201
|
Chae HK, Suh N, Jang MJ, Kim YS, Kim BH, Aum J, Shin HC, You D, Hong B, Park HK, Kim CS. Efficacy and Safety of Human Bone Marrow-Derived Mesenchymal Stem Cells according to Injection Route and Dose in a Chronic Kidney Disease Rat Model. Int J Stem Cells 2022; 16:66-77. [PMID: 35483715 PMCID: PMC9978839 DOI: 10.15283/ijsc21146] [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: 08/26/2021] [Revised: 02/21/2022] [Accepted: 03/20/2022] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives We compared the efficacy and safety of human bone marrow-derived mesenchymal stem cells (hBMSC), delivered at different doses and via different injection routes in an animal model of chronic kidney disease. Methods and Results A total of ninety 12-week-old rats underwent 5/6 nephrectomy and randomized among nine groups: sham, renal artery control (RA-C), tail vein control (TV-C), renal artery low dose (RA-LD) (0.5×106 cells), renal artery moderate dose (RA-MD) (1.0×106 cells), renal artery high dose (RA-HD) (2.0×106 cells), tail vein low dose (TV-LD) (0.5×106 cells), tail vein moderate dose (TV-MD) (1.0×106 cells), and tail vein high dose (TV-HD) (2.0×106 cells). Renal function and mortality of rats were evaluated after hBMSC injection. Serum blood urea nitrogen was significantly lower in the TV-HD group at 2 weeks (p<0.01), 16 weeks (p<0.05), and 24 weeks (p<0.01) than in the TV-C group, as determined by one-way ANOVA. Serum creatinine was significantly lower in the TV-HD group at 24 weeks (p<0.05). At 8 weeks, creatinine clearance was significantly higher in the TV-MD and TV-HD groups (p<0.01, p<0.05) than in the TV-C group. In the safety evaluation, we observed no significant difference among the groups. Conclusions Our findings confirm the efficacy and safety of high dose (2×106 cells) injection of hBMSC via the tail vein.
Collapse
Affiliation(s)
- Han Kyu Chae
- Department of Urology, Gangneung Asan Medical Center, University of Ulsan College of Medicine, Gangneung, Korea
| | - Nayoung Suh
- Department of Pharmaceutical Engineering, College of Medical Sciences and Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, Korea
| | - Myong Jin Jang
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Yu Seon Kim
- Department of Urology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bo Hyun Kim
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Joomin Aum
- Department of Urology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | - Dalsan You
- Department of Urology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bumsik Hong
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyung Keun Park
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Choung-Soo Kim
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea,Correspondence to Choung-Soo Kim, Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea, Tel: +82-2-3010-3734, Fax: +82-2-477-8928, E-mail:
| |
Collapse
|
202
|
Sanmartin MC, Borzone FR, Giorello MB, Yannarelli G, Chasseing NA. Mesenchymal Stromal Cell-Derived Extracellular Vesicles as Biological Carriers for Drug Delivery in Cancer Therapy. Front Bioeng Biotechnol 2022; 10:882545. [PMID: 35497332 PMCID: PMC9046597 DOI: 10.3389/fbioe.2022.882545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second leading cause of death worldwide, with 10.0 million cancer deaths in 2020. Despite advances in targeted therapies, some pharmacological drawbacks associated with anticancer chemo and immunotherapeutic agents include high toxicities, low bioavailability, and drug resistance. In recent years, extracellular vesicles emerged as a new promising platform for drug delivery, with the advantage of their inherent biocompatibility and specific targeting compared to artificial nanocarriers, such as liposomes. Particularly, mesenchymal stem/stromal cells were proposed as a source of extracellular vesicles for cancer therapy because of their intrinsic properties: high in vitro self-renewal and proliferation, regenerative and immunomodulatory capacities, and secretion of extracellular vesicles that mediate most of their paracrine functions. Moreover, extracellular vesicles are static and safer in comparison with mesenchymal stem/stromal cells, which can undergo genetic/epigenetic or phenotypic changes after their administration to patients. In this review, we summarize currently reported information regarding mesenchymal stem/stromal cell-derived extracellular vesicles, their proper isolation and purification techniques - from either naive or engineered mesenchymal stem/stromal cells - for their application in cancer therapy, as well as available downstream modification methods to improve their therapeutic properties. Additionally, we discuss the challenges associated with extracellular vesicles for cancer therapy, and we review some preclinical and clinical data available in the literature.
Collapse
Affiliation(s)
- María Cecilia Sanmartin
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro - CONICET, Buenos Aires, Argentina
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Belén Giorello
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gustavo Yannarelli
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro - CONICET, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
203
|
Li Y, Gao H, Brunner TM, Hu X, Yan Y, Liu Y, Qiao L, Wu P, Li M, Liu Q, Yang F, Lin J, Löhning M, Shen P. Menstrual blood-derived mesenchymal stromal cells efficiently ameliorate experimental autoimmune encephalomyelitis by inhibiting T cell activation in mice. Stem Cell Res Ther 2022; 13:155. [PMID: 35410627 PMCID: PMC8995916 DOI: 10.1186/s13287-022-02838-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/10/2021] [Indexed: 11/27/2022] Open
Abstract
Background Immunosuppressive properties grant mesenchymal stromal cells (MSCs) promising potential for treating autoimmune diseases. As autologous MSCs suffer from limited availability, the readily available allogeneic MSCs isolated from menstrual blood (MB-MSCs) donated by young, healthy individuals offer great potential. Here, we evaluate the therapeutic potential of MB-MSCs as ready-to-use allo-MSCs in multiple sclerosis, an autoimmune disease developed by the activation of myelin sheath-reactive Th1 and Th17 cells, by application in its animal model experimental autoimmune encephalomyelitis (EAE). Methods We assessed the therapeutic effect of MB-MSCs transplanted via either intravenous (i.v.) or intraperitoneal (i.p.) route in EAE in comparison with umbilical cord-derived MSCs (UC-MSCs). We used histology to assess myelin sheath integrity and infiltrated immune cells in CNS and flow cytometry to evaluate EAE-associated inflammatory T cells and antigen-presenting cells in lymphoid organs. Results We observed disease-ameliorating effects of MB-MSCs when transplanted at various stages of EAE (day − 1, 6, 10, and 19), via either i.v. or i.p. route, with a potency comparable to UC-MSCs. We observed reduced Th1 and Th17 cell responses in mice that had received MB-MSCs via either i.v. or i.p. injection. The repressed Th1 and Th17 cell responses were associated with a reduced frequency of plasmacytoid dendritic cells (pDCs) and a suppressed co-stimulatory capacity of pDCs, cDCs, and B cells. Conclusions Our data demonstrate that the readily available MB-MSCs significantly reduced the disease severity of EAE upon transplantation. Thus, they have the potential to be developed as ready-to-use allo-MSCs in MS-related inflammation. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02838-8.
Collapse
Affiliation(s)
- Yonghai Li
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Haiyao Gao
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tobias M Brunner
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117, Berlin, Germany.,Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Xiaoxi Hu
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yushan Yan
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yanli Liu
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liang Qiao
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Peihua Wu
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117, Berlin, Germany.,Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
| | - Meng Li
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Qing Liu
- Henan Key Lab of Biological Psychiatry, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Fen Yang
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China.,School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Juntang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Max Löhning
- Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117, Berlin, Germany. .,Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany.
| | - Ping Shen
- Stem Cell and Biotherapy Engineering Research Center of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, China. .,Pitzer Laboratory of Osteoarthritis Research, German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117, Berlin, Germany. .,Experimental Immunology and Osteoarthritis Research, Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany.
| |
Collapse
|
204
|
Fagoonee S, Shukla SP, Dhasmana A, Birbrair A, Haque S, Pellicano R. Routes of Stem Cell Administration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022:63-82. [PMID: 35389198 DOI: 10.1007/5584_2022_710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Stem cells are very promising for the treatment of a plethora of human diseases. Numerous clinical studies have been conducted to assess the safety and efficacy of various stem cell types. Factors that ensure successful therapeutic outcomes in patients are cell-based parameters such as source, viability, and number, as well as frequency and timing of intervention and disease stage. Stem cell administration routes should be appropriately chosen as these can affect homing and engraftment of the cells and hence reduce therapeutic effects, or compromise safety, resulting in serious adverse events. In this chapter, we will describe the use of stem cells in organ repair and regeneration, in particular, the liver and the available routes of cell delivery in the clinic for end-stage liver diseases. Factors affecting homing and engraftment of stem cells for each administration route will be discussed.
Collapse
Affiliation(s)
- Sharmila Fagoonee
- Institute of Biostructure and Bioimaging, National Research Council (CNR), Molecular Biotechnology Center, Turin, Italy.
| | - Shiv Poojan Shukla
- Department of Dermatology & Cutaneous Biology, Sydney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Anupam Dhasmana
- Department of Immunology and Microbiology and South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA
- Department of Biosciences and Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Bursa Uludağ University Faculty of Medicine, Nilüfer, Bursa, Turkey
| | | |
Collapse
|
205
|
Li J, Wang Q, An Y, Chen X, Xing Y, Deng Q, Li Z, Wang S, Dai X, Liang N, Hou Y, Yang H, Shang Z. Integrative Single-Cell RNA-Seq and ATAC-Seq Analysis of Mesenchymal Stem/Stromal Cells Derived from Human Placenta. Front Cell Dev Biol 2022; 10:836887. [PMID: 35450295 PMCID: PMC9017713 DOI: 10.3389/fcell.2022.836887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells derived from placenta (PMSCs) are an attractive source for regenerative medicine because of their multidifferentiation potential and immunomodulatory capabilities. However, the cellular and molecular heterogeneity of PMSCs has not been fully characterized. Here, we applied single-cell RNA sequencing (scRNA-seq) and assay for transposase-accessible chromatin sequencing (scATAC-seq) techniques to cultured PMSCs from human full-term placenta. Based on the inferred characteristics of cell clusters, we identify several distinct subsets of PMSCs with specific characteristics, including immunomodulatory-potential and highly proliferative cell states. Furthermore, integrative analysis of gene expression and chromatin accessibility showed a clearer chromatin accessibility signature than those at the transcriptional level on immunomodulatory-related genes. Cell cycle gene-related heterogeneity can be more easily distinguished at the transcriptional than the chromatin accessibility level in PMSCs. We further reveal putative subset-specific cis-regulatory elements regulating the expression of immunomodulatory- and proliferation-related genes in the immunomodulatory-potential and proliferative subpopulations, respectively. Moreover, we infer a novel transcription factor PRDM1, which might play a crucial role in maintaining immunomodulatory capability by activating PRDM1-regulon loop. Collectively, our study first provides a comprehensive and integrative view of the transcriptomic and epigenomic features of PMSCs, which paves the way for a deeper understanding of cellular heterogeneity and offers fundamental biological insight of PMSC subset-based cell therapy.
