on English-language literature that explored the relationship between DFU, MSCs, and related factors. Results and Discussion Numerous studies are being conducted and have demonstrated that MSCs can induce re-epithelialization and angiogenesis, decrease inflammation, contribute to immunological modulation, and subsequently promote DFU healing, making them a promising approach to treating DFU. This review article provides a general snapshot of DFU (including clinical presentation, risk factors and etiopathogenesis, and conventional treatment) and discusses the clinical progress of MSCs in the management of DFU, taking into consideration the side effects and challenges during the application of MSCs and how to overcome these challenges to achieve maximum benefits. Conclusion The incorporation of MSCs in the management of DFU highlights their potential as a feasible therapeutic strategy. Establishing a comprehensive understanding of the complex relationship between DFU pathophysiology, MSC therapies, and related obstacles is essential for optimizing therapy outcomes and maximizing patient benefits.
Collapse
Affiliation(s)
- Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative MedicineFaculty of Pharmacy, University of TabukTabukSaudi Arabia
- Department of Medical Microbiology and ImmunologyFaculty of Medicine, Assiut UniversityAssiutEgypt
| | - Alaa Elsaghir
- Department of Microbiology and ImmunologyFaculty of Pharmacy, Assiut UniversityAssiutEgypt
| | | | | | - Sayed A. Gad
- Faculty of Medicine, Assiut UniversityAssiutEgypt
| | | | - Mahlet S. Zeleke
- Menelik II Medical and Health Science College, Kotebe Metropolitan UniversityAddis AbabaEthiopia
| | - Fawaz E. Alanazi
- Department of Pharmacology and ToxicologyFaculty of Pharmacy, University of TabukTabukSaudi Arabia
| | | | - Yasmin N. Ramadan
- Department of Microbiology and ImmunologyFaculty of Pharmacy, Assiut UniversityAssiutEgypt
| |
Collapse
|
36
|
Wang J, Deng G, Wang S, Li S, Song P, Lin K, Xu X, He Z. Enhancing regenerative medicine: the crucial role of stem cell therapy. Front Neurosci 2024; 18:1269577. [PMID: 38389789 PMCID: PMC10881826 DOI: 10.3389/fnins.2024.1269577] [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: 07/30/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Stem cells offer new therapeutic avenues for the repair and replacement of damaged tissues and organs owing to their self-renewal and multipotent differentiation capabilities. In this paper, we conduct a systematic review of the characteristics of various types of stem cells and offer insights into their potential applications in both cellular and cell-free therapies. In addition, we provide a comprehensive summary of the technical routes of stem cell therapy and discuss in detail current challenges, including safety issues and differentiation control. Although some issues remain, stem cell therapy demonstrates excellent potential in the field of regenerative medicine and provides novel tactics and methodologies for managing a wider spectrum of illnesses and traumas.
Collapse
Affiliation(s)
- Jipeng Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gang Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuyi Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuang Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Peng Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kun Lin
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaoxiang Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zuhong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
37
|
Beauregard MA, Bedford GC, Brenner DA, Sanchez Solis LD, Nishiguchi T, Abhimanyu, Longlax SC, Mahata B, Veiseh O, Wenzel PL, DiNardo AR, Hilton IB, Diehl MR. Persistent tailoring of MSC activation through genetic priming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578489. [PMID: 38370626 PMCID: PMC10871228 DOI: 10.1101/2024.02.01.578489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Mesenchymal stem/stromal cells (MSCs) are an attractive platform for cell therapy due to their safety profile and unique ability to secrete broad arrays of immunomodulatory and regenerative molecules. Yet, MSCs are well known to require preconditioning or priming to boost their therapeutic efficacy. Current priming methods offer limited control over MSC activation, yield transient effects, and often induce expression of pro-inflammatory effectors that can potentiate immunogenicity. Here, we describe a 'genetic priming' method that can both selectively and sustainably boost MSC potency via the controlled expression of the inflammatory-stimulus-responsive transcription factor IRF1 (interferon response factor 1). MSCs engineered to hyper-express IRF1 recapitulate many core responses that are accessed by biochemical priming using the proinflammatory cytokine interferon-γ (IFNγ). This includes the upregulation of anti-inflammatory effector molecules and the potentiation of MSC capacities to suppress T cell activation. However, we show that IRF1-mediated genetic priming is much more persistent than biochemical priming and can circumvent IFNγ-dependent expression of immunogenic MHC class II molecules. Together, the ability to sustainably activate and selectively tailor MSC priming responses creates the possibility of programming MSC activation more comprehensively for therapeutic applications.
