1
|
Nagase K, Kuramochi H, Grainger DW, Takahashi H. Functional aligned mesenchymal stem cell sheets fabricated using micropatterned thermo-responsive cell culture surfaces. Mater Today Bio 2025; 32:101657. [PMID: 40166377 PMCID: PMC11957804 DOI: 10.1016/j.mtbio.2025.101657] [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: 12/23/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are frequently applied for cell transplantation and regenerative therapy because they secrete diverse therapeutic cytokines that prompt immuno-stimulatory and tissue repair processes. Furthermore, cultured MSC sheets exhibit enhanced cytokine secretion compared to their MSC suspensions, and represent a durable, versatile format for tissue engineering as singular, multi-layered, or multi-cell type sandwiched, transplantable constructs. Tissue engineered implants with various cellular orientations have been reported. In this study, patterned, temperature-responsive culture surfaces were used to prepare oriented MSC sheets. Patterned culture surfaces were fabricated by grafting polyacrylamide (PAAm) onto commercial poly(N-isopropylacrylamide) (PNIPAAm)-modified plastic via photopolymerization using a stripe-patterned photomask. Patterned surfaces were characterized using x-ray photoelectron spectroscopy, fluorescently labelled fibronectin and albumin adsorption assays, wetting (contact angle) measurements, atomic force microscopy, and scanning electron microscopy. Striped grafted patterns of PAAm were fabricated on the PNIPAAm-coated culture substrates, and PAAm polymerized within the PNIPAAm overlayer. Cell-aligned MSC sheets were then produced from MSC culture on this patterned surface, secreting higher amounts of therapeutic cytokines (vascular endothelial growth factor, hepatocyte growth factor, and transforming growth factor-β) than non-aligned MSC control sheets. In addition, aligned MSC sheets maintained enhanced cell multi-potent differentiation capabilities. New, aligned MSC sheets might exhibit improved functional properties for cell sheet transplant therapies.
Collapse
Affiliation(s)
- Kenichi Nagase
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City, Hiroshima, 734-8553, Japan
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo, 105-8512, Japan
| | - Hasumi Kuramochi
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo, 105-8512, Japan
| | - David W. Grainger
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Health Sciences, Salt Lake City, UT, 84112, USA
| | - Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku, Tokyo, 162-8666, Japan
| |
Collapse
|
2
|
Tang S, Zhang Y, Wang P, Tang Q, Liu Y, Lu F, Han M, Zhou M, Hu Q, Feng M, Liang D. NKG2D-CAR-targeted iPSC-derived MSCs efficiently target solid tumors expressing NKG2D ligand. iScience 2025; 28:112343. [PMID: 40276759 PMCID: PMC12020857 DOI: 10.1016/j.isci.2025.112343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Mesenchymal stem cells (MSCs) hold potential in cancer therapy; however, insufficient tumor homing ability and heterogeneity limit their therapeutic benefits. Obviously, the homogeneous induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) with enhanced ability of tumor targeting could be the solution. In this study, a CAR containing the NKG2D extracellular domain was targeted at the B2M locus of iPSCs to generate NKG2D-CAR-iPSCs, which were subsequently differentiated into NKG2D-CAR-iMSCs. In vitro, NKG2D-CAR significantly enhanced migration and adhesion of iMSCs to a variety of solid tumor cells expressing NKG2D ligands. RNA sequencing (RNA-seq) revealed significant upregulation of genes related to cell adhesion, migration, and binding in NKG2D-CAR-iMSCs. In A549 xenograft model, NKG2D-CAR-iMSCs demonstrated a 57% improvement in tumor-homing ability compared with iMSCs. In conclusion, our findings demonstrate enhanced targeting specificity of NKG2D-CAR-iMSCs to tumor cells expressing NKG2D ligands in vitro and in vivo, facilitating future investigation of iMSCs as an off-the-shelf living carrier for targeted delivery of anti-tumor agents.
Collapse
Affiliation(s)
- Shuqing Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Yusang Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Peiyun Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Qiyu Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Yating Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Fan Lu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Mengting Han
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Miaojin Zhou
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Qian Hu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Mai Feng
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410078, China
| | - Desheng Liang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
3
|
Wang Y, Wu B, Tao Y, Wang M, Wu D, Chen E, Tang H. Therapeutic effect of hUC-MSCs from different transplantation routes on acute liver failure in rats. Front Med (Lausanne) 2025; 12:1525719. [PMID: 40417693 PMCID: PMC12098624 DOI: 10.3389/fmed.2025.1525719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
Objective Acute liver failure (ALF) is a rare yet serious clinical syndrome. Recent studies have indicated that stem cells can effectively treat this condition. However, the optimal route for stem cell transplantation in the treatment of ALF remains unclear. This study aims to investigate the most effective transplantation route for stem cell therapy in ALF. Methods Human umbilical cord mesenchymal stem cells (hUC-MSCs) expressing both luciferase and green fluorescent protein were generated using a lentiviral vector. The hUC-MSCs were transplanted via the tail vein, portal vein, and abdominal cavity. The survival and distribution of the transplanted hUC-MSCs in rats were assessed through in vivo imaging and immunofluorescence. Furthermore, the therapeutic effects of hUC-MSCs transplanted via different routes on ALF were compared. Results The survival time of hUC-MSCs transplanted via the tail vein and portal vein was shorter compared to those transplanted intraperitoneally. The distribution of hUC-MSCs varied by transplantation route: those injected via the tail vein and portal vein were primarily found in the lungs and liver, respectively, while intraperitoneally transplanted hUC-MSCs predominantly localized in the abdominal cavity. In ALF rats, hUC-MSCs transplanted via the tail vein and portal vein improved survival rates, enhanced liver pathology, and reduced levels of inflammatory cytokines in liver tissue. In contrast, abdominal transplantation of hUC-MSCs showed no significant therapeutic effect. Conclusion hUC-MSCs transplanted via the tail vein and portal vein exhibited similar therapeutic effects on ALF; however, abdominal transplantation of hUC-MSCs showed no significant effect.
Collapse
Affiliation(s)
- Yonghong Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Bei Wu
- Department of Hepatology, Public Health Clinical Center of Chengdu, Chengdu, China
| | - Yachao Tao
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Menglan Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Wang J, Yan K, Ma L, Yan X, Meng Z, Li JA, Wang M, Du C, Yu Y. Notch Signaling Pathway Interfering as a Possible Asthma Treatment: A Narrative Review. J Asthma Allergy 2025; 18:437-446. [PMID: 40129884 PMCID: PMC11932036 DOI: 10.2147/jaa.s504925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
Asthma is a respiratory disease common among all age groups, and there is currently no cure that can be applied, as the patients react differently to the available treatments. Recent studies have shown that the Notch signaling pathway can regulate the dynamic balance between Th1 and Th2 cells, inhibit airway inflammation, reduce airway hyperresponsiveness and remodeling, and promote mesenchymal stem cell (MSCs) homing. This study conducted a comprehensive search of multiple large databases and provided a narrative review of the role of the Notch signaling pathway in asthma.
Collapse
Affiliation(s)
- Jingya Wang
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
- Baoding No. 1 hospital of Traditional Chinese Medicine, Baoding, Hebei, People’s Republic of China
| | - Kang Yan
- College of Traditional Chinese Medicine, Hebei University, Baoding, Hebei, People’s Republic of China
| | - Leilei Ma
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, People’s Republic of China
| | - Xianglei Yan
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
| | - Zihan Meng
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
| | - Ji-an Li
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
| | - Meng Wang
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
| | - Chenguang Du
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
| | - Yueyue Yu
- College of Traditional Chinese Medicine, North China University of Technology, Tangshan, Hebei, People’s Republic of China
| |
Collapse
|
5
|
Smallbone P, Kebriaei P, Mendt M, Shpall EJ, Olson AL, Fingrut WB. Mesenchymal stem cells in hematology: Therapeutic initiatives and future directions. Best Pract Res Clin Haematol 2025; 38:101613. [PMID: 40274341 DOI: 10.1016/j.beha.2025.101613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025]
Abstract
In recent years, the landscape of hematology has undergone rapid transformation, driven by innovative therapeutic strategies harnessing the properties of novel cellular therapies. Mesenchymal stem cells (MSCs) represent one of these promising therapies, with potential applications across a range of hematologic conditions. These cells are notable for their immunomodulatory properties, key role in supporting the hematopoietic micro-environment and capacity for multi-directional differentiation. This review will focus on the biologic mechanisms underlying MSC therapeutic use, current avenues of clinical investigation, and potential challenges and future directions for MSC derived therapies.
Collapse
Affiliation(s)
- Portia Smallbone
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayela Mendt
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amanda L Olson
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Warren B Fingrut
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
6
|
Mohan PP, Deo S, Liu ZJ, Dikici E, Kaneku H, Chang D, Garcia-Buitrago M, Jalaeian H, Zeynaloo E, Ortiz YY, Li Y, Bhatia S, Velazquez O, Daunert S. Liver Regeneration Following Thermal Ablation Using Nanocarrier Mediated Targeted Mesenchymal Stem Cell Therapy. Cardiovasc Intervent Radiol 2025; 48:233-243. [PMID: 39505737 PMCID: PMC11790787 DOI: 10.1007/s00270-024-03862-2] [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: 01/24/2024] [Accepted: 09/07/2024] [Indexed: 11/08/2024]
Abstract
PURPOSE To test the efficacy of nanocarrier (NC) mediated mesenchymal stem cell (MSC) therapy for liver regeneration following thermal ablation of porcine livers. MATERIALS AND METHODS Liver radiofrequency ablation was performed in 18 swines divided into MSC, MSC + NC and control groups. The test groups received infusion of MSC or MSC + NC labeled with enhanced green fluorescent protein (eGFP) via hepatic artery. MSC + NC group had MSCs coated with dendrimer nanocarrier complexed with I-Domain of lymphocyte function-associated antigen-1 (LFA-1). Nanocarriers direct homing of MSCs by binding to its counterpart protein, intercellular adhesion molecule-1 (ICAM-1), which is overexpressed at the periablation margins from inflammation. Ablation cavity reduction by CT volumetry was used as surrogate marker for liver regeneration. Cell proliferation was assessed with Ki67 and HepPar-1 stains. GFP identified MSC derived cells. RESULTS Total number of ablations in control animals were 13 across 4 animals. In the MSC group, there were 23 ablations across 6 animals, and in MSC + NC group there were 21 ablations across 6 animals. Ablation cavity volume reduction from day 0 to 30 were 64.4 ± 15.0%, 61.5 ± 12.9% and 80.3 ± 9.4% for control, MSC and MSC + NC groups, respectively (MSC + NC vs MSC: p < 0.001, MSC + NC vs. control: p = 0.001). GFP+ cell count at margins was 426.8 ± 193.2 for MSC group and 498.6 ± 235.2 for MSC + NC group (p = 0.01). The mean Ki67 and HepPar-1 staining at margins were 9.81 ± 4.5% and 6.12 ± 4.2% for MSC + NC group versus 7.59 ± 3.7% and 5.09 ± 3.7% for MSC group, respectively (P < 0.001 and P = 0.09, respectively). CONCLUSION Nanocarrier-mediated MSC therapy promotes liver regeneration by engrafting MSCs at ablation margins, potentially making liver-directed therapy viable for patients with severe liver dysfunction. This technology may also benefit other solid organs.
Collapse
Affiliation(s)
- Prasoon P Mohan
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Sapna Deo
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Zhao-Jun Liu
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Emre Dikici
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Hugo Kaneku
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Doyoung Chang
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Monica Garcia-Buitrago
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Hamed Jalaeian
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Elnaz Zeynaloo
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Yulexi Y Ortiz
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Yan Li
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Shivank Bhatia
- Department of Interventional Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Omaida Velazquez
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Interventional Radiology, UMHC-SCC, 1475 NW 12th Ave., Miami, FL, 33136, USA.
| |
Collapse
|
7
|
Zhang Y, Zhao Y, An C, Guo Y, Ma Y, Shao F, Zhang Y, Sun K, Cheng F, Ren C, Zhang L, Sun B, Zhang Y, Wang H. Material-driven immunomodulation and ECM remodeling reverse pulmonary fibrosis by local delivery of stem cell-laden microcapsules. Biomaterials 2025; 313:122757. [PMID: 39178558 DOI: 10.1016/j.biomaterials.2024.122757] [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: 06/20/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
Recent progress in stem cell therapy has demonstrated the therapeutic potential of intravenous stem cell infusions for treating the life-threatening lung disease of pulmonary fibrosis (PF). However, it is confronted with limitations, such as a lack of control over cellular function and rapid clearance by the host after implantation. In this study, we developed an innovative PF therapy through tracheal administration of microfluidic-templated stem cell-laden microcapsules, which effectively reversed the progression of inflammation and fibrotic injury. Our findings highlight that hydrogel microencapsulation can enhance the persistence of donor mesenchymal stem cells (MSCs) in the host while driving MSCs to substantially augment their therapeutic functions, including immunoregulation and matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) remodeling. We revealed that microencapsulation activates the MAPK signaling pathway in MSCs to increase MMP expression, thereby degrading overexpressed collagen accumulated in fibrotic lungs. Our research demonstrates the potential of hydrogel microcapsules to enhance the therapeutic efficacy of MSCs through cell-material interactions, presenting a promising yet straightforward strategy for designing advanced stem cell therapies for fibrotic diseases.