Collapse
Affiliation(s)
- Jinlu Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Quanlei Wang
- BGI-Shenzhen, Shenzhen, China
- Key Laboratory of Regenerative Medicine of Ministry of Education, Biology Postdoctoral Research Station, Jinan University, Guangzhou, China
| | | | | | - Yanan Xing
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Qiuting Deng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Zelong Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Shengpeng Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | - Xi Dai
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
| | | | | | - Huanming Yang
- BGI-Shenzhen, Shenzhen, China
- James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Zhouchun Shang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI-Shenzhen, Shenzhen, China
- BGI College, Northwest University, Xi’an, China
- *Correspondence: Zhouchun Shang,
| |
Collapse
|
206
|
Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther 2022; 13:146. [PMID: 35379361 PMCID: PMC8981790 DOI: 10.1186/s13287-022-02822-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/20/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based therapy has been considered as a promising approach targeting a variety of intractable diseases due to remarkable multiple effect of MSCs, such as multilineage differentiation, immunomodulatory property, and pro-regenerative capacity. However, poor engraftment, low survival rate of transplanted MSC, and impaired donor-MSC potency under host age/disease result in unsatisfactory therapeutic outcomes. Enhancement strategies, including genetic manipulation, pre-activation, and modification of culture method, have been investigated to generate highly functional MSC, and approaches for MSC pre-activation are highlighted. In this review, we summarized the current approaches of MSC pre-activation and further classified, analysed the scientific principles and main characteristics of these manipulations, and described the pros and cons of individual pre-activation strategies. We also discuss the specialized tactics to solve the challenges in this promising field so that it improves MSC therapeutic functions to serve patients better.
Collapse
Affiliation(s)
- Meirong Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| | - Yufeng Jiang
- Wound Repairing Department, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Qian Hou
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Yali Zhao
- Central Laboratory, Trauma Treatment Center, Chinese PLA General Hospital, Hainan Hospital, Sanya, China
| | - Lingzhi Zhong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| |
Collapse
|
207
|
Hu T, Liu Y, Li X, Li X, Liu Y, Wang Q, Huang J, Yu J, Wu Y, Chen S, Zeng T, Tan L. Tumor necrosis factor-alpha stimulated gene-6: A biomarker reflecting disease activity in rheumatoid arthritis. J Clin Lab Anal 2022; 36:e24395. [PMID: 35353944 PMCID: PMC9102767 DOI: 10.1002/jcla.24395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To explore the serum tumor necrosis factor-alpha stimulated gene-6 (TSG-6) level and its association with disease activity in rheumatoid arthritis (RA) patients. METHODS We recruited 176 RA patients, 178 non-RA patients (lupus erythematosus, osteoarthritis, ulcerative colitis, ankylosing spondylitis and psoriasis) and 71 healthy subjects. Serum TSG-6 levels were detected by enzyme-linked immunosorbent assay (ELISA). RA patients were divided into inactive RA and active RA groups by disease activity score of 28 joints based on C-reactive protein (DAS28-CRP). The receiver operating characteristic (ROC) curve and Spearman's rank correlation test analyzed the correlation between TSG-6 concentration and RA disease activity. RESULTS Tumor necrosis factor-alpha stimulated gene-6 levels in the RA group were increased (p < 0.01). TSG-6 concentrations indicated an upward tendency with increased disease activity; The area under the curve (AUC) of TSG-6 for diagnosing RA and assessing the severity of RA were 0.78 and 0.80, respectively; The combination of TSG-6 and anti-mutated citrullinated vimentin antibodies (anti-MCV) (sensitivity:98.4%)improved the diagnostic accuracy of RA. Binary logistic regression analysis showed that TSG-6 was an independent risk factor related to the severity of RA, and OR (95% CI) was 1.2 (1.003-1.453). CONCLUSION The TSG-6 levels in RA patients were elevated and related to disease activity. Therefore, TSG-6 may serve as a new potential biomarker for evaluating RA disease activity.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China.,School of Public Health of Nanchang University, Nanchang, China
| | - Yuhan Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China.,School of Public Health of Nanchang University, Nanchang, China
| | - Xu Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Xiaohang Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Yanzhao Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Qunxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Jiayi Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianlin Yu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Yang Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Simei Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| | - Tingting Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liming Tan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, China
| |
Collapse
|
208
|
Abstract
Human mesenchymal stem cells (MSCs), also known as mesenchymal stromal cells or medicinal signaling cells, are important adult stem cells for regenerative medicine, largely due to their regenerative characteristics such as self-renewal, secretion of trophic factors, and the capability of inducing mesenchymal cell lineages. MSCs also possess homing and trophic properties modulating immune system, influencing microenvironment around damaged tissues and enhancing tissue repair, thus offering a broad perspective in cell-based therapies. Therefore, it is not surprising that MSCs have been the broadly used adult stem cells in clinical trials. To gain better insights into the current applications of MSCs in clinical applications, we perform a comprehensive review of reported data of MSCs clinical trials conducted globally. We summarize the biological effects and mechanisms of action of MSCs, elucidating recent clinical trials phases and findings, highlighting therapeutic effects of MSCs in several representative diseases, including neurological, musculoskeletal diseases and most recent Coronavirus infectious disease. Finally, we also highlight the challenges faced by many clinical trials and propose potential solutions to streamline the use of MSCs in routine clinical applications and regenerative medicine.
Collapse
|
209
|
Han Y, Yang J, Fang J, Zhou Y, Candi E, Wang J, Hua D, Shao C, Shi Y. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduct Target Ther 2022; 7:92. [PMID: 35314676 PMCID: PMC8935608 DOI: 10.1038/s41392-022-00932-0] [Citation(s) in RCA: 265] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 11/18/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
AbstractMesenchymal stromal/stem cells (MSCs) possess multi-lineage differentiation and self-renewal potentials. MSCs-based therapies have been widely utilized for the treatment of diverse inflammatory diseases, due to the potent immunoregulatory functions of MSCs. An increasing body of evidence indicates that MSCs exert their therapeutic effects largely through their paracrine actions. Growth factors, cytokines, chemokines, extracellular matrix components, and metabolic products were all found to be functional molecules of MSCs in various therapeutic paradigms. These secretory factors contribute to immune modulation, tissue remodeling, and cellular homeostasis during regeneration. In this review, we summarize and discuss recent advances in our understanding of the secretory behavior of MSCs and the intracellular communication that accounts for their potential in treating human diseases.
Collapse
|
210
|
A Novel 3D Culture System Using a Chitin-Based Polysaccharide Material Produces High-Quality Allogeneic Human UCMSCs with Dispersed Sphere Morphology. Cells 2022; 11:cells11060995. [PMID: 35326446 PMCID: PMC8947357 DOI: 10.3390/cells11060995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/07/2022] [Accepted: 03/13/2022] [Indexed: 11/29/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation, in particular allogeneic transplantation, is a promising therapy for a variety of diseases. However, before performing allograft treatment it is necessary to find suitable donors, establish culture methods that maintain cell quality, and reduce cell production costs. Here, we present a new method of producing allogeneic MSCs combining human umbilical cord-derived mesenchymal stem cells (UCMSCs) and chitin-based polysaccharide fibers (Cellhesion® MS). UCMSC numbers significantly increased, and cells grew as dispersed spheres on Cellhesion® MS. Subsequent biological analyses showed that the expression levels of stemness-related and migration-related genes were significantly upregulated, including octamer-binding transcription factor 4 (OCT4), Nanog homeobox (NANOG), and C-X-C chemokine receptor type 4 (CXCR4). The secretion levels of paracrine factors such as prostaglandin E2 (PGE2), TNFα-stimulating gene (TSG)-6, fibroblast growth factor 2 (bFGF), and Angiogenin (Ang) from UCMSCs using Cellhesion® MS were significantly higher than with microcarrier and U-bottom plate culture. In addition, culture supernatant from UCMSCs with Cellhesion® MS had better angiogenic potential than that from monolayer cultured UCMSCs. Furthermore, we succeeded in a scaled-up culture of UCMSCs with Cellhesion® MS using a closed culture bag. Therefore, Cellhesion® MS is a key material for producing high-quality UCMSCs in a three-dimensional (3D) culture system.
Collapse
|
211
|
Poomani MS, Mariappan I, Perumal R, Regurajan R, Muthan K, Subramanian V. Mesenchymal Stem Cell (MSCs) Therapy for Ischemic Heart Disease: A Promising Frontier. Glob Heart 2022; 17:19. [PMID: 35342702 PMCID: PMC8916054 DOI: 10.5334/gh.1098] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/26/2022] [Indexed: 01/07/2023] Open
Abstract
Although tremendous progress has been made in conventional treatment for ischemic heart disease, it still remains a major cause of death and disability. Cell-based therapeutics holds an exciting frontier of research for complete cardiac recuperation. The capacity of diverse stem and progenitor cells to stimulate cardiac renewal has been analysed, with promising results in both pre-clinical and clinical trials. Mesenchymal stem cells have been ascertained to have regenerative ability via a variety of mechanisms, including differentiation from the mesoderm lineage, immunomodulatory properties, and paracrine effects. Also, their availability, maintenance, and ability to replenish endogenous stem cell niches have rendered them suitable for front-line research. This review schemes to outline the use of mesenchymal stem cell therapeutics for ischemic heart disease, their characteristics, the potent mechanisms of mesenchymal stem cell-based heart regeneration, and highlight preclinical data. Additionally, we discuss the results of the clinical trials to date as well as ongoing clinical trials on ischemic heart disease.
Collapse
Affiliation(s)
- Merlin Sobia Poomani
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli 627012, Tamil Nadu, India
| | - Iyyadurai Mariappan
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli 627012, Tamil Nadu, India
| | | | - Rathika Regurajan
- Center for Marine Science and Technology, Manonmaniam Sundaranar University, Tirunelveli 627012 Tamil Nadu, India
| | - Krishnaveni Muthan
- Center for Marine Science and Technology, Manonmaniam Sundaranar University, Tirunelveli 627012 Tamil Nadu, India
| | - Venkatesh Subramanian
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli 627012, Tamil Nadu, India
| |
Collapse
|
212
|
Yan W, Chen Y, Guo Y, Xia Y, Li C, Du Y, Lin C, Xu X, Qi T, Fan M, Zhang F, Hu G, Gao E, Liu R, Hai C, Tao L. Irisin Promotes Cardiac Homing of Intravenously Delivered MSCs and Protects against Ischemic Heart Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103697. [PMID: 35038246 PMCID: PMC8895138 DOI: 10.1002/advs.202103697] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/09/2021] [Indexed: 05/15/2023]
Abstract
Few intravenously administered mesenchymal stromal cells (MSCs) engraft to the injured myocardium, thereby limiting their therapeutic efficacy for the treatment of ischemic heart injury. Here, it is found that irisin pretreatment increases the cardiac homing of adipose tissue-derived MSCs (ADSCs) administered by single and multiple intravenous injections to mice with MI/R by more than fivefold, which subsequently increases their antiapoptotic, proangiogenic, and antifibrotic effects in rats and mice that underwent MI/R. RNA sequencing, Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway analysis, and loss-of-function studies identified CSF2RB as a cytokine receptor that facilitates the chemotaxis of irisin-treated ADSCs in the presence of CSF2, a chemokine that is significantly upregulated in the ischemic heart. Cardiac-specific CSF2 knockdown blocked the cardiac homing and cardioprotection abilities of intravenously injected irisin-treated ADSCs in mice subjected to MI/R. Moreover, irisin pretreatment reduced the apoptosis of hydrogen peroxide-induced ADSCs and increased the paracrine proangiogenic effect of ADSCs. ERK1/2-SOD2, and ERK1/2-ANGPTL4 are responsible for the antiapoptotic and paracrine angiogenic effects of irisin-treated ADSCs, respectively. Integrin αV/β5 is identified as the irisin receptor in ADSCs. These results provide compelling evidence that irisin pretreatment can be an effective means to optimize intravenously delivered MSCs as therapy for ischemic heart injury.