Collapse
Affiliation(s)
| | - Guy C. Bedford
- Department of Bioengineering, Rice University, Houston, TX, USA
| | | | | | - Tomoki Nishiguchi
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Abhimanyu
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Santiago Carrero Longlax
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Barun Mahata
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Pamela L. Wenzel
- Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Andrew R. DiNardo
- The Global Tuberculosis Program, Texas Children’s Hospital, Immigrant and Global Health, WTS Center for Human Immunobiology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Isaac B. Hilton
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Michael R. Diehl
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Chemistry, Rice University, Houston, TX, USA
| |
Collapse
|
38
|
Tao X, Wang J, Liu B, Cheng P, Mu D, Du H, Niu B. Plasticity and crosstalk of mesenchymal stem cells and macrophages in immunomodulation in sepsis. Front Immunol 2024; 15:1338744. [PMID: 38352879 PMCID: PMC10861706 DOI: 10.3389/fimmu.2024.1338744] [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: 11/15/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Sepsis is a multisystem disease characterized by dysregulation of the host immune response to infection. Immune response kinetics play a crucial role in the pathogenesis and progression of sepsis. Macrophages, which are known for their heterogeneity and plasticity, actively participate in the immune response during sepsis. These cells are influenced by the ever-changing immune microenvironment and exhibit two-sided immune regulation. Recently, the immunomodulatory function of mesenchymal stem cells (MSCs) in sepsis has garnered significant attention. The immune microenvironment can profoundly impact MSCs, prompting them to exhibit dual immunomodulatory functions akin to a double-edged sword. This discovery holds great importance for understanding sepsis progression and devising effective treatment strategies. Importantly, there is a close interrelationship between macrophages and MSCs, characterized by the fact that during sepsis, these two cell types interact and cooperate to regulate inflammatory processes. This review summarizes the plasticity of macrophages and MSCs within the immune microenvironment during sepsis, as well as the intricate crosstalk between them. This remains an important concern for the future use of these cells for immunomodulatory treatments in the clinic.
Collapse
Affiliation(s)
- Xingyu Tao
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Jialian Wang
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Bin Liu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Peifeng Cheng
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Dan Mu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
| | - Huimin Du
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bailin Niu
- Department of Critical Care Medicine, Chongqing Key Laboratory of Emergency Medicine, School of Medicine, Chongqing University Central Hospital, Chongqing University, Chongqing, China
- Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
39
|
Harland N, Knoll J, Amend B, Abruzzese T, Abele H, Jakubowski P, Stenzl A, Aicher WK. Xenogenic Application of Human Placenta-Derived Mesenchymal Stromal Cells in a Porcine Large Animal Model. Cell Transplant 2024; 33:9636897241226737. [PMID: 38323325 PMCID: PMC10851762 DOI: 10.1177/09636897241226737] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
In animal models, cell therapies for different diseases or injuries have been very successful. Preclinical studies with cells aiming at a stroke, heart attack, and other emergency situations were promising but sometimes failed translation in clinical situations. We, therefore, investigated if human placenta-derived mesenchymal stromal cells can be injected in pigs without provoking rejection to serve as a xenogenic transplantation model to bridge preclinical animal studies to more promising future preclinical studies. Male human placenta-derived mesenchymal stromal cells were isolated, expanded, and characterized by flow cytometry, in vitro differentiation, and quantitative reverse-transcription polymerase chain reaction to prove their nature. Such cells were injected into the sphincter muscle of the urethrae of female pigs under visual control by cystoscopy employing a Williams needle. The animals were observed over 7 days of follow-up. Reactions of the host to the xenogeneic cells were explored by monitoring body temperature, and inflammatory markers including IL-1ß, CRP, and haptoglobin in blood. After sacrifice on day 7, infiltration of inflammatory cells in the tissue targeted was investigated by histology and immunofluorescence. DNA of injected human cells was detected by PCR. Upon injection in vascularized porcine tissue, human placenta-derived mesenchymal stromal cells were tolerated, and systemic inflammatory parameters were not elevated. DNA of injected cells was detected in situ 7 days after injection, and moderate local infiltration of inflammatory cells was observed. The therapeutic potential of human placenta-derived mesenchymal stromal cells can be explored in porcine large animal models of injury or disease. This seems a promising strategy to explore technologies for cell injections in infarcted hearts or small organs and tissues in therapeutically relevant amounts requiring large animal models to yield meaningful outcomes.
