1
|
Chen Y, Xu Y, Cai J, Lauwers M, Xiang L, Zheng Y, Chu H, Chen X, Ker DFE, Zhang C, Wang DM, Zhang Z. Automated and Enclosed Three-Dimensional Biofabrication System for Mesenchymal Stem Cell Culture to Enhance Diabetic Wound Healing. Biomater Res 2025; 29:0205. [PMID: 40421058 PMCID: PMC12104559 DOI: 10.34133/bmr.0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/01/2025] [Accepted: 04/19/2025] [Indexed: 05/28/2025] Open
Abstract
The industrialization of mesenchymal stem cells for regenerative medicine faces substantial challenges, particularly in large-scale production. Conventional 2-dimensional (2D) culture systems demonstrate limitations in meeting clinical requirements, such as inadequate cell yield, and poor cell-cell and cell-matrix interactions. These challenges can potentially be addressed by employing a 3D culture platform, which offers higher cell yields and enhanced efficacy. Moreover, it is essential to conduct a systematic and rigorous evaluation of cells produced in 3D culture systems to ensure their successful clinical translation. In this study, we cultured human umbilical cord mesenchymal stem cells (hUCMSCs) using an automated, scalable, and enclosed 3D microcarrier-bioreactor system, and comprehensively investigated their biological characteristics and potential therapeutic effects for diabetic wound repair. Our findings revealed that hUCMSCs harvested from this 3D microcarrier-bioreactor system are genetically stable and maintain the trilineage differentiation potential. Compared to hUCMSCs expanded under 2D conditions, those cultured in 3D exhibited reduced senescence and enhanced capabilities in migration, angiogenesis, and anti-inflammatory responses across different passages in vitro. RNA-sequencing analysis showed higher expression levels of genes related to angiogenesis and anti-inflammatory pathways in hUCMSCs cultured in 3D compared to those in 2D, which was further validated using quantitative real-time polymerase chain reaction and Western blot analysis. Additionally, 3D-cultured hUCMSCs demonstrated superior therapeutic effects for diabetic wound repair in mice, potentially due to their enhanced angiogenetic and anti-inflammatory effects. Collectively, our finding showcases the high quality of hUCMSCs cultured using an automated and enclosed 3D microcarrier-bioreactor system and their promising potential for diabetic wound repair.
Collapse
Affiliation(s)
- Yanmei Chen
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yang Xu
- School of Biomedical Sciences, Faculty of Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiawei Cai
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Marianne Lauwers
- Centre for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
| | - Liwei Xiang
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yali Zheng
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hua Chu
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xianglong Chen
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dai Fei Elmer Ker
- Department of Biomedical Engineering, Faculty of Engineering, The
Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Cheng Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dan Michelle Wang
- School of Biomedical Sciences, Faculty of Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
- Institute for Tissue Engineering and Regenerative Medicine,
The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology,
The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhiyong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing, Department of Orthopaedic Surgery, Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Li JZ, Zhan X, Sun HB, Chi C, Zhang GF, Liu DH, Zhang WX, Sun LH, Kang K. L-arginine from elder human mesenchymal stem cells induces angiogenesis and enhances therapeutic effects on ischemic heart diseases. World J Stem Cells 2025; 17:103314. [PMID: 40308887 PMCID: PMC12038462 DOI: 10.4252/wjsc.v17.i4.103314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/06/2025] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapy may be a future treatment for myocardial infarction (MI). However, few studies have assessed the therapeutic efficacy of adipose tissue-derived MSCs (ADSCs) obtained from elderly patients in comparison to that of bone marrow-derived MSCs (BMSCs) from the same elderly patients. The metabolomics results revealed a significantly higher L-arginine excretion from aged ADSCs vs BMSCs in hypoxic conditions. This was hypothesized as the possible mechanism that ADSCs showed an improved angiogenic capacity and enhanced the therapeutic effect on ischemic heart diseases. AIM To investigate the role of L-arginine in enhancing angiogenesis and cardiac protection by comparing ADSCs and BMSCs in hypoxic conditions for MI therapy. METHODS Metabolomic profiling of supernatants from ADSCs and BMSCs under hypoxic conditions were performed. Then, arginine succinate lyase (ASL) overexpression and short hairpin RNA plasmid were prepared and transfected into BMSCs. Subsequently, in vitro wound healing and Matrigel tube formation assays were used to verify the proangiogenetic effects of ADSC positive control, BMSCs, BMSCs ASL short hairpin RNA, BMSCs ASL overexpressed, and BMSC negative control on cocultured human umbilical vein endothelial cells. All sample sizes, which were determined to meet the statistical requirements and be greater than 3, were established on the basis of previously established literature standards. The protein levels of vascular endothelial growth factor (VEGF), basic fibroblast growth factor, etc. were detected. In vivo, the five types of cells were transplanted into the infarcted area of MI rat models, and the therapeutic effects of the transplanted cells were evaluated by echocardiography on cardiac function and by Masson's staining/terminal-deoxynucleotidyl transferase mediated nick end labeling assay/immunofluorescence detection on the infarcted area. RESULTS Metabolomic analysis showed that L-arginine was increased. Using ASL gene transfection, we upregulated the production of L-arginine in aged patient-derived BMSCs in vitro, which in turn enhanced mitogen activated protein kinase and VEGF receptor 2 protein expression, VEGF and basic fibroblast growth factor secretion, and inductive angiogenesis to levels comparable to donor-matched ADSCs. After the cell transplantation in vivo, the modified BMSCs as well as ADSCs exhibited decreased apoptotic cells, enhanced vessel formation, reduced scar size, and improved cardiac function in the MI rat model. The therapeutic efficacy decreased by inhibiting L-arginine synthesis. CONCLUSION L-arginine is important for inducing therapeutic angiogenesis for ADSCs and BMSCs in hypoxic conditions. ADSCs have higher L-arginine secretion, which leads to better angiogenesis induction and cardiac protection. ADSC transplantation is a promising autologous cell therapy strategy in the context of the present aging society.
Collapse
Affiliation(s)
- Jian-Zhong Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Key Laboratory of Cell Transplantation of the National Ministry of Public Health, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710014, Shaanxi Province, China
| | - Xu Zhan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Key Laboratory of Cell Transplantation of the National Ministry of Public Health, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Hao-Bo Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Key Laboratory of Cell Transplantation of the National Ministry of Public Health, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Chao Chi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Guo-Fu Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Dong-Hui Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Wen-Xi Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Li-Hua Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University and Pharmacology Department of Pharmacy College of Harbin Medical University, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Kai Kang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Key Laboratory of Cell Transplantation of the National Ministry of Public Health, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, Heilongjiang Province, China.
| |
Collapse
|
3
|
Jain I, Chan AHP, Yang G, He H, Lam J, Sung K, Huang NF. Combinatorial extracellular matrix tissue chips for optimizing mesenchymal stromal cell microenvironment and manufacturing. NPJ Regen Med 2025; 10:21. [PMID: 40263357 PMCID: PMC12015357 DOI: 10.1038/s41536-025-00408-z] [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: 07/24/2024] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Despite the therapeutic potential of mesenchymal stromal cells (MSC), there is limited understanding of optimal extracellular matrix (ECM) environments to manufacture these cells. We developed tissue chips to study the effects of multi-factorial ECM environments under manufacturable stiffness ranges and multi-component ECM compositions. Manufacturing qualities of cell expansion potential, immunomodulation, and differentiation capacity were examined. The results show stiffness effects, with 900 kPa substrates supporting higher proliferation and osteogenic differentiation, along with anti-inflammatory IL-10 expression, whereas 150 kPa substrates promoted adipogenic differentiation at 150 kPa, suggesting that optimal ECM environments may differ based on manufacturing goals. ECM biochemistries containing fibronectin and laminin further modulated MSC manufacturing qualities across various stiffnesses. Proteomic and transcriptomic analyses revealed unique ECM combinations that induced higher levels of angiogenic and immunomodulatory cytokines, compared to single factor ECMs. These findings demonstrate that optimized ECM environments enhance MSC manufacturing quality.
Collapse
Affiliation(s)
- Ishita Jain
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Alex H P Chan
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Guang Yang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
- Epicrispr Biotechnologies, South San Francisco, CA, USA
| | - Hao He
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA
| | - Johnny Lam
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Kyung Sung
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA.
- Center for Tissue Regeneration, Repair and Restoration & Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
4
|
Park H, Kim HJ, Kim IG, Kim MJ, Kim Y, Eom SY, Seok J, Oh SH, Chung EJ. Esophageal Reconstruction with Myogenesis-Inducing Gene Transfected Mesenchymal Stem Cell-Seeded Film with Leaf-Stacked Structure. ACS Biomater Sci Eng 2025; 11:2274-2289. [PMID: 40183365 DOI: 10.1021/acsbiomaterials.4c02396] [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: 04/05/2025]
Abstract
In the domain of tissue engineering and regenerative medicine, artificial replacements have been developed as viable options for esophageal reconstruction and serve as alternatives to traditional surgical procedures. Restoration of smooth muscle functionality is crucial in esophageal regeneration. We evaluated the efficacy of esophageal reconstructions in an animal model, using tissue-engineered films with a leaf-stacked structure (FLSS), seeded with mesenchymal stem cells (MSCs), which were genetically modified with myogenic genes. Esophageal partial defects were variously reconstructed in animals (n = 8 per group, except the no-implantation group), categorized as (1) normal rats; (2) rats implanted with naked FLSS; (3) rats implanted with FLSS with MSCs; (4) rats implanted using FLSS with myogenesis-inducing gene transfected MSCs; and (5) rats without implantation at the defect site (n = 3). The FLSS exhibited appropriate mechanical characteristics for transplantation. Successful repair of esophageal defects was observed with significantly enhanced epithelial regeneration in the MSC-seeded FLSS group compared to that in the naked FLSS group. Moreover, smooth muscle regeneration was notably higher in the FLSS with myogenesis-inducing gene transfected MSCs than in the group without myogenic gene transfection. The myogenesis-inducing gene-transfected MSC-seeded FLSS group showed a tendency toward increased smooth muscle regeneration, this indicates that FLSS with myogenesis-inducing genes transfected MSC may contribute positively to the maintenance of function in the reconstructed esophagus.
Collapse
Affiliation(s)
- Hanaro Park
- Department of Otorhinolaryngology- Head and Neck Surgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon 51353, Republic of Korea
| | - Hye-Joung Kim
- Institute of Chemical Engineering Convergence System, Korea University, Seoul 02841, Republic of Korea
| | - In Gul Kim
- Department of Otorhinolaryngology- Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Min Ji Kim
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Yewon Kim
- Department of Otorhinolaryngology- Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - So Young Eom
- Institute of Chemical Engineering Convergence System, Korea University, Seoul 02841, Republic of Korea
| | - Jungirl Seok
- Department of Otorhinolaryngology- Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Se Heang Oh
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Republic of Korea
| | - Eun-Jae Chung
- Department of Otorhinolaryngology- Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
5
|
Xiang YY, Won JH, Kim JS, Baek KW. Transplantation of Exercise-Enhanced Mesenchymal Stem Cells Improves Obesity and Glucose Tolerance via Immune Modulation in Adipose Tissue. Stem Cell Rev Rep 2025:10.1007/s12015-025-10881-0. [PMID: 40227488 DOI: 10.1007/s12015-025-10881-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Exercise-conditioned mesenchymal stem cells (MSCs) may modulate immune responses and improve white adipose tissue (WAT) function. While MSCs are known to reduce inflammation, it remains unclear if exercise-stimulated MSCs can improve obesity-related dysfunctions. This study is the first to explore how exercise-conditioned MSCs may influence adipose tissue inflammation and remodeling in the context of obesity. MSCs were isolated from exercised- and sedentary donor mice, then cultured in vitro. After culture, MSCs were assessed for differentiation capacity and cytokine gene expression, including Il10, as indicators of immune modulation. Exercise-conditioned MSCs were then transplanted into obese recipient mice. Following transplantation, immune cell profiles, inflammatory markers, and adipocyte morphology in recipient WAT were analyzed. Flow cytometry was used to quantify macrophage subtypes (pro-inflammatory and anti-inflammatory), and histological analysis was performed to measure changes in adipocyte size. Exercise-activated MSCs showed a ± 35% increase in Il10 expression and a ± 20% enhancement in differentiation capacity compared to controls, indicating improved immunomodulatory potential. In recipient mice, transplantation led to a ± 25% reduction in pro-inflammatory macrophages (CD86+ CD206-) and a 15% decrease in adipocyte size within WAT. Additionally, WAT in treated mice showed balanced inflammatory profiles and reduced adipose hypertrophy, suggesting restored immune balance and metabolic health. These findings suggest that exercise-modified MSCs exhibit enhanced immunomodulatory and metabolic regulatory properties. This study provides evidence that exercise enhances MSC characteristics, potentially improving their capacity to modulate adipose tissue immune balance and metabolic function in obesity. Exercise-conditioned MSCs may serve as a foundation for future strategies that integrate exercise-induced stem cell modifications to modulate obesity-related metabolic dysfunction.
Collapse
Affiliation(s)
- Ying-Ying Xiang
- Department of Physical Education, Gyeongsang National University, Jinju, 52828, Korea
| | - Jong-Hwa Won
- Department of Physical Education, Gyeongsang National University, Jinju, 52828, Korea
| | - Ji-Seok Kim
- Department of Physical Education, Gyeongsang National University, Jinju, 52828, Korea
- Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, 52828, Korea
| | - Kyung-Wan Baek
- Department of Physical Education, Gyeongsang National University, Jinju, 52828, Korea.
- Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, 52828, Korea.
| |
Collapse
|
6
|
Lee E, Lim GH, An JH, Ryu MO, Seo KW, Youn HY. Enhanced immunomodulatory effects of canine adipose tissue-derived mesenchymal stem cells in 3D culture. Front Vet Sci 2025; 12:1500267. [PMID: 40206260 PMCID: PMC11979191 DOI: 10.3389/fvets.2025.1500267] [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: 09/23/2024] [Accepted: 03/03/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) have been introduced as a treatment for dogs owing to their immunomodulatory effects. In humans, 3D-cultured MSCs have recently been applied in treating various conditions, including myocardial infarction, liver disease, and kidney disease. This study aimed to evaluate whether the immunomodulatory effects of canine adipose tissue-derived MSCs (cAT-MSCs) are enhanced when cultured in a 3D environment compared to conventional 2D culture. Methods cAT-MSC spheroids were generated using ultra-low-adhesion plates. The structural and hypoxic characteristics of these spheroids were assessed via confocal imaging. The expression levels of the stemness markers SOX2 and OCT4 were examined through western blotting. Additionally, the expression of inflammatory factors within the cAT-MSC spheroids was analyzed using RT-PCR and ELISA. The immunomodulatory effects were further evaluated in canine macrophages (DH82) treated with conditioned media (CM) from cAT-MSC spheroids, using RT-PCR and flow cytometry. Results 3D culture induced hypoxic conditions within the cAT-MSC spheroids and significantly increased the expression of SOX2 and OCT4 (p < 0.05). Moreover, the expression of inflammation-associated factors, including TGF-β1, TSG-6, COX-2, PGE2, and IL-10, was upregulated in the 3D culture (p < 0.05). Treatment of DH82 cells with CM from the cAT-MSC spheroids led to a significant reduction in the expression of pro-inflammatory factors such as TNF-α, IL-1β, and IL-6 (p < 0.01). Additionally, M1 polarization was diminished in DH82 cells exposed to the CM from the cAT-MSC spheroids (p < 0.0001). And M2 polarization was increased in DH82 cells exposed to the CM from the cAT-MSC spheroids (p < 0.0001). Conclusion This study confirms that the immunomodulatory effects of MSCs are enhanced in 3D culture. Therefore, 3D cultured MSCs may offer a more effective therapeutic approach than conventional 2D-cultured MSCs for treating canine inflammatory diseases.
Collapse
Affiliation(s)
- Eunbi Lee
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ga-Hyun Lim
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- VIP Animal Medical Center, Seoul, Republic of Korea
| | - Ju-Hyun An
- Department of Veterinary Emergency and Critical Care Medicine and Institute of Veterinary Science, College of Veterinary Medicine, Kangwon National University, Chuncheon-si, Republic of Korea
| | - Min-Ok Ryu
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyoung-Won Seo
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Cho GH, Bae HC, Lee YJ, Yang HR, Kang H, Park HJ, Wang SY, Kim YJ, Kang HS, Kim IG, Choi BS, Han HS. Insulin-Like Growth Factor 2 Secreted from Mesenchymal Stem Cells with High Glutathione Levels Alleviates Osteoarthritis via Paracrine Rejuvenation of Senescent Chondrocytes. Biomater Res 2025; 29:0152. [PMID: 39990979 PMCID: PMC11842674 DOI: 10.34133/bmr.0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/25/2025] Open
Abstract
Senescent chondrocytes, which are increased in osteoarthritic (OA) cartilage, promote cartilage defects and the senescent knee microenvironment by inducing senescence to surrounding normal chondrocytes by secreting senescence-associated secretory proteins. Many studies have used mesenchymal stem cells (MSCs) to treat OA, but MSC treatment remains challenging for clinical application owing to MSC quality control, engraftment, and fibrocartilage regeneration. Here, rather than relying on the direct regeneration of MSCs, we present a novel strategy to suppress OA by MSC-mediated senescent chondrocyte targeting via the paracrine activity of MSCs, thereby improving the knee microenvironment. First, to enable quality control of umbilical cord MSCs, priming MSCs by supplementing human platelet lysate (hPL) greatly enhanced MSC functions by increasing cellular glutathione levels throughout serial passaging. Intra-articular injection of primed MSCs successfully suppressed OA progression and senescent chondrocyte accumulation without direct regeneration. Indirect coculture with primed MSCs using transwell ameliorated the senescence phenotypes in OA chondrocytes, suggesting paracrine rejuvenation. Based on secretome analysis, we identified insulin-like growth factor 2 (IGF2) as a key component that induces paracrine rejuvenation by primed MSCs. The rejuvenation effects of IGF2 act through autophagy activation through the up-regulation of autophagy-related gene expression and autophagic flux. To cross-validate the effects of secreted IGF2 in vivo, knockdown of IGF2 in primed MSCs substantially abolished its therapeutic efficacy in a rabbit OA model. Collectively, these findings demonstrate that hPL supplementation enables MSC quality control by increasing MSC glutathione levels. The therapeutic mechanism of primed MSCs was secreted IGF2, which induces paracrine rejuvenation of senescent OA chondrocytes by activating autophagy.
Collapse
Affiliation(s)
- Gun Hee Cho
- Interdisciplinary Programs: Stem Cell Biology, College of Medicine,
Seoul National University, Seoul 03080, Korea
- Department of Orthopedic Surgery, College of Medicine,
Seoul National University, Seoul 03080, Korea
| | - Hyun Cheol Bae
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| | - Yu Jeong Lee
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| | - Ha Ru Yang
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| | - Hyewon Kang
- Laboratory for Cellular Response to Oxidative Stress, Cell2in Inc., Seoul 03127, Korea
| | - Hee Jung Park
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| | - Sun Young Wang
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| | - You Jung Kim
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| | - Heun-Soo Kang
- Laboratory for Cellular Response to Oxidative Stress, Cell2in Inc., Seoul 03127, Korea
| | - In Gyu Kim
- Laboratory for Cellular Response to Oxidative Stress, Cell2in Inc., Seoul 03127, Korea
| | - Byung Sun Choi
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| | - Hyuk-Soo Han
- Interdisciplinary Programs: Stem Cell Biology, College of Medicine,
Seoul National University, Seoul 03080, Korea
- Department of Orthopedic Surgery, College of Medicine,
Seoul National University, Seoul 03080, Korea
- Department of Orthopedic Surgery,
Seoul National University Hospital, Seoul 110-744, Korea
| |
Collapse
|
8
|
Ibrahim E, Gaafar AH, Mehanna RA, El Achy S, Taha AH. Effect of Application of Bone Marrow Mesenchymal Stem Cells After Subglottic Injury: Animal Study. Laryngoscope 2025. [DOI: 10.1002/lary.32070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/05/2025] [Indexed: 05/03/2025]
Abstract
ABSTRACTObjectiveAcquired laryngotracheal stenosis (LTS) is notorious for recurrence and the need for repeated procedures to achieve a satisfactory outcome. This limitation is, at least partly, due to the suboptimal healing process of the airway following trauma. Consequently, innovative interventions are required to improve wound healing, which would reflect on improving the outcome of the management of LTS. The objective of this study is to examine the effect of the application of BM‐MSCs in preventing subglottic acute stenosis following trauma.MethodsTwenty‐four Sprague–Dawley rats were divided into three equal groups (n = 8): two intervention groups and one control group. Subglottic trauma was performed in the three groups. In the intervention groups, bone marrow‐derived mesenchymal stem cells (BM‐MSCs) were applied either systemically (TS group) or via local injection (TL group). Subglottic specimens from the three groups were compared by histopathological appearance, lamina propria (LP) thickness, and luminal anteroposterior (AP) diameter.ResultsAn intense inflammatory reaction was observed in the control specimens, while a mild inflammatory reaction was detected in the two intervention groups. The LP thickness and the AP diameter were significantly different between the control group and the two intervention groups.ConclusionsIn the present animal model, stem cell application, either systemic or local application, can mitigate subglottic inflammation and stenosis after trauma.
Collapse
Affiliation(s)
| | | | | | - Samar El Achy
- Faculty of Medicine Alexandria University Alexandria Egypt
| | | |
Collapse
|
9
|
Soltanmohammadi F, Mahmoudi Gharehbaba A, Alizadeh E, Javadzadeh Y. Innovative approaches to tissue engineering: Utilizing decellularized extracellular matrix hydrogels for mesenchymal stem cell transport. Int J Biol Macromol 2025; 290:138893. [PMID: 39706433 DOI: 10.1016/j.ijbiomac.2024.138893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
In recent years, the realm of tissue regeneration experienced significant advancements, leading to the development of innovative therapeutic agents. The systemic delivery of mesenchymal stem cells (MSCs) emerged as a promising strategy for promoting tissue regeneration. However, this approach is hindered by hurdles such as poor cell survival, limited cell propagation, and inadequate cell integration. Decellularized extracellular matrix (dECM) hydrogel serves as an innovative carrier that protects MSCs from the detrimental effects of the hostile microenvironment, facilitates their localization and retention at the injection site, and preserves their viability. Regarding its low immunogenicity, low cytotoxicity, high biocompatibility, and its ability to mimic natural extracellular matrix (ECM), this natural hydrogel offers a new avenue for systemic delivery of MSCs. This review digs into the properties of dECM hydrogels (dECMHs), the methods employed for decellularization and the utilization of dECMH as carriers for various types of MSCs for tissue regeneration purposes. This review also sheds light on the benefits of hybrid hydrogels composed of dECMH and other components such as proteins and polysaccharides. By addressing the limitations of conventional hydrogels and enhancing efficacy of cell therapy, dECMH opens new pathways for the future of tissue regeneration.
Collapse
Affiliation(s)
- Fatemeh Soltanmohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Adel Mahmoudi Gharehbaba
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Effat Alizadeh
- Endocrin Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Yousef Javadzadeh
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Heo S, Noh M, Kim Y, Park S. Stem Cell-Laden Engineered Patch: Advances and Applications in Tissue Regeneration. ACS APPLIED BIO MATERIALS 2025; 8:62-87. [PMID: 39701826 DOI: 10.1021/acsabm.4c01427] [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: 12/21/2024]
Abstract
Stem cell-based therapies are emerging as significant approaches in tissue engineering and regenerative medicine, applicable to both fundamental scientific research and clinical practice. Despite remarkable results in clinical studies, challenges such as poor standardization of graft tissues, limited sources, and reduced functionality have hindered the effectiveness of these therapies. In this review, we summarize the engineering approaches involved in fabricating stem cell assisted patches and the substantial strategies for designing stem cell-laden engineered patches (SCP) to complement the existing stem cell-based therapies. We then outline the potential applications of SCP in advancing tissue regeneration and regenerative medicine. By combining living stem cells with engineered patches, SCP can enhance the functions of both components, particularly for tissue engineering applications. Finally, we addressed current challenges, such as ethical considerations, high costs, and regulatory hurdles and proposed future research directions to overcome these barriers.
Collapse
Affiliation(s)
- Seyeong Heo
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Minhyeok Noh
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Yeonseo Kim
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| | - Sunho Park
- Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang 50463, Republic of Korea
| |
Collapse
|
11
|
Lica JJ, Jakóbkiewicz-Banecka J, Hellmann A. In Vitro models of leukemia development: the role of very small leukemic stem-like cells in the cellular transformation cascade. Front Cell Dev Biol 2025; 12:1463807. [PMID: 39830209 PMCID: PMC11740207 DOI: 10.3389/fcell.2024.1463807] [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: 07/15/2024] [Accepted: 11/28/2024] [Indexed: 01/22/2025] Open
Abstract
Recent experimental findings indicate that cancer stem cells originate from transformed very small embryonic-like stem cells. This finding represents an essential advancement in uncovering the processes that drive the onset and progression of cancer. In continuously growing cell lines, for the first time, our team's follow-up research on leukemia, lung cancer, and healthy embryonic kidney cells revealed stages that resembles very small precursor stem cells. This review explores the origin of leukemic stem-like cells from very small leukemic stem-like cells establish from transformed very small embryonic-like stem cells. We explore theoretical model of acute myeloid leukemia initiation and progresses through various stages, as well basing the HL60 cell line, present its hierarchical stage development in vitro, highlighting the role of these very small precursor primitive stages. We also discuss the potential implications of further research into these unique cellular stages for advancing leukemia and cancer treatment and prevention.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Department Medical Biology and Genetics, Faculty of Biology, University of Gdansk, Gdansk, Poland
- Department Health Science; Powiśle University, Gdańsk, Poland
| | | | - Andrzej Hellmann
- Department of Hematology and Transplantology, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
12
|
Mercurio M, Minici R, Spina G, Cofano E, Laganà D, Familiari F, Galasso O, Gasparini G. Clinical and Radiological Outcomes of combined Arthroscopic Microfracture and Mesenchymal Stem Cell Injection Versus Isolated Microfracture for Osteochondral Lesions of the Talus: A Meta-Analysis of Comparative Studies. J Foot Ankle Surg 2025; 64:103-109. [PMID: 39182822 DOI: 10.1053/j.jfas.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/01/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
We aimed to systematically evaluate the clinical and radiological outcomes reported in comparative studies evaluating combined arthroscopic microfracture and mesenchymal stem cell (MSC) injection versus isolated microfracture for osteochondral lesions of the talus (OLT). A total of 5 studies were included. Demographics, American Orthopaedic Foot and Ankle Society (AOFAS) score, Tegner activity scale score, Foot and Ankle Outcome Scale (FAOS), visual analogue scale (VAS), and Magnetic Resonance Observation of Cartilage Repair Tissue Score (MOCART) were analyzed. A total of 348 patients were evaluated, 171 of whom underwent combined microfracture and MSC injection and 177 of whom underwent isolated microfracture. The frequency-weighted mean ages were 38.9 ± 13.5 and 36.9 ± 11.4 years and the mean follow-up were 36.7 ± 13.3 and 36.2 ± 16.2 months in the combined microfracture and MSC injection and isolated microfracture groups, respectively. The combined microfracture and MSC injection group showed significantly better postoperative AOFAS score (81.5 ± 7.4 vs 68.2 ± 5.1, p < .001), and MOCART score (74.3 ± 16.3 vs 63.9 ± 15.5, p < .001) with differences beyond the minimum clinically important difference. The combination of arthroscopic microfracture and MSC injection significantly improved functionality and radiological outcomes compared to those of isolated microfracture for OLT.