Collapse
Affiliation(s)
- Wenjun Yan
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Youhu Chen
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Yongzhen Guo
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Yunlong Xia
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Congye Li
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Yunhui Du
- Beijing Anzhen HospitalCapital Medical UniversityBeijing Institute of Heart, Lung and Blood Vessel DiseasesBeijing100029China
| | - Chen Lin
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Xiaoming Xu
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Tingting Qi
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Miaomiao Fan
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Fuyang Zhang
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Guangyu Hu
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Erhe Gao
- Center for Translational MedicineTemple UniversityPhiladelphiaPA19104USA
| | - Rui Liu
- Department of ToxicologyShanxi Key Lab of Free Radical Biology and MedicineSchool of Public HealthThe Fourth Military Medical UniversityXi'an710032China
| | - Chunxu Hai
- Department of ToxicologyShanxi Key Lab of Free Radical Biology and MedicineSchool of Public HealthThe Fourth Military Medical UniversityXi'an710032China
| | - Ling Tao
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'an710032China
| |
Collapse
|
213
|
Razi S, Molavi Z, Mirmotalebisohi SA, Niknam Z, Sameni M, Niazi V, Adibi A, Yazdani M, Ranjbar MM, Zali H. Mesenchymal Stem Cells in the Treatment of New Coronavirus Pandemic: A Novel Promising Therapeutic Approach. Adv Pharm Bull 2022; 12:206-216. [PMID: 35620342 PMCID: PMC9106958 DOI: 10.34172/apb.2022.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/12/2021] [Accepted: 03/25/2021] [Indexed: 11/09/2022] Open
Abstract
After severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) outbreaks, coronavirus disease 2019 (COVID-19) is the third coronavirus epidemic that soon turned into a pandemic. This virus causes acute respiratory syndrome in infected people. The mortality rate of SARS-CoV-2 infection will probably rise unless efficient treatments or vaccines are developed. The global funding and medical communities have started performing more than five hundred clinical examinations on a broad spectrum of repurposed drugs to acquire effective treatments. Besides, other novel treatment approaches have also recently emerged, including cellular host-directed therapies. They counteract the unwanted responses of the host immune system that led to the severe pathogenesis of SARS-CoV-2. This brief review focuses on mesenchymal stem cell (MSC) principles in treating the COVID-19. The US clinical trials database and the world health organization database for clinical trials have reported 82 clinical trials (altogether) exploring the effects of MSCs in COVID-19 treatment. MSCs also had better be tried for treating other pathogens worldwide. MSC treatment may have the potential to end the high mortality rate of COVID-19. Besides, it also limits the long-term inability of survivors.
Collapse
Affiliation(s)
- Sara Razi
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Molavi
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Seyed Amir Mirmotalebisohi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Niknam
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Marzieh Sameni
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirjafar Adibi
- Departments of Orthopedics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Yazdani
- Institute of Biochemistry and Biophysics, Tehran University, Tehran, Iran
| | - Mohammad Mehdi Ranjbar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
214
|
Mesenchymal Stem Cell-Derived Extracellular Vesicles: Immunomodulatory Effects and Potential Applications in Intervertebral Disc Degeneration. Stem Cells Int 2022; 2022:7538025. [PMID: 35222648 PMCID: PMC8881131 DOI: 10.1155/2022/7538025] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 11/25/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Intervertebral disc (IVD) degenerative disease is a common health problem worldwide. Administration of mesenchymal stem cells (MSCs) in intervertebral disc degeneration (IVDD) has been widely explored in recent years. However, transplantation of MSCs is restricted by several factors. Currently, paracrine signaling is one of the main mechanisms by which MSCs play a therapeutic role in disc regeneration. Extracellular vehicles (EVs) are the main paracrine products of MSCs. They show great potential as an effective alternative to MSCs and play immunomodulation roles such as anti-inflammatory effects, antioxidative stress, antiapoptosis, and antiextracellular matrix (ECM) degradation during treatment of IVDD. This review focuses on the immunomodulatory effect of MSC EVs and their potential applications.
Collapse
|
215
|
Liang Q, Li Q, Ren B, Yin ZQ. Intravenous infusion of small umbilical cord mesenchymal stem cells could enhance safety and delay retinal degeneration in RCS rats. BMC Ophthalmol 2022; 22:67. [PMID: 35144581 PMCID: PMC8832832 DOI: 10.1186/s12886-021-02171-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
Background Human umbilical cord mesenchymal stem cells (UCMSCs) transplantation is a promising therapy for the treatment of retinitis pigmentosa (RP). However, intravenously infused cells may be blocked in the lung, increasing the risk of vascular obstruction, which needs to be optimized to further improve safety and efficacy. Methods We derived small UCMSCs (S-UCMSCs) from filtering UCMSCs with a 10-μm filter, and compared with UCMSCs by flow cytometry, directional differentiation culture and transcriptome sequencing. Then the S-UCMSCs and UCMSCs were intravenously infused in the Royal College Surgeons (RCS) rats to evaluate the safety and the efficacy. Results The diameter of S-UCMSCs ranged from 5.568 to 17.231 μm, with an average diameter of 8.636 ± 2.256 μm, which was significantly smaller than that of UCMSCs. Flow cytometry, immunofluorescence and transcriptome sequencing demonstrated that the S-UCMSCs and UCMSCs were the same kind of MSCs, and the S-UCMSCs were more proliferative. After the S-UCMSCs and UCMSCs were intravenously infused into the Royal College of Surgeons (RCS) rats at a dose of 1 × 106 cells/rat, the S-UCMSCs blocked in the lungs were significantly fewer and disappeared more quickly than UCMSCs. The b wave of the flash electroretinogram was improved at 7 d, and the retinal outer nuclear layer thickness was thicker at 7 d and 14 d. The expression level of inflammation was inhibited, and the expression level of neurotrophic factors was upregulated in the retina and serum after transplantation. Conclusions S-UCMSCs intravenous infusion was safer than UCMSCs and could delay retinal degeneration and protect visual function in RCS rats, which may be a preferable therapeutic approach for RP. Supplementary Information The online version contains supplementary material available at 10.1186/s12886-021-02171-3.
Collapse
Affiliation(s)
- Qingling Liang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration, Chongqing, 400038, China.
| | - Qiyou Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration, Chongqing, 400038, China
| | - Bangqi Ren
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration, Chongqing, 400038, China
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Amy Medical University), Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration, Chongqing, 400038, China.
| |
Collapse
|
216
|
Floriano JF, Emanueli C, Vega S, Barbosa AMP, Oliveira RGD, Floriano EAF, Graeff CFDO, Abbade JF, Herculano RD, Sobrevia L, Rudge MVC. Pro-angiogenic approach for skeletal muscle regeneration. Biochim Biophys Acta Gen Subj 2022; 1866:130059. [PMID: 34793875 DOI: 10.1016/j.bbagen.2021.130059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
The angiogenesis process is a phenomenon in which numerous molecules participate in the stimulation of the new vessels' formation from pre-existing vessels. Angiogenesis is a crucial step in tissue regeneration and recovery of organ and tissue function. Muscle diseases affect millions of people worldwide overcome the ability of skeletal muscle to self-repair. Pro-angiogenic therapies are key in skeletal muscle regeneration where both myogenesis and angiogenesis occur. These therapies have been based on mesenchymal stem cells (MSCs), exosomes, microRNAs (miRs) and delivery of biological factors. The use of different calls of biomaterials is another approach, including ceramics, composites, and polymers. Natural polymers are use due its bioactivity and biocompatibility in addition to its use as scaffolds and in drug delivery systems. One of these polymers is the natural rubber latex (NRL) which is biocompatible, bioactive, versatile, low-costing, and capable of promoting tissue regeneration and angiogenesis. In this review, the advances in the field of pro-angiogenic therapies are discussed.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sofia Vega
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | | | | | | | | | - Joelcio Francisco Abbade
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil
| | | | - Luis Sobrevia
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland, Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD, 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands.
| | | |
Collapse
|
217
|
Abstract
Mesenchymal stem cells (MSCs) exhibit regenerative and reparative properties. However, most MSC-related studies remain to be translated for regular clinical usage, partly due to challenges in pre-transplantation cell labelling and post-transplantation cell tracking. Amidst this, there are growing concerns over the toxicity of commonly used gadolinium-based contrast agents that mediate in-vivo cell detection via MRI. This urges to search for equally effective but less toxic alternatives that would facilitate and enhance MSC detection post-administration and provide therapeutic benefits in-vivo. MSCs labelled with iron oxide nanoparticles (IONPs) have shown promising results in-vitro and in-vivo. Thus, it would be useful to revisit these studies before inventing new labelling approaches. Aiming to inform regenerative medicine and augment clinical applications of IONP-labelled MSCs, this review collates and critically evaluates the utility of IONPs in enhancing MSC detection and therapeutics. It explains the rationale, principle, and advantages of labelling MSCs with IONPs, and describes IONP-induced intracellular alterations and consequent cellular manifestations. By exemplifying clinical pathologies, it examines contextual in-vitro, animal, and clinical studies that used IONP-labelled bone marrow-, umbilical cord-, adipose tissue- and dental pulp-derived MSCs. It compiles and discusses studies involving MSC-labelling of IONPs in combinations with carbohydrates (Venofer, ferumoxytol, dextran, glucosamine), non-carbohydrate polymers [poly(L-lysine), poly(lactide-co-glycolide), poly(L-lactide), polydopamine], elements (ruthenium, selenium, gold, zinc), compounds/stains (silica, polyethylene glycol, fluorophore, rhodamine B, DAPI, Prussian blue), DNA, Fibroblast growth Factor-2 and the drug doxorubicin. Furthermore, IONP-labelling of MSC exosomes is reviewed. Also, limitations of IONP-labelling are addressed and methods of tackling those challenges are suggested.
Collapse
|
218
|
Zhang C, Han X, Liu J, Chen L, Lei Y, Chen K, Si J, Wang TY, Zhou H, Zhao X, Zhang X, An Y, Li Y, Wang QF. Single-cell Transcriptomic Analysis Reveals the Cellular Heterogeneity of Mesenchymal Stem Cells. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:70-86. [PMID: 35123072 PMCID: PMC9510874 DOI: 10.1016/j.gpb.2022.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 01/04/2022] [Accepted: 01/27/2022] [Indexed: 01/04/2023]
Abstract
Ex vivo-expanded mesenchymal stem cells (MSCs) have been demonstrated to be a heterogeneous mixture of cells exhibiting varying proliferative, multipotential, and immunomodulatory capacities. However, the exact characteristics of MSCs remain largely unknown. By single-cell RNA sequencing of 61,296 MSCs derived from bone marrow and Wharton’s jelly, we revealed five distinct subpopulations. The developmental trajectory of these five MSC subpopulations was mapped, revealing a differentiation path from stem-like active proliferative cells (APCs) to multipotent progenitor cells, followed by branching into two paths: 1) unipotent preadipocytes or 2) bipotent prechondro-osteoblasts that were subsequently differentiated into unipotent prechondrocytes. The stem-like APCs, expressing the perivascular mesodermal progenitor markers CSPG4/MCAM/NES, uniquely exhibited strong proliferation and stemness signatures. Remarkably, the prechondrocyte subpopulation specifically expressed immunomodulatory genes and was able to suppress activated CD3+ T cell proliferation in vitro, supporting the role of this population in immunoregulation. In summary, our analysis mapped the heterogeneous subpopulations of MSCs and identified two subpopulations with potential functions in self-renewal and immunoregulation. Our findings advance the definition of MSCs by identifying the specific functions of their heterogeneous cellular composition, allowing for more specific and effective MSC application through the purification of their functional subpopulations.