Collapse
Affiliation(s)
- Yujie Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Yuan Zhao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Chuanfeng An
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Yiyang Guo
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, PR China
| | - Yubin Ma
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, PR China
| | - Fei Shao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Yonggang Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Kai Sun
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China
| | - Fang Cheng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China
| | - Changle Ren
- Faculty of Medicine, Dalian University of Technology, Dalian, 116023, PR China; Department of Joint Surgery, Dalian Municipal Central Hospital, Dalian, 116044, PR China
| | - Lijun Zhang
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116024, PR China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China; School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, PR China
| | - Yang Zhang
- School of Dentistry, Health Science Center, Shenzhen University, Shenzhen, 518015, PR China
| | - Huanan Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, PR China; State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, PR China.
| |
Collapse
|
8
|
Wan R, Liu Y, Yan J, Lin J. Cell therapy: A beacon of hope in the battle against pulmonary fibrosis. FASEB J 2025; 39:e70356. [PMID: 39873972 DOI: 10.1096/fj.202402790r] [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: 11/10/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive interstitial lung disease characterized by abnormal activation of myofibroblasts and pathological remodeling of the extracellular matrix, with a poor prognosis and limited treatment options. Lung transplantation is currently the only approach that can extend the life expectancy of patients; however, its applicability is severely restricted due to donor shortages and patient-specific limitations. Therefore, the search for novel therapeutic strategies is imperative. In recent years, stem cells have shown great promise in the field of regenerative medicine due to their self-renewal capacity and multidirectional differentiation potential, and a growing body of literature supports the efficacy of stem cell therapy in PF treatment. This paper systematically summarizes the research progress of various stem cell types in the treatment of PF. Furthermore, it discusses the primary methods and clinical outcomes of stem cell therapy in PF, based on both preclinical and clinical data. Finally, the current challenges and key factors to consider in stem cell therapy for PF are objectively analyzed, and future directions for improving this therapy are proposed, providing new insights and references for the clinical treatment of PF patients.
Collapse
Affiliation(s)
- Ruyan Wan
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jingwen Yan
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
9
|
Giesen M, Fleck E, Scheele J, Hartmann TN, Henschler R. Rap1 Guanosine Triphosphate Hydrolase (GTPase) Regulates Shear Stress-Mediated Adhesion of Mesenchymal Stromal Cells. BIOLOGY 2025; 14:96. [PMID: 39857326 PMCID: PMC11762871 DOI: 10.3390/biology14010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Intravenously transplanted mesenchymal stromal cells (MSCs) have been shown to interact with endothelial cells and to migrate to tissues. However, intracellular signals regulating MSC migration are still incompletely understood. Here, we analyzed the role of Rap1 GTPase in the migration of human bone marrow-derived MSCs in vitro and in short-term homing in mice in vivo. MSCs expressed both Rap1A and Rap1B mRNAs, which were downregulated after treatment with siRNA against Rap1A and/or B. In a flow chamber model with pre-established human umbilical vein endothelial cells (HUVECs), Rap1A/B downregulated MSCs interacted for longer distances before arrest, indicating adhesion defects. CXCL12-induced adhesion of MSCs on immobilized Vascular Cell Adhesion Molecule (VCAM)-1 was also decreased after the downregulation of Rap1A, Rap1B, or both, as was CXCL12-induced transwell migration. In a competitive murine short-term homing model with i.v. co-injection of Rap1A+B siRNA-treated and control MSCs that were labeled with PKH 26 and PKH 67 fluorescent dyes, the Rap1A+B siRNA-treated MSCs were detected at increased frequencies in blood, liver, and spleen compared to control MSCs. Thus, Rap1 GTPase modulates the adhesion and migration of MSCs in vitro and may increase the bio-availability of i.v.-transplanted MSCs in tissues in a murine model.
Collapse
Affiliation(s)
- Melanie Giesen
- Institute for Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service, Clinics of the Goethe University Frankfurt (Main), 60528 Frankfurt am Main, Germany; (M.G.); (E.F.)
| | - Erika Fleck
- Institute for Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Service, Clinics of the Goethe University Frankfurt (Main), 60528 Frankfurt am Main, Germany; (M.G.); (E.F.)
| | - Jürgen Scheele
- Department of Medicine I, Medical Center University of Freiburg and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (J.S.); (T.N.H.)
| | - Tanja Nicole Hartmann
- Department of Medicine I, Medical Center University of Freiburg and Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (J.S.); (T.N.H.)
| | - Reinhard Henschler
- Institute for Transfusion Medicine, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
10
|
Pierro M, Thébaud B. Cell-based strategies for the treatment of injury to the developing lung. THE LUNG 2025:403-426. [DOI: 10.1016/b978-0-323-91824-4.00020-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
11
|
Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Golshekan M, Abedinzade M, Ahmadi E, Neha S, Najafi M. Revolutionizing Cancer Treatment: Harnessing the Power of Mesenchymal Stem Cells for Precise Targeted Therapy in the Tumor Microenvironment. Curr Top Med Chem 2025; 25:243-262. [PMID: 38797895 DOI: 10.2174/0115680266299112240514103048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
In recent years, mesenchymal stem cells (MSCs) have emerged as promising anti-- cancer mediators with the potential to treat several cancers. MSCs have been modified to produce anti-proliferative, pro-apoptotic, and anti-angiogenic molecules that could be effective against a variety of malignancies. Additionally, customizing MSCs with cytokines that stimulate pro-tumorigenic immunity or using them as vehicles for traditional chemical molecules with anti-cancer characteristics. Even though the specific function of MSCs in tumors is still challenged, promising outcomes from preclinical investigations of MSC-based gene therapy for a variety of cancers inspire the beginning of clinical trials. In addition, the tumor microenvironment (TME) could have a substantial influence on normal tissue stem cells, which can affect the treatment outcomes. To overcome the complications of TME in cancer development, MSCs could provide some signs of hope for converting TME into unequivocal therapeutic tools. Hence, this review focuses on engineered MSCs (En-MSCs) as a promising approach to overcoming the complications of TME.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Rostamzadeh
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Seyed Mohammad Amini
- Radiation Biology Research center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mostafa Golshekan
- Guilan Road Trauma Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahmoud Abedinzade
- Department of Medical Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Elham Ahmadi
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Singh Neha
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, Connecticut, USA
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
12
|
Mani Giri P, Banerjee A, Ghosal A, Salu P, Reindl K, Layek B. Mesenchymal stem cell-delivered paclitaxel nanoparticles exhibit enhanced efficacy against a syngeneic orthotopic mouse model of pancreatic cancer. Int J Pharm 2024; 666:124753. [PMID: 39321899 PMCID: PMC11760187 DOI: 10.1016/j.ijpharm.2024.124753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Pancreatic cancer is considered the deadliest among various solid tumors, with a five-year survival rate of 13 %. One of the major challenges in the management of advanced pancreatic cancer is the inefficient delivery of chemotherapeutics to the tumor site. Even though nanocarriers have been developed to improve tumoral delivery of chemotherapeutics, less than 1 % of the drugs reach tumors, rendering inadequate concentration for effective inhibition of tumors. As a potential alternative, mesenchymal stem cells (MSCs) can effectively deliver their cargo to tumor sites because of their resistance to chemotherapeutics and inherent tumor tropism. In this study, we used MSCs for the delivery of dibenzocyclooctyne (DBCO)-functionalized paclitaxel (PTX)-loaded poly(lactide-co-glycolide)-b-poly (ethylene glycol) (PLGA) nanoparticles. MSCs were modified to generate artificial azide groups on their surface, allowing nanoparticle loading via endocytosis and surface conjugation via click chemistry. This dual drug loading strategy significantly improves the PTX-loading capacity of azide-expressed MSCs (MSC-Az, 55.4 pg/cell) compared to unmodified MSCs (28.1 pg/cell). The in vitro studies revealed that PTX-loaded MSC-Az, nano-MSCs, exhibited cytotoxic effects against pancreatic cancer without altering their inherent phenotype, differentiation abilities, and tumor tropism. In an orthotopic pancreatic tumor model, nano-MSCs demonstrated significant inhibition of tumor growth (p < 0.05) and improved survival (p < 0.0001) compared to PTX solution, PTX nanocarriers, and Abraxane. Thus, nano-MSCs could be an effective delivery system for targeted pancreatic cancer chemotherapy and other solid tumors.
Collapse
Affiliation(s)
- Paras Mani Giri
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Anurag Banerjee
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Arpita Ghosal
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Philip Salu
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Katie Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States.
| |
Collapse
|
13
|
Gao P, Kajiya M, Motoike S, Ikeya M, Yang J. Application of mesenchymal stem/stromal cells in periodontal regeneration: Opportunities and challenges. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:95-108. [PMID: 38314143 PMCID: PMC10837070 DOI: 10.1016/j.jdsr.2024.01.001] [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: 08/08/2023] [Revised: 12/06/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Guided tissue regeneration (GTR) has been widely used in the periodontal treatment of intrabony and furcation defects for nearly four decades. The treatment outcomes have shown effectiveness in reducing pocket depth, improving attachment gain and bone filling in periodontal tissue. Although applying GTR could reconstruct the periodontal tissue, the surgical indications are relatively narrow, and some complications and race ethic problems bring new challenges. Therefore, it is challenging to achieve a consensus concerning the clinical benefits of GTR. With the appearance of stem cell-based regenerative medicine, mesenchymal stem/stromal cells (MSCs) have been considered a promising cell resource for periodontal regeneration. In this review, we highlight preclinical and clinical periodontal regeneration using MSCs derived from distinct origins, including non-odontogenic and odontogenic tissues and induced pluripotent stem cells, and discuss the transplantation procedures, therapeutic mechanisms, and concerns to evaluate the effectiveness of MSCs.
Collapse
Affiliation(s)
- Pan Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Souta Motoike
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jingmei Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
14
|
Kwon J, Kim MY, Lee JI, Kim W, Hyun JE, Yoon HY. Medication effects on pulmonary thromboembolism in mice intravenously transplanted with canine adipose tissue-derived mesenchymal stem cells. J Vet Sci 2024; 25:e80. [PMID: 39608774 PMCID: PMC11611486 DOI: 10.4142/jvs.24163] [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: 06/03/2024] [Revised: 08/12/2024] [Accepted: 08/23/2024] [Indexed: 11/30/2024] Open
Abstract
IMPORTANCE The intravenous administration of adipose tissue-derived mesenchymal stem cells (AdMSCs) in veterinary medicine is a promising regenerative therapy, but it can lead to severe complications, including pulmonary thromboembolism (PTE). OBJECTIVE As part of an ongoing study, this study examined the impact of medications, such as heparin, aspirin, and sodium nitroprusside (SNP), on the factors linked to PTE after an intravenous injection of canine mesenchymal stem cell into experimental animals. METHODS Fluorescently labeled canine AdMSCs were administered intravenously into the tail veins of five-week-old male BALB/c hairless mice. This study compared the survival rates, biodistribution, platelet counts, D-dimer levels, and histological examination results among the drug treatment experimental and the control groups. RESULTS The final survival rates in the SNP, control aspirin, and heparin groups were 25%, 33%, 50%, and 100%, respectively. Ex vivo imaging confirmed fluorescence exclusively in the lungs of all subjects who died during the injection, with no fluorescence detected in the other organs. On the other hand, in the heparin experimental group, the surviving individuals exhibited fluorescence in the lungs and the liver on day one. Histological biopsies revealed PTE in all deceased individuals within the medication experimental groups (p = 0.029). CONCLUSIONS AND RELEVANCE Heparin was highly effective, with no PTE-related deaths observed when used alongside cell injections. Aspirin revealed moderate effectiveness, surpassing the control group. On the other hand, the efficacy of SNP was inferior to that of the other two drugs.