Collapse
Affiliation(s)
- Michele Mercurio
- Department of Orthopaedic and Trauma Surgery, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100, Catanzaro, Italy; Research Center on Musculoskeletal Health, MusculoSkeletalHealth@UMG, Magna Graecia University, 88100 Catanzaro, Italy.
| | - Roberto Minici
- Radiology Unit, Department of Experimental and Clinical Medicine, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100 Catanzaro, Italy
| | - Giovanna Spina
- Department of Orthopaedic and Trauma Surgery, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100, Catanzaro, Italy
| | - Erminia Cofano
- Department of Orthopaedic and Trauma Surgery, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100, Catanzaro, Italy
| | - Domenico Laganà
- Radiology Unit, Department of Experimental and Clinical Medicine, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100 Catanzaro, Italy
| | - Filippo Familiari
- Department of Orthopaedic and Trauma Surgery, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100, Catanzaro, Italy; Research Center on Musculoskeletal Health, MusculoSkeletalHealth@UMG, Magna Graecia University, 88100 Catanzaro, Italy
| | - Olimpio Galasso
- Department of Orthopaedic and Trauma Surgery, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100, Catanzaro, Italy; Department of Medicine, Surgery and Dentistry, University of Salerno, 84081, Baronissi, Salerno, Italy
| | - Giorgio Gasparini
- Department of Orthopaedic and Trauma Surgery, "Magna Græcia" University, "Renato Dulbecco" University Hospital, 88100, Catanzaro, Italy; Research Center on Musculoskeletal Health, MusculoSkeletalHealth@UMG, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
13
|
Atta H, Kassem DH, Kamal MM, Hamdy NM. Harnessing the ubiquitin proteasome system as a key player in stem cell biology. Biofactors 2025; 51:e2157. [PMID: 39843166 DOI: 10.1002/biof.2157] [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: 08/31/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025]
Abstract
Intracellular proteins take part in almost every body function; thus, protein homeostasis is of utmost importance. The ubiquitin proteasome system (UPS) has a fundamental role in protein homeostasis. Its main role is to selectively eradicate impaired or misfolded proteins, thus halting any damage that could arise from the accumulation of these malfunctioning proteins. Proteasomes have a critical role in controlling protein homeostasis in all cell types, including stem cells. We will discuss the role of UPS enzymes as well as the 26S proteasome complex in stem cell biology from several angles. First, we shall overview common trends of proteasomal activity and gene expression of different proteasomal subunits and UPS enzymes upon passaging and differentiation of stem cells toward various cell lineages. Second, we shall explore the effect of modulating proteasomal activity in stem cells and navigate through the interrelation between proteasomes' activity and various proteasome-related transcription factors. Third, we will shed light on curated microRNAs and long non-coding RNAs using various bioinformatics tools that might have a possible role in regulating UPS in stem cells and possibly, upon manipulation, can enhance the differentiation process into different lineages and/or delay senescence upon cell passaging. This will help to decipher the role played by individual UPS enzymes and subunits as well as various interrelated molecular mediators in stem cells' maintenance and/or differentiation and open new avenues in stem cell research. This can ultimately provide a leap toward developing novel therapeutic interventions related to proteasome dysregulation.
Collapse
Affiliation(s)
- Hind Atta
- School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, Cairo, Egypt
| | - Dina H Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed M Kamal
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Drug Research and Development Group, Health Research Center of Excellence, The British University in Egypt, Cairo, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
14
|
Hu C, Wang L. Advances in the treatment of liver injury based on mesenchymal stem cell-derived exosomes. Stem Cell Res Ther 2024; 15:474. [PMID: 39696473 DOI: 10.1186/s13287-024-04087-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have shown a great potential role in treating liver injury. MSCs can promote liver regeneration by differentiating into hepatocytes, and can also secrete exosomes to participate in the repair of liver injury. Increasing evidence has shown that mesenchymal stem cell-derived exosomes (MSC-EXOs) play an important role in treating liver injury. In this review, the biogenesis and function of exosomes and the characteristics of MSC-EXOs were analyzed based on recent research results. MSC-EXOs are significant in liver injuries such as liver fibrosis, liver failure, hepatocellular carcinoma, oxidative stress, and lipid steatosis, and participate in the process of liver regeneration.
Collapse
Affiliation(s)
- Changlong Hu
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, 710000, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, 710000, China.
| |
Collapse
|
15
|
Tang XL, Wysoczynski M, Gumpert AM, Solanki M, Li Y, Wu WJ, Zheng S, Ruble H, Li H, Stowers H, Zheng S, Ou Q, Tanveer N, Slezak J, Kalra DK, Bolli R. Intravenous infusions of mesenchymal stromal cells have cumulative beneficial effects in a porcine model of chronic ischaemic cardiomyopathy. Cardiovasc Res 2024; 120:1939-1952. [PMID: 39163570 PMCID: PMC11630033 DOI: 10.1093/cvr/cvae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
AIMS The development of cell therapy as a widely available clinical option for ischaemic cardiomyopathy is hindered by the invasive nature of current cell delivery methods. Furthermore, the rapid disappearance of cells after transplantation provides a cogent rationale for using repeated cell doses, which, however, has not been done thus far in clinical trials because it is not feasible with invasive approaches. The goal of this translational study was to test the therapeutic utility of the intravenous route for cell delivery. METHODS AND RESULTS Pigs with chronic ischaemic cardiomyopathy induced by myocardial infarction received one or three intravenous doses of allogeneic bone marrow mesenchymal stromal cells (MSCs) or placebo 35 days apart. Rigour guidelines, including blinding and randomization, were strictly followed. A comprehensive assessment of left ventricular (LV) function was conducted with three independent methods (echocardiography, magnetic resonance imaging, and haemodynamic studies). The results demonstrate that three doses of MSCs improved both load-dependent and independent indices of LV function and reduced myocardial hypertrophy and fibrosis; in contrast, one dose failed to produce most of these benefits. CONCLUSIONS To our knowledge, this is the first study to show that intravenous infusion of a cell product improves LV function and structure in a large animal model of chronic ischaemic cardiomyopathy and that repeated infusions are necessary to produce robust effects. This study, conducted in a clinically relevant model, supports a new therapeutic strategy based on repeated intravenous infusions of allogeneic MSCs and provides a foundation for a first-in-human trial testing this strategy in patients with chronic ischaemic cardiomyopathy.
Collapse
Affiliation(s)
- Xian-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Anna M Gumpert
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Mitesh Solanki
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Yan Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Wen-Jian Wu
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Shirong Zheng
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Halina Ruble
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Hong Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Heather Stowers
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Shengnan Zheng
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Qinghui Ou
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Nida Tanveer
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Bratislava, Slovakia
| | - Dinesh K Kalra
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY 40202
| |
Collapse
|
16
|
Razavi Z, Soltani M, Souri M, van Wijnen AJ. CRISPR innovations in tissue engineering and gene editing. Life Sci 2024; 358:123120. [PMID: 39426588 DOI: 10.1016/j.lfs.2024.123120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/22/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
The CRISPR/Cas9 system is a powerful tool for genome editing, utilizing the Cas9 nuclease and programmable single guide RNA (sgRNA). However, the Cas9 nuclease activity can be disabled by mutation, resulting in catalytically deactivated Cas9 (dCas9). By combining the customizable sgRNA with dCas9, researchers can inhibit specific gene expression (CRISPR interference, CRISPRi) or activate the expression of a target gene (CRISPR activation, CRISPRa). In this review, we present the principles and recent advancements of these CRISPR technologies, as well as their delivery vectors. We also explore their applications in stem cell engineering and regenerative medicine, with a focus on in vitro stem cell fate manipulation and in vivo treatments. These include the prevention of retinal and muscular degeneration, neural regeneration, bone regeneration, cartilage tissue engineering, and the treatment of blood, skin, and liver diseases. Furthermore, we discuss the challenges of translating CRISPR technologies into regenerative medicine and provide future perspectives. Overall, this review highlights the potential of CRISPR in advancing regenerative medicine and offers insights into its application in various areas of research and therapy.
Collapse
Affiliation(s)
- ZahraSadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Biochemistry Research Center, Iran University Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada; Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada; Centre for Sustainable Business, International Business University, Toronto, Canada.
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Andre J van Wijnen
- Department of Biochemistry, University of Vermont, Burlington, VT, USA; Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
17
|
Liu S, Ren J, Hu Y, Zhou F, Zhang L. TGFβ family signaling in human stem cell self-renewal and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:26. [PMID: 39604763 PMCID: PMC11602941 DOI: 10.1186/s13619-024-00207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Human stem cells are undifferentiated cells with the capacity for self-renewal and differentiation into distinct cell lineages, playing important role in the development and maintenance of diverse tissues and organs. The microenvironment of stem cell provides crucial factors and components that exert significant influence over the determination of cell fate. Among these factors, cytokines from the transforming growth factor β (TGFβ) superfamily, including TGFβ, bone morphogenic protein (BMP), Activin and Nodal, have been identified as important regulators governing stem cell maintenance and differentiation. In this review, we present a comprehensive overview of the pivotal roles played by TGFβ superfamily signaling in governing human embryonic stem cells, somatic stem cells, induced pluripotent stem cells, and cancer stem cells. Furthermore, we summarize the latest research and advancements of TGFβ family in various cancer stem cells and stem cell-based therapy, discussing their potential clinical applications in cancer therapy and regeneration medicine.
Collapse
Affiliation(s)
- Sijia Liu
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiang Ren
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanmei Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Long Zhang
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
18
|
Safi R, Mohsen-Kanson T, Kouzi F, El-Saghir J, Dermesrobian V, Zugasti I, Zibara K, Menéndez P, El Hajj H, El-Sabban M. Direct Interaction Between CD34 + Hematopoietic Stem Cells and Mesenchymal Stem Cells Reciprocally Preserves Stemness. Cancers (Basel) 2024; 16:3972. [PMID: 39682159 DOI: 10.3390/cancers16233972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES A specialized microenvironment in the bone marrow, composed of stromal cells including mesenchymal stem cells (MSCs), supports hematopoietic stem cell (HSC) self-renewal, and differentiation bands play an important role in leukemia development and progression. The reciprocal direct interaction between MSCs and CD34+ HSCs under physiological and pathological conditions is yet to be fully characterized. METHODS Here, we established a direct co-culture model between MSCs and CD34+ HSCs or MSCs and acute myeloid leukemia cells (THP-1, Molm-13, and primary cells from patients) to study heterocellular communication. RESULTS Following MSCs-CD34+ HSCs co-culture, the expression of adhesion markers N-Cadherin and connexin 43 increased in both cell types, forming gap junction channels. Moreover, the clonogenic potential of CD34+ HSCs was increased. However, direct contact of acute myeloid leukemia cells with MSCs reduced the expression levels of connexin 43 and N-Cadherin in MSCs. The impairment in gap junction formation may potentially be due to a defect in the acute myeloid leukemia-derived MSCs. Interestingly, CD34+ HSCs and acute myeloid leukemia cell lines attenuated MSC osteoblastic differentiation upon prolonged direct cell-cell contact. CONCLUSIONS In conclusion, under physiological conditions, connexin 43 and N-Cadherin interaction preserves stemness of both CD34+ HSCs and MSCs, a process that is compromised in acute myeloid leukemia, pointing to the possible role of gap junctions in modulating stemness.
Collapse
Affiliation(s)
- Rémi Safi
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
| | - Tala Mohsen-Kanson
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Farah Kouzi
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Jamal El-Saghir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vera Dermesrobian
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Laboratory of Adaptive Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Inés Zugasti
- Department of Hematology, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Kazem Zibara
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, 28029 Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), 08028 Barcelona, Spain
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
| |
Collapse
|
19
|
Wang CC, Hu XM, Long YF, Huang HR, He Y, Xu ZR, Qi ZQ. Treatment of Parkinson's disease model with human umbilical cord mesenchymal stem cell-derived exosomes loaded with BDNF. Life Sci 2024; 356:123014. [PMID: 39182566 DOI: 10.1016/j.lfs.2024.123014] [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: 10/30/2023] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
AIMS Parkinson's disease (PD) is a common neurodegenerative disease that has received widespread attention; however, current clinical treatments can only relieve its symptoms, and do not effectively protect dopaminergic neurons. The purpose of the present study was to investigate the therapeutic effects of human umbilical cord mesenchymal stem cell-derived exosomes loaded with brain-derived neurotrophic factor (BDNF-EXO) on PD models and to explore the underlying mechanisms of these effects. MAIN METHODS 6-Hydroxydopamine was used to establish in vivo and in vitro PD models. Western blotting, flow cytometry, and immunofluorescence were used to detect the effects of BDNF-EXO on apoptosis and ferroptosis in SH-SY5Y cells. The in vivo biological distribution of BDNF-EXO was detected using a small animal imaging system, and dopaminergic neuron improvements in brain tissue were detected using western blotting, immunofluorescence, immunohistochemistry, and Nissl and Prussian blue staining. KEY FINDINGS BDNF-EXO effectively suppressed 6-hydroxydopamine-induced apoptosis and ferroptosis in SH-SY5Y cells. Following intravenous administration, BDNF-EXO crossed the blood-brain barrier to reach afflicted brain regions in mice, leading to a notable enhancement in neuronal survival. Furthermore, BDNF-EXO modulated microtubule-associated protein 2 and phosphorylated tau expression, thereby promoting neuronal cytoskeletal stability. Additionally, BDNF-EXO bolstered cellular antioxidant defense mechanisms through the activation of the nuclear factor erythroid 2-related factor 2 signaling pathway, thereby conferring neuroprotection against damage. SIGNIFICANCE The novel drug delivery system, BDNF-EXO, had substantial therapeutic effects in both in vivo and in vitro PD models, and may represent a new treatment strategy for PD.