Collapse
Affiliation(s)
- Chen Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China; Qingdao Key Lab of Mitochondrial Medicine, Qingdao 266035, China
| | - Xueshuai Han
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingkun Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Lei
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kunying Chen
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia Si
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tian-Yi Wang
- International Department, Liangxiang Campus, Beijing University of Chinese Medicine, Beijing 102401, China
| | - Hui Zhou
- Yihua Biotechnology Co., Ltd., Beijing 100041, China
| | - Xiaoyun Zhao
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China; Qingdao Key Lab of Mitochondrial Medicine, Qingdao 266035, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China
| | - Yihua An
- Department of Functional Neurosurgery, Third Medical Center, General Hospital of Chinese PLA, Beijing 100039, China
| | - Yueying Li
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qian-Fei Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
219
|
Tang XL, Wysoczynski M, Gumpert AM, Li Y, Wu WJ, Li H, Stowers H, Bolli R. Effect of intravenous cell therapy in rats with old myocardial infarction. Mol Cell Biochem 2022; 477:431-444. [PMID: 34783963 PMCID: PMC8896398 DOI: 10.1007/s11010-021-04283-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 10/21/2021] [Indexed: 10/19/2022]
Abstract
Mounting evidence shows that cell therapy provides therapeutic benefits in experimental and clinical settings of chronic heart failure. However, direct cardiac delivery of cells via transendocardial injection is logistically complex, expensive, entails risks, and is not amenable to multiple dosing. Intravenous administration would be a more convenient and clinically applicable route for cell therapy. Thus, we determined whether intravenous infusion of three widely used cell types improves left ventricular (LV) function and structure and compared their efficacy. Rats with a 30-day-old myocardial infarction (MI) received intravenous infusion of vehicle (PBS) or 1 of 3 types of cells: bone marrow mesenchymal stromal cells (MSCs), cardiac mesenchymal cells (CMCs), and c-kit-positive cardiac cells (CPCs), at a dose of 12 × 106 cells. Rats were followed for 35 days after treatment to determine LV functional status by serial echocardiography and hemodynamic studies. Blood samples were collected for Hemavet analysis to determine inflammatory cell profile. LV ejection fraction (EF) dropped ≥ 20 points in all hearts at 30 days after MI and deteriorated further at 35-day follow-up in the vehicle-treated group. In contrast, deterioration of EF was halted in rats that received MSCs and attenuated in those that received CMCs or CPCs. None of the 3 types of cells significantly altered scar size, myocardial content of collagen or CD45-positive cells, or Hemavet profile. This study demonstrates that a single intravenous administration of 3 types of cells in rats with chronic ischemic cardiomyopathy is effective in attenuating the progressive deterioration in LV function. The extent of LV functional improvement was greatest with CPCs, intermediate with CMCs, and least with MSCs.
Collapse
Affiliation(s)
- Xian-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Anna M Gumpert
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Yan Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Wen-Jian Wu
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Hong Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Heather Stowers
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA.
| |
Collapse
|
220
|
Tu C, Wang Z, Xiang E, Zhang Q, Zhang Y, Wu P, Li C, Wu D. Human Umbilical Cord Mesenchymal Stem Cells Promote Macrophage PD-L1 Expression and Attenuate Acute Lung Injury in Mice. Curr Stem Cell Res Ther 2022; 17:564-575. [PMID: 35086457 DOI: 10.2174/1574888x17666220127110332] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) remains a serious clinical problem but has no approved pharmacotherapy. Mesenchymal stem cells (MSCs) represent an attractive therapeutic tool for tissue damage and inflammation owing to their unique immunomodulatory properties. The present study aims to explore the therapeutic effect and underlying mechanisms of human umbilical cord MSCs (UC-MSCs) in ALI mice. OBJECTIVE In this study, we identify a novel mechanism for human umbilical cord-derived MSCs (UC-MSCs)-mediated immunomodulation through PGE2-dependent reprogramming of host macrophages to promote their PD-L1 expression. Our study suggests that UC-MSCs or primed-UC-MSCs offer new therapeutic approaches for lung inflammatory diseases. METHODS Lipopolysaccharide (LPS)-induced ALI mice were injected with 5×105 UC-MSCs via the tail vein after 4 hours of LPS exposure. After 24 hours of UC-MSC administration, the total protein concentration and cell number in the bronchoalveolar lavage fluid (BALF), and cytokine levels in the lung tissue were measured. Lung pathological changes and macrophage infiltration after UC-MSC treatment were analyzed. Moreover, in vitro co-culture experiments were performed to analyze cytokine levels of RAW264.7 cells and Jurkat T cells. RESULTS UC-MSC treatment significantly improved LPS-induced ALI, as indicated by decreased total protein exudation concentration and cell number in BALF, and reduced pathological damage in ALI mice. UC-MSCs could inhibit pro-inflammatory cytokine levels (IL-1β, TNF-α, MCP-1, IL-2, and IFN-γ), whereas enhancing anti-inflammatory cytokine IL-10 expression, as well as reduced macrophage infiltration into the injured lung tissue. Importantly, UC-MSC administration increased programmed cell death protein ligand 1 (PD-L1) expression in the lung macrophages. Mechanistically, UC-MSCs upregulated cyclooxygenase-2 (COX2) expression and prostaglandin E2 (PGE2) secretion in response to LPS stimulation. UC-MSCs reduced the inflammatory cytokine levels in murine macrophage Raw264.7 through the COX2/PGE2 axis. Furthermore, UC-MSC-derived PGE2 enhanced PD-L1 expression in RAW264.7 cells, which in turn promoted programmed cell death protein 1 (PD-1) expression and reduced IL-2 and IFN-γ production in Jurkat T cells. CONCLUSION Our results suggest that UC-MSCs attenuate ALI via PGE2-dependent reprogramming of macrophages to promote their PD-L1 expression.
Collapse
Affiliation(s)
- Chengshu Tu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan, China
| | | | - E Xiang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Quan Zhang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
| | - Yaqi Zhang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Ping Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan, China
| | - Changyong Li
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Dongcheng Wu
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Guangzhou Hamilton Biotechnology-Co., Ltd, Guangzhou, China
| |
Collapse
|
221
|
Anggelia MR, Cheng HY, Lai PC, Hsieh YH, Lin CH, Lin CH. Cell Therapy in Vascularized Composite Allotransplantation. Biomed J 2022; 45:454-464. [PMID: 35042019 PMCID: PMC9422067 DOI: 10.1016/j.bj.2022.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/02/2021] [Accepted: 01/10/2022] [Indexed: 11/18/2022] Open
Abstract
Allograft rejection is one of the obstacles in achieving a successful vascularized composite allotransplantation (VCA). Treatments of graft rejection with lifelong immunosuppression (IS) subject the recipients to a lifelong risk of cancer development and opportunistic infections. Cell therapy has recently emerged as a promising strategy to modulate the immune system, minimize immunosuppressant drug dosages, and induce allograft tolerance. In this review, the recent works regarding the use of cell therapy to improve allograft outcomes are discussed. The current data supports the safety of cell therapy. The suitable type of cell therapy in allotransplantation is clinically dependent. Bone marrow cell therapy is more suitable for the induction phase, while other cell therapies are more feasible in either the induction or maintenance phase, or for salvage of allograft rejection. Immune cell therapy focuses on modulating the immune response, whereas stem cells may have an additional role in promoting structural regenerations, such as nerve regeneration. Source, frequency, dosage, and route of cell therapy delivery are also dependent on the specific need in the clinical setting.
Collapse
Affiliation(s)
- Madonna Rica Anggelia
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ping-Chin Lai
- The Kidney Institute and Division of Nephrology, China Medical University Hospital, Taichung, Taiwan
| | - Yun-Huan Hsieh
- Department of Plastic and Reconstructive Surgery, Epworth Eastern Hospital, Victoria, Australia
| | - Chih-Hung Lin
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
222
|
Costa A, Quarto R, Bollini S. Small Extracellular Vesicles from Human Amniotic Fluid Samples as Promising Theranostics. Int J Mol Sci 2022; 23:ijms23020590. [PMID: 35054775 PMCID: PMC8775841 DOI: 10.3390/ijms23020590] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 02/05/2023] Open
Abstract
Since the first evidence that stem cells can provide pro-resolving effects via paracrine secretion of soluble factors, growing interest has been addressed to define the most ideal cell source for clinical translation. Leftover or clinical waste samples of human amniotic fluid obtained following prenatal screening, clinical intervention, or during scheduled caesarean section (C-section) delivery at term have been recently considered an appealing source of mesenchymal progenitors with peculiar regenerative capacity. Human amniotic fluid stem cells (hAFSC) have been demonstrated to support tissue recovery in several preclinical models of disease by exerting paracrine proliferative, anti-inflammatory and regenerative influence. Small extracellular vesicles (EVs) concentrated from the hAFSC secretome (the total soluble trophic factors secreted in the cell-conditioned medium, hAFSC-CM) recapitulate most of the beneficial cell effects. Independent studies in preclinical models of either adult disorders or severe diseases in newborns have suggested a regenerative role of hAFSC-EVs. EVs can be eventually concentrated from amniotic fluid (hAF) to offer useful prenatal information, as recently suggested. In this review, we focus on the most significant aspects of EVs obtained from either hAFSC and hAF and consider the current challenges for their clinical translation, including isolation, characterization and quantification methods.
Collapse
Affiliation(s)
- Ambra Costa
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
| | - Rodolfo Quarto
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Sveva Bollini
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
- Correspondence: ; Tel.: +39-010-555-8394
| |
Collapse
|
223
|
Zheng D, Wang X, Zhang Z, Li E, Yeung C, Borkar R, Qin G, Wu Y, Xu RH. Engineering of human mesenchymal stem cells resistant to multiple natural killer subtypes. Int J Biol Sci 2022; 18:426-440. [PMID: 34975342 PMCID: PMC8692142 DOI: 10.7150/ijbs.64640] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/25/2021] [Indexed: 12/04/2022] Open
Abstract
Mesenchymal stem cells (MSCs) as a therapeutic promise are often quickly cleared by innate immune cells of the host including natural killer (NK) cells. Efforts have been made to generate immune-escaping human embryonic stem cells (hESCs) where T cell immunity is evaded by defecting β-2-microglobulin (B2M), a common unit for human leukocyte antigen (HLA) class I, and NK cells are inhibited via ectopic expression of HLA-E or -G. However, NK subtypes vary among recipients and even at different pathologic statuses. It is necessary to dissect and optimize the efficacy of the immune-escaping cells against NK subtypes. Here, we first generated B2M knockout hESCs and differentiated them to MSCs (EMSCs) and found that NK resistance occurred with B2M-/- EMSCs expressing HLA-E and -G only when they were transduced via an inducible lentiviral system in a dose-dependent manner but not when they were inserted into a safe harbor. HLA-E and -G expressed at high levels together in transduced EMSCs inhibited three major NK subtypes, including NKG2A+/LILRB1+, NKG2A+/LILRB1-, and NKG2A-/LILRB1+, which was further potentiated by IFN-γ priming. Thus, this study engineers MSCs with resistance to multiple NK subtypes and underscores that dosage matters when a transgene is used to confer a novel effect to host cells, especially for therapeutic cells to evade immune rejection.