Collapse
Affiliation(s)
- Niklas Harland
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Jasmin Knoll
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Bastian Amend
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Tanja Abruzzese
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Harald Abele
- Department of Gynecology and Obstetrics, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Peter Jakubowski
- Department of Gynecology and Obstetrics, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Arnulf Stenzl
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Wilhelm K. Aicher
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| |
Collapse
|
40
|
Liao W, Li X, Tang X. Human Umbilical Cord Mesenchymal Stem Cells Alleviate Chronic Salpingitis by Modulating Macrophage-Associated Inflammatory Factors. Curr Stem Cell Res Ther 2024; 19:1442-1448. [PMID: 38173206 DOI: 10.2174/011574888x261128231108043931] [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: 06/11/2023] [Revised: 09/24/2023] [Accepted: 10/04/2023] [Indexed: 01/05/2024]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) have been widely studied because of their established anti-inflammatory properties. During chronic salpingitis (CS), infiltrated macrophages have vital roles in inflammation and tissue remodeling. METHODS We employed the type of MSCs, human umbilical cord (huc) MSCs in an experimental CS model and therapeutic efficacy was assessed. hucMSCs exerted this therapeutic effect by regulating macrophage function. To verify the regulatory effects of hucMSCs on the macrophage, macrophage line RAW264.7 markers were analyzed under LPS stimulation with or without co-culturing with hucMSCs for 12h and 24h. In addition, flow cytometry analysis was applied to reveal the interaction of co-culture. For animal studies, CS was induced by the MoPn strain Chlamydia trachomatis (CT), hucMSCs were intravaginally injected in the CS, and we analyzed the infiltrated macrophage by immunofluorescence. RESULTS We found the markers IL-10 was markedly increased and IL-1β, caspase-1 was notably downregulated after co-culturing with hucMSCs by RT-PCR. hucMSCs promote macrophage line RAW264.7 apoptosis. We also found that hucMSCs treatment can alleviate CS by decreasing the mRNA expression of IL-1β, caspase-1 and MCP-1 in the tubal tissue by RT-PCR and decreasing the protein expression of IL-1β, caspase-1 and TGF-β by western blotting. CONCLUSION These results suggest that macrophage function may be related to the immune-modulating characteristics of hucMSCs that contribute to CS.
Collapse
Affiliation(s)
- Wenjuan Liao
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Sun Yat-Sen University, 600, Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Xiaomao Li
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Sun Yat-Sen University, 600, Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Xinrang Tang
- Department of Obstetrics and Gynecology, Third Affiliated Hospital of Sun Yat-Sen University, 600, Tianhe Road, Tianhe District, Guangzhou, 510630, Guangdong, China
| |
Collapse
|
41
|
Xia M, Tong S, Gao L. Identification of MDK as a Hypoxia- and Epithelial-Mesenchymal Transition-Related Gene Biomarker of Glioblastoma Based on a Novel Risk Model and In Vitro Experiments. Biomedicines 2024; 12:92. [PMID: 38255198 PMCID: PMC10813330 DOI: 10.3390/biomedicines12010092] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/23/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Tumor cells are commonly exposed to a hypoxic environment, which can easily induce the epithelial-mesenchymal transition (EMT) of tumor cells, further affecting tumor proliferation, invasion, metastasis, and drug resistance. However, the predictive role of hypoxia and EMT-related genes in glioblastoma (GBM) has not been investigated. METHODS Intersection genes were identified by weighted correlation network analysis (WGCNA) and differential expression analyses, and a risk model was further constructed by LASSO and Cox analyses. Clinical, immune infiltration, tumor mutation, drug treatment, and enrichment profiles were analyzed based on the risk model. The expression level of the MDK gene was tested using RT-PCR, immunohistochemistry, and immunofluorescence. CCK8 and EdU were employed to determine the GBM cells' capacity for proliferation while the migration and invasion ability were detected by a wound healing assay and transwell assay, respectively. RESULTS Based on the GBM data of the TCGA and GTEx databases, 58 intersection genes were identified, and a risk model was constructed. The model was verified in the CGGA cohort, and its accuracy was confirmed by the ROC curve (AUC = 0.807). After combining clinical subgroups, univariate and multivariate Cox regression analyses showed that risk score and age were independent risk factors for GBM patients. Furthermore, our subsequent analysis of immune infiltration, tumor mutation, and drug treatment showed that risk score and high- and low-risk groups were associated with multiple immune cells, mutated genes, and drugs. Enrichment analysis indicated that the differences between high- and low-risk groups were manifested in tumor-related pathways, including the PI3K-AKT and JAK-STAT pathways. Finally, in vivo experiments proved that the hypoxia environment promoted the expression of MDK, and MDK knockdown reduced the proliferation, migration, and EMT of GBM cells induced by hypoxia. CONCLUSIONS Our novel prognostic correlation model provided more potential treatment strategies for GBM patients.