Collapse
Affiliation(s)
- Can-Can Wang
- Medical College, Guangxi University, Da-Xue-Dong Road No.100, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Xin-Mei Hu
- Medical College, Guangxi University, Da-Xue-Dong Road No.100, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Yu-Fei Long
- Medical College, Guangxi University, Da-Xue-Dong Road No.100, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Hong-Ri Huang
- GuangXi TaiMeiRenSheng Biotechnology Co., LTD., Nanning, Guangxi 530000, China
| | - Ying He
- Medical College, Guangxi University, Da-Xue-Dong Road No.100, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Ran Xu
- Translational Medicine Research Center, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi 530011, China
| | - Zhong-Quan Qi
- Medical College, Guangxi University, Da-Xue-Dong Road No.100, Nanning 530004, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
20
|
Kou Y, Li J, Zhu Y, Liu J, Ren R, Jiang Y, Wang Y, Qiu C, Zhou J, Yang Z, Jiang T, Huang J, Ren X, Li S, Qiu C, Wei X, Yu L. Human Amniotic Epithelial Stem Cells Promote Colonic Recovery in Experimental Colitis via Exosomal MiR-23a-TNFR1-NF-κB Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401429. [PMID: 39378064 PMCID: PMC11600273 DOI: 10.1002/advs.202401429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/20/2024] [Indexed: 11/28/2024]
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, manifests as chronic intestinal inflammation with debilitating symptoms, posing a significant burden on global healthcare. Moreover, current therapies primarily targeting inflammation can lead to immunosuppression-related complications. Human amniotic epithelial stem cells (hAESCs), which exhibit low immunogenicity and ethical acceptability, have gained attention as potential therapeutics. In this study, it is demonstrated that their encapsulation in a hydrogel and administration via anal injection enhanced the colonic mucosal barrier repair in a murine colitis model induced by dextran sodium sulfate during the recovery phase. The underlying mechanism involved the release of exosomes from hAESCs enriched with microRNA-23a-3p, which post-transcriptionally reduced tumor necrosis factor receptor 1 expression, suppressing the nuclear factor-κB pathway in colonic epithelial cells, thus played a key role in inflammation. The novel approach shows potential for IBD treatment by restoring intestinal epithelial homeostasis without the immunosuppressive therapy-associated risks. Furthermore, the approach provides an alternative strategy to target the key molecular pathways involved in inflammation and promotes intestinal barrier function using hAESCs and their secreted exosomes. Overall, this study provides key insights to effectively treat IBD, addresses the unmet needs of patients, and reduces related healthcare burden.
Collapse
Affiliation(s)
- Yaohui Kou
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Jinying Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Yingyi Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Jia Liu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Ruizhe Ren
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Yuanqing Jiang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Yunyun Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Chen Qiu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Jiayi Zhou
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Zhuoheng Yang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Tuoying Jiang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Jianan Huang
- Eye Center the Second Affiliated HospitalSchool of MedicineZhejiang Provincial Key Laboratory of OphthalmologyZhejiang Provincial Clinical Research Center for Eye DiseasesZhejiang Provincial Engineering Institute on Eye DiseasesZhejiang UniversityHangzhouZhejiang310009China
| | - Xiangyi Ren
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Shiguang Li
- Department of ObstetricsWomen's HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310006China
| | - Cong Qiu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| | - Xiyang Wei
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineLiangzhu LaboratoryZhejiang UniversityHangzhouZhejiang310012China
| | - Luyang Yu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw HospitalMOE Laboratory of Biosystems Homeostasis & Protection of College of Life SciencesZhejiang UniversityHangzhouZhejiang310058China
- College of Life Sciences‐iCell Biotechnology Regenerative Biomedicine LaboratoryZhejiang University‐Lishui Joint Innovation Center for Life and HealthLishuiZhejiang323010China
| |
Collapse
|
21
|
Zhao Q, Mo Z, Zeng L, Yuan Y, Wang Y, Wang Y. Construction and Evaluation of Hepatic Targeted Drug Delivery System with Hydroxycamptothecin in Stem Cell-Derived Exosomes. Molecules 2024; 29:5174. [PMID: 39519815 PMCID: PMC11547497 DOI: 10.3390/molecules29215174] [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] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Hydroxycamptothecin (HCPT) is commonly used in the treatment of liver cancer; however, its low water solubility and poor stability significantly limit its clinical application. In recent years, research on exosomes has deepened considerably. Exosomes possess a unique phospholipid bilayer structure, enabling them to traverse tissue barriers, which provides natural advantages as drug carriers. Nevertheless, delivering exosomes safely and efficiently to target cells remains a major challenge. In this study, we utilized the affinity of the SP94 peptide for human liver cancer cell receptors. HCPT was coated with exosomes in our experimental design, and the exosome membrane was modified with SP94 peptide to facilitate drug delivery to liver cancer cells. Exosomes were purified from bone marrow mesenchymal stem cells, and targeted peptides were attached to their surfaces via post-insertion techniques. Subsequently, HCPT was incorporated into the exosomes through electroporation. Using the HepG2 hepatoma cell line, we evaluated a series of in vitro pharmacodynamics and studied pharmacokinetics and tissue distribution in animal models. The results indicated that ligand-targeted, modified drug-carrying exosomes significantly enhance drug bioavailability, prolong retention time in vivo, and facilitate liver targeting. Moreover, this approach reduces drug nephrotoxicity, enhances anti-tumor efficacy, and lays the groundwork for the development of novel liver cancer-targeting agents.
Collapse
Affiliation(s)
- Qiongjun Zhao
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Q.Z.); (Z.M.); (L.Z.); (Y.Y.)
| | - Zixuan Mo
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Q.Z.); (Z.M.); (L.Z.); (Y.Y.)
| | - Liuting Zeng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Q.Z.); (Z.M.); (L.Z.); (Y.Y.)
| | - Yue Yuan
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Q.Z.); (Z.M.); (L.Z.); (Y.Y.)
| | - Yan Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China; (Q.Z.); (Z.M.); (L.Z.); (Y.Y.)
| | - Ying Wang
- Teaching and Experimental Center, Guangdong Pharmaceutical University, Zhongshan 528453, China
| |
Collapse
|
22
|
Kobal N, Marzidovšek M, Schollmayer P, Maličev E, Hawlina M, Marzidovšek ZL. Molecular and Cellular Mechanisms of the Therapeutic Effect of Mesenchymal Stem Cells and Extracellular Vesicles in Corneal Regeneration. Int J Mol Sci 2024; 25:11121. [PMID: 39456906 PMCID: PMC11507649 DOI: 10.3390/ijms252011121] [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: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
The cornea is a vital component of the visual system, and its integrity is crucial for optimal vision. Damage to the cornea resulting from trauma, infection, or disease can lead to blindness. Corneal regeneration using mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (MSC-EVs) offers a promising alternative to corneal transplantation. MSCs are multipotent stromal cells that can differentiate into various cell types, including corneal cells. They can also secrete a variety of anti-inflammatory cytokines and several growth factors, promoting wound healing and tissue reconstruction. This review summarizes the current understanding of the molecular and cellular mechanisms by which MSCs and MSC-EVs contribute to corneal regeneration. It discusses the potential of MSCs and MSC-EV for treating various corneal diseases, including corneal epithelial defects, dry eye disease, and keratoconus. The review also highlights finalized human clinical trials investigating the safety and efficacy of MSC-based therapy in corneal regeneration. The therapeutic potential of MSCs and MSC-EVs for corneal regeneration is promising; however, further research is needed to optimize their clinical application.
Collapse
Affiliation(s)
- Nina Kobal
- Eye Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (N.K.)
| | - Miha Marzidovšek
- Eye Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (N.K.)
| | - Petra Schollmayer
- Eye Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (N.K.)
| | - Elvira Maličev
- Blood Transfusion Centre of Slovenia, 1000 Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Marko Hawlina
- Eye Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (N.K.)
- Medical Faculty, Department of Ophthalmology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Zala Lužnik Marzidovšek
- Eye Hospital, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (N.K.)
- Medical Faculty, Department of Ophthalmology, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
23
|
Yuan Y, Liu T. Influence of mesenchymal stem cells from different origins on the therapeutic effectiveness of systemic lupus erythematosus. Exp Cell Res 2024; 442:114263. [PMID: 39307406 DOI: 10.1016/j.yexcr.2024.114263] [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: 06/17/2024] [Revised: 08/09/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is a chronic autoimmune inflammatory disorder characterized by alterations in the balance between inflammatory and regulatory cytokines. Mesenchymal stem cells (MSCs), which are non-hematopoietic stem cells with multipotent differentiation potential, due to their immunomodulatory, tissue repair, low immunogenicity, and chemotactic properties, have garnered increasing interest in SLE treatment. Studies increasingly reveal the heterogeneous nature of MSC populations. With sources including dental pulp, adipose tissue, bone marrow, and umbilical cord, the therapeutic effects of MSCs on SLE vary depending on their origin. This review consolidates clinical research on MSCs from different sources in treating SLE and analyzes the possible causes underlying these variable outcomes. Additionally, it elucidates five potential factors impacting the outcomes of MSC therapy in SLE: the influence of the microenvironment on MSCs, the complexity and paradoxical aspects of MSC mechanisms in SLE treatment, the heterogeneity of MSCs, the in vivo differentiation potential and post-transplant survival rates of MSCs, and disparities in MSC preparation conditions.
Collapse
Affiliation(s)
- Yuan Yuan
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China.
| | - Tong Liu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China
| |
Collapse
|
24
|
Mili B, Choudhary OP. Advancements and mechanisms of stem cell-based therapies for spinal cord injury in animals. Int J Surg 2024; 110:6182-6197. [PMID: 38265419 PMCID: PMC11486964 DOI: 10.1097/js9.0000000000001074] [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: 10/23/2023] [Accepted: 12/24/2023] [Indexed: 01/25/2024]
Abstract
Spinal cord injury (SCI) is a neurodegenerative disorder of the central nervous system that can lead to permanent loss of sensation and voluntary movement beyond the affected area. Extensive preclinical and clinical trials have been conducted to evaluate the safety and effectiveness of stem cells for the treatment of various central nervous system diseases or disorders, including SCI. However, several challenges hinder nerve cell regeneration in the injured spinal cord, such as extensive cell loss, limited neural cell regeneration capacity, axonal disruption, and the presence of growth-inhibiting molecules, particularly astroglial scarring or glial scars at the injury site in chronic cases. These obstacles pose significant challenges for physicians in restoring normal motor and sensory nerve function in both humans and animals following SCI. This review focuses on SCI pathogenesis, the mechanisms underlying the therapeutic potential of mesenchymal stem cells in SCI, and the potential of stem cell-based therapies as promising avenues for treatment. This review article also included relevant preclinical and clinical data from animal studies.
Collapse
Affiliation(s)
- Bhabesh Mili
- Department of Veterinary Physiology and Biochemistry, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Jalukie, Peren, Nagaland
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda, Punjab, India
| |
Collapse
|
25
|
Rusch RM, Inagaki E, Taniguchi H, Sakakura S, Tamai R, Nonaka H, Shimizu S, Sato S, Ogawa Y, Masatoshi H, Negishi K, Okano H, Shimmura S. Adipose-derived mesenchymal stromal cells: A study on safety and efficacy in ocular inflammation. Ocul Surf 2024; 34:523-534. [PMID: 39542088 DOI: 10.1016/j.jtos.2024.11.001] [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: 06/12/2024] [Revised: 10/28/2024] [Accepted: 11/02/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE This study explores the application of adipose-derived mesenchymal stromal cells (adMSCs) as a therapy for ocular inflammatory diseases utilizing a chronic GVHD model. METHODS Human adMSCs were administered via subconjunctival injection into mice with chronic ocular GVHD. Clinical scores and changes in T cell populations were analyzed. RESULTS The study showed significant improvement in corneal integrity, including epithelial damage, opacity, thickness, and structure, after subconjunctival adMSC transplantation. Additionally, adMSC transplantation increased CD45+ and Foxp3+ Tregs while decreasing CD4+ T cells, 1IL17A+ Th17 cells, and IFNγ+ Th1 cells in local cervical lymph nodes. Moreover, adMSC-conditioned media enhanced wound closure and cell migration toward the wound bed in vitro. The cells disappeared within a week suggesting that trophic factors were involved. CONCLUSION The dual benefit of adMSCs in immune-related ocular disorders underscores their potential for clinical application. This study focuses on subconjunctival delivery, effects of adMSCs and migration post-injection, with implications for optimizing cellular therapy application. The observed dual action, combining immunomodulation and tissue repair enhancement, underscores holistic approach of adMSC therapy in regenerative medicine, making it a potent treatment for diseases involving inflammation and tissue damage in the ocular surface.