Collapse
Affiliation(s)
- Dejin Zheng
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Xiaoyan Wang
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Zhenwu Zhang
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Enqin Li
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Cheungkwan Yeung
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Roma Borkar
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Guihui Qin
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| | - Yaojiong Wu
- The Shenzhen Key Laboratory of Health Sciences and Technology, International Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Ren-He Xu
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau, China
| |
Collapse
|
224
|
Kim JC, Min K, Tae G. The effect of the surface coating of human adipose-derived stem cells by various GAGs on the biodistribution of them upon intravenous administration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 135:112671. [DOI: 10.1016/j.msec.2022.112671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/22/2021] [Accepted: 01/16/2022] [Indexed: 12/17/2022]
|
225
|
Malhotra P, Shukla M, Meena P, Kakkar A, Khatri N, Nagar RK, Kumar M, Saraswat SK, Shrivastava S, Datt R, Pandey S. Mesenchymal stem cells are prospective novel off-the-shelf wound management tools. Drug Deliv Transl Res 2022; 12:79-104. [PMID: 33580481 DOI: 10.1007/s13346-021-00925-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Chronic/non-healing cutaneous wounds pose a debilitating burden on patients and healthcare system. Presently, treatment modalities are rapidly shifting pace from conventional methods to advanced wound care involving cell-based therapies. Mesenchymal stem cells (MSCs) have come across as a prospective option due to its pleiotropic functions viz. non-immunogenicity, multipotency, multi-lineage plasticity and secretion of growth factors, cytokines, microRNAs (miRNA), exosomes, and microvesicles as part of their secretome for assisting wound healing. We outline the therapeutic role played by MSCs and its secretome in suppressing tissue inflammation, causing immunomodulation, aiding angiogenesis and assisting in scar-free wound healing. We further assess the mechanism of action by which MSCs contribute in manifesting tissue repair. The review flows ahead in exploring factors that influence healing behavior including effect of multiple donor sites, donor age and health status, tissue microenvironment, and in vitro expansion capability. Moving ahead, we overview the advancements achieved in extending the lifespan of cells upon implantation, influence of genetic modifications aimed at altering MSC cargo, and evaluating bioengineered matrix-assisted delivery methods toward faster healing in preclinical and clinical models. We also contribute toward highlighting the challenges faced in commercializing cell-based therapies as standard of care treatment regimens. Finally, we strongly advocate and highlight its application as a futuristic technology for revolutionizing tissue regeneration.
Collapse
Affiliation(s)
- Poonam Malhotra
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Manish Shukla
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Poonam Meena
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Anupama Kakkar
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Nitin Khatri
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Rakesh K Nagar
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Mukesh Kumar
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Sumit K Saraswat
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Supriya Shrivastava
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Rajan Datt
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India
| | - Siddharth Pandey
- Department of Life Sciences, Datt Mediproducts Private Ltd, Roz Ka Meo Industrial Area, Distt. Mewat, Nuh, 122103, Haryana, India.
| |
Collapse
|
226
|
Zhao L, Li Y, Kou X, Chen B, Cao J, Li J, Zhang J, Wang H, Zhao J, Shi S. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:778-789. [PMID: 35608372 PMCID: PMC9299510 DOI: 10.1093/stcltm/szac028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/01/2022] [Indexed: 11/14/2022] Open
Abstract
Mesenchymal stem cell-based therapy has emerged as a great potential approach to treat individuals with autism spectrum disorders (ASD), a group of developmental disabilities characterized by impairments in social interaction and communication. Stem cells from human exfoliated deciduous teeth (SHED), holding earlier developing characteristics, have immune-modulatory and anti-inflammatory properties. To investigate whether SHED transplantation can rescue autistic-like symptoms in SHANK3 mutant beagle dogs, 12 SHANK3 mutant beagle dogs were randomly assigned into 2 groups according to their behavior evaluated by social interaction tests. Six mutant dogs received 6 intravenous infusions of SHED and were followed up for 3 months by testing social interaction and inflammatory cytokine levels. We found that infusion of SHED significantly improved impaired social novel preference of SHANK3 mutant beagle dogs at 1- and 3-month follow-ups. Social intimacies (following, sniffing, and licking) between mutant beagle dogs and human experimenters were partly improved. Stressed tail posture, indicating social stress, was also significantly alleviated. In addition, we showed that the levels of serum interferon-γ and interleukin-10 were notably increased and decreased, respectively, in SHANK3 mutant beagle dogs. Infusion of SHED was able to rescue altered interferon-γ and interleukin-10 levels. We failed to observe any serious adverse events after infusion of SHED. In summary, SHED transplantation may be a safe and effective therapy for ASD. The correction in the levels of serum interferon-γ and interleukin-10 may serve as an index to predict autistic severity and therapeutic outcomes.
Collapse
Affiliation(s)
- Lu Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yuan Li
- Beijing Sinogene Biotechnology Co. Ltd, Changping District, Beijing, People’s Republic of China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Benchi Chen
- Beijing Sinogene Biotechnology Co. Ltd, Changping District, Beijing, People’s Republic of China
| | - Jing Cao
- CAR-T (Shanghai) Biotechnology Co. Ltd, Yangpu District, Shanghai, People’s Republic of China
| | - Jun Li
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Jianqi Zhang
- Beijing Sinogene Biotechnology Co. Ltd, Changping District, Beijing, People’s Republic of China
| | - Heng Wang
- Beijing Sinogene Biotechnology Co. Ltd, Changping District, Beijing, People’s Republic of China
| | - Jianping Zhao
- Beijing Sinogene Biotechnology Co. Ltd, Changping District, Beijing, People’s Republic of China
| | - Songtao Shi
- Corresponding author: Songtao Shi, South China Center of Craniofacial Stem Cell Research, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, 74 Zhongshan 2 Road, Guangzhou, Guangdong 510080, People’s Republic of China. Tel: +86 020 83811509; Fax: +86 020 83811509;
| |
Collapse
|
227
|
Advances in Targeting ACE2 for Developing COVID-19 Therapeutics. Ann Biomed Eng 2022; 50:1734-1749. [PMID: 36261668 PMCID: PMC9581451 DOI: 10.1007/s10439-022-03094-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 01/01/2023]
Abstract
Since the onset of the coronavirus pandemic in December 2019, the SARS-CoV-2 virus has accounted for over 6.3 million lives resulting in the demand to develop novel therapeutic approaches to target and treat SARS-CoV-2. Improved understanding of viral entry and infection mechanisms has led to identifying different target receptors to mitigate infection in the host. Researchers have been working on identifying and targeting potential therapeutic target receptors utilizing different candidate drugs. Angiotensin-converting enzyme-2 (ACE2) has been known to perform critical functions in maintaining healthy cardiorespiratory function. However, ACE2 also functions as the binding site for the spike protein of SARS-CoV-2, allowing the virus to enter the cells and ensue infection. Therefore, drugs targeting ACE2 receptors can be considered as therapeutic candidates. Strategies targeting the level of ACE2 expression have been investigated and compared to other potential therapeutic targets, such as TMPRSS2, RdRp, and DPP4. This mini review discusses the key therapeutic approaches that target the ACE2 receptor, which is critical to the cellular entry and propagation of the novel SARS-CoV-2. In addition, we summarize the main advantages of ACE2 targeting against alternative approaches for the treatment of COVID-19.
Collapse
|
228
|
Inhibition of aberrant tissue remodelling by mesenchymal stromal cells singly coated with soft gels presenting defined chemomechanical cues. Nat Biomed Eng 2022; 6:54-66. [PMID: 34083763 PMCID: PMC8908879 DOI: 10.1038/s41551-021-00740-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
The precise understanding and control of microenvironmental cues could be used to optimize the efficacy of cell therapeutics. Here, we show that mesenchymal stromal cells (MSCs) singly coated with a soft conformal gel presenting defined chemomechanical cues promote matrix remodelling by secreting soluble interstitial collagenases in response to the presence of tumour necrosis factor alpha (TNF-α). In mice with fibrotic lung injury, treatment with the coated MSCs maintained normal collagen levels, fibre density and microelasticity in lung tissue, and the continuous presentation of recombinant TNF-α in the gel facilitated the reversal of aberrant tissue remodelling by the cells when inflammation subsided in the host. Gel coatings with predefined chemomechanical cues could be used to tailor cells with specific mechanisms of action for desired therapeutic outcomes.
Collapse
|
229
|
Belkozhayev AM, Al-Yozbaki M, George A, Niyazova RY, Sharipov KO, Byrne LJ, Wilson CM. Extracellular Vesicles, Stem Cells and the Role of miRNAs in Neurodegeneration. Curr Neuropharmacol 2022; 20:1450-1478. [PMID: 34414870 PMCID: PMC9881087 DOI: 10.2174/1570159x19666210817150141] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/16/2021] [Accepted: 06/14/2021] [Indexed: 11/22/2022] Open
Abstract
There are different modalities of intercellular communication governed by cellular homeostasis. In this review, we will explore one of these forms of communication called extracellular vesicles (EVs). These vesicles are released by all cells in the body and are heterogeneous in nature. The primary function of EVs is to share information through their cargo consisting of proteins, lipids and nucleic acids (mRNA, miRNA, dsDNA etc.) with other cells, which have a direct consequence on their microenvironment. We will focus on the role of EVs of mesenchymal stem cells (MSCs) in the nervous system and how these participate in intercellular communication to maintain physiological function and provide neuroprotection. However, deregulation of this same communication system could play a role in several neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, multiple sclerosis, prion disease and Huntington's disease. The release of EVs from a cell provides crucial information to what is happening inside the cell and thus could be used in diagnostics and therapy. We will discuss and explore new avenues for the clinical applications of using engineered MSC-EVs and their potential therapeutic benefit in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Ayaz M. Belkozhayev
- Al-Farabi Kazakh National University, Faculty of Biology and Biotechnology, Almaty, Republic of Kazakhstan
- Structural and Functional Genomics Laboratory of M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Republic of Kazakhstan
| | - Minnatallah Al-Yozbaki
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Alex George
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
- Jubilee Centre for Medical Research, Jubilee Mission Medical College & Research Institute, Thrissur, Kerala, India
| | - Raigul Ye Niyazova
- Al-Farabi Kazakh National University, Faculty of Biology and Biotechnology, Almaty, Republic of Kazakhstan
| | - Kamalidin O. Sharipov
- Structural and Functional Genomics Laboratory of M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Republic of Kazakhstan
| | - Lee J. Byrne
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Cornelia M. Wilson
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| |
Collapse
|
230
|
Romecín PA, Vinyoles M, López-Millán B, de la Guardia RD, Atucha NM, Querol S, Bueno C, Benitez R, Gonzalez-Rey E, Delgado M, Menéndez P. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:88-96. [PMID: 35641173 PMCID: PMC8895490 DOI: 10.1093/stcltm/szab007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
Mesenchymal stromal stem/cells (MSC) therapies are clinically used in a wide range of disorders based on their robust HLA-independent immunosuppressive and anti-inflammatory properties. However, the mechanisms underlying MSC therapeutic activity remain elusive as demonstrated by the unpredictable therapeutic efficacy of MSC infusions reported in multiple clinical trials. A seminal recent study showed that infused MSCs are actively induced to undergo apoptosis by recipient cytotoxic T cells, a mechanism that triggers in vivo recipient-induced immunomodulation by such apoptotic MSCs, and the need for such recipient cytotoxic cell activity could be replaced by the administration of ex vivo-generated apoptotic MSCs. Moreover, the use of MSC-derived extracellular vesicles (MSC-EVs) is being actively explored as a cell-free therapeutic alternative over the parental MSCs. We hypothesized that the introduction of a “suicide gene” switch into MSCs may offer on-demand in vivo apoptosis of transplanted MSCs. Here, we prompted to investigate the utility of the iCasp9/AP1903 suicide gene system in inducing apoptosis of MSCs. iCasp9/AP1903-induced apoptotic MSCs (MSCiCasp9+) were tested in vitro and in in vivo models of acute colitis. Our data show a very similar and robust immunosuppressive and anti-inflammatory properties of both “parental” alive MSCGFP+ cells and apoptotic MSCiCasp9+ cells in vitro and in vivo regardless of whether apoptosis was induced in vivo or in vitro before administering MSCiCasp9+ lysates. This development of an efficient iCasp9 switch may potentiate the safety of MSC-based therapies in the case of an adverse event and, will also circumvent current logistic technical limitations and biological uncertainties associated to MSC-EVs.