Collapse
Affiliation(s)
- Minqi Xia
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shiao Tong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
42
|
Sarvari A, Niasari-Naslaji A, Shirazi A, Heidari B, Boroujeni SB, Moradi MH, Naderi MM, Behzadi B, Mehrazar MM, Dehghan MM. Effect of Intra-ovarian Injection of Mesenchymal Stem Cells or its Conditioned Media on Repeated OPU-IVEP Outcomes in Jersey Heifers and Its Relationship with Follicular Fluid Inflammatory Markers. Avicenna J Med Biotechnol 2024; 16:16-28. [PMID: 38605741 PMCID: PMC11005394 DOI: 10.18502/ajmb.v16i1.14167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/26/2023] [Indexed: 04/13/2024] Open
Abstract
Background Repeated Ovum Pick Up (OPU) could have a detrimental effect on ovarian function, reducing In Vitro Embryo Production (IVEP). The present study examined the therapeutic effect of adipose-derived Mesenchymal Stem Cells (MSCs) or its Conditioned Medium (ConM) on ovarian trauma following repeated OPU. Resolvin E1 (RvE1) and Interleukin-12 (IL-12) were investigated as biomarkers. Methods Jersey heifers (n=8) experienced 11 OPU sessions including 5 pre-treatment and 6 treatment sessions. Heifers received intra-ovarian administration of MSCs or ConM (right ovary) and Dulbecco's Modified Phosphate Buffer Saline (DMPBS; left ovary) after OPU in sessions 5 and 8 and 2 weeks after session 11. The concentrations of RvE1 and IL-12 in follicular fluid was evaluated on sessions 1, 5, 6, 9, and 4 weeks after session 11. Following each OPU session, the IVEP parameters were recorded. Results Intra-ovarian administration of MSCs, ConM, and DMPBS did not affect IVEP parameters (p>0.05). The concentration of IL-12 in follicular fluid increased at the last session of pre-treatment (Session 5; p<0.05) and remained elevated throughout the treatment period. There was no correlation between IL-12 and IVEP parameters (p>0.05). However, RvE1 remained relatively high during the pre-treatment and decreased toward the end of treatment period (p<0.05). This in turn was associated with decline in some IVEP parameters (p<0.05). Conclusion Intra-ovarian administration of MSCs or ConM during repeated OPU did not enhance IVEP outcomes in Bos taurus heifers. The positive association between RvE1 and some of IVEP parameters could nominate RvE1 as a promising biomarker to predict IVEP parameters following repeated OPU.
Collapse
Affiliation(s)
- Ali Sarvari
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir Niasari-Naslaji
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abolfazl Shirazi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Banafsheh Heidari
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Sara Borjian Boroujeni
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Moradi
- Department of Animal Sciences, Faculty of Agriculture and Natural Resources, Arak University, Arak, Iran
| | - Mohammad-Mahdi Naderi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Bahareh Behzadi
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad-Mahdi Mehrazar
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
43
|
Derakhshani A, Taheri F, Geraminia N, Mohammadipoor-ghasemabad L, Sabzalizadeh M, Vafee F, Afarinesh MR, Sheibani V. Amelioration of behavioral and histological impairments in somatosensory cortex injury rats by limbal mesenchymal stem cell transplantation. Transl Neurosci 2024; 15:20220346. [PMID: 39156044 PMCID: PMC11330157 DOI: 10.1515/tnsci-2022-0346] [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: 03/10/2024] [Revised: 06/22/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Cortical lesions can cause major sensory and motor impairments, representing a significant challenge in neuroscience and clinical medicine. Limbal mesenchymal stem cells (LMSCs), renowned for their remarkable ability to proliferate and distinct characteristics within the corneal epithelium, offer a promising opportunity for regenerative treatments. This study aimed to assess whether the transplantation of LMSCs could improve tactile ability in rats with lesions of the barrel cortex. Methods In this experimental study, we divided 21 rats into three groups: a control group, a lesion group with cortical cold lesion induction but no stem cell treatment, and a group receiving LMSC transplantation following cold lesion induction. We conducted 3-week sensory assessments using a texture discrimination test and an open-field test. We also performed Nissl staining to assess changes on the cellular level. Results Rats in the LMSC transplantation group demonstrated significant improvements in their ability to discrimination textures during the second and third weeks compared to those in the lesion group. The open-field test results showed an increased exploratory behavior of rats in the LMSC transplantation group by the third week compared to the lesion group. Additionally, Nissl staining revealed cellular alterations in the damaged cortex, with a significant distinction observed between rats in the LMSCs and lesion group. Conclusion The findings suggest that LMSC transplantation enhances sensory recovery in rats with cortical lesions, particularly their ability to discriminate textures. LMSC transplantation benefits brain tissue reparation after a cold lesion on the somatosensory cortex.