Collapse
Affiliation(s)
- Jaeyeon Kwon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Mu-Young Kim
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Jeong-Ik Lee
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, School of Medicine, Konkuk University, Seoul 05029, Korea
| | - Woosuk Kim
- Department of Anatomy, College of Veterinary Medicine, and Veterinary Science Research Institute, Konkuk University, Seoul 05030, Korea
| | - Jae-Eun Hyun
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Hun-Young Yoon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- KU Center for Animal Blood Medical Science, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
15
|
Wei N, Chen X, Liu D, Bu X, Wang G, Sun X, Zhang J. A multi-modality imaging strategy to determine the multiple in vivo fates of human umbilical cord mesenchymal stem cells at different periods of acute liver injury treatment. J Mater Chem B 2024; 12:9213-9228. [PMID: 39041357 DOI: 10.1039/d4tb00914b] [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: 07/24/2024]
Abstract
Human umbilical cord mesenchymal stem cells (HUCMSCs) are applied for disease therapy as a new type of drug in many countries. Their effects are not only presented by live cells, but also apoptotic bodies or cell fragments of dead cells. Therefore, it is meaningful to determine the multiple fates of HUCMSCs in vivo. Although various probes combining different imaging modalities have been developed to label and trace transplanted HUCMSCs in vivo, the status of the cells (live, dead, or apoptotic) was not distinguished, and a thorough understanding of the multiple fates of HUCMSCs after transplantation in vivo is lacking. Therefore, a magnetic resonance (MR)/near infrared fluorescent (NIRF)/bioluminescence (BI) multi-modality imaging strategy was developed. Iron oxide nanoparticles (IONPs) were assembled into 100 nm nanoparticles using epigallocatechin gallate as a chemical linker to increase the MR signal and reduce the exocytosis of IONPs for direct cell labeling and longitudinal MR imaging tracking. Fluorescent probes for apoptosis (DEVD-Cy-OH) were also loaded in the above assemblies to monitor the cell status. Meanwhile, the cell surface was labeled with the fluorescent dye Cy7 via bioorthogonal reactions to visualize the NIRF signal. Luciferase was lentivirally transfected into live cells to generate bioluminescence. Such labeling did not affect either the viability, proliferation, migration, differentiation characteristics of HUCMSCs or their therapeutic effects on acute liver injury mice in vivo. The in vivo fates of HUCMSCs were monitored via MR/NIRF/BI multi-modality imaging in acute liver injury mice. Although MR and Cy7 signals aggregated in injured liver for 7 days, the BI signals persisted for less than 24 hours. There was an increase in DEVD-Cy-OH signals in the injured liver, but they were almost at the basal level. That means that HUCMSCs survive in mice for a short time, and the dead form of HUCMSCs accumulated in a large quantity and sustained for a long time, which might contribute to their therapeutic effect.
Collapse
Affiliation(s)
- Naijie Wei
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaoyang Chen
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| | - Danchen Liu
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiangchao Bu
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiaolian Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Quality Control and Pharmacovigilance, Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, 211198, China.
| | - Jingwei Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
16
|
Li Z, Han Z, Han ZC. Mesenchymal Stem Cells in Clinical Trials for Immune Disorders. Glob Med Genet 2024; 11:196-199. [PMID: 38947762 PMCID: PMC11210999 DOI: 10.1055/s-0044-1788044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Affiliation(s)
- Zongjin Li
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin, China
| | - Zhibo Han
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, China
| | - Zhong-Chao Han
- Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, China
| |
Collapse
|
17
|
Pharoun J, Berro J, Sobh J, Abou-Younes MM, Nasr L, Majed A, Khalil A, Joseph, Stephan, Faour WH. Mesenchymal stem cells biological and biotechnological advances: Implications for clinical applications. Eur J Pharmacol 2024; 977:176719. [PMID: 38849038 DOI: 10.1016/j.ejphar.2024.176719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem cells that are able to differentiate into multiple lineages including bone, cartilage, muscle and fat. They hold immunomodulatory properties and therapeutic ability to treat multiple diseases, including autoimmune and chronic degenerative diseases. In this article, we reviewed the different biological properties, applications and clinical trials of MSCs. Also, we discussed the basics of manufacturing conditions, quality control, and challenges facing MSCs in the clinical setting. METHODS Extensive review of the literature was conducted through the databases PubMed, Google Scholar, and Cochrane. Papers published since 2015 and covering the clinical applications and research of MSC therapy were considered. Furthermore, older papers were considered when referring to pioneering studies in the field. RESULTS The most widely studied stem cells in cell therapy and tissue repair are bone marrow-derived mesenchymal stem cells. Adipose tissue-derived stem cells became more common and to a lesser extent other stem cell sources e.g., foreskin derived MSCs. MSCs therapy were also studied in the setting of COVID-19 infections, ischemic strokes, autoimmune diseases, tumor development and graft rejection. Multiple obstacles, still face the standardization and optimization of MSC therapy such as the survival and the immunophenotype and the efficiency of transplanted cells. MSCs used in clinical settings displayed heterogeneity in their function despite their extraction from healthy donors and expression of similar surface markers. CONCLUSION Mesenchymal stem cells offer a rising therapeutic promise in various diseases. However, their potential use in clinical applications requires further investigation.
Collapse
Affiliation(s)
- Jana Pharoun
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jana Berro
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Jeanine Sobh
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | | | - Leah Nasr
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Ali Majed
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Alia Khalil
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Joseph
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Stephan
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, LAU, Byblos, Lebanon, P.O. Box 36.
| |
Collapse
|
18
|
Jiang Z, Chen L, Huang L, Yu S, Lin J, Li M, Gao Y, Yang L. Bioactive Materials That Promote the Homing of Endogenous Mesenchymal Stem Cells to Improve Wound Healing. Int J Nanomedicine 2024; 19:7751-7773. [PMID: 39099796 PMCID: PMC11297574 DOI: 10.2147/ijn.s455469] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 08/06/2024] Open
Abstract
Endogenous stem cell homing refers to the transport of endogenous mesenchymal stem cells (MSCs) to damaged tissue. The paradigm of using well-designed biomaterials to induce resident stem cells to home in to the injured site while coordinating their behavior and function to promote tissue regeneration is known as endogenous regenerative medicine (ERM). ERM is a promising new avenue in regenerative therapy research, and it involves the mobilizing of endogenous stem cells for homing as the principal means through which to achieve it. Comprehending how mesenchymal stem cells home in and grasp the influencing factors of mesenchymal stem cell homing is essential for the understanding and design of tissue engineering. This review summarizes the process of MSC homing, the factors influencing the homing process, analyses endogenous stem cell homing studies of interest in the field of skin tissue repair, explores the integration of endogenous homing promotion strategies with cellular therapies and details tissue engineering strategies that can be used to modulate endogenous homing of stem cells. In addition to providing more systematic theories and ideas for improved materials for endogenous tissue repair, this review provides new perspectives to explore the complex process of tissue remodeling to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Shengxiang Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jiabao Lin
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Mengyao Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
19
|
Shaw TD, Krasnodembskaya AD, Schroeder GN, Doherty DF, Silva JD, Tandel SM, Su Y, Butler D, Ingram RJ, O'Kane CM. Human mesenchymal stromal cells inhibit Mycobacterium avium replication in clinically relevant models of lung infection. Thorax 2024; 79:778-787. [PMID: 38508718 PMCID: PMC11287638 DOI: 10.1136/thorax-2023-220819] [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: 08/07/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Novel therapeutic strategies are urgently needed for Mycobacterium avium complex pulmonary disease (MAC-PD). Human mesenchymal stromal cells (MSCs) can directly inhibit MAC growth, but their effect on intracellular bacilli is unknown. We investigated the ability of human MSCs to reduce bacterial replication and inflammation in MAC-infected macrophages and in a murine model of MAC-PD. METHODS Human monocyte-derived macrophages (MDMs) were infected with M. avium Chester strain and treated with human bone marrow-derived MSCs. Intracellular and extracellular colony-forming units (CFUs) were counted at 72 hours. Six-week-old female balb/c mice were infected by nebulisation of M. avium Chester. Mice were treated with 1×106 intravenous human MSCs or saline control at 21 and 28 days post-infection. Lungs, liver and spleen were harvested 42 days post-infection for bacterial counts. Cytokines were quantified by ELISA. RESULTS MSCs reduced intracellular bacteria in MDMs over 72 hours (median 35% reduction, p=0.027). MSC treatment increased extracellular concentrations of prostaglandin E2 (PGE2) (median 10.1-fold rise, p=0.002) and reduced tumour necrosis factor-α (median 28% reduction, p=0.025). Blocking MSC PGE2 production by cyclo-oxygenase-2 (COX-2) inhibition with celecoxib abrogated the antimicrobial effect, while this was restored by adding exogenous PGE2. MSC-treated mice had lower pulmonary CFUs (median 18% reduction, p=0.012), but no significant change in spleen or liver CFUs compared with controls. CONCLUSION MSCs can modulate inflammation and reduce intracellular M. avium growth in human macrophages via COX-2/PGE2 signalling and inhibit pulmonary bacterial replication in a murine model of chronic MAC-PD.
Collapse
Affiliation(s)
- Timothy D Shaw
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | | | - Gunnar N Schroeder
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | - Declan F Doherty
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | - Johnatas Dutra Silva
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | - Shikha M Tandel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | - Yue Su
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | - David Butler
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | - Rebecca J Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK
| |
Collapse
|
20
|
Garza Treviño EN, Quiroz Reyes AG, Delgado Gonzalez P, Rojas Murillo JA, Islas JF, Alonso SS, Gonzalez Villarreal CA. Applications of Modified Mesenchymal Stem Cells as Targeted Systems against Tumor Cells. Int J Mol Sci 2024; 25:7791. [PMID: 39063032 PMCID: PMC11276748 DOI: 10.3390/ijms25147791] [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: 05/30/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Combined gene and cell therapy are promising strategies for cancer treatment. Given the complexity of cancer, several approaches are actively studied to fight this disease. Using mesenchymal stem cells (MSCs) has demonstrated dual antitumor and protumor effects as they exert massive immune/regulatory effects on the tissue microenvironment. MSCs have been widely investigated to exploit their antitumor target delivery system. They can be genetically modified to overexpress genes and selectively or more efficiently eliminate tumor cells. Current approaches tend to produce more effective and safer therapies using MSCs or derivatives; however, the effect achieved by engineered MSCs in solid tumors is still limited and depends on several factors such as the cell source, transgene, and tumor target. This review describes the progress of gene and cell therapy focused on MSCs as a cornerstone against solid tumors, addressing the different MSC-engineering methods that have been approached over decades of research. Furthermore, we summarize the main objectives of engineered MSCs against the most common cancers and discuss the challenges, limitations, risks, and advantages of targeted treatments combined with conventional ones.
Collapse
Affiliation(s)
- Elsa N. Garza Treviño
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Adriana G. Quiroz Reyes
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Paulina Delgado Gonzalez
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Juan Antonio Rojas Murillo
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Jose Francisco Islas
- Laboratorio de Terapia Celular, Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Dr. José Eleuterio González 235, Monterrey 64460, Nuevo León, Mexico; (E.N.G.T.); (A.G.Q.R.); (P.D.G.); (J.A.R.M.); (J.F.I.)
| | - Santiago Saavedra Alonso
- Departamento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico
| | - Carlos A. Gonzalez Villarreal
- Departamento de Ciencias Básicas, Vicerrectoría de Ciencias de la Salud, Universidad de Monterrey, Ignacio Morones Prieto 4500, Jesus M. Garza, San Pedro Garza García 66238, Nuevo León, Mexico
| |
Collapse
|
21
|
Botelho BF, Barreira AL, Filippo MG, Asensi KD, Faccioli LAP, dos Santos Salgado AB, de Salles EF, Marques CEC, Silva PL, dos Santos Goldenberg RC, Maiolino A, Gutfilen B, de Souza SAL, Junior ML, Morales MM. Safety and Biodistribution of an Autologous Bone Marrow-Derived Mononuclear Cell Infusion into Renal Arteries in Patients with Focal Segmental Glomerulosclerosis: A Phase 1 Study. Stem Cells Int 2024; 2024:2385568. [PMID: 39015674 PMCID: PMC11251782 DOI: 10.1155/2024/2385568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 04/26/2024] [Accepted: 06/12/2024] [Indexed: 07/18/2024] Open
Abstract
Patients with focal segmental glomerulosclerosis (FSGS) who are refractory to drug treatment may present progressive loss of kidney function, leading to chronic kidney disease stage 5 under dialysis treatment. The safety of systemic administration of bone marrow-derived mononuclear cells (BMDMCs) has been shown in different preclinical models of kidney diseases. However, to date, no study has evaluated the safety and biodistribution of BMDMCs after infusion in renal arteries in patients with FSGS. We used a prospective, non-randomized, single-center longitudinal design to investigate this approach. Five patients with refractory FSGS and an estimated glomerular filtration rate (eGFR) between 20 and 40 ml/min/1.73 m2 underwent bone marrow aspiration and received an arterial infusion of autologous BMDMCs (5 × 107) for each kidney. In addition, BMDMCs labeled with technetium-99m (99mTc-BMDMCs) were used to assess the biodistribution by scintigraphy. All patients completed the 270-day follow-up protocol with no serious adverse events. A transient increase in creatinine was observed after the cell therapy, with improvement on day 30. 99mTc-BMDMCs were detected in both kidneys and counts were higher after 2 hr compared with 24 hr. The arterial infusion of BMDMCs in both kidneys of patients with FSGS was considered safe with stable eGFR at the end of follow-up. This trial is registered with NCT02693366.