Collapse
Affiliation(s)
- Paola Alejandra Romecín
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- Paola Alejandra Romecin, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Carrer Casanova 143, 4º floor, 08036, Barcelona, Spain. Tel: (+34) 93 5572810;
| | | | - Belén López-Millán
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- GENYO, Centro Pfizer-Universidad de Granada-Junta de Andalucía de Genómica e Investigación Oncológica, Granada, Spain
| | - Rafael Diaz de la Guardia
- GENYO, Centro Pfizer-Universidad de Granada-Junta de Andalucía de Genómica e Investigación Oncológica, Granada, Spain
| | - Noemi M Atucha
- Departamento de Fisiologia Humana, Facultad de Medicina, Murcia, Spain
| | - Sergi Querol
- RICORS-TERAV, ISCIII, Madrid, Spain
- Banc de Sang i Teixits, Barcelona, Spain
| | - Clara Bueno
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- CIBERONC, ISCIII, Barcelona, Spain
| | - Raquel Benitez
- Instituto de Parasitologia y Biomedicina López-Neyra (IPBLN-CSIC), Armilla, Granada, Spain
| | - Elena Gonzalez-Rey
- Instituto de Parasitologia y Biomedicina López-Neyra (IPBLN-CSIC), Armilla, Granada, Spain
| | - Mario Delgado
- Instituto de Parasitologia y Biomedicina López-Neyra (IPBLN-CSIC), Armilla, Granada, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-TERAV, ISCIII, Madrid, Spain
- CIBERONC, ISCIII, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Corresponding author: Pablo Menéndez, Josep Carreras Leukemia Research Institute, School of Medicine, University of Barcelona, Carrer Casanova 143, 4º floor, 08036, Barcelona, Spain. Tel: (+34) 93 5572810;
| |
Collapse
|
231
|
Taechangam N, Kol A, Arzi B, Borjesson DL. Multipotent Stromal Cells and Viral Interaction: Current Implications for Therapy. Stem Cell Rev Rep 2022; 18:214-227. [PMID: 34347271 PMCID: PMC8335712 DOI: 10.1007/s12015-021-10224-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2021] [Indexed: 12/29/2022]
Abstract
Multipotent stromal cells (MSCs) are widely utilized in therapy for their immunomodulatory properties, but their usage in infectious viral diseases is less explored. This review aimed to collate the current novel use of MSCs in virus-associated conditions, including MSC's susceptibility to virus infection, antiviral properties of MSCs and their effects on cell-based immune response and implementation of MSC therapy in animal models and human clinical trials of viral diseases. Recent discoveries shed lights on MSC's capability in suppressing viral replication and augmenting clearance through enhancement of antiviral immunity. MSC therapy may maintain a crucial balance between aiding pathogen clearance and suppressing hyperactive immune response.
Collapse
Affiliation(s)
- Nopmanee Taechangam
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Amir Kol
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Dori L. Borjesson
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| |
Collapse
|
232
|
Bagno LL, Salerno AG, Balkan W, Hare JM. Mechanism of Action of Mesenchymal Stem Cells (MSCs): impact of delivery method. Expert Opin Biol Ther 2021; 22:449-463. [PMID: 34882517 DOI: 10.1080/14712598.2022.2016695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs; AKA mesenchymal stem cells) stimulate healing and reduce inflammation. Promising therapeutic responses are seen in many late-phase clinical trials, but others have not satisfied their primary endpoints, making translation of MSCs into clinical practice difficult. These inconsistencies may be related to the route of MSC delivery, lack of product optimization, or varying background therapies received in clinical trials over time. AREAS COVERED Here we discuss the different routes of MSC delivery, highlighting the proposed mechanism(s) of therapeutic action as well as potential safety concerns. PubMed search criteria used: MSC plus: local administration; routes of administration; delivery methods; mechanism of action; therapy in different diseases. EXPERT OPINION Direct injection of MSCs using a controlled local delivery approach appears to have benefits in certain disease states, but further studies are required to make definitive conclusions regarding the superiority of one delivery method over another.
Collapse
Affiliation(s)
- Luiza L Bagno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alessandro G Salerno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami
| |
Collapse
|
233
|
Pourgholaminejad A, Pahlavanneshan S, Basiri M. COVID-19 immunopathology with emphasis on Th17 response and cell-based immunomodulation therapy: Potential targets and challenges. Scand J Immunol 2021; 95:e13131. [PMID: 34936112 DOI: 10.1111/sji.13131] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/04/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022]
Abstract
The role of the immune system against coronavirus disease 2019 (COVID-19) is unknown in many aspects, and the protective or pathologic mechanisms of the immune response are poorly understood. Pro-inflammatory cytokine release and a consequent cytokine storm can lead to acute respiratory distress syndrome (ARDS) and result in multi-organ failure. There are many T cell subsets during anti-viral immunity. The Th17-associated response, as a pro-inflammatory pathway, and its consequent outcomes in many autoimmune disorders play a fundamental role in progression of systemic hyper-inflammation during COVID-19. Therapeutic strategies based on immunomodulation therapy could be helpful for targeting hyper-inflammatory immune responses in COVID-19, especially Th17-related inflammation and hyper-cytokinemia. Cell-based immunotherapeutic approaches including mesenchymal stem cells (MSCs), tolerogenic dendritic cells (tolDCs) and regulatory T cells (Tregs) seem to be promising strategies as orchestrators of the immune response against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review, we highlight Th17-related immunopathology of SARS-CoV-2 infection and discuss cell-based immunomodulatory strategies and their mechanisms for regulation of the hyper-inflammation during COVID-19.
Collapse
Affiliation(s)
- Arash Pourgholaminejad
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Saghar Pahlavanneshan
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
234
|
Liu CW, Su BC, Chen JY. Tilapia Piscidin 4 (TP4) Reprograms M1 Macrophages to M2 Phenotypes in Cell Models of Gardnerella vaginalis-Induced Vaginosis. Front Immunol 2021; 12:773013. [PMID: 34925343 PMCID: PMC8674419 DOI: 10.3389/fimmu.2021.773013] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/15/2021] [Indexed: 01/24/2023] Open
Abstract
Gardnerella vaginalis is associated with bacterial vaginosis (BV). The virulence factors produced by G. vaginalis are known to stimulate vaginal mucosal immune response, which is largely driven by activated macrophages. While Tilapia piscidin 4 (TP4), an antimicrobial peptide isolated from Nile tilapia, is known to display a broad range of antibacterial functions, it is unclear whether TP4 can affect macrophage polarization in the context of BV. In this study, we used the culture supernatants from G. vaginalis to stimulate differentiation of THP-1 and RAW264.7 cells to an M1 phenotype. The treatment activated the NF-κB/STAT1 signaling pathway, induced reactive nitrogen and oxygen species, and upregulated inflammatory mediators. We then treated the induced M1 macrophages directly with a non-toxic dose of TP4 or co-cultured the M1 macrophages with TP4-treated vaginal epithelial VK2 cells. The results showed that TP4 could not only decrease pro-inflammatory mediators in the M1 macrophages, but it also enriched markers of M2 macrophages. Further, we found that direct treatment with TP4 switched M1 macrophages toward a resolving M2c phenotype via the MAPK/ERK pathway and IL-10-STAT3 signaling. Conversely, tissue repair M2a macrophages were induced by TP4-treated VK2 cells; TP4 upregulated TSG-6 in VK2 cells, which subsequently activated STAT6 and M2a-related gene expression in the macrophages. In conclusion, our results imply that TP4 may be able to attenuate the virulence of G. vaginalis by inducing resolving M2c and tissue repair M2a macrophage polarizations, suggesting a novel strategy for BV therapy.
Collapse
Affiliation(s)
- Chia-Wen Liu
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Taiwan
| | - Bor-Chyuan Su
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Jiaushi, Taiwan.,The iEGG and Animal Biotechnology Center, The Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
235
|
Luo DS, Li YQ, Deng ZQ, Liu GH. Progress and prospect of stem cell therapy for diabetic erectile dysfunction. World J Diabetes 2021; 12:2000-2010. [PMID: 35047115 PMCID: PMC8696650 DOI: 10.4239/wjd.v12.i12.2000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/18/2021] [Accepted: 10/31/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetic erectile dysfunction (DED) is a common complication of diabetes mellitus, significantly impairing the quality of life of patients. The conventional clinical treatment still has limitations. Stem cells (SCs), as a type of cells with multidirectional or directional differentiation capability and sustainable self-renewal potential, are widely used in regenerative medicine and tissue engineering. With the continuous update of regenerative medicine theory and the success of animal experiments, SCs as a treatment for male erectile dysfunction, especially DED, have attracted widespread attention because of curable possibility. This review focus on the current progress in the clinical application of SC treatment for DED. Moreover, we summarize the development prospects of SCs in the field of DMED therapy.
Collapse
Affiliation(s)
- Dao-Sheng Luo
- Department of Urology, Dongguan People’s Hospital, Dongguan 523000, Guangdong Province, China
| | - Yan-Qing Li
- Reproductive Centre, Sun Yat-Sen University, The Sixth Affiliated Hospital, Guangzhou 510000, Guangdong Province, China
| | - Zhi-Quan Deng
- Department of Urology, Dongguan People’s Hospital, Dongguan 523000, Guangdong Province, China
| | - Gui-Hua Liu
- Reproductive Centre, Sun Yat-Sen University, The Sixth Affiliated Hospital, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
236
|
Van Hoecke L, Van Cauwenberghe C, Börger V, Bruggeman A, Castelein J, Van Imschoot G, Van Wonterghem E, Dittrich R, Claeys W, Xie J, Giebel B, Vandenbroucke RE. Anti-Inflammatory Mesenchymal Stromal Cell-Derived Extracellular Vesicles Improve Pathology in Niemann-Pick Type C Disease. Biomedicines 2021; 9:1864. [PMID: 34944681 PMCID: PMC8698931 DOI: 10.3390/biomedicines9121864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022] Open
Abstract
Niemann-Pick type C (NPC) disease is a rare neurovisceral lipid storage disease with progressive neurodegeneration, leading to premature death. The disease is caused by loss-of-function mutations either in the NPC1 or NPC2 gene which results in lipid accumulation in the late endosomes and lysosomes. The involved disease mechanisms are still incompletely understood, making the design of a rational treatment very difficult. Since the disease is characterized by peripheral inflammation and neuroinflammation and it is shown that extracellular vesicles (EVs) obtained from mesenchymal stromal cells (MSCs) provide immunomodulatory capacities, we tested the potential of MSC-EV preparations to alter NPC1 disease pathology. Here, we show that the administration of an MSC-EV preparation with in vitro and in vivo confirmed immune modulatory capabilities is able to reduce the inflammatory state of peripheral organs and different brain regions of NPC1-diseased mice almost to normal levels. Moreover, a reduction of foamy cells in different peripheral organs was observed upon MSC-EV treatment of NPC1-/- mice. Lastly, the treatment was able to decrease microgliosis and astrogliosis, typical features of NPC1 patients that lead to neurodegeneration. Altogether, our results reveal the therapeutic potential of MSC-EVs as treatment for the genetic neurovisceral lipid storage disease NPC, thereby counteracting both central and peripheral features.