Collapse
Affiliation(s)
- Ali Derakhshani
- Hydatid Disease Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Farahnaz Taheri
- Neurology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Nima Geraminia
- Kerman Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, P. O. Box: 76198-13159, Kerman, Iran
| | - Lily Mohammadipoor-ghasemabad
- Kerman Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, P. O. Box: 76198-13159, Kerman, Iran
| | - Mansoureh Sabzalizadeh
- Kerman Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, P. O. Box: 76198-13159, Kerman, Iran
- Cognitive Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, Kerman, Iran
| | - Farzaneh Vafee
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Afarinesh
- Kerman Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, P. O. Box: 76198-13159, Kerman, Iran
- Cognitive Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Kerman Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, P. O. Box: 76198-13159, Kerman, Iran
- Cognitive Neuroscience Research Center, Institute of Neuropharmachology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
44
|
Xie W, Luo T, Ma Z, Xue S, Jia X, Yang T, Song Z. Tumor Necrosis Factor Alpha Preconditioned Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Enhance the Inhibition of Necroptosis of Acinar cells in Severe Acute Pancreatitis. Tissue Eng Part A 2023; 29:607-619. [PMID: 37565286 DOI: 10.1089/ten.tea.2023.0139] [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] [Indexed: 08/12/2023] Open
Abstract
Severe acute pancreatitis (SAP) is a common abdominal emergency with a high mortality rate and a lack of effective therapeutic options. Although mesenchymal stem cell (MSC) transplantation is a potential treatment for SAP, the mechanism remains unclear. It has been suggested that MSCs may act mainly through paracrine effects; therefore, we aimed to demonstrate the therapeutic efficacy of extracellular vesicles (EVs) derived from human umbilical cord mesenchymal stem cells (UCMSCs) for SAP. Na-taurocholate was used to induce a rat SAP model through retrograde injection into the common biliopancreatic duct. After 72 h of EVs transplantation, pancreatic pathological damage was alleviated, along with a decrease in serum amylase activity and pro-inflammatory cytokine levels. Interestingly, when UCMSCs were preconditioned with 10 ng/mL tumor necrosis factor alpha (TNF-α) for 48 h, the obtained EVs (named TNF-α-EVs) performed an enhanced efficacy. Furthermore, both animal and cellular experiments showed that TNF-α-EVs alleviated the necroptosis of acinar cells of SAP through RIPK3/MLKL axis. In conclusion, our study demonstrated that TNF-α-EVs were able to enhance the therapeutic effect on SAP by inhibiting necroptosis compared to normal EVs. This study heralds that TNF-α-EVs may be a promising therapeutic approach for SAP in the future.
Collapse
Affiliation(s)
- Wangcheng Xie
- Department of General Surgery and Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Hepatic-Biliary-Pancreatic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingyi Luo
- Department of General Surgery and Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Hepatic-Biliary-Pancreatic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhilong Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaobo Xue
- Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuyang Jia
- Department of Metabolic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingsong Yang
- Department of General Surgery and Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenshun Song
- Department of Hepatic-Biliary-Pancreatic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
45
|
Zhao X, Hu Y, Xiao W, Ma Y, Shen D, Jiang Y, Shen Y, Wang S, Ma J. Efficacy of mesenchymal stromal cells in the treatment of unexplained recurrent spontaneous abortion in mice: An analytical and systematic review of meta-analyses. PLoS One 2023; 18:e0294855. [PMID: 38011163 PMCID: PMC10681256 DOI: 10.1371/journal.pone.0294855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVES Unexplained recurrent spontaneous abortion (URSA) remains an intractable reproductive dilemma due to the lack of understanding of the pathogenesis. This study aimed to evaluate the preclinical evidence for the mesenchymal stromal cell (MSC) treatment for URSA. METHODS A meticulous literature search was independently performed by two authors across the Cochrane Library, EMBASE, and PubMed databases from inception to April 9, 2023. Each study incorporated was assessed using the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) risk of bias tool. The amalgamated standardized mean difference (SMD) accompanied by 95% confidence interval (CI) were deduced through a fixed-effects or random-effects model analysis. RESULTS A total of ten studies incorporating 140 mice were subjected to data analysis. The MSC treatment yielded a significant reduction in the abortion rate within the URSA model (OR = 0.23, 95%CI [0.17, 0.3], P<0.00001). Moreover, it elicited a positive modulatory impact on the expression profiles of several inflammatory cytokines in the decidual tissue of URSA murine models, inclusive of IL4 (SMD 1.63, 95% CI [0.39, 2.86], P = 0.01), IL10 (SMD 1.60, 95% CI [0.58, 2.61], P = 0.002), IFN-γ (SMD -1.66, 95%CI [-2.79, -0.52], P = 0.004), and TNF-α (SMD -1.98, 95% CI [-2.93, -1.04], P< 0.0001). Subgroup analyses underscored that the administration mode of intraperitoneal and uterine horn injections, and sources of bone MSCs and adipose-derived MSCs contributed positively to the expression of IL4, IL10, and decreased the expression of IFN-γ in decidual tissue of URSA (P<0.05). Conversely, the tail vein injections subgroup was observed with no statistical significance (P>0.05). CONCLUSIONS The findings underscore the considerable potential of MSCs in URSA therapy. Nonetheless, the demand for enhanced transparency in research design and direct comparisons between various MSC sources and administration routes in URSA is paramount to engendering robust evidence that could pave the way for successful clinical translation.