Collapse
Affiliation(s)
- Bruno Freire Botelho
- Department of NephrologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Luis Barreira
- Department of NephrologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcio Gomes Filippo
- Department of Vascular SurgeryClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina Dutra Asensi
- Cellular and Molecular Cardiology LaboratoryCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lanuza A P Faccioli
- Cellular and Molecular Cardiology LaboratoryCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Beatriz dos Santos Salgado
- Cellular and Molecular Cardiology LaboratoryCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elizabeth Figueiredo de Salles
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Eduardo Cruz Marques
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Coeli dos Santos Goldenberg
- Precision Medicine Research CenterCarlos Chagas Filho Institute of Biophysics e Brazilian Institute of Science and Technology—INCT REGENERA, Rio de Janeiro, Brazil
| | - Angelo Maiolino
- Department of HematologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bianca Gutfilen
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio Augusto Lopes de Souza
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maurilo Leite Junior
- Department of NephrologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Marcos Morales
- Laboratory of Cellular and Molecular PhysiologyCarlos Chagas Filho Biophysics InstituteHealth Sciences CenterFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Mahida RY, Yuan Z, Kolluri KK, Scott A, Parekh D, Hardy RS, Matthay MA, Perkins GD, Janes SM, Thickett DR. 11β hydroxysteroid dehydrogenase type 1 transgenic mesenchymal stem cells attenuate inflammation in models of sepsis. Front Bioeng Biotechnol 2024; 12:1422761. [PMID: 39036559 PMCID: PMC11257926 DOI: 10.3389/fbioe.2024.1422761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/12/2024] [Indexed: 07/23/2024] Open
Abstract
Background Human bone marrow mesenchymal stem cell (MSC) administration reduces inflammation in pre-clinical models of sepsis and sepsis-related lung injury, however clinical efficacy in patients has not yet been demonstrated. We previously showed that Alveolar Macrophage (AM) 11β-hydroxysteroid dehydrogenase type-1 (HSD-1) autocrine signalling is impaired in critically ill sepsis patients, which promotes inflammatory injury. Administration of transgenic MSCs (tMSCs) which overexpress HSD-1 may enhance the anti-inflammatory effects of local glucocorticoids and be more effective at reducing inflammation in sepsis than cellular therapy alone. Methods MSCs were transfected using a recombinant lentiviral vector containing the HSD-1 and GPF transgenes under the control of a tetracycline promoter. Thin layer chromatography assessed HSD-1 reductase activity in tMSCs. Mesenchymal stem cell phenotype was assessed by flow cytometry and bi-lineage differentiation. HSD-1 tMSCs were co-cultured with LPS-stimulated monocyte-derived macrophages (MDMs) from healthy volunteers prior to assessment of pro-inflammatory cytokine release. HSD-1 tMSCs were administered intravenously to mice undergoing caecal ligation and puncture (CLP). Results MSCs were transfected with an efficiency of 91.1%, and maintained an MSC phenotype. Functional HSD-1 activity was demonstrated in tMSCs, with predominant reductase cortisol activation (peak 8.23 pM/hour/100,000 cells). HSD-1 tMSC co-culture with LPS-stimulated MDMs suppressed TNFα and IL-6 release. Administration of transgene activated HSD-1 tMSCs in a murine model of CLP attenuated neutrophilic inflammation more effectively than transgene inactive tMSCs (medians 0.403 v 1.36 × 106/ml, p = 0.033). Conclusion The synergistic impact of HSD-1 transgene expression and MSC therapy attenuated neutrophilic inflammation in a mouse model of peritoneal sepsis more effectively than MSC therapy alone. Future studies investigating the anti-inflammatory capacity of HSD-1 tMSCs in models of sepsis-related direct lung injury and inflammatory diseases are required.
Collapse
Affiliation(s)
- Rahul Y. Mahida
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Zhengqiang Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Krishna K. Kolluri
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Dhruv Parekh
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Rowan S. Hardy
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Michael A. Matthay
- Cardiovascular Research Institute, Department of Medicine and Department of Anaesthesia, University of California San Francisco, San Francisco, CA, United States
| | - Gavin D. Perkins
- Warwick Medical School, University of Warwick, Warwick, United Kingdom
| | - Sam M. Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom
| | - David R. Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
23
|
Lightner AL, Irving PM, Lord GM, Betancourt A. Stem Cells and Stem Cell-Derived Factors for the Treatment of Inflammatory Bowel Disease with a Particular Focus on Perianal Fistulizing Disease: A Minireview on Future Perspectives. BioDrugs 2024; 38:527-539. [PMID: 38914783 PMCID: PMC11247053 DOI: 10.1007/s40259-024-00661-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/26/2024]
Abstract
Inflammatory bowel disease remains a difficult disease to effectively treat, especially fistulizing Crohn's disease. Perianal fistulas in the setting of Crohn's disease remain an area of unmet need with significant morbidity in this patient population. Up to one third of Crohn's patients will have perianal fistulizing disease and current medical and surgical interventions are of limited efficacy. Thus, most patients experience significant morbidity, narcotic use, and loss of employment and end up with multiple surgical interventions. Mesenchymal stem cells (MSCs) have shown efficacy in phase 3 clinical trials, but considerable infrastructure challenges make MSCs limited with regard to scalability in clinical practice. Extracellular vesicles, being derived from MSCs and capturing the secretome functionality of MSCs, offer similar physiological utility regarding mechanism, while also providing an off the shelf regenerative medicine product that could be widely used in daily clinical practice.
Collapse
Affiliation(s)
- Amy L Lightner
- Surgery, Scripps Clinic, 10667 N Torrey Pines Rd, La Jolla, CA, 92037, USA.
- Molecular Medicine, Scripps Research Institute, La Jolla, USA.
| | - Peter M Irving
- Guy's and St Thomas' Hospital, London, UK
- King's College London, London, UK
| | | | - Aline Betancourt
- Vitabolus Inc, San Diego, CA, USA
- Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
24
|
Martínez-Muñoz ME, Payares-Herrera C, Lipperheide I, Malo de Molina R, Salcedo I, Alonso R, Martín-Donaire T, Sánchez R, Zafra R, García-Berciano M, Trisán-Alonso A, Pérez-Torres M, Ramos-Martínez A, Ussetti P, Rubio JJ, Avendaño-Solà C, Duarte RF. Mesenchymal stromal cell therapy for COVID-19 acute respiratory distress syndrome: a double-blind randomised controlled trial. Bone Marrow Transplant 2024; 59:777-784. [PMID: 38409332 DOI: 10.1038/s41409-024-02230-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/28/2024]
Abstract
Mesenchymal stromal cells (MSC) have immunomodulatory and tissue-regenerative properties and have shown promising results in acute respiratory distress syndrome (ARDS) of multiple causes, including COVID-19. We conducted a randomised (1:1), placebo-controlled, double-blind clinical trial to assess the efficacy and safety of one bone marrow-derived MSC infusion in twenty patients with moderate to severe ARDS caused by COVID-19. The primary endpoint (increase in PaO2/FiO2 ratio from baseline to day 7, MSC 83.3 versus placebo 57.6) was not statistically significant, although a clinical improvement at day 7 in the WHO scale was observed in MSC patients (5, 50% vs 0, 0%, p = 0.033). Median time to discontinuation of supplemental oxygen was also shorter in the experimental arm (14 versus 23 days, p = 0.007), resulting in a shorter hospital stay (17.5 versus 28 days, p = 0.042). No significant differences were observed for other efficacy or safety secondary endpoints. No infusion or treatment-related serious adverse events occurred during the one-year follow-up. This study did not meet the primary endpoint of PaO2/FiO2 increase by day 7, although it suggests that MSC are safe in COVID-19 ARDS and may accelerate patients' clinical recovery and hospital discharge. Larger studies are warranted to elucidate their role in ARDS and other inflammatory lung disorders.Trial Registration: EudraCT Number: 2020-002193-27, registered on July 14th, 2020, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-002193-27/ES . NCT number: NCT04615429, registered on November 4th, 2020, https://clinicaltrials.gov/ct2/show/NCT04615429 .
Collapse
Affiliation(s)
- María E Martínez-Muñoz
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Concepción Payares-Herrera
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Clinical Pharmacology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Inés Lipperheide
- Intensive Care Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Rosa Malo de Molina
- Department of Pneumology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Isabel Salcedo
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Rosalía Alonso
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Trinidad Martín-Donaire
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Rocío Sánchez
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Rocío Zafra
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Miguel García-Berciano
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Andrea Trisán-Alonso
- Department of Pneumology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Manuel Pérez-Torres
- Intensive Care Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Antonio Ramos-Martínez
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Internal Medicine and Infectious Diseases, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Piedad Ussetti
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Pneumology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Juan J Rubio
- Intensive Care Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Cristina Avendaño-Solà
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Clinical Pharmacology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Rafael F Duarte
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain.
| |
Collapse
|
25
|
Mello DB, Mesquita FCP, Silva dos Santos D, Asensi KD, Dias ML, Campos de Carvalho AC, Goldenberg RCDS, Kasai-Brunswick TH. Mesenchymal Stromal Cell-Based Products: Challenges and Clinical Therapeutic Options. Int J Mol Sci 2024; 25:6063. [PMID: 38892249 PMCID: PMC11173248 DOI: 10.3390/ijms25116063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Mesenchymal stromal cell (MSC)-based advanced therapy medicinal products (ATMPs) are being tried in a vast range of clinical applications. These cells can be isolated from different donor tissues by using several methods, or they can even be derived from induced pluripotent stem cells or embryonic stem cells. However, ATMP heterogeneity may impact product identity and potency, and, consequently, clinical trial outcomes. In this review, we discuss these topics and the need to establish minimal criteria regarding the manufacturing of MSCs so that these innovative therapeutics may be better positioned to contribute to the advancement of regenerative medicine.
Collapse
Affiliation(s)
- Debora B. Mello
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
| | | | - Danúbia Silva dos Santos
- Center of Cellular Technology, National Institute of Cardiology, INC, Rio de Janeiro 22240-002, Brazil;
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
| | - Karina Dutra Asensi
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Marlon Lemos Dias
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Antonio Carlos Campos de Carvalho
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Regina Coeli dos Santos Goldenberg
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Tais Hanae Kasai-Brunswick
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
26
|
Kwon J, Kim MY, Lee S, Lee J, Yoon HY. Pulmonary passage of canine adipose tissue-derived mesenchymal stem cells through intravenous transplantation in mouse model. J Vet Sci 2024; 25:e36. [PMID: 38834506 PMCID: PMC11156597 DOI: 10.4142/jvs.23300] [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: 12/06/2023] [Revised: 01/16/2024] [Accepted: 03/29/2024] [Indexed: 06/06/2024] Open
Abstract
IMPORTANCE The intravenous administration of adipose tissue-derived mesenchymal stem cells (AdMSCs) in veterinary medicine is an attractive treatment option. On the other hand, it can result in severe complications, including pulmonary thromboembolism (PTE). OBJECTIVE The present study assessed the occurrence of PTE after the intravenous infusion of canine AdMSCs (cAdMSCs) into experimental animals. METHODS Five-week-old male BALB/c hairless mice were categorized into groups labeled A to G. In the control group (A), fluorescently stained 2 × 106 cAdMSCs were diluted in 200 μL of suspension and injected into the tail vein as a single bolus. The remaining groups included the following: group B with 5 × 106 cells, group C with 3 × 106 cells, group D with 1 × 106 cells, group E with 1 × 106 cells injected twice with a one-day interval, group F with 2 × 106 cells in 100 μL of suspension, and group G with 2 × 106 cells in 300 μL of suspension. RESULTS Group D achieved a 100% survival rate, while none of the subjects in groups B and C survived (p = 0.002). Blood tests revealed a tendency for the D-dimer levels to increase as the cell dose increased (p = 0.006). The platelet count was higher in the low cell concentration groups and lower in the high cell concentration groups (p = 0.028). A histological examination revealed PTE in most deceased subjects (96.30%). CONCLUSIONS AND RELEVANCE PTE was verified, and various variables were identified as potential contributing factors, including the cell dose, injection frequency, and suspension volume.
Collapse
Affiliation(s)
- Jaeyeon Kwon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
| | - Mu-Young Kim
- Department of Small Animal Clinical Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL 32611, USA
| | - Soojung Lee
- Department of Companion Animal Health, Yeonsung University, Anyang 14011, Korea
| | - Jeongik Lee
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, School of Medicine, Konkuk University, Seoul 05029, Korea
| | - Hun-Young Yoon
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- KU Center for Animal Blood Medical Science, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
27
|
Zhang C. Exosomes Derived from Mesenchymal Stem Cells: Therapeutic Opportunities for Spinal Cord Injury. Bull Exp Biol Med 2024; 176:716-721. [PMID: 38888648 DOI: 10.1007/s10517-024-06095-y] [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: 05/15/2023] [Indexed: 06/20/2024]
Abstract
Spinal cord injury (SCI) is a serious neurological condition comprising primary and secondary injury and causing severe neurological impairments. The effect of the conventional treatment is limited, including supportive therapy and emergency surgery. Exosomes derived from mesenchymal stem cells (MSCs-Exos) were previously reported to exert its potential therapeutic effects on SCI. Compared with mesenchymal stem cells (MSCs) transplantation for SCI, MSC-Exos showed several superiorities. In the present review, we summarized the revealed data of mechanisms underlying MSC-Exos repairing of SCI and discussed the issues of MSC-Exos use. Thus, in this review we summarized the latest studies on MSCs-Exos in the therapy of SCI and discussed whether MSCs-Exos can be applied to SCI and the prospects of transformation application.