Collapse
Affiliation(s)
- Robert M Rusch
- Department of Clinical Regenerative Medicine, Fujita Health University, Japan
| | - Emi Inagaki
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Hiroko Taniguchi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Saki Sakakura
- Department of Clinical Regenerative Medicine, Fujita Health University, Japan
| | | | | | - Shota Shimizu
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Hirayama Masatoshi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Japan
| | - Shigeto Shimmura
- Department of Clinical Regenerative Medicine, Fujita Health University, Japan.
| |
Collapse
|
26
|
Chatzianagnosti S, Dermitzakis I, Theotokis P, Kousta E, Mastorakos G, Manthou ME. Application of Mesenchymal Stem Cells in Female Infertility Treatment: Protocols and Preliminary Results. Life (Basel) 2024; 14:1161. [PMID: 39337944 PMCID: PMC11433628 DOI: 10.3390/life14091161] [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: 07/22/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Infertility is a global phenomenon that impacts people of both the male and the female sex; it is related to multiple factors affecting an individual's overall systemic health. Recently, investigators have been using mesenchymal stem cell (MSC) therapy for female-fertility-related disorders such as polycystic ovarian syndrome (PCOS), premature ovarian failure (POF), endometriosis, preeclampsia, and Asherman syndrome (AS). Studies have shown promising results, indicating that MSCs can enhance ovarian function and restore fertility for affected individuals. Due to their regenerative effects and their participation in several paracrine pathways, MSCs can improve the fertility outcome. However, their beneficial effects are dependent on the methodologies and materials used from isolation to reimplantation. In this review, we provide an overview of the protocols and methods used in applications of MSCs. Moreover, we summarize the findings of published preclinical studies on infertility treatments and discuss the multiple properties of these studies, depending on the isolation source of the MSCs used.
Collapse
Affiliation(s)
- Sofia Chatzianagnosti
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Eleni Kousta
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George Mastorakos
- Department of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
27
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [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: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
28
|
Gunes EG, Gunes M, Yu J, Janakiram M. Targeting cancer stem cells in multiple myeloma. Trends Cancer 2024; 10:733-748. [PMID: 38971642 DOI: 10.1016/j.trecan.2024.06.002] [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: 02/07/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 07/08/2024]
Abstract
Multiple myeloma (MM) is a hematological malignancy of bone marrow (BM) plasma cells with excessive clonal expansion and is associated with the overproduction of light-chain or monoclonal immunoglobulins (Igs). MM remains incurable, with high rates of relapses and refractory disease after first-line treatment. Cancer stem cells (CSCs) have been implicated in drug resistance in MM; however, the evidence for CSCs in MM is not adequate, partly due to a lack of uniformity in the definitions of multiple myeloma stem cells (MMSCs). We review advances in understanding MMSCs and their role in drug resistance to MM therapies. We also discuss novel therapeutic strategies to overcome MMSC-mediated relapses and drug resistance.
Collapse
Affiliation(s)
- Emine Gulsen Gunes
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Los Angeles, CA 91010, USA; Toni Stephenson Lymphoma Center, City of Hope, Los Angeles, CA 91010, USA.
| | - Metin Gunes
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Department of Immuno-Oncology, Beckman Research Institute, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA
| | - Murali Janakiram
- Department of Hematology, Division of Myeloma, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| |
Collapse
|
29
|
Deng Y, Liu Z, Lu M. Extracellular vesicles deviced from hypoxia-3D-GMSCs rescue the mitochondrial dysfunction of aging-GMSCs. Biochem Biophys Res Commun 2024; 717:150021. [PMID: 38718565 DOI: 10.1016/j.bbrc.2024.150021] [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: 03/26/2024] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/21/2024]
Abstract
Mesenchymal stem cells (MSCs) are ubiquitous multipotent cells exhibiting significant therapeutic potential for various diseases. It is generally accepted that clinical application requires massive expansion of MSCs, which is often accompanied by the occurrence of replicative senescence. Additionally, senescent MSCs exhibit significantly reduced proliferation, differentiation, and therapeutic potential. The scale-up of MSCs production and cellular senescence are major challenges for translational applications. This study first collected extracellular vesicles (EVs) from gingival MSCs (GMSCs) under hypoxia preconditioning combined with 3D dynamic culture (obtained EVs designed as H-3D-EVs). Subsequently, we further explored the effects and mechanisms of H-3D-EVs on aging-GMSCs. The results showed that H-3D-EVs improved the proliferation ability and cell activity of aging-GMSCs, and ameliorated their senescence. mRNA sequencing reveals transcriptomic changes in aging-GMSCs. It was found that H-3D-EVs up-regulated genes related to mitochondrial dynamics, cell cycle, and DNA repair, while down-regulated aging-related genes. Furthermore, we verified that H-3D-EVs corrected the mitochondrial dysfunction of aging-GMSCs by improving mitochondrial dynamics. In summary, this study provides a promising strategy for improving the culture methods of GMSCs and avoiding its senescence in large-scale production.
Collapse
Affiliation(s)
- Yujia Deng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, China; First Clinical Department of Changsha Medical University, The 1501 Leifeng Road in Wangcheng District, Changsha, 410219, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Ming Lu
- Hunan Provincical Key Laboratory of Neurorestoratology, The Second Affiliated Hospital, Hunan Normal University, Changsha, 410003, China.
| |
Collapse
|
30
|
Shojaporian S, Mahmoudian-Sani MR, Khodadadi A, Dehcheshmeh MG, Amari A. Effect of Priming With Toll-Like Receptor 3 Agonist on Expression of Long Noncoding RNAs in Human Wharton Jelly Mesenchymal Stem Cells. EXP CLIN TRANSPLANT 2024; 22:551-558. [PMID: 39223813 DOI: 10.6002/ect.2024.0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Mesenchymal stem cells are gaining attention in medicine because of their anti-inflammatory and immunosuppressive properties. Inflammatory conditions can modulate immune responses in mesenchymal stem cells.We investigated the expression of long noncoding RNAs (RMRP, MALT1, NKILA,THRIL, and Linc-MAF-4) in humanWharton jelly mesenchymal stem cells primed with polyinosinicpolycytidylic acid. MATERIALS AND METHODS Mesenchymal stem cells were isolated from human Wharton jelly by the explant method. To determine the stem nature of the cells, we performed a differentiation test on bone and fat cells. We used flow cytometry analysis to determine surface markers. Umbilical cord mesenchymal stem cells (1 × 105) were cultured in T75 culture flasks in Dulbecco's modified Eagle medium containing 10% fetal bovine serum. After cells reached approximately 80% confluency, cells were exposed to 50 µg/mL of polyinosinic-polycytidylic acid, a Toll-like receptor 3 ligand, for 24, 48, and 72 hours. The control group were cells not exposed to polyinosinic-polycytidylic acid. Real-time polymerase chain reaction evaluated RMRP, MALAT1, NKILA, THRIL, and Linc-MAF-4 long noncoding RNAs. RESULTS We observed significantly increased expression of NKILA inWharton jelly mesenchymal stem cells stimulated with polyinosinic-polycytidylic acid at 72 hours compared with expression level in the control group (P < .001). CONCLUSIONS Results indicated that a potential mechanism by which the Toll-like receptor 3 ligand improves immunosuppression of mesenchymal stem cells can be attributed to the regulatory role of long noncoding RNAs, possibly through increased expression of anti-inflammatory long noncoding RNAs such as NKILA.
Collapse
Affiliation(s)
- Samira Shojaporian
- >From the Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | | | | |
Collapse
|
31
|
Zheng T, Li S, Zhang T, Fu W, Liu S, He Y, Wang X, Ma T. Exosome-shuttled miR-150-5p from LPS-preconditioned mesenchymal stem cells down-regulate PI3K/Akt/mTOR pathway via Irs1 to enhance M2 macrophage polarization and confer protection against sepsis. Front Immunol 2024; 15:1397722. [PMID: 38957471 PMCID: PMC11217356 DOI: 10.3389/fimmu.2024.1397722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
Rationale Sepsis is a life-threatening organ dysfunction and lack of effective measures in the current. Exosomes from mesenchymal stem cells (MSCs) reported to alleviate inflammation during sepsis, and the preconditioning of MSCs could enhance their paracrine potential. Therefore, this study investigated whether exosomes secreted by lipopolysaccharide (LPS)-pretreated MSCs exert superior antiseptic effects, and explored the underlying molecular mechanisms. Methods Exosomes were isolated and characterized from the supernatants of MSCs. The therapeutic efficacy of normal exosomes (Exo) and LPS-pretreated exosomes (LPS-Exo) were evaluated in terms of survival rates, inflammatory response, and organ damage in an LPS-induced sepsis model. Macrophages were stimulated with LPS and treated with Exo or LPS-Exo to confirm the results of the in vivo studies, and to explain the potential mechanisms. Results LPS-Exo were shown to inhibit aberrant pro-inflammatory cytokines, prevent organ damages, and improve survival rates of the septic mice to a greater extent than Exo. In vitro, LPS-Exo significantly promoted the M2 polarization of macrophages exposed to inflammation. miRNA sequencing and qRT-PCR analysis identified the remarkable expression of miR-150-5p in LPS-Exo compared to that in Exo, and exosomal miR-150-5p was transferred into recipient macrophages and mediated macrophage polarization. Further investigation demonstrated that miR-150-5p targets Irs1 in recipient macrophages and subsequently modulates macrophage plasticity by down-regulating the PI3K/Akt/mTOR pathway. Conclusion The current findings highly suggest that exosomes derived from LPS pre-conditioned MSCs represent a promising cell-free therapeutic method and highlight miR-150-5p as a novel molecular target for regulating immune hyperactivation during sepsis.
Collapse
Affiliation(s)
- Ting Zheng
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Sipeng Li
- Department of Orthopedics, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Teng Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuchang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuxin He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Ma
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
32
|
Lavi Arab F, Hoseinzadeh A, Hafezi F, Sadat Mohammadi F, Zeynali F, Hadad Tehran M, Rostami A. Mesenchymal stem cell-derived exosomes for management of prostate cancer: An updated view. Int Immunopharmacol 2024; 134:112171. [PMID: 38701539 DOI: 10.1016/j.intimp.2024.112171] [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: 02/25/2024] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 05/05/2024]
Abstract
Prostate cancer represents the second most prevalent form of cancer found in males, and stands as the fifth primary contributor to cancer-induced mortality on a global scale. Research has shown that transplanted mesenchymal stem cells (MSCs) can migrate by homing to tumor sites in the body. In prostate cancer, researchers have explored the fact that MSC-based therapies (including genetically modified delivery vehicles or vectors) and MSC-derived exosomes are emerging as attractive options to improve the efficacy and safety of traditional cancer therapies. In addition, researchers have reported new insights into the application of extracellular vesicle (EV)-MSC therapy as a novel treatment option that could provide a more effective and targeted approach to prostate cancer treatment. Moreover, the new generation of exosomes, which contain biologically functional molecules as signal transducers between cells, can simultaneously deliver different therapeutic agents and induce an anti-tumor phenotype in immune cells and their recruitment to the tumor site. The results of the current research on the use of MSCs in the treatment of prostate cancer may be helpful to researchers and clinicians working in this field. Nevertheless, it is crucial to emphasize that although dual-role MSCs show promise as a therapeutic modality for managing prostate cancer, further investigation is imperative to comprehensively grasp their safety and effectiveness. Ongoing clinical trials are being conducted to assess the viability of MSCs in the management of prostate cancer. The results of these trials will help determine the viability of this approach. Based on the current literature, engineered MSCs-EV offer great potential for application in targeted tumor therapy.
Collapse
Affiliation(s)
- Fahimeh Lavi Arab
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Akram Hoseinzadeh
- Department of Immunology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.; Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Hafezi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Sadat Mohammadi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farid Zeynali
- Department of Urology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Melika Hadad Tehran
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Amirreza Rostami
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
33
|
Endo N, Matsumoto T, Kazama T, Kano K, Shimizu M, Ryu K, Tokuhashi Y, Nakanishi K. Therapeutic potential of dedifferentiated fat cells in a rat model of osteoarthritis of the knee. Regen Ther 2024; 26:50-59. [PMID: 38859891 PMCID: PMC11163150 DOI: 10.1016/j.reth.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024] Open
Abstract
Introduction Mature adipocyte-derived dedifferentiated fat cells (DFATs) represent a subtype of multipotent cells that exhibit comparable phenotypic and functional characteristics to adipose-derived stem cells (ASCs). In this study, we assessed the chondroprotective properties of intra-articularly administrated DFATs in a rat model of osteoarthritis (OA). We also investigated in vitro the expression of anti-inflammatory and chondroprotective genes in DFATs prepared from the infrapatellar fat pad (IFP) and subcutaneous adipose-tissue (SC) of human origin. Methods In the cell transplantation experiment, rats were assigned to the DFAT and Control group (n = 10 in each group) and underwent anterior cruciate ligament transection (ACLT) accompanied by medial meniscus resection (MMx) to induce OA. One week later, they received intra-articular injections of 1 × 106 DFATs (DFAT group) or PBS (control group) four times, with a weekly administration frequency. Macroscopic and microscopic evaluations were conducted five weeks post-surgery. In the in vitro experiments. DFATs derived from the IFP (IFP-DFATs) and SC (SC-DFATs) were prepared from donor-matched tissue samples (n = 3). The gene expression of PTGS2, TNFAIP6, PRG4, BMP2, and BMP6 under TNF-α or IFN-γ stimulation in these cells was evaluated using RT-PCR. Furthermore, the effect of co-culturing synovial fibroblasts with DFATs on the gene expression of ADAMTS4 and IL-6 were evaluated. Results Intra-articular injections of DFATs significantly inhibited cartilage degeneration in the rat OA model induced by ACLT and MMx. RT-PCR analysis revealed that both IFP-DFATs and SC-DFATs upregulated the expression of genes involved in immune regulation, anti-inflammation, and cartilage protection such as PTGS2, TNFAIP6, and BMP2, under stimulation by inflammatory cytokines. Co-culture with DFATs suppressed the expression of ADAMTS4 and IL6 in synovial fibroblasts. Conclusions The intra-articular injection of DFATs resulted in chondroprotective effects in the rat OA model. Both SC-DFATs and IFP-DFATs induced the expression of anti-inflammatory and chondroprotective genes in vitro. These results indicate that DFATs appear to possess therapeutic potential in inhibiting cartilage degradation and could serve as a promising cellular resource for OA treatment.