Collapse
Affiliation(s)
- Xiaoxuan Zhao
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Yijie Hu
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Wenjun Xiao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Yiming Ma
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Dan Shen
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Yuepeng Jiang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310053, China
| | - Yi Shen
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Suxia Wang
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| | - Jing Ma
- Department of Traditional Chinese Medicine (TCM) Gynecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang Province, Hangzhou, 310007, China
| |
Collapse
|
46
|
Trevizani M, Leal LL, Rettore JVP, Macedo GC, Alves CCDS, de Castro SBR, do Carmo AMR, da Silva SA, Maranduba CMDC, Silva FDS. Tumor necrosis factor α, and agonist and antagonists of cannabinoid receptor type 1 and type 2 alter the immunophenotype of stem cells from human exfoliated deciduous teeth. EINSTEIN-SAO PAULO 2023; 21:eAO0405. [PMID: 37970951 PMCID: PMC10631756 DOI: 10.31744/einstein_journal/2023ao0405] [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: 12/01/2022] [Accepted: 03/19/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVE To verify the involvement of the endocannabinoid system in the immunomodulatory profile of stem cells from human exfoliated deciduous teeth, in the presence or absence of TNF-α, and agonist and antagonists of CB1 and CB2. METHODS Stem cells from human exfoliated deciduous teeth were cultured in the presence or absence of an agonist, anandamide, and two antagonists, AM251 and SR144528, of CB1 and CB2 receptors, with or without TNF-α stimulation. For analysis of immunomodulation, surface molecules linked to immunomodulation, namely human leukocyte antigen-DR isotype (HLA-DR), and programmed death ligands 1 (PD-L1) and 2 (PD-L2) were measured using flow cytometry. RESULTS The inhibition of endocannabinoid receptors together with the proinflammatory effect of TNF-α resulted in increased HLA-DR expression in stem cells from human exfoliated deciduous teeth, as well as, in these cells acquiring an anti-inflammatory profile by enhancing the expression of PD-L1 and PD-L2. CONCLUSION Stem cells from human exfoliated deciduous teeth respond to the endocannabinoid system and TNF-α by altering key immune response molecules. Inhibition of endocannabinoid receptors and TNF-α led to an increase in HLA-DR, PD-L1, and PD-L2 levels in stem cells from human exfoliated deciduous teeth. This study shows the interaction between mesenchymal stromal cells and the immune and endocannabinoid systems.
Collapse
Affiliation(s)
- Marizia Trevizani
- Instituto de Ciências BiológicasUniversidade Federal de Juiz de ForaJuiz de ForaMGBrazil Instituto de Ciências Biológicas , Universidade Federal de Juiz de Fora , Juiz de Fora , MG , Brazil .
| | - Laís Lopardi Leal
- Instituto de Ciências BiológicasUniversidade Federal de Juiz de ForaJuiz de ForaMGBrazil Instituto de Ciências Biológicas , Universidade Federal de Juiz de Fora , Juiz de Fora , MG , Brazil .
| | - João Vitor Paes Rettore
- Instituto de Ciências BiológicasUniversidade Federal de Juiz de ForaJuiz de ForaMGBrazil Instituto de Ciências Biológicas , Universidade Federal de Juiz de Fora , Juiz de Fora , MG , Brazil .
| | - Gilson Costa Macedo
- Instituto de Ciências BiológicasUniversidade Federal de Juiz de ForaJuiz de ForaMGBrazil Instituto de Ciências Biológicas , Universidade Federal de Juiz de Fora , Juiz de Fora , MG , Brazil .
| | - Caio César de Souza Alves
- Faculdade de Medicina do MucuriUniversidade Federal dos Vales do Jequitinhonha e MucuriTeófilo OtoniMGBrazil Faculdade de Medicina do Mucuri , Universidade Federal dos Vales do Jequitinhonha e Mucuri , Teófilo Otoni , MG , Brazil .
| | - Sandra Bertelli Ribeiro de Castro
- Faculdade de Medicina do MucuriUniversidade Federal dos Vales do Jequitinhonha e MucuriTeófilo OtoniMGBrazil Faculdade de Medicina do Mucuri , Universidade Federal dos Vales do Jequitinhonha e Mucuri , Teófilo Otoni , MG , Brazil .
| | - Antônio Márcio Resende do Carmo
- Instituto de Ciências BiológicasUniversidade Federal de Juiz de ForaJuiz de ForaMGBrazil Instituto de Ciências Biológicas , Universidade Federal de Juiz de Fora , Juiz de Fora , MG , Brazil .
| | - Silvioney Augusto da Silva
- Instituto de Ciências BiológicasUniversidade Federal de Juiz de ForaJuiz de ForaMGBrazil Instituto de Ciências Biológicas , Universidade Federal de Juiz de Fora , Juiz de Fora , MG , Brazil .
| | - Carlos Magno da Costa Maranduba
- Instituto de Ciências BiológicasUniversidade Federal de Juiz de ForaJuiz de ForaMGBrazil Instituto de Ciências Biológicas , Universidade Federal de Juiz de Fora , Juiz de Fora , MG , Brazil .
| | - Fernando de Sá Silva
- Universidade Federal de Juiz de ForaGovernador ValadaresMGBrazil Universidade Federal de Juiz de Fora , Governador Valadares , MG , Brazil .