Collapse
Affiliation(s)
- C Zhang
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
| |
Collapse
|
28
|
Maličev E, Jazbec K. An Overview of Mesenchymal Stem Cell Heterogeneity and Concentration. Pharmaceuticals (Basel) 2024; 17:350. [PMID: 38543135 PMCID: PMC10975472 DOI: 10.3390/ph17030350] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest in cell therapies due to the immunomodulatory and other effects they have after autologous or allogeneic transplantation. In most clinical applications, a high number of MSCs is required; therefore, the isolated MSC population must be expanded in the cell culture until the desired number is reached. Analysing freshly isolated MSCs is challenging due to their rareness and heterogeneity, which is noticeable among donors, tissues, and cell subpopulations. Although the phenotype of MSCs in tissue can differ from those of cultured cells, phenotyping and counting are usually performed only after MSC proliferation. As MSC applicability is a developing and growing field, there is a need to implement phenotyping and counting methods for freshly isolated MSCs, especially in new one-step procedures where isolated cells are implanted immediately without cell culturing. Only by analysing harvested cells can we correctly evaluate such studies. This review describes multilevel heterogeneity and concentrations of MSCs and different strategies for phenotype determination and enumeration of freshly isolated MSCs.
Collapse
Affiliation(s)
- Elvira Maličev
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| | - Katerina Jazbec
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
| |
Collapse
|
29
|
Greco R, Alexander T, Del Papa N, Müller F, Saccardi R, Sanchez-Guijo F, Schett G, Sharrack B, Snowden JA, Tarte K, Onida F, Sánchez-Ortega I, Burman J, Castilla Llorente C, Cervera R, Ciceri F, Doria A, Henes J, Lindsay J, Mackensen A, Muraro PA, Ricart E, Rovira M, Zuckerman T, Yakoub-Agha I, Farge D. Innovative cellular therapies for autoimmune diseases: expert-based position statement and clinical practice recommendations from the EBMT practice harmonization and guidelines committee. EClinicalMedicine 2024; 69:102476. [PMID: 38361991 PMCID: PMC10867419 DOI: 10.1016/j.eclinm.2024.102476] [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: 11/22/2023] [Revised: 01/05/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by loss of immune tolerance, high chronicity, with substantial morbidity and mortality, despite conventional immunosuppression (IS) or targeted disease modifying therapies (DMTs), which usually require repeated administration. Recently, novel cellular therapies (CT), including mesenchymal stromal cells (MSC), Chimeric Antigen Receptors T cells (CART) and regulatory T cells (Tregs), have been successfully adopted in ADs. An international expert panel of the European Society for Blood and Marrow Transplantation and the International Society for the Cell and Gene Therapy, reviewed all available evidence, based on the current literature and expert practices, on use of MSC, CART and Tregs, in AD patients with rheumatological, neurological, and gastroenterological indications. Expert-based consensus and recommendations for best practice and quality of patient care were developed to support clinicians, scientists, and their multidisciplinary teams, as well as patients and care providers and will be regularly updated.
Collapse
Affiliation(s)
- Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
- Co-Chair of the Practice Harmonization and Guidelines Committee of EBMT and Chair of the ADWP of the EBMT, Barcelona, Spain
| | - Tobias Alexander
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - Nicoletta Del Papa
- Scleroderma Clinic, Rheumatology Department, ASST G. Pini-CTO, Università degli Studi di Milano, Milano, Italy
| | - Fabian Müller
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany
- Bayrisches Zentrum für Krebsforschung (BZKF) Erlangen, Germany
| | - Riccardo Saccardi
- Cellular Therapies and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Fermin Sanchez-Guijo
- Department of Hematology, IBSAL-University Hospital of Salamanca and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen- Nürnberg, Erlangen, Germany
| | - Basil Sharrack
- Department of Neuroscience and Sheffield NIHR Translational Neuroscience BRC, Sheffield Teaching Hospitals NHS Foundation Trust & University of Sheffield, Sheffield, England, United Kingdom
| | - John A. Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Karin Tarte
- SITI Lab, CHU Rennes, EFS Bretagne, University Rennes, Rennes, France
| | - Francesco Onida
- Hematology & ASCT Unit, ASST Fatebenefratelli-Sacco, University of Milan, Italy
- Co-Chair of the Practice Harmonization and Guidelines Committee of EBMT, Spain
| | - Isabel Sánchez-Ortega
- Secretary of the Practice Harmonization and Guidelines Committee of EBMT, Barcelona, Spain
- EBMT Medical Officer, Executive Office, Barcelona, Spain
| | - Joachim Burman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC, CSUR) of the Catalan and Spanish Health Systems/Member of ERN-ReCONNET, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DiMED), University of Padua, Padua, Italy
| | - Jörg Henes
- Center for Interdisciplinary Rheumatology, Immunology and Autoimmune diseases and Department of Internal Medicine II (Haematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Germany
| | - James Lindsay
- Department of Gastroenterology, The Royal London Hospital, Barts Health NHS Trust, London, UK
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany
- Bayrisches Zentrum für Krebsforschung (BZKF) Erlangen, Germany
| | - Paolo A. Muraro
- Department of Brain Sciences, Imperial College London, London, UK
| | - Elena Ricart
- Gastroenterology Department. Hospital Clínic Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Montserrat Rovira
- BMT Unit, Haematology Department, Institute of Haematology and Oncology, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Foundation, Spain
| | - Tsila Zuckerman
- Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ibrahim Yakoub-Agha
- CHU de Lille, University Lille, INSERM U1286, Infinite, 59000, Lille, France
- Chair of the Practice Harmonization and Guidelines Committee of EBMT, Spain
| | - Dominique Farge
- Internal Medicine Unit (04): CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies auto-immunes systémiques Rares d’Ile-de-France, AP-HP, St-Louis Hospital Paris-Cite University, France
- Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
30
|
Shin MJ, Park JY, Park JY, Lim SH, Lim H, Choi JK, Park CK, Kang YJ, Khang D. Inflammation-Targeting Mesenchymal Stem Cells Combined with Photothermal Treatment Attenuate Severe Joint Inflammation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304333. [PMID: 38096399 DOI: 10.1002/adma.202304333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/07/2023] [Indexed: 12/23/2023]
Abstract
Current clinical therapeutic efficacy for the treatment of osteo- and rheumatoid-arthritis is obviously limited. Although mesenchymal stem cells (MSCs) are considered as a source of promising regenerative therapy, un-modified or genetically engineered MSCs injected in vivo restrict their clinical utility because of the low drug efficacy and unpredicted side effect, respectively. Herein, a strategy to enhance the migration efficacy of MSCs to inflamed joints via an inflammation-mediated education process is demonstrated. To reinforce the limited anti-inflammatory activity of MSCs, gold nanostar loaded with triamcinolone is conjugated to MSC. Furthermore, near-infrared laser-assisted photothermal therapy (PTT) induced by gold nanostar significantly elevates the anti-inflammatory efficacy of the developed drugs, even in advanced stage arthritis model. An immunological regulation mechanism study of PTT is first suggested in this study; the expression of the interleukin 22 receptor, implicated in the pathogenesis of arthritis, is downregulated in T lymphocytes by PTT, and Th17 differentiation from naïve CD4 T cell is inhibited. Collectively, inflammation-targeting MSCs conjugated with triamcinolone-loaded gold nanostar (Edu-MSCs-AuS-TA) promote the repolarization of macrophages and decrease neutrophil recruitment in joints. In addition, Edu-MSCs-AuS-TA significantly alleviate arthritis-associated pain, improve general locomotor activity, and more importantly, induce cartilage regeneration even for severe stages of arthritis model.
Collapse
Affiliation(s)
- Min Jun Shin
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Jun-Young Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jun Young Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Su Hyun Lim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Hyoungsub Lim
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jin Kyeong Choi
- Department of Immunology, School of Medicine, Jeonbuk National University, Jeonju, 54907, South Korea
| | - Chul-Kyu Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
| | - Youn Joo Kang
- Department of Rehabilitation Medicine, Eulji Hospital, School of Medicine, Eulji University, Seoul, 01830, South Korea
| | - Dongwoo Khang
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, School of Medicine, Gachon University, Incheon, 21999, South Korea
- Ectosome Inc., Incheon, 21999, South Korea
| |
Collapse
|
31
|
Zhou Y, Zhao Y, Wu Y, Chen J, Wu H, Wei W, Yan S. Human Umbilical Cord Mesenchymal Stem Cells Alleviate Rat Knee Osteoarthritis via Activating Wnt/ β-catenin Signaling Pathway. Curr Stem Cell Res Ther 2024; 19:234-244. [PMID: 37132309 DOI: 10.2174/1574888x18666230428094400] [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: 11/03/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic disease characterized by joint cartilage degeneration, destruction, and osteogenic hyperplasia. Human umbilical cord mesenchymal stem cells (hUCMSCs) have attracted increasing research interest due to their high clonogenic, proliferative, and migratory potential, as well as their improved secretion of relevant chondrogenic factors. This study evaluated the therapeutic potential and underlying mechanism of hUC-MSCs in alleviating pathological symptoms of OA. METHODS For the in vivo study, OA rats were established by the Hulth method to observe the therapeutic effect of intra-articular injection of hUC-MSCs. X-ray tests, gross observations, and histological and immunohistochemical assessments were conducted in rats. Levels of interleukin-1 beta (IL-1β), IL-6, matrix metalloproteinase-13 (MMP-13), and tissue inhibitor matrix metalloproteinase-1 in rats' synovial fluid were measured using enzyme-linked immunosorbent assay kits. For the in vitro study, hUC-MSCs and chondrocytes were cultured to explore the effect and underlying mechanisms of hUC-MSCs on OA. Apoptosis, proliferation, and glycosaminoglycan (GAG) were measured in the chondrocytes. The relative expression of aggrecan, COL-2, and SOX-9 mRNA was quantified by real-time polymerase chain reaction. Expressions of Wnt/β-catenin signaling molecules were measured by Western blot. RESULTS We found that intra-articular injection of hUC-MSCs reduced the combined score, increased the expression of collagen II, and decreased the expression of MMP-13, IL-1β, and IL-6 in rat knee joints. Additionally, hUC-MSCs increased the content of GAGs, inhibited chondrocyte apoptosis, and promoted chondrocyte proliferation. The expression of aggrecan, COL-2, and SOX-9 mRNA in chondrocytes was promoted by hUC-MSCs via activation of the Wnt/β-catenin signaling pathway. CONCLUSION Overall, this study demonstrated that hUC-MSCs induce the secretion of some cytokines via the paracrine function to activate the Wnt/β-catenin signaling pathway to reduce the pathological condition of OA and maintain the proper expression of cytokines and extracellular matrix proteins.
Collapse
Affiliation(s)
- Yue Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drug Collaborative Innovation Center, Hefei, 230032, Anhui Province, China
- Rheumatoid Arthritis Research Center, Anhui Medical University, Hefei, 230032, China
- The First Hospital of Anhui University of Science and Technology, Huainan, 232007, China
| | - Yingjie Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drug Collaborative Innovation Center, Hefei, 230032, Anhui Province, China
- Rheumatoid Arthritis Research Center, Anhui Medical University, Hefei, 230032, China
| | - Yujiao Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drug Collaborative Innovation Center, Hefei, 230032, Anhui Province, China
- Rheumatoid Arthritis Research Center, Anhui Medical University, Hefei, 230032, China
| | - Jingyu Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drug Collaborative Innovation Center, Hefei, 230032, Anhui Province, China
- Rheumatoid Arthritis Research Center, Anhui Medical University, Hefei, 230032, China
| | - Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drug Collaborative Innovation Center, Hefei, 230032, Anhui Province, China
- Rheumatoid Arthritis Research Center, Anhui Medical University, Hefei, 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drug Collaborative Innovation Center, Hefei, 230032, Anhui Province, China
- Rheumatoid Arthritis Research Center, Anhui Medical University, Hefei, 230032, China
| | - Shangxue Yan
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
- Anti-inflammatory Immune Drug Collaborative Innovation Center, Hefei, 230032, Anhui Province, China
- Rheumatoid Arthritis Research Center, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
32
|
Xia Z, Mu W, Yuan S, Fu S, Liu Y, Zhang N. Cell Membrane Biomimetic Nano-Delivery Systems for Cancer Therapy. Pharmaceutics 2023; 15:2770. [PMID: 38140108 PMCID: PMC10748133 DOI: 10.3390/pharmaceutics15122770] [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: 10/24/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Nano-delivery systems have demonstrated great promise in the therapy of cancer. However, the therapeutic efficacy of conventional nanomedicines is hindered by the clearance of the blood circulation system and the physiological barriers surrounding the tumor. Inspired by the unique capabilities of cells within the body, such as immune evasion, prolonged circulation, and tumor-targeting, there has been a growing interest in developing cell membrane biomimetic nanomedicine delivery systems. Cell membrane modification on nanoparticle surfaces can prolong circulation time, activate tumor-targeting, and ultimately improve the efficacy of cancer treatment. It shows excellent development potential. This review will focus on the advancements in various cell membrane nano-drug delivery systems for cancer therapy and the obstacles encountered during clinical implementation. It is hoped that such discussions will inspire the development of cell membrane biomimetic nanomedical systems.
Collapse
Affiliation(s)
- Zhenxing Xia
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Weiwei Mu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shijun Yuan
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Shunli Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Yongjun Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| | - Na Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Jinan 250012, China; (Z.X.); (W.M.); (S.Y.); (S.F.)