Collapse
Affiliation(s)
- Noriyuki Endo
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Taro Matsumoto
- Department of Functional Morphology, Division of Cell Regeneration and Transplantation, Nihon University School of Medicine, Tokyo, Japan
| | - Tomohiko Kazama
- Department of Functional Morphology, Division of Cell Regeneration and Transplantation, Nihon University School of Medicine, Tokyo, Japan
| | - Koichiro Kano
- Laboratory of Cell and Tissue Biology, College of Bioresource Science, Nihon University, Fujisawa, Japan
| | - Manabu Shimizu
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Keinosuke Ryu
- Department of Orthopaedic Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yasuaki Tokuhashi
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Kazuyoshi Nakanishi
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Zhang XD, Luo Q, Du Y, Yang L, Yu LC, Feng L, Rao D, Tang JX, Tan HM, Guo XY, Tang SS, Liu T, Yue F, Huang HX. The allostery and modification of hGHRH molecules and specific dimer produced significant fertility effect by proliferating and activating in-situ ovarian mesenchymal stem cells. Eur J Pharm Sci 2024; 197:106768. [PMID: 38643940 DOI: 10.1016/j.ejps.2024.106768] [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: 02/05/2024] [Revised: 03/23/2024] [Accepted: 04/13/2024] [Indexed: 04/23/2024]
Abstract
The negative coordination of growth hormone secretagogue receptor (GHS-R) and growth hormone-releasing hormone receptor (GHRH-R) involves in the repair processes of cellular injury. The allosteric U- or H-like modified GHRH dimer Grinodin and 2Y were comparatively evaluated in normal Kunming mice and hamster infertility models induced by CPA treatment. 1-3-9 µg of Grinodin or 2Y per hamster stem-cell-exhaustion model was subcutaneously administered once a week, respectively inducing 75-69-46 or 45-13-50 % of birth rates. In comparison, the similar mole of human menopausal gonadotropin (hMG) or human growth hormone (hGH) was administered once a day but caused just 25 or 20 % of birth rates. Grinodin induced more big ovarian follicles and corpora lutea than 2Y, hMG, hGH. The hMG-treated group was observed many distorted interstitial cells and more connective tissues and the hGH-treated group had few ovarian follicles. 2Y had a plasma lifetime of 21 days and higher GH release in mice, inducing lower birth rate and stronger individual specificity in reproduction as well as only promoting the proliferation of mesenchymal-stem-cells (MSCs) in the models. In comparison, Grinodin had a plasma lifetime of 30 days and much lower GH release in mice. It significantly promoted the proliferation and activation of ovarian MSCs together with the development of follicles in the models by increasing Ki67 and GHS-R expressions, and decreasing GHRH-R expression in a dose-dependent manner. However, the high GH and excessive estrogen levels in the models showed a dose-dependent reduction in fertility. Therefore, unlike 2Y, the low dose of Grinodin specifically shows low GHS-R and high GHRH-R expressions thus evades GH and estrogen release and improves functions of organs, resulting in an increase of fertility.
Collapse
Affiliation(s)
- Xu-Dong Zhang
- Department of Clinical Laboratories & Pathology, Guangdong Provincial Cops Hospital of Chinese People's Armed Police Forces, Guangzhou 510507, China
| | - Qun Luo
- Research & Development Department, Shenzhen Nafe Biopharmaceutical Company LTD, Shenzhen 518107, China
| | - Yan Du
- Department of Clinical Laboratories & Pathology, Guangdong Provincial Cops Hospital of Chinese People's Armed Police Forces, Guangzhou 510507, China
| | - Li Yang
- Department of Digestive & Endocrinology, Guangdong Provincial Cops Hospital of Chinese People's Armed Police Forces, Guangzhou 510507, China
| | - Li-Cheng Yu
- Department of Clinical Laboratories & Pathology, Guangdong Provincial Cops Hospital of Chinese People's Armed Police Forces, Guangzhou 510507, China
| | - Lan Feng
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Dan Rao
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jing-Xuan Tang
- Department of Chemistry, College of Literature, Science, and the Arts, University of Michigan-Ann Arbor, Ann Arbor 48109, United States
| | - Hong-Mei Tan
- Department of Clinical Laboratories, Luopu Street Lijiang Community Health Service Station, Guangzhou 511431, China
| | - Xiao-Yuan Guo
- Department of Pathology, Sanya People's Hospital, Sanya City 572000, Hainan Province, China
| | - Song-Shan Tang
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Feng Yue
- Department of Clinical Laboratories, Guangzhou Tianhe District Hospital of Traditional Chinese Medicine, Guangzhou 510655, China
| | - Hui-Xian Huang
- Department of Clinical Laboratories & Pathology, Guangdong Provincial Cops Hospital of Chinese People's Armed Police Forces, Guangzhou 510507, China
| |
Collapse
|
35
|
Tahmasebi F, Asl ER, Vahidinia Z, Barati S. Stem Cell-Derived Exosomal MicroRNAs as Novel Potential Approach for Multiple Sclerosis Treatment. Cell Mol Neurobiol 2024; 44:44. [PMID: 38713302 PMCID: PMC11076329 DOI: 10.1007/s10571-024-01478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by inflammation and demyelination of CNS neurons. Up to now, there are many therapeutic strategies for MS but they are only being able to reduce progression of diseases and have not got any effect on repair and remyelination. Stem cell therapy is an appropriate method for regeneration but has limitations and problems. So recently, researches were used of exosomes that facilitate intercellular communication and transfer cell-to-cell biological information. MicroRNAs (miRNAs) are a class of short non-coding RNAs that we can used to their dysregulation in order to diseases diagnosis. The miRNAs of microvesicles obtained stem cells may change the fate of transplanted cells based on received signals of injured regions. The miRNAs existing in MSCs may be displayed the cell type and their biological activities. Current studies show also that the miRNAs create communication between stem cells and tissue-injured cells. In the present review, firstly we discuss the role of miRNAs dysregulation in MS patients and miRNAs expression by stem cells. Finally, in this study was confirmed the relationship of microRNAs involved in MS and miRNAs expressed by stem cells and interaction between them in order to find appropriate treatment methods in future for limit to disability progression.
Collapse
Affiliation(s)
- Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
36
|
Zhang M, Xing J, Zhao S, Lu M, Liu Y, Lin L, Gao W, Chen L, Li W, Shang J, Zhou J, Yin X, Zhu X. Exosomal YB-1 facilitates ovarian restoration by MALAT1/miR-211-5p/FOXO 3 axis. Cell Biol Toxicol 2024; 40:29. [PMID: 38700571 PMCID: PMC11068691 DOI: 10.1007/s10565-024-09871-8] [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: 11/20/2023] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
Premature ovarian failure (POF) affects many adult women less than 40 years of age and leads to infertility. Mesenchymal stem cells-derived small extracellular vesicles (MSCs-sEVs) are attractive candidates for ovarian function restoration and folliculogenesis for POF due to their safety and efficacy, however, the key mediator in MSCs-sEVs that modulates this response and underlying mechanisms remains elusive. Herein, we reported that YB-1 protein was markedly downregulated in vitro and in vivo models of POF induced with H2O2 and CTX respectively, accompanied by granulosa cells (GCs) senescence phenotype. Notably, BMSCs-sEVs transplantation upregulated YB-1, attenuated oxidative damage-induced cellular senescence in GCs, and significantly improved the ovarian function of POF rats, but that was reversed by YB-1 depletion. Moreover, YB-1 showed an obvious decline in serum and GCs in POF patients. Mechanistically, YB-1 as an RNA-binding protein (RBP) physically interacted with a long non-coding RNA, MALAT1, and increased its stability, further, MALAT1 acted as a competing endogenous RNA (ceRNA) to elevate FOXO3 levels by sequestering miR-211-5p to prevent its degradation, leading to repair of ovarian function. In summary, we demonstrated that BMSCs-sEVs improve ovarian function by releasing YB-1, which mediates MALAT1/miR-211-5p/FOXO3 axis regulation, providing a possible therapeutic target for patients with POF.
Collapse
Affiliation(s)
- Mengxue Zhang
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jie Xing
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Department of Obstetrics and Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Shijie Zhao
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Department of Obstetrics and Gynecology, The Fourth Hospital of Changsha, Changsha, People's Republic of China
| | - Minjun Lu
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Yueqin Liu
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
| | - Li Lin
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Wujiang Gao
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Lu Chen
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Wenxin Li
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Junyu Shang
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Jiamin Zhou
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Xinming Yin
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China
| | - Xiaolan Zhu
- Reproductive Center, The Fourth Affiliated Hospital of Jiangsu University, 20 Zhengdong Road, Zhenjiang, Jiangsu, 212001, People's Republic of China.
- Institute of Reproductive Sciences, Jiangsu University, Zhenjiang, 212001, Jiangsu, People's Republic of China.
| |
Collapse
|
37
|
Bou Ghanem GO, Wareham LK, Calkins DJ. Addressing neurodegeneration in glaucoma: Mechanisms, challenges, and treatments. Prog Retin Eye Res 2024; 100:101261. [PMID: 38527623 DOI: 10.1016/j.preteyeres.2024.101261] [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: 12/28/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
Glaucoma is the leading cause of irreversible blindness globally. The disease causes vision loss due to neurodegeneration of the retinal ganglion cell (RGC) projection to the brain through the optic nerve. Glaucoma is associated with sensitivity to intraocular pressure (IOP). Thus, mainstay treatments seek to manage IOP, though many patients continue to lose vision. To address neurodegeneration directly, numerous preclinical studies seek to develop protective or reparative therapies that act independently of IOP. These include growth factors, compounds targeting metabolism, anti-inflammatory and antioxidant agents, and neuromodulators. Despite success in experimental models, many of these approaches fail to translate into clinical benefits. Several factors contribute to this challenge. Firstly, the anatomic structure of the optic nerve head differs between rodents, nonhuman primates, and humans. Additionally, animal models do not replicate the complex glaucoma pathophysiology in humans. Therefore, to enhance the success of translating these findings, we propose two approaches. First, thorough evaluation of experimental targets in multiple animal models, including nonhuman primates, should precede clinical trials. Second, we advocate for combination therapy, which involves using multiple agents simultaneously, especially in the early and potentially reversible stages of the disease. These strategies aim to increase the chances of successful neuroprotective treatment for glaucoma.
Collapse
Affiliation(s)
- Ghazi O Bou Ghanem
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Lauren K Wareham
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - David J Calkins
- Vanderbilt Eye Institute, Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
38
|
Buruiană A, Gheban BA, Gheban-Roșca IA, Georgiu C, Crișan D, Crișan M. The Tumor Stroma of Squamous Cell Carcinoma: A Complex Environment That Fuels Cancer Progression. Cancers (Basel) 2024; 16:1727. [PMID: 38730679 PMCID: PMC11083853 DOI: 10.3390/cancers16091727] [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: 04/08/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
The tumor microenvironment (TME), a complex assembly of cellular and extracellular matrix (ECM) components, plays a crucial role in driving tumor progression, shaping treatment responses, and influencing metastasis. This narrative review focuses on the cutaneous squamous cell carcinoma (cSCC) tumor stroma, highlighting its key constituents and their dynamic contributions. We examine how significant changes within the cSCC ECM-specifically, alterations in fibronectin, hyaluronic acid, laminins, proteoglycans, and collagens-promote cancer progression, metastasis, and drug resistance. The cellular composition of the cSCC TME is also explored, detailing the intricate interplay of cancer-associated fibroblasts (CAFs), mesenchymal stem cells (MSCs), endothelial cells, pericytes, adipocytes, and various immune cell populations. These diverse players modulate tumor development, angiogenesis, and immune responses. Finally, we emphasize the TME's potential as a therapeutic target. Emerging strategies discussed in this review include harnessing the immune system (adoptive cell transfer, checkpoint blockade), hindering tumor angiogenesis, disrupting CAF activity, and manipulating ECM components. These approaches underscore the vital role that deciphering TME interactions plays in advancing cSCC therapy. Further research illuminating these complex relationships will uncover new avenues for developing more effective treatments for cSCC.
Collapse
Affiliation(s)
- Alexandra Buruiană
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Bogdan-Alexandru Gheban
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Emergency Clinical County Hospital, 400347 Cluj-Napoca, Romania
| | - Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400129 Cluj-Napoca, Romania;
| | - Carmen Georgiu
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Doința Crișan
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Maria Crișan
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
39
|
Ramachandran A, Dhar R, Devi A. Stem Cell-Derived Exosomes: An Advanced Horizon to Cancer Regenerative Medicine. ACS APPLIED BIO MATERIALS 2024; 7:2128-2139. [PMID: 38568170 DOI: 10.1021/acsabm.4c00089] [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: 04/16/2024]
Abstract
Cancer research has made significant progress in recent years, and extracellular vesicles (EVs) based cancer investigation reveals several facts about cancer. Exosomes are a subpopulation of EVs. In the present decade, exosomes is mostly highlighted for cancer theranostic research. Tumor cell derived exosomes (TEXs) promote cancer but there are multiple sources of exosomes that can be used as cancer therapeutic agents (plant exosomes, stem cell-derived exosomes, modified or synthetic exosomes). Stem cells based regenerative medicine faces numerous challenges, such as promote tumor development, cellular reprogramming etc., and therefore addressing these complications becomes essential. Stem cell-derived exosomes serves as an answer to these problems and offers a better solution. Global research indicates that stem cell-derived exosomes also play a dual role in the cellular system by either inhibiting or promoting cancer. Modified exosomes which are genetically engineered exosomes or surface modified exosomes to increase the efficacy of the therapeutic properties can also be considered to target the above concerns. However, the difficulties associated with the exosomes include variations in exosomes heterogenity, isolation protocols, large scale production, etc., and these have to be managed effectively. In this review, we explore exosomes biogenesis, multiple stem cell-derived exosome sources, drug delivery, modified stem cells exosomes, clinical trial of stem cells exosomes, and the related challenges in this domain and future orientation. This article may encourage researchers to explore stem cell-derived exosomes and develop an effective and affordable cancer therapeutic solution.