| |
Collapse
|
47
|
Cehakova M, Ivanisova D, Strecanska M, Plava J, Varchulova Novakova Z, Nicodemou A, Harsanyi S, Culenova M, Bernatova S, Danisovic L. Rheumatoid Synovial Fluid and Acidic Extracellular pH Modulate the Immunomodulatory Activity of Urine-Derived Stem Cells. Int J Mol Sci 2023; 24:15856. [PMID: 37958839 PMCID: PMC10648750 DOI: 10.3390/ijms242115856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Urine-derived stem cells (UdSCs) possess a remarkable anti-inflammatory and immune-modulating activity. However, the clinical significance of UdSCs in autoimmune inflammatory diseases such as rheumatoid arthritis (RA) is yet to be explored. Hence, we tested the UdSCs response to an articular RA microenvironment. To simulate the inflamed RA joint more authentically in vitro, we treated cells with rheumatoid synovial fluids (RASFs) collected from RA patients, serum deprivation, acidosis (pH 7.0 and 6.5), and their combinations. Firstly, the RASFs pro-inflammatory status was assessed by cytokine quantification. Then, UdSCs were exposed to the RA environmental factors for 48 h and cell proliferation, gene expression and secretion of immunomodulatory factors were evaluated. The immunosuppressive potential of pre-conditioned UdSCs was also assessed via co-cultivation with activated peripheral blood mononuclear cells (PBMCs). In all experimental conditions, UdSCs' proliferation was not affected. Conversely, extracellular acidosis considerably impaired the viability/proliferation of adipose tissue-derived stem cells (ATSCs). In the majority of cases, exposure to RA components led to the upregulated expression of IL-6, TSG6, ICAM-1, VCAM-1, and PD-L1, all involved in immunomodulation. Upon RASFs and acidic stimulation, UdSCs secreted higher levels of immunomodulatory cytokines: IL-6, IL-8, MCP-1, RANTES, GM-CSF, and IL-4. Furthermore, RASFs and combined pretreatment with RASFs and acidosis promoted the UdSCs-mediated immunosuppression and the proliferation of activated PBMCs was significantly inhibited. Altogether, our data indicate that the RA microenvironment certainly has the capacity to enhance UdSCs' immunomodulatory function. For potential preclinical/clinical applications, the intra-articular injection might be a reasonable approach to maximize UdSCs' therapeutic efficiency in the RA treatment.
Collapse
Affiliation(s)
- Michaela Cehakova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.S.); (Z.V.N.); (A.N.); (S.H.); (M.C.); (L.D.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Dana Ivanisova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Magdalena Strecanska
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.S.); (Z.V.N.); (A.N.); (S.H.); (M.C.); (L.D.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Jana Plava
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
- Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Varchulova Novakova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.S.); (Z.V.N.); (A.N.); (S.H.); (M.C.); (L.D.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Andreas Nicodemou
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.S.); (Z.V.N.); (A.N.); (S.H.); (M.C.); (L.D.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Stefan Harsanyi
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.S.); (Z.V.N.); (A.N.); (S.H.); (M.C.); (L.D.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Martina Culenova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.S.); (Z.V.N.); (A.N.); (S.H.); (M.C.); (L.D.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Sona Bernatova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| | - Lubos Danisovic
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.S.); (Z.V.N.); (A.N.); (S.H.); (M.C.); (L.D.)
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (D.I.); (J.P.); (S.B.)
| |
Collapse
|
48
|
Haddouti EM, Reinhardt N, Ossendorff R, Burger C, Wirtz DC, de la Fuente M, Schildberg FA. Effects of single and repeated shock wave application on the osteogenic differentiation potential of human primary mesenchymal stromal cells and the osteoblastic cell line MG63 in vitro. Front Bioeng Biotechnol 2023; 11:1207655. [PMID: 37901841 PMCID: PMC10602737 DOI: 10.3389/fbioe.2023.1207655] [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: 04/17/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction: Extracorporeal shock wave therapy is a non-invasive and effective option for treating various musculoskeletal disorders. Recent literature indicates that the parameters for extracorporeal shock wave therapy, such as the optimal intensity, treatment frequency, and localization, are yet to be determined. Studies reporting on the effects of shock wave application on primary mesenchymal stromal cells (MSCs) as well as osteoblastic cell lines in vitro are barely available and not standardized. Methods: In this study, we designed a special setup to precisely expose primary MSCs and the osteoblastic cell line MG63 to shock waves and subsequently analyzed the resulting cellular responses using standardized protocols to investigate their viability, proliferation behavior, cytokine secretion, and osteogenic differentiation potential in vitro. The shock wave transducer was coupled to a specifically designed water bath containing a 5 mL tube holder. Primary human MSCs and MG63 cells were trypsinated and centrifuged in a 5 mL tube and exposed to single and repeated shock wave application using different intensities and numbers of pulses. Results: Single treatment of MSCs using intensities 5, 10, 15, and 20 and pulse numbers 100, 250, 500, 750, and 1,000 at a constant pulse repetition frequency of 1 Hz resulted in a decreased viability and proliferation of both cell types with an increase in the intensity and number of pulses compared to controls. No significant difference in the osteogenic differentiation was observed at different time intervals in both cell types when a single shock wave application was performed. However, repeated shock wave sessions over three consecutive days of primary MSCs using low intensity levels 0.1 and 1 showed significant osteogenic differentiation 4-fold higher than that of the extracted Alizarin Red S at day 14, whereas MG63 cells showed no significant osteogenic differentiation compared to their corresponding controls. More specifically, repeated shock wave application triggered a significant downregulation of COL1A1, upregulation of RUNX2, and sustained increase of OCN in primary MSCs but not in the cell line MG63 when induced toward the osteogenic differentiation. Discussion: The effects of shock wave application on MSCs make it an effective therapy in regenerative medicine. We established a protocol to analyze a standardized shock wave application on MSCs and were able to determine conditions that enhance the osteogenic differentiation of MSCs in vitro.