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan 250012, China
| |
Collapse
|
33
|
Dubský M, Husáková J, Sojáková D, Fejfarová V, Jude EB. Cell Therapy of Severe Ischemia in People with Diabetic Foot Ulcers-Do We Have Enough Evidence? Mol Diagn Ther 2023; 27:673-683. [PMID: 37740111 PMCID: PMC10590286 DOI: 10.1007/s40291-023-00667-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 09/24/2023]
Abstract
This current opinion article critically evaluates the efficacy of autologous cell therapy (ACT) for chronic limb-threatening ischemia (CLTI), especially in people with diabetes who are not candidates for standard revascularization. This treatment approach has been used in 'no-option' CLTI in the last two decades and more than 1700 patients have received ACT worldwide. Here we analyze the level of published evidence of ACT as well as our experience with this treatment method. Many studies have shown that ACT is safe and an effective method for patients with the most severe lower limb ischemia. However, some trials did not show any benefit of ACT, and there is some heterogeneity in the types of injected cells, route of administration and assessed endpoints. Nevertheless, we believe that ACT plays an important role in a comprehensive treatment of patients with diabetic foot and severe ischemia.
Collapse
Affiliation(s)
- Michal Dubský
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- First Faculty of Medicine, Charles Universtiy, Prague, Czech Republic.
| | - Jitka Husáková
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- First Faculty of Medicine, Charles Universtiy, Prague, Czech Republic
| | - Dominika Sojáková
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- First Faculty of Medicine, Charles Universtiy, Prague, Czech Republic
| | | | - Edward B Jude
- Diabetes Center, Tameside and Glossop Integrated Care NHS Foundation Trust, Ashton Under Lyne, UK.
- University of Manchester, Lancashire, UK.
| |
Collapse
|
34
|
Grishin OV, Shushunova NA, Bratashov DN, Prikhozhdenko ES, Verkhovskii RA, Kozlova AA, Abdurashitov AS, Sindeeva OA, Karavaev AS, Kulminskiy DD, Shashkov EV, Inozemtseva OA, Tuchin VV. Effect of pulsed laser parameters on photoacoustic flow cytometry efficiency in vitro and in vivo. Cytometry A 2023; 103:868-880. [PMID: 37455600 DOI: 10.1002/cyto.a.24778] [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: 12/06/2022] [Revised: 03/07/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Photoacoustic flow cytometry is one of the most effective approaches to detect "alien" objects in the bloodstream, including circulating tumor cells, blood clots, parasites, and emboli. However, the possibility of detecting high-amplitude signals from these objects against the background of blood depends on the parameters of the laser pulse. So, the dependencies of photoacoustic signals amplitude and number on laser pulse energy (5-150 μJ), pulse length (1, 2, 5 ns), and pulse repetition rate (2, 5, 10 kHz) for the melanoma cells were investigated. First, the PA responses of a melanoma cell suspension in vitro were measured to directly assess the efficiency of converting laser light into an acoustic signal. After it, the same dependence with the developed murine model based on constant rate melanoma cell injection into the animal blood flow was tested. Both in vivo and in vitro experiments show that signal generation efficiency increases with laser pulse energy above 15 μJ. Shorter pulses, especially 1 ns, provide more efficient signal generation as well as higher pulse rates. A higher pulse rate also provides more efficient signal generation, but also leads to overheating of the skin. The results show the limits where the photoacoustic flow cytometry system can be effectively used for the detection of circulating tumor cells in undiluted blood both for in vitro experiments and for in vivo murine models.
Collapse
Affiliation(s)
- Oleg V Grishin
- Science Medical Center, Saratov State University, Saratov, Russia
| | | | | | | | | | | | - Arkady S Abdurashitov
- A.V. Zelmann Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Olga A Sindeeva
- A.V. Zelmann Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Anatoly S Karavaev
- Science Medical Center, Saratov State University, Saratov, Russia
- Laboratory of Nonlinear Dynamics Modeling, Saratov Branch of the Institute of Radio-Engineering and Electronics of Russian Academy of Sciences, Saratov, Russia
- Department of Innovative Cardiological Information Technology, Institute of Cardiological Research, Saratov State Medical University, Saratov, Russia
| | - Danil D Kulminskiy
- Laboratory of Nonlinear Dynamics Modeling, Saratov Branch of the Institute of Radio-Engineering and Electronics of Russian Academy of Sciences, Saratov, Russia
- Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, Sochi, Russia
| | - Evgeny V Shashkov
- Pico-Femtoseconds Laser Laboratory, Photoelectronics Department, Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| | | | - Valery V Tuchin
- Science Medical Center, Saratov State University, Saratov, Russia
- Laboratory of Laser Molecular Imaging and Machine Learning, Tomsk State University, Tomsk, Russia
- Institute of Precision Mechanics and Control, FRC "Saratov Scientific Centre of the Russian Academy of Sciences", Saratov, Russia
- Bach Institute of Biochemistry, FRC "Fundamentals of Biotechnology of the Russian Academy of Sciences", Moscow, Russia
| |
Collapse
|
35
|
Mubarak HA, Kamal MM, Mahmoud Y, Abd-Elsamea FS, Abdelbary E, Gamea MG, El-Mahdy RI. The ameliorating effects of mesenchymal stem cells compared to α-tocopherol on apoptosis and autophagy in streptozotocin-induced diabetic rats: Implication of PI3K/Akt signaling pathway and entero-insular axis. J Cell Biochem 2023; 124:1705-1719. [PMID: 37796145 DOI: 10.1002/jcb.30482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/21/2023] [Accepted: 09/17/2023] [Indexed: 10/06/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) are considered a novel regenerative therapy that holds much potential. This study aimed to examine and compare the ameliorative effects of BM-MSCs compared to α-tocopherol (α-Toc) on apoptosis, autophagy, and β-cell function in a rat model of streptozotocin (STZ)-induced diabetes and further analyzed the implications and interrelations of the entero-insular axis, and type I phosphoinositide 3-kinase (PI3K)/Akt signaling. Forty adult male albino rats were categorized into four groups (n = 10, in each): control group, STZ-induced diabetic group (single i.p. injection of STZ 45 mg/kg), diabetic and treated with BM-MSCs injection, diabetic and treatment with α-Toc p.o. The serum glucose, insulin, nitric oxide (NO), and catalase (CAT) were measured. Histopathological examination of the pancreas, the expression levels of insulin, CD44, caspase-3, autophagy markers, P13K/Akt, and pancreas/duodenum homeobox protein 1, in pancreatic tissue, and glucose-dependent insulinotropic polypeptide (GIP) in the duodenum were detected by hematoxylin and eosin staining, immunofluorescence labeling, and by quantitative real-time polymerase chain reaction. The diabetic rats showed reduced insulin, hyperglycemia, nitrosative stress (NO, CAT), augmented apoptosis (caspase 3), impaired autophagy (p62/SQSTM1, LC3), downregulated PI3K/Akt pathway and increased GIP expression, and degeneration of pancreatic islets. Treatment with either BM-MSCs or α-Toc suppressed the nitrosative stress, reduced apoptosis, recovered autophagy, upregulated PI3K/Akt pathway, and subsequently increased insulin levels, decreased blood glucose, and downregulated GIP expression with partial restoration of pancreatic islets. Based on our findings, the cytoprotective effects of BM-MSCs and α-Toc in type 1-induced diabetes appeared to be related to repaired autophagy and recovered PI3K/Akt signaling. Moreover, we reported their novel effects on reversing intestinal GIP expression level. The effect of BM-MSCs was notably superior to that of α-Toc.
Collapse
Affiliation(s)
- Heba A Mubarak
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Manal M Kamal
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Yossra Mahmoud
- Department of Clinical Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Fatma S Abd-Elsamea
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Eman Abdelbary
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Marwa G Gamea
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Reham I El-Mahdy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Biochemistry and Physiology, West of Assiut, New Naser City, Badr University, Assiut, Egypt
| |
Collapse
|
36
|
Jin Y, Tang Z, Shang S, Chen Y, Han G, Song M, Zhou J, Zhang H, Ding Y. A Nanodisc-Paved Biobridge Facilitates Stem Cell Membrane Fusogenicity for Intracerebral Shuttling and Bystander Effects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302367. [PMID: 37543432 DOI: 10.1002/adma.202302367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/21/2023] [Indexed: 08/07/2023]
Abstract
Mesenchymal stem cell (MSC) therapies experience steadfast clinical advances but are still hindered by inefficient site-specific migration. An adaptable MSC membrane fusogenicity technology is conceptualized for lipid raft-mediated targeting ligand embedding by using toolkits of discoidal high-density lipoprotein (HDL)-containing biomimicking 4F peptides. According to the pathological clues of brain diseases, the vascular cell adhesion molecule 1 specialized VBP peptide is fused with 4F to assemble 4F-VBP (HDL), which acts as a biobridge and transfers VBP onto the living cell membrane via lipid rafts for surface engineering of MSCs in suspension. When compared with the membrane-modifying strategies of PEGylated phospholipids, 4F-VBP (HDL) provides a 3.86 higher linkage efficiency to obtain MSCs4F-VBP(HDL) , which can recognize and adhere to the inflammatory endothelium for efficient blood-brain barrier crossing and brain accumulation. In APPswe/PSEN1dE9 mice with Alzheimer's disease (AD), the transcriptomic analysis reveals that systemic administration of MSCs4F-VBP(HDL) can activate pathways associated with neuronal activity and diminish neuroinflammation for rewiring AD brains. This customizable HDL-mediated membrane fusogenicity platform primes MSC inflammatory brain delivery, which can be expanded to other disease treatments by simply fusing 4F with relevant ligands for living cell engineering.
Collapse
Affiliation(s)
- Yi Jin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Shibeilei Shang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yun Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Guochen Han
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Mingjie Song
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Huaqing Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| |
Collapse
|
37
|
Zhu G, Zhou Y, Xu Y, Wang L, Han M, Xi K, Tang J, Li Z, Kou Y, Zhou X, Feng Y, Gu Y, Chen L. Functionalized acellular periosteum guides stem cell homing to promote bone defect repair. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2000-2020. [PMID: 37071056 DOI: 10.1080/09205063.2023.2204779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/17/2023] [Indexed: 04/19/2023]
Abstract
The periosteum plays a key role in bone tissue regeneration, especially in the promotion and protection of new bones. However, among the bone repair materials, many biomimetic artificial periosteum lack the natural periosteal structure, stem cells, and immunoregulation required for bone regeneration. In this study, we used natural periosteum to produce acellular periosteum. To retain the appropriate cell survival structure and immunomodulatory proteins, we grafted the functional polypeptide SKP on the surface collagen of the periosteum via an amide bond, providing the acellular periosteum with the ability to recruit mesenchymal stem cells. Thus, we developed a biomimetic periosteum (DP-SKP) with the ability to promote stem cell homing and immunoregulation in vivo. Compared to the blank and simple decellularized periosteum groups, DP-SKP was more conducive to stem cell adhesion, growth, and osteogenic differentiation in vitro. Additionally, compared with the other two groups, DP-SKP significantly promoted mesenchymal stem cell homing to the periosteal transplantation site, improved the bone immune microenvironment, and accelerated new lamellar bone formation in the critical size defect of rabbit skulls in vivo. Therefore, this acellular periosteum with a mesenchymal stem cell homing effect is expected to be used as an extracellular artificial periosteum in clinical practice.
Collapse
Affiliation(s)
- Guoqing Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Suzhou Municipal Hospital, Suzhou, China
| | - Yidi Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingjun Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Han
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Kou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yu Feng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Hsueh PR, Ho SJ, Hsieh PC, Liu IM, Jean SS. Use of Multiple Doses of Intravenous Infusion of Umbilical Cord-Mesenchymal Stem Cells for the Treatment of Adult Patients with Severe COVID-19-Related Acute Respiratory Distress Syndrome: Literature Review. Stem Cells Int 2023; 2023:7179592. [PMID: 37638334 PMCID: PMC10457163 DOI: 10.1155/2023/7179592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
Objectives Acute respiratory distress syndrome (ARDS) is a critical complication in severe COVID-19 patients. The intravenous infusion (IVF) of umbilical cord- (UC-) mesenchymal stem cells (MSCs), validated to substantially reduce the release of several inflammatory cytokines in vivo, was also shown to exhibit benefits in improving hypoxemia among severe COVID-19 patients. A single dose of IVF-UC-MSCs therapy for severe COVID-19 patients was shown to alleviate the initial ARDS severity, but have 50%-67% case-fatality rates. In Taiwan, few adult patients with severe COVID-19-induced ARDS receiving compassionate adjuvant treatment consisting of either a single dose (1-10 × 106 cells/kg body weight (kg BW)) or three doses (5 × 106 cells/kg BW in each dose) of IVF-UC-MSCs had good outcomes. However, the optimal dosage and rounds of IVF-UC-MSCs administration for the treatment of severe COVID-19 patients with ARDS are undetermined. Methods We reviewed the 2020-2022 PubMed literature database concerning the clinical efficacy of IVF-UC-MSCs among severe COVID-19 patients. Results The data of COVID-19 case series in the PubMed literature revealed a notable heterogeneity in the therapeutic dosage (a single dose: 1-10 × 106 cells/kg BW; and three doses: 50-200 × 106 cells/kg BW in each dose) and the post-ARDS days of IVF-UC-MSCs administration (a single dose: 1-12; and multiple doses: 5-14) for the treatment of severe COVID-19-associated ARDS. The survival rates among these severe COVID-19 patients ranged from 50% to 76%. However, an overall rate of 93.1% of significant improvement in hypoxemia was observed for the COVID-19 survivors receiving IVF-UC-MSCs at the initial ARDS stage. Conclusions According to our analysis, the ideal treatment dosage of IVF-UC-MSCs for severe COVID-19-induced ARDS is likely 5 × 106 cells/kg BW for three cycles within 5 days of ARDS onset in severe COVID-19 patients.