Collapse
Affiliation(s)
- Aparna Ramachandran
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| |
Collapse
|
40
|
Chen L, Xiong Y, Chopp M, Zhang Y. Engineered exosomes enriched with select microRNAs amplify their therapeutic efficacy for traumatic brain injury and stroke. Front Cell Neurosci 2024; 18:1376601. [PMID: 38566841 PMCID: PMC10985177 DOI: 10.3389/fncel.2024.1376601] [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: 01/25/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Traumatic brain injury (TBI) and stroke stand as prominent causes of global disability and mortality. Treatment strategies for stroke and TBI are shifting from targeting neuroprotection toward cell-based neurorestorative strategy, aiming to augment endogenous brain remodeling, which holds considerable promise for the treatment of TBI and stroke. Compelling evidence underscores that the therapeutic effects of cell-based therapy are mediated by the active generation and release of exosomes from administered cells. Exosomes, endosomal derived and nano-sized extracellular vesicles, play a pivotal role in intercellular communication. Thus, we may independently employ exosomes to treat stroke and TBI. Systemic administration of mesenchymal stem cell (MSC) derived exosomes promotes neuroplasticity and neurological functional recovery in preclinical animal models of TBI and stroke. In this mini review, we describe the properties of exosomes and recent exosome-based therapies of TBI and stroke. It is noteworthy that the microRNA cargo within exosomes contributes to their therapeutic effects. Thus, we provide a brief introduction to microRNAs and insight into their key roles in mediating therapeutic effects. With the increasing knowledge of exosomes, researchers have "engineered" exosome microRNA content to amplify their therapeutic benefits. We therefore focus our discussion on the therapeutic benefits of recently employed microRNA-enriched engineered exosomes. We also discuss the current opportunities and challenges in translating exosome-based therapy to clinical applications.
Collapse
Affiliation(s)
- Liang Chen
- Department of Neurosurgery, Henry Ford Health, Detroit, MI, United States
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Health, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Health, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Yanlu Zhang
- Department of Neurosurgery, Henry Ford Health, Detroit, MI, United States
| |
Collapse
|
41
|
Cai X, Li Y, Gao F, Muhammad B, Yang H. Therapeutic effect and study of human umbilical cord blood mononuclear cells in patients with ischaemic bowel disease. Sci Rep 2024; 14:6121. [PMID: 38480861 PMCID: PMC10937724 DOI: 10.1038/s41598-024-56720-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/10/2024] [Indexed: 03/17/2024] Open
Abstract
Ischaemic bowel disease (ICBD) is a group of intestinal ischaemia syndromes caused by various aetiologies of reduced intestinal blood flow or vascular occlusion. ICBD can present as abdominal pain, bloody stool, and diarrhoea. This disease often occurs in middle-aged and elderly individuals with cardiovascular and cerebrovascular diseases. The incidence of ischaemic bowel disease has been increasing for decades, and it is difficult to diagnose, resulting in rapid disease progression and a high mortality rate. Therefore, fully understanding this disease, improving the diagnosis rate of this disease, and finding appropriate treatment methods are urgently needed to improve the condition and prognosis of patients. Umbilical cord blood stem cells are accessible, have weak immunogenicity, and have various biological functions, such as angiogenesis, inflammation and immune regulation. Many studies have confirmed that cord blood stem cells can relieve ischaemia, and these cells have attracted tremendous amounts of attention in regenerative medicine in recent years. In this paper, we discuss the clinical characteristics of ICBD, analyse the characteristics of human umbilical cord blood mononuclear cells (HUCB-MNCs), and use its to treat ischaemic bowel disease. Additionally, we compare the clinical manifestations and related indicators before and after treatment to evaluate the efficacy and safety of these methods.
Collapse
Affiliation(s)
- Xiaoxiao Cai
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238 Jingshi East Road, Jinan, Shandong, China
- Graduate Department of Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, Shandong, China
| | - Yonghao Li
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238 Jingshi East Road, Jinan, Shandong, China
- Graduate Department of Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, Shandong, China
| | - Fengyu Gao
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238 Jingshi East Road, Jinan, Shandong, China
| | - Bilal Muhammad
- Graduate Department of Shandong First Medical University & Shandong Academy of Medical Sciences, No. 6699 Qingdao Road, Jinan, Shandong, China
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766 Jingshi Road, Jinan, Shandong, China
| | - Hongli Yang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766 Jingshi Road, Jinan, Shandong, China.
| |
Collapse
|
42
|
Kafili G, Niknejad H, Tamjid E, Simchi A. Amnion-derived hydrogels as a versatile platform for regenerative therapy: from lab to market. Front Bioeng Biotechnol 2024; 12:1358977. [PMID: 38468689 PMCID: PMC10925797 DOI: 10.3389/fbioe.2024.1358977] [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: 12/20/2023] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
In recent years, the amnion (AM) has emerged as a versatile tool for stimulating tissue regeneration and has been of immense interest for clinical applications. AM is an abundant and cost-effective tissue source that does not face strict ethical issues for biomedical applications. The outstanding biological attributes of AM, including side-dependent angiogenesis, low immunogenicity, anti-inflammatory, anti-fibrotic, and antibacterial properties facilitate its usage for tissue engineering and regenerative medicine. However, the clinical usage of thin AM sheets is accompanied by some limitations, such as handling without folding or tearing and the necessity for sutures to keep the material over the wound, which requires additional considerations. Therefore, processing the decellularized AM (dAM) tissue into a temperature-sensitive hydrogel has expanded its processability and applicability as an injectable hydrogel for minimally invasive therapies and a source of bioink for the fabrication of biomimetic tissue constructs by recapitulating desired biochemical cues or pre-defined architectural design. This article reviews the multi-functionality of dAM hydrogels for various biomedical applications, including skin repair, heart treatment, cartilage regeneration, endometrium regeneration, vascular graft, dental pulp regeneration, and cell culture/carrier platform. Not only recent and cutting-edge research is reviewed but also available commercial products are introduced and their main features and shortcomings are elaborated. Besides the great potential of AM-derived hydrogels for regenerative therapy, intensive interdisciplinary studies are still required to modify their mechanical and biological properties in order to broaden their therapeutic benefits and biomedical applications. Employing additive manufacturing techniques (e.g., bioprinting), nanotechnology approaches (e.g., inclusion of various bioactive nanoparticles), and biochemical alterations (e.g., modification of dAM matrix with photo-sensitive molecules) are of particular interest. This review article aims to discuss the current function of dAM hydrogels for the repair of target tissues and identifies innovative methods for broadening their potential applications for nanomedicine and healthcare.
Collapse
Affiliation(s)
- Golara Kafili
- Center for Nanoscience and Nanotechnology, Institute for Convergence Science and Technology, Sharif University of Technology, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elnaz Tamjid
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolreza Simchi
- Center for Nanoscience and Nanotechnology, Institute for Convergence Science and Technology, Sharif University of Technology, Tehran, Iran
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
- Center for Bioscience and Technology, Institute for Convergence Science and Technology, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
43
|
Mehta JM, Hiremath SC, Chilimba C, Ghasemi A, Weaver JD. Translation of cell therapies to treat autoimmune disorders. Adv Drug Deliv Rev 2024; 205:115161. [PMID: 38142739 PMCID: PMC10843859 DOI: 10.1016/j.addr.2023.115161] [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: 09/15/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Autoimmune diseases are a diverse and complex set of chronic disorders with a substantial impact on patient quality of life and a significant global healthcare burden. Current approaches to autoimmune disease treatment comprise broadly acting immunosuppressive drugs that lack disease specificity, possess limited efficacy, and confer undesirable side effects. Additionally, there are limited treatments available to restore organs and tissues damaged during the course of autoimmune disease progression. Cell therapies are an emergent area of therapeutics with the potential to address both autoimmune disease immune dysfunction as well as autoimmune disease-damaged tissue and organ systems. In this review, we discuss the pathogenesis of common autoimmune disorders and the state-of-the-art in cell therapy approaches to (1) regenerate or replace autoimmune disease-damaged tissue and (2) eliminate pathological immune responses in autoimmunity. Finally, we discuss critical considerations for the translation of cell products to the clinic.
Collapse
Affiliation(s)
- Jinal M Mehta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Chishiba Chilimba
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Azin Ghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
44
|
Aglan HA, Kotob SE, Mahmoud NS, Kishta MS, Ahmed HH. Bone marrow stem cell-derived β-cells: New issue for diabetes cell therapy. Tissue Cell 2024; 86:102280. [PMID: 38029457 DOI: 10.1016/j.tice.2023.102280] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
This investigation aimed to establish the promising role of insulin-producing cells (IPCs) growing from bone marrow-mesenchymal stem cells (BM-MSCs) in relieving hyperglycemia induced in rats. BM-MSCs were differentiated into IPCs using three different protocols. The efficiency of BM-MSCs differentiation into IPCs in vitro was confirmed by detecting IPCs specific gene expression (Foxa-2, PDX-1 and Ngn-3) and insulin release assay. The in vivo study design included 3 groups of male Wistar rats; negative control group, diabetic group and IPCs-transfused group (5 ×106 cells of the most functional IPCs/rat). One month after IPCs infusion, serum glucose, insulin, c-peptide and visfatin levels as well as pancreatic glucagon level were quantified. Gene expression analysis of pancreatic Foxa-2 and Sox-17, IGF-1 and FGF-10 was done. Additionally, histological investigation of pancreatic tissue sections was performed. Our data clarified that, the most functional IPCs are those generated from BM-MSCs using differentiation protocol 3 as indicated by the significant up-regulation of Foxa-2, PDX-1 and Ngn-3 gene expression levels. These findings were further emphasized by releasing of a significant amount of insulin in response to glucose load. The transplantation of the IPCs in diabetic rats elicited significant decline in serum glucose, visfatin and pancreatic glucagon levels along with significant rise in serum insulin and c-peptide levels. Moreover, it triggered significant up-regulation in the expression levels of pancreatic Foxa-2, Sox-17, IGF-1 and FGF-10 genes versus the untreated diabetic counterpart. The histopathological examination of pancreatic tissue almost assisted the biochemical and molecular genetic analyses. These results disclose that the cell therapy holds potential to develop a new cure for DM based on the capability of BM-MSCs to generate β-cell phenotype using specific protocol.
Collapse
Affiliation(s)
- Hadeer A Aglan
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt.
| | - Soheir E Kotob
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | - Nadia S Mahmoud
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| | - Hanaa H Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt; Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
45
|
Shi Y, Wang S, Liu D, Wang Z, Zhu Y, Li J, Xu K, Li F, Wen H, Yang R. Exosomal miR-4645-5p from hypoxic bone marrow mesenchymal stem cells facilitates diabetic wound healing by restoring keratinocyte autophagy. BURNS & TRAUMA 2024; 12:tkad058. [PMID: 38250706 PMCID: PMC10796268 DOI: 10.1093/burnst/tkad058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Refractory diabetic wounds are a common occurrence in patients with diabetes and epidermis-specific macroautophagy/autophagy impairment has been implicated in their pathogenesis. Therefore, identifying and developing treatment strategies capable of normalizing epidermis-specific macroautophagy/autophagy could facilitate diabetic wound healing. The study aims to investigate the potential of bone marrow mesenchymal stem cell-derived exosomes (BMSC-exos) from hypoxic conditions as a treatment to normalize epidermis-specific autophagy for diabetic wound healing. METHODS We compared the effects of bone marrow mesenchymal stem cell (BMSC)-sourced exosomes (BMSC-Exos) from hypoxic conditions to those of BMSC in normoxic conditions (noBMSC-Exos). Our studies involved morphometric assessment of the exosomes, identification of the microRNA (miRNA) responsible for the effects, evaluation of keratinocyte functions and examination of effects of the exosomes on several molecules involved in the autophagy pathway such as microtubule-associated protein 1 light chain 3 beta, beclin 1, sequestosome 1, autophagy-related 5 and autophagy-related 5. The experiments used human BMSCs from the American Type Culture Collection, an in vivo mouse model of diabetes (db/db) to assess wound healing, as well as the human keratinocyte HaCaT cell line. In the methodology, the authors utilized an array of approaches that included electron microscopy, small interfering RNA (siRNA) studies, RNA in situ hybridization, quantitative real-time reverse transcription PCR (qRT-PCR), the isolation, sequencing and differential expression of miRNAs, as well as the use of miR-4645-5p-specific knockdown with an inhibitor. RESULTS Hypoxia affected the release of exosomes from hypoxic BMSCs (hy-BMSCs) and influenced the size and morphology of the exosomes. Moreover, hyBMSC-Exo treatment markedly improved keratinocyte function, including keratinocyte autophagy, proliferation and migration. miRNA microarray and bioinformatics analysis showed that the target genes of the differentially expressed miRNAs were mainly enriched in 'autophagy' and 'process utilizing autophagic mechanism' in the 'biological process' category and miR-4645-5p as a major contributor to the pro-autophagy effect of hyBMSC-Exos. Moreover, mitogen-activated protein kinase-activated protein kinase 2 (MAPKAPK2) was identified as a potential target of exosomal miR-4645-5p; this was confirmed using a dual luciferase assay. Exosomal miR-4645-5p mediates the inactivation of the MAPKAPK2-induced AKT kinase group (comprising AKT1, AKT2, and AKT3), which in turn suppresses AKT-mTORC1 signaling, thereby facilitating miR-4645-5p-mediated autophagy. CONCLUSIONS Overall, the results of this study showed that hyBMSC-Exo-mediated transfer of miR-4645-5p inactivated MAPKAPK2-induced AKT-mTORC1 signaling in keratinocytes, which activated keratinocyte autophagy, proliferation and migration, resulting in diabetic wound healing in mice. Collectively, the findings could aid in the development of a novel therapeutic strategy for diabetic wounds.