Collapse
Affiliation(s)
- Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Caroline Van Cauwenberghe
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Verena Börger
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.B.); (R.D.); (B.G.)
| | - Arnout Bruggeman
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Jonas Castelein
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Robin Dittrich
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.B.); (R.D.); (B.G.)
| | - Wouter Claeys
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Liver Research Center Ghent, Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | - Junhua Xie
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany; (V.B.); (R.D.); (B.G.)
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (L.V.H.); (C.V.C.); (A.B.); (J.C.); (G.V.I.); (E.V.W.); (W.C.); (J.X.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
237
|
Chen J, Zhou D, Nie Z, Lu L, Lin Z, Zhou D, Zhang Y, Long X, Fan S, Xu T. A scalable coaxial bioprinting technology for mesenchymal stem cell microfiber fabrication and high extracellular vesicle yield. Biofabrication 2021; 14:015012. [PMID: 34798619 DOI: 10.1088/1758-5090/ac3b90] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/19/2021] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) are promising candidates for regenerative medicine; however, the lack of scalable methods for high quantity EV production limits their application. In addition, signature EV-derived proteins shared in 3D environments and 2D surfaces, remain mostly unknown. Herein, we present a platform combining MSC microfiber culture with ultracentrifugation purification for high EV yield. Within this platform, a high quantity MSC solution (∼3 × 108total cells) is encapsulated in a meter-long hollow hydrogel-microfiber via coaxial bioprinting technology. In this 3D core-shell microfiber environment, MSCs express higher levels of stemness markers (Oct4, Nanog, Sox2) than in 2D culture, and maintain their differentiation capacity. Moreover, this platform enriches particles by ∼1009-fold compared to conventional 2D culture, while preserving their pro-angiogenic properties. Liquid chromatography-mass spectrometry characterization results demonstrate that EVs derived from our platform and conventional 2D culturing have unique protein profiles with 3D-EVs having a greater variety of proteins (1023 vs 605), however, they also share certain proteins (536) and signature MSC-EV proteins (10). This platform, therefore, provides a new tool for EV production using microfibers in one culture dish, thereby reducing space, labor, time, and cost.
Collapse
Affiliation(s)
- Jianwei Chen
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
| | - Duchao Zhou
- East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Zhenguo Nie
- Department of Orthopedics, Fourth Medical Center of PLA general hospital, 100048 Beijing, People's Republic of China
| | - Liang Lu
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, People's Republic of China
| | - Zhidong Lin
- The Second Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, 510006 Guangzhou, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Yi Zhang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
| | - Xiaoyan Long
- East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Siyang Fan
- Heart Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Tao Xu
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| |
Collapse
|
238
|
Kamiyama Y, Naritomi Y, Moriya Y, Yamamoto S, Kitahashi T, Maekawa T, Yahata M, Hanada T, Uchiyama A, Noumaru A, Koga Y, Higuchi T, Ito M, Komatsu H, Miyoshi S, Kimura S, Umeda N, Fujita E, Tanaka N, Sugita T, Takayama S, Kurogi A, Yasuda S, Sato Y. Biodistribution studies for cell therapy products: Current status and issues. Regen Ther 2021; 18:202-216. [PMID: 34307798 PMCID: PMC8282960 DOI: 10.1016/j.reth.2021.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
Information on the biodistribution (BD) of cell therapy products (CTPs) is essential for prediction and assessment of their efficacy and toxicity profiles in non-clinical and clinical studies. To conduct BD studies, it is necessary to understand regulatory requirements, implementation status, and analytical methods. This review aimed at surveying international and Japanese trends concerning the BD study for CTPs and the following subjects were investigated, which were considered particularly important: 1) comparison of guidelines to understand the regulatory status of BD studies in a global setting; 2) case studies of the BD study using databases to understand its current status in cell therapy; 3) case studies on quantitative polymerase chain reaction (qPCR) used primarily in non-clinical BD studies for CTPs; and 4) survey of imaging methods used for non-clinical and clinical BD studies. The results in this review will be a useful resource for implementing BD studies.
Collapse
Affiliation(s)
- Yoshiteru Kamiyama
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Yoichi Naritomi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Yuu Moriya
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Syunsuke Yamamoto
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Tsukasa Kitahashi
- Bioscience & Engineering Laboratory, FUJIFILM Corp., 577 Ushijima, Kaisei-Machi, Ashigarakami-gun, Kanagawa, Japan
| | - Toshihiko Maekawa
- Bioscience & Engineering Laboratory, FUJIFILM Corp., 577 Ushijima, Kaisei-Machi, Ashigarakami-gun, Kanagawa, Japan
| | - Masahiro Yahata
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, Japan
| | - Takeshi Hanada
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo.Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, Japan
| | - Asako Uchiyama
- Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories, Ltd., Kagoshima, Kagoshima, Japan
| | - Akari Noumaru
- Kumamoto Laboratories, LSIM Safety Institute Corporation, 1285 Kurisaki-machi, Uto, Kumamoto, Japan
| | - Yoshiyuki Koga
- Kumamoto Laboratories, LSIM Safety Institute Corporation, 1285 Kurisaki-machi, Uto, Kumamoto, Japan
| | - Tomoaki Higuchi
- Non-clinical Development, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Masahiko Ito
- Tsukuba Research Institute, BoZo Research Center Inc., 8 Okubo, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Komatsu
- Science BD Department, CMIC Pharma Science Co., Ltd., 1-1-1 Shibaura, Minato-ku, Tokyo, Japan
| | - Sosuke Miyoshi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Sadaaki Kimura
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Nobuhiro Umeda
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Eriko Fujita
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Naoko Tanaka
- Evaluation Center, Terumo Corporation, 1500 Inokuchi, Nakai-machi, Ashigarakami-gun, Kanagawa, Japan
| | - Taku Sugita
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Satoru Takayama
- Cell Therapy Technology, Healthcare R&D Center, Asahi Kasei Corporation, 2-1 Samejima, Fuji-Shi, Shizuoka, Japan
| | - Akihiko Kurogi
- Regenerative Medicine Research & Planning Division, ROHTO Pharmaceutical Co., Ltd., Osaka, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| |
Collapse
|
239
|
Ebrahimi M, Rad MTS, Zebardast A, Ayyasi M, Goodarzi G, Tehrani SS. The critical role of mesenchymal stromal/stem cell therapy in COVID-19 patients: An updated review. Cell Biochem Funct 2021; 39:945-954. [PMID: 34545605 PMCID: PMC8652792 DOI: 10.1002/cbf.3670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 12/20/2022]
Abstract
New coronavirus disease 2019 (COVID-19), as a pandemic disaster, has drawn the attention of researchers in various fields to discover suitable therapeutic approaches for the management of COVID-19 patients. Currently, there are many worries about the rapid spread of COVID-19; there is no approved treatment for this infectious disease, despite many efforts to develop therapeutic procedures for COVID-19. Emerging evidence shows that mesenchymal stromal/stem cell (MSC) therapy can be a suitable option for the management of COVID-19. These cells have many biological features (including the potential of differentiation, high safety and effectiveness, secretion of trophic factors and immunoregulatory features) that make them suitable for the treatment of various diseases. However, some studies have questioned the positive role of MSC therapy in the treatment of COVID-19. Accordingly, in this paper, we will focus on the therapeutic impacts of MSCs and their critical role in cytokine storm of COVID-19 patients.
Collapse
Affiliation(s)
- Mohsen Ebrahimi
- Neonatal and Child Health Research CenterGolestan University of Medical SciencesGorganIran
| | - Mohammad Taha Saadati Rad
- Psychiatric and Behavioral Sciences Research Center, Addiction Research InstituteMazandaran University of Medical SciencesSariIran
| | - Arghavan Zebardast
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Mitra Ayyasi
- Critical Care NursingIslamic Azad University, Sari BranchSariIran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
- Scientific Research CenterTehran University of Medical SciencesTehranIran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
- Scientific Research CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
240
|
Suksatan W, Chupradit S, Yumashev AV, Ravali S, Shalaby MN, Mustafa YF, Kurochkin A, Siahmansouri H. Immunotherapy of multisystem inflammatory syndrome in children (MIS-C) following COVID-19 through mesenchymal stem cells. Int Immunopharmacol 2021; 101:108217. [PMID: 34627083 PMCID: PMC8487784 DOI: 10.1016/j.intimp.2021.108217] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new type of coronavirus causing coronavirus 2019 (COVID-19) that was first observed in Wuhan, China, in Dec. 2019. An inflammatory immune response targeting children appeared during the pandemic, which was associated with COVID-19 named multisystem inflammatory syndrome in children (MIS-C). Characteristics of MIS-C include the classic inflammation findings, multi-organ dysfunction, and fever as the cardinal feature. Up to now, no specific therapy has been identified for MIS-C. Currently, considerable progress has been obtained in the MIS-C treatment by cell therapy, specially Mesenchymal stem cells (MSCs). Unique properties have been reported for MSCs, such as various resources for purification of cell, high proliferation, self-renewal, non-invasive procedure, tissue regenerator, multidirectional differentiation, and immunosuppression. As indicated by a recent clinical research, MSCs have the ability of reducing disease inflammation and severity in children with MIS-C. In the present review study, the benefits and characteristics of MSCs and exosomes are discussed for treating patients with MIS-C.
Collapse
Affiliation(s)
- Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Sahithya Ravali
- Department of Pharmacy Practice, SRM College of Pharmacy, SRM Institute of Science and Technology, Chennai, India
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | | | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
241
|
Immunotherapy of multisystem inflammatory syndrome in children (MIS-C) following COVID-19 through mesenchymal stem cells. Int Immunopharmacol 2021. [DOI: oi.org/10.1016/j.intimp.2021.108217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
242
|
Pereira DR, Silva-Correia J, Oliveira JM, Reis RL, Pandit A. Macromolecular modulation of a 3D hydrogel construct differentially regulates human stem cell tissue-to-tissue interface. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 133:112611. [DOI: 10.1016/j.msec.2021.112611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/25/2021] [Accepted: 12/11/2021] [Indexed: 01/21/2023]
|
243
|
Suksatan W, Chupradit S, Yumashev AV, Ravali S, Shalaby MN, Mustafa YF, Kurochkin A, Siahmansouri H. Immunotherapy of multisystem inflammatory syndrome in children (MIS-C) following COVID-19 through mesenchymal stem cells. Int Immunopharmacol 2021. [DOI: https://doi.org/10.1016/j.intimp.2021.108217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
244
|
Cao H, Wu L, Tian X, Zheng W, Yuan M, Li X, Tian X, Wang Y, Song H, Shen Z. HO-1/BMMSC perfusion using a normothermic machine perfusion system reduces the acute rejection of DCD liver transplantation by regulating NKT cell co-inhibitory receptors in rats. Stem Cell Res Ther 2021; 12:587. [PMID: 34819139 PMCID: PMC8611848 DOI: 10.1186/s13287-021-02647-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 10/31/2021] [Indexed: 01/14/2023] Open
Abstract
Background Liver transplantation (LT) is required in many end-stage liver diseases. Donation after cardiac death (DCD) livers are often used, and treatment of acute rejection (ACR) requires the use of immunosuppressive drugs that are associated with complications. Bone marrow mesenchymal stem cells (BMMSCs) are used in treatment following LT; however, they have limitations, including low colonization in the liver. An optimized BMMSC application method is required to suppress ACR. Methods BMMSCs were isolated and modified with the heme oxygenase 1 (HO-1) gene. HO-1/BMMSCs were perfused into donor liver in vitro using a normothermic machine perfusion (NMP) system, followed by LT into rats. The severity of ACR was evaluated based on liver histopathology. Gene chip technology was used to detect differential gene expression, and flow cytometry to analyze changes in natural killer (NK) T cells. Results NMP induced BMMSCs to colonize the donor liver during in vitro preservation. The survival of HO-1/BMMSCs in liver grafts was significantly longer than that of unmodified BMMSCs. When the donor liver contained HO-1/BMMSCs, the local immunosuppressive effect was improved and prolonged, ACR was controlled, and survival time was significantly prolonged. The application of HO-1/BMMSCs reduced the number of NKT cells in liver grafts, increased the expression of NKT cell co-inhibitory receptors, and reduced NKT cell expression of interferon-γ. Conclusions NK cell and CD8+ T cell activation was inhibited by application of HO-1/BMMSCs, which reduced ACR of transplanted liver. This approach could be developed to enhance the success rate of LT. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02647-5.