Collapse
Affiliation(s)
- Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Departments of Laboratory Medicine and Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Jung Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan
| | - Po-Chuen Hsieh
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - I-Min Liu
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Shio-Shin Jean
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
- Departments of Internal Medicine and Critical Care Medicine, Min-Sheng General Hospital, Taoyuan, Taiwan
| |
Collapse
|
39
|
Yao Y, Zhang L, Cheng F, Jiang Q, Ye Y, Ren Y, He Y, Su D, Cheng L, Shi G, Dai L, Deng H. PPARγ-dependent hepatic macrophage switching acts as a central hub for hUCMSC-mediated alleviation of decompensated liver cirrhosis in rats. Stem Cell Res Ther 2023; 14:184. [PMID: 37501214 PMCID: PMC10375757 DOI: 10.1186/s13287-023-03416-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 07/17/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Decompensated liver cirrhosis (DLC), a terminal-stage complication of liver disease, is a major cause of morbidity and mortality in patients with hepatopathies. Human umbilical cord mesenchymal stem cell (hUCMSC) therapy has emerged as a novel treatment alternative for the treatment of DLC. However, optimized therapy protocols and the associated mechanisms are not entirely understood. METHODS We constructed a DLC rat model consistent with the typical clinical characteristics combined use of PB and CCL4. Performing dynamic detection of liver morphology and function in rats for 11 weeks, various disease characteristics of DLC and the therapeutic effect of hUCMSCs on DLC in experimental rats were thoroughly investigated, according to ascites examination, histopathological, and related blood biochemical analyses. Flow cytometry analysis of rat liver, immunofluorescence, and RT-qPCR was performed to examine the changes in the liver immune microenvironment after hucMSCs treatment. We performed RNA-seq analysis of liver and primary macrophages and hUCMSCs co-culture system in vitro to explore possible signaling pathways. PPARγ antagonist, GW9662, and clodronate liposomes were used to inhibit PPAR activation and pre-exhaustion of macrophages in DLC rats' livers, respectively. RESULTS We found that changing the two key issues, the frequency and initial phase of hUCMSCs infusion, can affect the efficacy of hUCMSCs, and the optimal hUCMSCs treatment schedule is once every week for three weeks at the early stage of DLC progression, providing the best therapeutic effect in reducing mortality and ascites, and improving liver function in DLC rats. hUCMSCs treatment skewed the macrophage phenotype from M1-type to M2-type by activating the PPARγ signaling pathway in the liver, which was approved by primary macrophages and hUCMSCs co-culture system in vitro. Both inhibition of PPARγ activation with GW9662 and pre-exhaustion of macrophages in DLC rats' liver abolished the regulation of hUCMSCs on macrophage polarization, thus attenuating the beneficial effect of hUCMSCs treatment in DLC rats. CONCLUSIONS These data demonstrated that the optimal hUCMSCs treatment effectively inhibits the ascites formation, prolongs survival and significantly improves liver structure and function in DLC rats through the activation of the PPARγ signaling pathway within liver macrophages. Our study compared the efficacy of different hUCMSCs infusion regimens for DLC, providing new insights on cell-based therapies for regenerative medicine.
Collapse
Affiliation(s)
- Yunqi Yao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Lin Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Fuyi Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Qingyuan Jiang
- Department of Obstetrics, Sichuan Provincial Hospital for Women and Children, Chengdu, People's Republic of China
| | - Yixin Ye
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Yushuang Ren
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Yuting He
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dongsheng Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Ke-yuan Road 4, No.1, Gao-peng Street, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
40
|
Yoon J, Lee SK, Park A, Lee J, Jung I, Song KB, Choi EJ, Kim S, Yu J. Exosome from IFN-γ-Primed Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Improved Skin Inflammation and Barrier Function. Int J Mol Sci 2023; 24:11635. [PMID: 37511392 PMCID: PMC10380988 DOI: 10.3390/ijms241411635] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/09/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The pathogenesis of atopic dermatitis (AD) is multifactorial, including immune dysregulation and epidermal barrier defects, and a novel therapeutic modality that can simultaneously target multiple pathways is needed. We investigated the therapeutic effects of exosomes (IFN-γ-iExo) secreted from IFN-γ-primed induced pluripotent stem cell-derived mesenchymal stem cells (iMSC) in mice with Aspergillus fumigatus-induced AD. IFN-γ-iExo was epicutaneously administered to mice with AD-like skin lesions. The effects of IFN-γ-iExo treatment were investigated through clinical scores, transepidermal water loss (TEWL) measurements, and histopathology. To elucidate the therapeutic mechanism, we used an in vitro model of human keratinocyte HaCaT cells stimulated with IL-4 and IL-13 and performed extensive bioinformatics analysis of skin mRNA from mice. The expression of indoleamine 2,3-dioxygenase was higher in IFN-γ primed iMSCs than in iMSCs. In human keratinocyte HaCaT cells, treatment with IFN-γ-iExo led to decreases in the mRNA expression of thymic stromal lymphopoietin, IL-25, and IL-33 and increases in keratin 1, keratin 10, desmoglein 1, and ceramide synthase 3. IFN-γ-iExo treatment significantly improved clinical and histological outcomes in AD mice, including clinical scores, TEWL, inflammatory cell infiltration, and epidermal thickness. Bioinformatics analysis of skin mRNA from AD mice showed that IFN-γ-iExo treatment is predominantly involved in skin barrier function and T cell immune response. Treatment with IFN-γ-iExo improved the clinical and histological outcomes of AD mice, which were likely mediated by restoring proper skin barrier function and suppressing T cell-mediated immune response.
Collapse
Affiliation(s)
- Jin Yoon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea; (J.Y.); (A.P.); (J.L.)
| | - Seul Ki Lee
- Brexogen Research Center, Brexogen Inc., Seoul 05855, Republic of Korea;
| | - Arum Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea; (J.Y.); (A.P.); (J.L.)
| | - Jiho Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Republic of Korea; (J.Y.); (A.P.); (J.L.)
| | - Inuk Jung
- School of Computer Science and Engineering, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Kun Baek Song
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (K.B.S.); (E.J.C.)
| | - Eom Ji Choi
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (K.B.S.); (E.J.C.)
| | - Soo Kim
- Brexogen Research Center, Brexogen Inc., Seoul 05855, Republic of Korea;
| | - Jinho Yu
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (K.B.S.); (E.J.C.)
| |
Collapse
|
41
|
Wu SCM, Zhu M, Chik SCC, Kwok M, Javed A, Law L, Chan S, Boheler KR, Liu YP, Chan GCF, Poon ENY. Adipose tissue-derived human mesenchymal stromal cells can better suppress complement lysis, engraft and inhibit acute graft-versus-host disease in mice. Stem Cell Res Ther 2023; 14:167. [PMID: 37357314 DOI: 10.1186/s13287-023-03380-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 05/18/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Acute graft-versus-host disease (aGvHD) is a life-threatening complication of allogeneic hematopoietic stem cell transplantation (HSCT). Transplantation of immunosuppressive human mesenchymal stromal cells (hMSCs) can protect against aGvHD post-HSCT; however, their efficacy is limited by poor engraftment and survival. Moreover, infused MSCs can be damaged by activated complement, yet strategies to minimise complement injury of hMSCs and improve their survival are limited. METHODS Human MSCs were derived from bone marrow (BM), adipose tissue (AT) and umbilical cord (UC). In vitro immunomodulatory potential was determined by co-culture experiments between hMSCs and immune cells implicated in aGvHD disease progression. BM-, AT- and UC-hMSCs were tested for their abilities to protect aGvHD in a mouse model of this disease. Survival and clinical symptoms were monitored, and target tissues of aGvHD were examined by histopathology and qPCR. Transplanted cell survival was evaluated by cell tracing and by qPCR. The transcriptome of BM-, AT- and UC-hMSCs was profiled by RNA-sequencing. Focused experiments were performed to compare the expression of complement inhibitors and the abilities of hMSCs to resist complement lysis. RESULTS Human MSCs derived from three tissues divergently protected against aGvHD in vivo. AT-hMSCs preferentially suppressed complement in vitro and in vivo, resisted complement lysis and survived better after transplantation when compared to BM- and UC-hMSCs. AT-hMSCs also prolonged survival and improved the symptoms and pathological features of aGvHD. We found that complement-decay accelerating factor (CD55), an inhibitor of complement, is elevated in AT-hMSCs and contributed to reduced complement activation. We further report that atorvastatin and erlotinib could upregulate CD55 and suppress complement in all three types of hMSCs. CONCLUSION CD55, by suppressing complement, contributes to the improved protection of AT-hMSCs against aGvHD. The use of AT-hMSCs or the upregulation of CD55 by small molecules thus represents promising new strategies to promote hMSC survival to improve the efficacy of transplantation therapy. As complement injury is a barrier to all types of hMSC therapy, our findings are of broad significance to enhance the use of hMSCs for the treatment of a wide range of disorders.
Collapse
Affiliation(s)
- Stanley Chun Ming Wu
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Manyu Zhu
- Department of Orthopaedics and Traumatology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Pathology, The Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Stanley C C Chik
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Maxwell Kwok
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong, Kowloon Bay, Hong Kong SAR, China
| | - Asif Javed
- School of Biomedical Science, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Laalaa Law
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Shing Chan
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kenneth R Boheler
- Division of Cardiology, Department of Medicine and Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yin Ping Liu
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Godfrey Chi Fung Chan
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
- , Doctors' Office, 9/F, Tower B, Hong Kong Children's Hospital, 1 Shing Cheong Road, Kowloon Bay, Hong Kong SAR, China.
| | - Ellen Ngar-Yun Poon
- Hong Kong Hub of Paediatric Excellence (HK HOPE), The Chinese University of Hong Kong, Kowloon Bay, Hong Kong SAR, China.
- The School of Biomedical Sciences, The Chinese University of Hong Kong, Rm 226A, 2/F, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Shatin, Hong Kong SAR, China.
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
42
|
Farid A, El-Alfy L, Madbouly N. Bone marrow-derived mesenchymal stem cells transplantation downregulates pancreatic NF-κB and pro-inflammatory cytokine profile in rats with type I and type II-induced diabetes: a comparison study. Biologia (Bratisl) 2023; 78:3165-3177. [DOI: 10.1007/s11756-023-01436-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/12/2023] [Indexed: 10/04/2024]
Abstract
AbstractDiabetes mellitus (DM) is a set of metabolic diseases defined by a persistently high blood sugar level. Mesenchymal stem cells (MSCs) are a novel potential therapeutic intervention in treatments of various diseases, which is also referred to as regenerative medicine. We aimed to compare the pro-inflammatory cytokines’ levels during bone marrow mesenchymal stem cells (BM-MSCs) transplantation in rats with induced type I (T1D) and type II diabetes (T2D). Thirty-five male Sprague dawley rats were divided into: Group I: the healthy control group, group II: untreated rats with streptozotocin (STZ)-induced T1D (65 mg/kg), group III: BM-MSCs treated rats with STZ-induced T1D, group IV: untreated rats with high-fat diet (HFD)/STZ-induced T2D (40 mg/kg), group V: BM-MSCs-treated rats with HFD/STZ-induced T2D. Biochemical, histopathological and immunohistochemical studies were applied. Our results showed that transplantation reduced hyperglycemia and increased insulin levels in both induced T1D and T2D. Also, reductions in the levels of inflammatory markers were noticed after transplantation that was coincided with nuclear factor-kappa B (NF-кB) immunohistochemical results; which showed negative or moderate cytoplasmic reactivity in treated groups III and V. These results indicated the ability of BM-MSCs transplantation to modulate the pro-inflammatory cytokine profile during treatment of both T1D and T2D.
Collapse
|
43
|
Desai N, Rana D, Pande S, Salave S, Giri J, Benival D, Kommineni N. "Bioinspired" Membrane-Coated Nanosystems in Cancer Theranostics: A Comprehensive Review. Pharmaceutics 2023; 15:1677. [PMID: 37376125 DOI: 10.3390/pharmaceutics15061677] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Achieving precise cancer theranostics necessitates the rational design of smart nanosystems that ensure high biological safety and minimize non-specific interactions with normal tissues. In this regard, "bioinspired" membrane-coated nanosystems have emerged as a promising approach, providing a versatile platform for the development of next-generation smart nanosystems. This review article presents an in-depth investigation into the potential of these nanosystems for targeted cancer theranostics, encompassing key aspects such as cell membrane sources, isolation techniques, nanoparticle core selection, approaches for coating nanoparticle cores with the cell membrane, and characterization methods. Moreover, this review underscores strategies employed to enhance the multi-functionality of these nanosystems, including lipid insertion, membrane hybridization, metabolic engineering, and genetic modification. Additionally, the applications of these bioinspired nanosystems in cancer diagnosis and therapeutics are discussed, along with the recent advances in this field. Through a comprehensive exploration of membrane-coated nanosystems, this review provides valuable insights into their potential for precise cancer theranostics.