Collapse
Affiliation(s)
- Yan Shi
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yongwaizheng Road, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yongwaizheng Road, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Zhengguang Wang
- Department of Orthopaedics, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191 China
| | - Yihan Zhu
- Department of Plastic and Aesthetic Surgery, Jiangxi Maternal and Child Health Hospital, Bayidadao Road, Donghu District, Nanchang 330006, China
| | - Jun Li
- HaploX Biotechnology Co., Ltd., Songpingshan Road, Nanshan District, Shenzhen 518057, Guangdong China
| | - Kui Xu
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine,Qianjiang Road, Yaohai District, Hefei 230038, Anhui, P. R. China
| | - Furong Li
- Translational Medicine Collaborative Innovation Center, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affifiliated Hospital, Southern University of Science and Technology), Dongmenbei Road, Luohu District, Shenzhen 518020, Guangdong, China
| | - Huicai Wen
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Yongwaizheng Road, Donghu District, Nanchang, Jiangxi, 330006, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Panfu Road, Yuexiu District, Guangzhou, Guangdong, 510180, China
| |
Collapse
|
46
|
Soltani L, Varmira K, Nazari M. Comparison of the differentiation of ovine fetal bone-marrow mesenchymal stem cells towards osteocytes on chitosan/alginate/CuO-NPs and chitosan/alginate/FeO-NPs scaffolds. Sci Rep 2024; 14:161. [PMID: 38168144 PMCID: PMC10762099 DOI: 10.1038/s41598-023-50664-6] [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] [Received: 09/26/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
In the current study, the creation of a chitosan/alginate scaffold hydrogel with and without FeO-NPs or CuO-NPs was studied. From fetal ovine bone marrow mesenchymal stem cells (BM-MSCs) were isolated and cultivated. Their differentiation into osteocyte and adipose cells was investigated. Also, on the scaffolds, cytotoxicity and apoptosis were studied. To investigate the differentiation, treatment groups include: (1) BM-MSCs were plated in DMEM culture medium with high glucose containing 10% FBS and antibiotics (negative control); (2) BM-MSCs were plated in osteogenic differentiation medium (positive control); (3) positive control group + FeO-NPs, (4) positive control group + CuO-NPs; (5) BM-MSCs were plated in osteogenic differentiation medium on chitosan/alginate scaffold; (6) BM-MSCs were plated in osteogenic differentiation medium on chitosan/alginate/FeO-NPs scaffold; and (7) BM-MSCs were plated in osteogenic differentiation medium on chitosan/alginate/CuO-NPs scaffold. Alkaline phosphatase enzyme concentrations, mineralization rate using a calcium kit, and mineralization measurement by alizarin staining quantification were evaluated after 21 days of culture. In addition, qRT-PCR was used to assess the expression of the ALP, ColA, and Runx2 genes. When compared to other treatment groups, the addition of CuO-NPs in the chitosan/alginate hydrogel significantly increased the expression of the ColA and Runx2 genes (p < 0.05). However, there was no significant difference between the chitosan/alginate hydrogel groups containing FeO-NPs and CuO-NPs in the expression of the ALP gene. It appears that the addition of nanoparticles, in particular CuO-NPs, has made the chitosan/alginate scaffold more effective in supporting osteocyte differentiation.
Collapse
Affiliation(s)
- Leila Soltani
- Department of Animal Sciences, College of Agriculture and Natural Resources, Razi University, Kermanshah, 67144-14971, Iran.
| | - Kambiz Varmira
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Maryam Nazari
- Applied Chemistry Department, Faculty of Chemistry, Razi University, Kermanshah, Iran
| |
Collapse
|
47
|
Kaviarasan V, Deka D, Balaji D, Pathak S, Banerjee A. Signaling Pathways in Trans-differentiation of Mesenchymal Stem Cells: Recent Advances. Methods Mol Biol 2024; 2736:207-223. [PMID: 37140811 DOI: 10.1007/7651_2023_478] [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: 05/05/2023]
Abstract
Mesenchymal stem cells are a group of multipotent cells that can be induced to differentiate into other cell types. The cells fate is decided by various signaling pathways, growth factors, and transcription factors in differentiation. The proper coordination of these factors will result in cell specification. MSCs are capable of being differentiated into osteogenic, chondrogenic, and adipogenic lineages. Different conditions induces the MSCs into particular phenotypes. The MSC trans-differentiation ensues as a response to environmental factors or due to circumstances that prove to favor trans-differentiation. Depending on the stage at which they are expressed, and the genetic alterations they undergo prior to their expression, transcription factors can accelerate the process of trans-differentiation. Further research has been conducted on the challenging aspect of MSCs being developed into non-mesenchymal lineage. The cells that are differentiated in this way maintain their stability even after being induced in animals. The recent advancements in the trans-differentiation capacities of MSCs on induction with chemicals, growth inducers, improved differentiation mediums, growth factors from plant extracts, and electrical stimulation are discussed in this paper. Signaling pathways have a great effect on MSCs trans-differentiation and they need to be better understood for their applications in therapeutic techniques. So, this paper tends to review the major signaling pathways that play a vital role in the trans-differentiation of MSC.
Collapse
Affiliation(s)
- Vaishak Kaviarasan
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Dikshita Deka
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Darshini Balaji
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India.
| |
Collapse
|
48
|
Zhang W, Lee PL, Li J, Komatsu C, Wang Y, Sun H, DeSanto M, Washington K, Gorantla V, Kokai L, Solari MG. Local Delivery of Adipose Stem Cells Promotes Allograft Survival in a Rat Hind-Limb Model of Vascularized Composite Allotransplantation. Plast Reconstr Surg 2024; 153:79e-90e. [PMID: 37014960 DOI: 10.1097/prs.0000000000010510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
BACKGROUND Adipose stem cells (ASCs) are a promising cell-based immunotherapy because of their minimally invasive harvest, high yield, and immunomodulatory capacity. In this study, the authors investigated the effects of local versus systemic ASC delivery on vascularized composite allotransplant survival and alloimmune regulation. METHODS Lewis rats received hind-limb transplants from Brown Norway rats and were administered donor-derived ASCs (passage 3 or 4, 1 × 10 6 cells/rat) locally in the allograft, or contralateral limb, or systemically at postoperative day 1. Recipients were treated intraperitoneally with rabbit anti-rat lymphocyte serum on postoperative days 1 and 4 and daily tacrolimus for 21 days. Limb allografts were monitored for clinical signs of rejection. Donor cell chimerism, immune cell differentiation, and cytokine expression in recipient lymphoid organs were measured by flow cytometric analysis. The immunomodulation function of ASCs was tested by mixed lymphocyte reaction assay and ASC stimulation studies. RESULTS Local-ASC-treated recipients achieved significant prolonged allograft survival (85.7% survived >130 days; n = 6) compared with systemic-ASC and contralateral-ASC groups. Secondary donor skin allografts transplanted to the local-ASC long-term surviving recipients accepted permanently without additional immunosuppression. The increases in donor cell chimerism and regulatory T-cells were evident in blood and draining lymph nodes of the local-ASC group. Moreover, mixed lymphocyte reaction showed that ASCs inhibited donor-specific T-cell proliferation independent of direct ASC-T-cell contact. ASCs up-regulated antiinflammatory molecules in response to cytokine stimulation in vitro. CONCLUSION Local delivery of ASCs promoted long-term survival and modulated alloimmune responses in a full major histocompatibility complex-mismatched vascularized composite allotransplantation model and was more effective than systemic administration. CLINICAL RELEVANCE STATEMENT ASCs are a readily available and abundant source of therapeutic cells that could decrease the amount of systemic immunosuppression required to maintain limb and face allografts.
Collapse
Affiliation(s)
- Wensheng Zhang
- From the Department of Plastic Surgery
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Wilford Hall Ambulatory Surgical Center, 59th Medical Wing Office of Science and Technology, Joint Base San Antonio
| | | | - Jingjing Li
- From the Department of Plastic Surgery
- Department of Burn and Plastic Surgery, Xiangya Hospital, Central South University
| | | | - Yong Wang
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of Colorado Anschutz Medical Campus
| | | | - Marisa DeSanto
- Ohio University Heritage College of Osteopathic Medicine
| | - Kia Washington
- Department of Surgery, Division of Plastic and Reconstructive Surgery, University of Colorado Anschutz Medical Campus
| | - Vijay Gorantla
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Institute for Regenerative Medicine, Wake Forest School of Medicine
| | - Lauren Kokai
- From the Department of Plastic Surgery
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
| | - Mario G Solari
- From the Department of Plastic Surgery
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
| |
Collapse
|
49
|
Sarabia-Sánchez MA, Robles-Flores M. WNT Signaling in Stem Cells: A Look into the Non-Canonical Pathway. Stem Cell Rev Rep 2024; 20:52-66. [PMID: 37804416 PMCID: PMC10799802 DOI: 10.1007/s12015-023-10610-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 10/09/2023]
Abstract
Tissue homeostasis is crucial for multicellular organisms, wherein the loss of cells is compensated by generating new cells with the capacity for proliferation and differentiation. At the origin of these populations are the stem cells, which have the potential to give rise to cells with both capabilities, and persevere for a long time through the self-renewal and quiescence. Since the discovery of stem cells, an enormous effort has been focused on learning about their functions and the molecular regulation behind them. Wnt signaling is widely recognized as essential for normal and cancer stem cell. Moreover, β-catenin-dependent Wnt pathway, referred to as canonical, has gained attention, while β-catenin-independent Wnt pathways, known as non-canonical, have remained conspicuously less explored. However, recent evidence about non-canonical Wnt pathways in stem cells begins to lay the foundations of a conceivably vast field, and on which we aim to explain this in the present review. In this regard, we addressed the different aspects in which non-canonical Wnt pathways impact the properties of stem cells, both under normal conditions and also under disease, specifically in cancer.
Collapse
Affiliation(s)
- Miguel Angel Sarabia-Sánchez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Martha Robles-Flores
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.
| |
Collapse
|
50
|
Hoseinzadeh A, Mahmoudi M, Rafatpanah H, Rezaieyazdi Z, Tavakol Afshari J, Hosseini S, Esmaeili SA. A new generation of mesenchymal stromal/stem cells differentially trained by immunoregulatory probiotics in a lupus microenvironment. Stem Cell Res Ther 2023; 14:358. [PMID: 38072921 PMCID: PMC10712058 DOI: 10.1186/s13287-023-03578-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Increasing evidence suggests that multipotent mesenchymal stem/stromal cells (MSCs) are a promising intervention strategy in treating autoimmune inflammatory diseases. It should be stated that systemic immunoregulation is increasingly recognized among the beneficial effects of MSCs and probiotics in treating morbid autoimmune disorders such as lupus. This study aimed to determine if immunoregulatory probiotics L. rhamnosus or L. delbrueckii can change the immunomodulatory effects of MSCs in lupus-like disease. METHODS Pristane-induced lupus (PIL) mice model was created via intraperitoneal injection of Pristane and then confirmed. Naïve MSCs (N-MSCs) were coincubated with two Lactobacillus strains, rhamnosus (R-MSCs) or delbrueckii (D-MSCs), and/or a combination of both (DR-MSCs) for 48 h, then administrated intravenously in separate groups. Negative (PBS-treated normal mice) and positive control groups (PBS-treated lupus mice) were also investigated. At the end of the study, flow cytometry and enzyme-linked immunosorbent assay (ELISA) analysis were used to determine the percentage of Th cell subpopulations in splenocytes and the level of their master cytokines in sera, respectively. Moreover, lupus nephritis was investigated and compared. Analysis of variance (ANOVA) was used for multiple comparisons. RESULTS Abnormalities in serum levels of anti-dsDNA antibodies, creatinine, and urine proteinuria were significantly suppressed by MSCs transplantation, whereas engrafted MSCs coincubation with both L. strains did a lesser effect on anti-dsDNA antibodies. L. rhamnosus significantly escalated the ability of MSCs to scale down the inflammatory cytokines (IFN-ɣ, IL-17), while L. delbrueckii significantly elevated the capacity of MSCs to scale down the percentage of Th cell subpopulations. However, incubation with both strains induced MSCs with augmented capacity in introducing inflammatory cytokines (IFN-ɣ, IL-17). Strikingly, R-MSCs directly restored the serum level of TGF-β more effectively and showed more significant improvement in disease parameters than N-MSCs. These results suggest that R-MSCs significantly attenuate lupus disease by further skew the immune phenotype of MSCs toward increased immunoregulation. CONCLUSIONS Results demonstrated that Lactobacillus strains showed different capabilities in training/inducing new abilities in MSCs, in such a way that pretreated MSCs with L. rhamnosus might benefit the treatment of lupus-like symptoms, given their desirable properties.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Centre, Division of Inflammation and Inflammatory Diseases, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol Afshari
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Hosseini
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|