Collapse
Affiliation(s)
- El-Mustapha Haddouti
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Nina Reinhardt
- Chair of Medical Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Matias de la Fuente
- Chair of Medical Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
49
|
Ou L, Tan X, Qiao S, Wu J, Su Y, Xie W, Jin N, He J, Luo R, Lai X, Liu W, Zhang Y, Zhao F, Liu J, Kang Y, Shao L. Graphene-Based Material-Mediated Immunomodulation in Tissue Engineering and Regeneration: Mechanism and Significance. ACS NANO 2023; 17:18669-18687. [PMID: 37768738 DOI: 10.1021/acsnano.3c03857] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Tissue engineering and regenerative medicine hold promise for improving or even restoring the function of damaged organs. Graphene-based materials (GBMs) have become a key player in biomaterials applied to tissue engineering and regenerative medicine. A series of cellular and molecular events, which affect the outcome of tissue regeneration, occur after GBMs are implanted into the body. The immunomodulatory function of GBMs is considered to be a key factor influencing tissue regeneration. This review introduces the applications of GBMs in bone, neural, skin, and cardiovascular tissue engineering, emphasizing that the immunomodulatory functions of GBMs significantly improve tissue regeneration. This review focuses on summarizing and discussing the mechanisms by which GBMs mediate the sequential regulation of the innate immune cell inflammatory response. During the process of tissue healing, multiple immune responses, such as the inflammatory response, foreign body reaction, tissue fibrosis, and biodegradation of GBMs, are interrelated and influential. We discuss the regulation of these immune responses by GBMs, as well as the immune cells and related immunomodulatory mechanisms involved. Finally, we summarize the limitations in the immunomodulatory strategies of GBMs and ideas for optimizing GBM applications in tissue engineering. This review demonstrates the significance and related mechanism of the immunomodulatory function of GBM application in tissue engineering; more importantly, it contributes insights into the design of GBMs to enhance wound healing and tissue regeneration in tissue engineering.
Collapse
Affiliation(s)
- Lingling Ou
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiner Tan
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shijia Qiao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junrong Wu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuan Su
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528399, China
| | - Wenqiang Xie
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Nianqiang Jin
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jiankang He
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ruhui Luo
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xuan Lai
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wenjing Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fujian Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiyuan Kang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
50
|
Ren X, Zhuang H, Zhang Y, Zhou P. Cerium oxide nanoparticles-carrying human umbilical cord mesenchymal stem cells counteract oxidative damage and facilitate tendon regeneration. J Nanobiotechnology 2023; 21:359. [PMID: 37789395 PMCID: PMC10546722 DOI: 10.1186/s12951-023-02125-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Tendon injuries have a high incidence and limited treatment options. Stem cell transplantation is essential for several medical conditions like tendon injuries. However, high local concentrations of reactive oxygen species (ROS) inhibit the activity of transplanted stem cells and hinder tendon repair. Cerium oxide nanoparticles (CeONPs) have emerged as antioxidant agents with reproducible reducibility. RESULTS In this study, we synthesized polyethylene glycol-packed CeONPs (PEG-CeONPs), which were loaded into the human umbilical cord mesenchymal stem cells (hUCMSCs) to counteract oxidative damage. H2O2 treatment was performed to evaluate the ROS scavenging ability of PEG-CeONPs in hUCMSCs. A rat model of patellar tendon defect was established to assess the effect of PEG-CeONPs-carrying hUCMSCs in vivo. The results showed that PEG-CeONPs exhibited excellent antioxidant activity both inside and outside the hUCMSCs. PEG-CeONPs protect hUCMSCs from senescence and apoptosis under excessive oxidative stress. Transplantation of hUCMSCs loaded with PEG-CeONPs reduced ROS levels in the tendon injury area and facilitated tendon healing. Mechanistically, NFκB activator tumor necrosis factor α and MAPK activator dehydrocrenatine, reversed the therapeutic effect of PEG-CeONPs in hUCMSCs, indicating that PEG-CeONPs act by inhibiting the NFκB and MAPK signaling pathways. CONCLUSIONS The carriage of the metal antioxidant oxidase PEG-CeONPs maintained the ability of hUCMSCs in the injured area, reduced the ROS levels in the microenvironment, and facilitated tendon regeneration. The data presented herein provide a novel therapeutic strategy for tendon healing and new insights into the use of stem cells for disease treatment.
Collapse
Affiliation(s)
- Xunshan Ren
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huangming Zhuang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuelong Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Panghu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|