Collapse
Affiliation(s)
- Nimeet Desai
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Shreya Pande
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, India
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, India
| | | |
Collapse
|
44
|
Wang X, Hu S, Zhu D, Li J, Cheng K, Liu G. Comparison of extruded cell nanovesicles and exosomes in their molecular cargos and regenerative potentials. NANO RESEARCH 2023; 16:7248-7259. [PMID: 37223430 PMCID: PMC9971669 DOI: 10.1007/s12274-023-5374-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 05/25/2023]
Abstract
Extracellular vesicles (EVs) generated from mesenchymal stem cells (MSCs) play an essential role in modulating cell-cell communication and tissue regeneration. The clinical translation of EVs is constrained by the poor yield of EVs. Extrusion has recently become an effective technique for producing a large scale of nanovesicles (NVs). In this study, we systematically compared MSC NVs (from extrusion) and EVs (from natural secretion). Proteomics and RNA sequencing data revealed that NVs resemble MSCs more closely than EVs. Additionally, microRNAs in NVs are related to cardiac repair, fibrosis repression, and angiogenesis. Lastly, intravenous delivery of MSC NVs improved heart repair and cardiac function in a mouse model of myocardial infarction. Electronic Supplementary Material Supplementary material (Figs. S1-S4) is available in the online version of this article at 10.1007/s12274-023-5374-3.
Collapse
Affiliation(s)
- Xianyun Wang
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000 China
- Scientific Research Data Center, The First Hospital of Hebei Medical University, Shijiazhuang, 050000 China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, 050000 China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, 050000 China
- Department of Molecular Biomedical Science, North Carolina State University, Raleigh, North Carolina 27607 USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, North Carolina 27607 USA
| | - Shiqi Hu
- Department of Molecular Biomedical Science, North Carolina State University, Raleigh, North Carolina 27607 USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, North Carolina 27607 USA
| | - Dashuai Zhu
- Department of Molecular Biomedical Science, North Carolina State University, Raleigh, North Carolina 27607 USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, North Carolina 27607 USA
| | - Junlang Li
- Department of Molecular Biomedical Science, North Carolina State University, Raleigh, North Carolina 27607 USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, North Carolina 27607 USA
| | - Ke Cheng
- Department of Molecular Biomedical Science, North Carolina State University, Raleigh, North Carolina 27607 USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill and North Carolina State University, Raleigh, North Carolina 27607 USA
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050000 China
- Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, 050000 China
- Hebei International Joint Research Center for Structural Heart Disease, Shijiazhuang, 050000 China
| |
Collapse
|
45
|
Miyake K, Azuma N, Rinoie C, Maeda S, Harada A, Li L, Minami I, Miyagawa S, Sawa Y. Regenerative Effect of Umbilical Cord-Derived Mesenchymal Stromal Cells in a Rat Model of Established Limb Ischemia. Circ J 2023; 87:412-420. [PMID: 36171115 DOI: 10.1253/circj.cj-22-0257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Although regenerative cell therapy is expected to be an alternative treatment for peripheral artery disease (PAD), many regenerative cell therapies have failed to show sufficient efficacy in clinical trials. Most preclinical studies have used acute ischemia models, despite PAD being a chronic disease. In addition, aging and atherosclerosis decrease the quality of a patient's stem cells. Therefore, using a non-acute ischemic preclinical model and stem cells with high regenerative potency are important for the development of effective regenerative therapy. In this study, we assessed the tissue regenerative potential of umbilical cord-derived mesenchymal stromal cells (UCMSCs), which could potentially be an ideal cell source, in a rat model of established ischemia. METHODS AND RESULTS The regenerative capacity of UCMSCs was analyzed in terms of angiogenesis and muscle regeneration. In vitro analysis showed that UCMSCs secrete high amounts of cytokines associated with angiogenesis and muscle regeneration. In vivo experiments in a rat non-acute ischemia model showed significant improvement in blood perfusion after intravenous injection of UCMSCs compared with injection of culture medium or saline. Histological analysis revealed UCMSCs injection enhanced angiogenesis, with an increased number of von Willebrand factor-positive microcapillaries, and improved muscle regeneration. CONCLUSIONS These results suggest that intravenous administration of UCMSCs may be useful for treating patients with PAD.
Collapse
Affiliation(s)
- Keisuke Miyake
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Nobuyoshi Azuma
- Department of Vascular Surgery, Asahikawa Medical University
| | | | - Shusaku Maeda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Liu Li
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | | | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | - Yoshiki Sawa
- Department of Future Medicine, Division of Health Science, Osaka University Graduate School of Medicine
| |
Collapse
|
46
|
Najera J, Hao J. Recent advance in mesenchymal stem cells therapy for atopic dermatitis. J Cell Biochem 2023; 124:181-187. [PMID: 36576973 DOI: 10.1002/jcb.30365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/07/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells found in a variety of tissues in the body, including but not limited to bone marrow, adipose tissue, umbilical cord, and umbilical cord blood. Given their immunomodulatory effect and ability to be readily isolated from several tissues, they have great potential to be used as a therapeutic agent in a variety of immune-mediated disorders. Atopic dermatitis (AD) is a persistent and relapsing immune skin condition that has recently become more common in several species such as humans, canines, equines, and felines. The use of MSCs to treat AD has piqued the great interest of researchers in recent years. In this article, we review the recent understanding of AD pathology and advances in preclinical and clinical studies of MSCs, MSCs-derived conditional media and exosomes as therapeutic tools to treat AD.
Collapse
Affiliation(s)
- Jonathan Najera
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, California, USA.,Department of Biology, College of Science, California State University Polytechnic University, Pomona, California, USA
| | - Jijun Hao
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
47
|
Human-derived hair follicle stem cells and hydrogen sulfide on focal cerebral ischemia model: A comparative evaluation of radiologic, neurobehavioral and immunohistochemical results. Brain Res 2023; 1799:148170. [PMID: 36410427 DOI: 10.1016/j.brainres.2022.148170] [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: 08/20/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
The present study investigated the effects of intracerebral human-derived hair follicle stem cells (HFBSCs), whether alone or in combination with hydrogen sulfide (H2S) in a rat model of focal cerebral ischemia. The rats were randomly assigned into 4 groups (n = 10): Control (phosphate buffered saline (PBS)), Group A (at 24 h post-middle cerebral artery occlusion(MCAo), stereotaxic intracerebral, 1,0 × 106, total 10 μL HFBSCs), Group B (3-14 d post-MCAo, intraperitoneal (i.p.), 25 μM/kg/day H2S), Group AB (HFBSCs + H2S). Cranial magnetic resonance images were recorded on postoperative 1st and 28th days. Three dimensional analysis was performed to calculate the infarct volumes. Rotarod and cylinder tests were performed after MCAo and finally all rats were euthanized by cardiac perfusion at 28 days after MCAo for immunohistochemical analysis. The reduction in infarct volumes of rats receiving HFBSC was significant. The cranial infarct volume on the postoperative 28th day was significantly higher in the group in which H2S was administered alone compared to the HFBSC alone group. All animals showed steadily improved spontaneous locomotor activity from day 7 post-MCAo on rotarod test, from day 1 on cylinder test, but showed no significant differences at all times. In all groups, the grading scores of CD34, CD5, CD11b and GFAP immunohistochemical markers did not differ significantly. In conclusion, intracerebral HFBSC treatment after 24 h of ischemic stroke may be an effective way to reduce the cranial infarct volume, whereas H2S treatment alone or in combination with HFBSC may not be sufficient for ischemic brain injury.
Collapse
|
48
|
Allogeneic Mesenchymal Stromal Cells as a Global Pediatric Prospective Approach in the Treatment of Respiratory Failure Associated with Surfactant Protein C Dysfunction. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10010162. [PMID: 36670712 PMCID: PMC9857592 DOI: 10.3390/children10010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/02/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023]
Abstract
Mesenchymal stromal cells (MSCs) have been proposed as a new therapeutic strategy to treat congenital and acquired respiratory system diseases. We describe a case report of an 18-month-old male patient with progressive chronic respiratory failure, associated with mutations of the surfactant protein C gene (SFTPC) due to c.289G > T variant p.Gly97Ser (rs927644577) and c.176A > G variant (p.His59Arg), submitted to repeated intravenous infusions of allogeneic bone marrow (BM) MSCs. The clinical condition of the patient was monitored. Immunologic studies before and during MSC treatment were performed. No adverse events related to the MSC infusions were recorded. Throughout the MSC treatment period, the patient showed a growth recovery. Starting the second infusion, the patient experienced an improvement in his respiratory condition, with progressive adaptation to mechanical ventilation. After the third infusion, five hours/die of spontaneous breathing was shown, and after infusion IV, spontaneous ventilation for 24/24 h was recorded. A gradual decrease of lymphocytes and cell subpopulations was observed. No variations in the in vitro T cell response to PHA were determined by MSC treatment as well as for the in vitro B cell response. A decrease in IFN-γ, TNF-α, and IL-10 levels was also detected. Even though we cannot exclude an improvement of pulmonary function due to the physiological maturation, the well-known action of MSCs in the repair of lung tissue, together with the sequence of events observed in our patient, may support the therapeutic role of MSCs in this clinical condition. However, further investigations are necessary to confirm the result and long-term follow-up will be mandatory to confirm the benefits on the pulmonary condition.
Collapse
|
49
|
Lee SH, Choung JS, Kim JM, Kim H, Kim M. Distribution of Embryonic Stem Cell-Derived Mesenchymal Stem Cells after Intravenous Infusion in Hypoxic-Ischemic Encephalopathy. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010227. [PMID: 36676176 PMCID: PMC9861288 DOI: 10.3390/life13010227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Systemic administration of mesenchymal stem cells (MSCs) has been reported to improve neurological function in brain damage, including hypoxic-ischemic encephalopathy (HIE), though the action mechanisms have not been fully elucidated. In this study, the cells were tracked live using a Pearl Trilogy Small Animal fluorescence imaging system after human embryonic stem Cell-Derived MSCs (ES-MSCs) infusion for an HIE mouse model. ES-MSC-treated HIE mice showed neurobehavioral improvement. In vivo imaging showed similar sequential migration of ES-MSCs from lungs, liver, and spleen within 7 days in both HIE and normal mice with the exception of lungs, where there was higher entrapment in the HIE 1 h after infusion. In addition, ex vivo experiments confirmed time-dependent infiltration of ES-MSCs into the organs, with similar findings in vivo, although lungs and brain revealed small differences. ES-MSCs seemed to remain in the brain only in the case of HIE on day 14 after the cell infusion. The homing effect in the host brain was confirmed with immunofluorescence staining, which showed that grafted cells remained in the brain tissue at the lesion area with neurorestorative findings. Further research should be carried out to elucidate the role of each host organ's therapeutic effects when stem cells are systemically introduced.
Collapse
Affiliation(s)
- Su Hyun Lee
- School of Medicine, CHA University, Pocheon 13496, Republic of Korea
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam 13488, Republic of Korea
| | - Jin Seung Choung
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea
| | - Jong Moon Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea
| | - Hyunjin Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea
| | - MinYoung Kim
- Rehabilitation and Regeneration Research Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13488, Republic of Korea
- Department of Biomedical Science, CHA University, Seongnam 13488, Republic of Korea
- Correspondence: ; Tel.: +82-31-780-1872
| |
Collapse
|
50
|
Botelho BF, Barreira AL, Leite M, Morales MM. Chronic Kidney Disease: Challenges in Translational Medicine. Methods Mol Biol 2023; 2575:61-75. [PMID: 36301471 DOI: 10.1007/978-1-0716-2716-7_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Chronic kidney disease (CKD) has long been recognized as a state of progressive decline in renal function. Morbidity and mortality are well correlated to the stage of renal function decline. Approximately one million deaths are estimated to be related to CKD worldwide. They are mostly associated with cardiovascular disease as a result of concurrent hypertension, accelerated atherosclerosis, and volume overload. Even with the best current treatment, disease progression is the general rule with a small fraction who reach CKD stage 5 requiring kidney transplantation or dialysis. Transplant patients show substantial reductions in mortality and cardiovascular events, as well as improvements in quality of life. However, the capacity of health systems to deliver kidney transplantation varies worldwide with worse indicators in low-income countries. Consequently, exploring novel and better therapeutic options for CKD is mandatory. Cell-based therapy is a promising strategy for treating CKD in preclinical models, and several clinical trials involving kidney disease exhibit a favorable safety profile. This chapter aims to provide an overview of CKD and the recent results of clinical trials of cell therapy in kidney diseases.
Collapse
Affiliation(s)
- Bruno Freire Botelho
- Department of Nephrology, School of Medicine, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | - André Luis Barreira
- Department of Nephrology, School of Medicine, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | - Maurilo Leite
- Department of Nephrology, School of Medicine, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, Brazil
- Hospital Universitário Clementino Fraga Filho Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | - Marcelo Marcos Morales
- Laboratory of Cellular and Molecular Physiology, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Ilha do Fundão, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|