Collapse
Affiliation(s)
- Huan Cao
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Longlong Wu
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Xuan Tian
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.,NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, People's Republic of China
| | - Mengshu Yuan
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Xiang Li
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Xiaorong Tian
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yuxin Wang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China. .,Tianjin Key Laboratory of Organ Transplantation, Tianjin, People's Republic of China.
| | - Zhongyang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China. .,Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, People's Republic of China.
| |
Collapse
|
245
|
Pelizzo G, Silvestro S, Avanzini MA, Zuccotti G, Mazzon E, Calcaterra V. Mesenchymal Stromal Cells for the Treatment of Interstitial Lung Disease in Children: A Look from Pediatric and Pediatric Surgeon Viewpoints. Cells 2021; 10:3270. [PMID: 34943779 PMCID: PMC8699409 DOI: 10.3390/cells10123270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/11/2021] [Accepted: 11/21/2021] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been proposed as a potential therapy to treat congenital and acquired lung diseases. Due to their tissue-regenerative, anti-fibrotic, and immunomodulatory properties, MSCs combined with other therapy or alone could be considered as a new approach for repair and regeneration of the lung during disease progression and/or after post- surgical injury. Children interstitial lung disease (chILD) represent highly heterogeneous rare respiratory diseases, with a wild range of age of onset and disease expression. The chILD is characterized by inflammatory and fibrotic changes of the pulmonary parenchyma, leading to gas exchange impairment and chronic respiratory failure associated with high morbidity and mortality. The therapeutic strategy is mainly based on the use of corticosteroids, hydroxychloroquine, azithromycin, and supportive care; however, the efficacy is variable, and their long-term use is associated with severe toxicity. The role of MSCs as treatment has been proposed in clinical and pre-clinical studies. In this narrative review, we report on the currently available on MSCs treatment as therapeutical strategy in chILD. The progress into the therapy of respiratory disease in children is mandatory to ameliorate the prognosis and to prevent the progression in adult age. Cell therapy may be a future therapy from both a pediatric and pediatric surgeon's point of view.
Collapse
Affiliation(s)
- Gloria Pelizzo
- Pediatric Surgery Department, Children’s Hospital “Vittore Buzzi”, 20154 Milano, Italy
- Department of Biomedical and Clinical Sciences-L. Sacco, University of Milan, 20157 Milan, Italy;
| | - Serena Silvestro
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (E.M.)
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Sciences-L. Sacco, University of Milan, 20157 Milan, Italy;
- Department of Pediatrics, Children’s Hospital “Vittore Buzzi”, 20154 Milano, Italy;
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (S.S.); (E.M.)
| | - Valeria Calcaterra
- Department of Pediatrics, Children’s Hospital “Vittore Buzzi”, 20154 Milano, Italy;
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
246
|
A Novel Cellular Therapy to Treat Pancreatic Pain in Experimental Chronic Pancreatitis Using Human Alpha-1 Antitrypsin Overexpressing Mesenchymal Stromal Cells. Biomedicines 2021; 9:biomedicines9111695. [PMID: 34829924 PMCID: PMC8615652 DOI: 10.3390/biomedicines9111695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 01/13/2023] Open
Abstract
Chronic pancreatitis (CP) is characterized by pancreatic inflammation, fibrosis, and abdominal pain that is challenging to treat. Mesenchymal stromal cells (MSCs) overexpressing human alpha-1 antitrypsin (hAAT-MSCs) showed improved mobility and protective functions over native MSCs in nonobese diabetic mice. We investigated whether hAAT-MSCs could mitigate CP and its associated pain using trinitrobenzene sulfonic acid (TNBS)-induced CP mouse models. CP mice were given native human MSCs or hAAT-MSCs (0.5 × 106 cells/mouse, i.v., n = 6–8/group). The index of visceral pain was measured by graduated von Frey filaments. Pancreatic morphology and pancreatic mast cell count were analyzed by morphological stains. Nociceptor transient receptor potential vanilloid 1 (TRPV1) expression in dorsal root ganglia (DRG) was determined by immunohistochemistry. hAAT-MSC-treated CP mice best preserved pancreatic morphology and histology. MSC or hAAT-MSC infusion reduced abdominal pain sensitivities. hAAT-MSC therapy also suppressed TRPV1 expression in DRG and reduced pancreatic mast cell density induced by TNBS. Overall, hAAT-MSCs reduced pain and mitigated pancreatic inflammation in CP equal to MSCs with a trend toward a higher pancreatic weight and better pain relief in the hAAT-MSC group compared to the MSC group. Both MSCs and hAAT-MSCs might be used as a novel therapeutic tool for CP-related pain.
Collapse
|
247
|
Mesenchymal stromal cell apoptosis is required for their therapeutic function. Nat Commun 2021; 12:6495. [PMID: 34764248 PMCID: PMC8586224 DOI: 10.1038/s41467-021-26834-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) ameliorate a wide range of diseases in preclinical models, but the lack of clarity around their mechanisms of action has impeded their clinical utility. The therapeutic effects of MSCs are often attributed to bioactive molecules secreted by viable MSCs. However, we found that MSCs underwent apoptosis in the lung after intravenous administration, even in the absence of host cytotoxic or alloreactive cells. Deletion of the apoptotic effectors BAK and BAX prevented MSC death and attenuated their immunosuppressive effects in disease models used to define MSC potency. Mechanistically, apoptosis of MSCs and their efferocytosis induced changes in metabolic and inflammatory pathways in alveolar macrophages to effect immunosuppression and reduce disease severity. Our data reveal a mode of action whereby the host response to dying MSCs is key to their therapeutic effects; findings that have broad implications for the effective translation of cell-based therapies. Mesenchymal stromal cells (MSCs) demonstrate therapeutic benefits in multiple diseases, but the mechanisms remain unclear as infused MSCs do not persist in the body. Here, the authors show that MSC apoptosis is an important mechanistic element, as MSCs rendered genetically incapable of apoptosis lose their ability to ameliorate disease.
Collapse
|
248
|
Dunbar H, Weiss DJ, Rolandsson Enes S, Laffey JG, English K. The Inflammatory Lung Microenvironment; a Key Mediator in MSC Licensing. Cells 2021; 10:cells10112982. [PMID: 34831203 PMCID: PMC8616504 DOI: 10.3390/cells10112982] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Recent clinical trials of mesenchymal stromal cell (MSC) therapy for various inflammatory conditions have highlighted the significant benefit to patients who respond to MSC administration. Thus, there is strong interest in investigating MSC therapy in acute inflammatory lung conditions, such as acute respiratory distress syndrome (ARDS). Unfortunately, not all patients respond, and evidence now suggests that the differential disease microenvironment present across patients and sub-phenotypes of disease or across disease severities influences MSC licensing, function and therapeutic efficacy. Here, we discuss the importance of licensing MSCs and the need to better understand how the disease microenvironment influences MSC activation and therapeutic actions, in addition to the need for a patient-stratification approach.
Collapse
Affiliation(s)
- Hazel Dunbar
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland
| | - Daniel J Weiss
- Department of Medicine, 226 Health Science Research Facility, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Sara Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22100 Lund, Sweden;
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, H91 W2TY Galway, Ireland;
- Department of Anaesthesia, Galway University Hospitals, SAOLTA University Health Group, H91 YR71 Galway, Ireland
| | - Karen English
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland
- Correspondence: ; Tel.: +353-1-7086290
| |
Collapse
|
249
|
Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 2021; 11:187. [PMID: 34727974 PMCID: PMC8561357 DOI: 10.1186/s13578-021-00698-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapeutics is already available for treatment of a range of diseases or medical conditions. Autologous or allogeneic MSCs obtained from self or donors have their own advantages and disadvantages in their medical practice. Therapeutic benefits of using autologous vs. allogeneic MSCs are inconclusive. Transplanted MSCs within the body interact with their physical microenvironment or niche, physiologically or pathologically, and such cells in a newly established tissue microenvironment may be impacted by the pathological harmful environmental factors to alter their unique biological behaviors. Meanwhile, a temporary microenvironment/niche may be also altered by the resident or niche-surrounding MSCs. Therefore, the functional plasticity and heterogeneity of MSCs caused by different donors and subpopulations of MSCs may result in potential uncertainty in their safe and efficacious medical practice. Acknowledging a connection between MSCs' biology and their existing microenvironment, donor-controlled clinical practice for the long-term therapeutic benefit is suggested to further consider minimizing MSCs potential harm for MSC-based individual therapies. In this review, we summarize the advantages and disadvantages of autologous vs. allogeneic MSCs in their therapeutic applications. Among other issues, we highlight the importance of better understanding of the various microenvironments that may affect the properties of niche-surrounding MSCs and discuss the clinical applications of MSCs within different contexts for treatment of different diseases including cardiomyopathy, lupus and lupus nephritis, diabetes and diabetic complications, bone and cartilage repair, cancer and tissue fibrosis.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| | - Hua Zhao
- Institute of Reproductive Medicine, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Linna Cheng
- Institute of Hematology, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Bin Wang
- Department of Neurosurgery, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
250
|
Kang IH, Baliga UK, Wu Y, Mehrotra S, Yao H, LaRue AC, Mehrotra M. Hematopoietic stem cell-derived functional osteoblasts exhibit therapeutic efficacy in a murine model of osteogenesis imperfecta. Stem Cells 2021; 39:1457-1477. [PMID: 34224636 DOI: 10.1002/stem.3432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/03/2021] [Accepted: 06/08/2021] [Indexed: 11/08/2022]
Abstract
Currently, there is no cure for osteogenesis imperfecta (OI)-a debilitating pediatric skeletal dysplasia. Herein we show that hematopoietic stem cell (HSC) therapy holds promise in treating OI. Using single-cell HSC transplantation in lethally irradiated oim/oim mice, we demonstrate significant improvements in bone morphometric, mechanics, and turnover parameters. Importantly, we highlight that HSCs cause these improvements due to their unique property of differentiating into osteoblasts/osteocytes, depositing normal collagen-an attribute thus far assigned only to mesenchymal stem/stromal cells. To confirm HSC plasticity, lineage tracing was done by transplanting oim/oim with HSCs from two specific transgenic mice-VavR, in which all hematopoietic cells are GFP+ and pOBCol2.3GFP, where GFP is expressed only in osteoblasts/osteocytes. In both models, transplanted oim/oim mice demonstrated GFP+ HSC-derived osteoblasts/osteocytes in bones. These studies unequivocally establish that HSCs differentiate into osteoblasts/osteocytes, and HSC transplantation can provide a new translational approach for OI.
Collapse
Affiliation(s)
- In-Hong Kang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Uday K Baliga
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yongren Wu
- Department of Orthopedics, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
- Clemson-MUSC Joint Bioengineering Program, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Hai Yao
- Department of Orthopedics, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
- Clemson-MUSC Joint Bioengineering Program, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Amanda C LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Meenal Mehrotra
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
- Center for Oral Health Research, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|