1
|
Silva Couto P, Stibbs DJ, Rotondi MC, Khalife R, Wolf D, Takeuchi Y, Rafiq QA. Biological differences between adult and perinatal human mesenchymal stromal cells and their impact on the manufacturing processes. Cytotherapy 2024; 26:1429-1441. [PMID: 38970611 DOI: 10.1016/j.jcyt.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 07/08/2024]
Abstract
The biological properties of human mesenchymal stromal cells (hMSCs) have been explored in over a thousand clinical trials in the last decade. Although hMSCs can be isolated from multiple sources, the degree of biological similarity between cell populations from these sources remains to be determined. A comparative study was performed investigating the growth kinetics and functionality of hMSCs isolated from adipose tissue (AT), bone marrow (BM) and umbilical cord tissue (UCT) expanded in monolayer over five passages. Adult hMSCs (AT, BM) had a slower proliferation ability than the UCT-hMSCs, with no apparent differences in their glucose consumption profile. BM-hMSCs produced higher concentrations of endogenous vascular endothelial growth factor (VEGF) compared to AT- and UCT-hMSCs. This study also revealed that UCT-hMSCs were more efficiently transduced by a lentiviral vector carrying a VEGF gene than their adult counterparts. Following cellular immunophenotypic characterization, no differences across the sources were found in the expression levels of the typical markers used to identify hMSCs. This work established a systematic approach for cell source selection depending on the hMSC's intended clinical application.
Collapse
Affiliation(s)
- Pedro Silva Couto
- Department of Biochemical Engineering, University College London, London, UK
| | - Dale J Stibbs
- Department of Biochemical Engineering, University College London, London, UK
| | - Marco C Rotondi
- Department of Biochemical Engineering, University College London, London, UK
| | - Rana Khalife
- Department of Biochemical Engineering, University College London, London, UK
| | | | - Yasuhiro Takeuchi
- Division of Infection and Immunity, University College London, London, UK; Biotherapeutics and Advanced Therapies, Scientific Research and Innovation, Medicines and Healthcare products Regulatory Agency, Potters Bar, UK
| | - Qasim A Rafiq
- Department of Biochemical Engineering, University College London, London, UK.
| |
Collapse
|
2
|
Muntiu A, Papait A, Vincenzoni F, Rossetti DV, Romele P, Cargnoni A, Silini A, Parolini O, Desiderio C. Proteomic analysis of the human amniotic mesenchymal stromal cell secretome by integrated approaches via filter-aided sample preparation. J Proteomics 2024; 310:105339. [PMID: 39448028 DOI: 10.1016/j.jprot.2024.105339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The immunomodulatory, anti-inflammatory and regenerative properties of the human amniotic mesenchymal stromal cells (hAMSCs) secretome are acknowledged but the understanding of the specific bioactive components remains incomplete. To address these limitations, the present investigation aimed to profile the proteins and peptides content of the hAMSC secretome through sample pretreatment and fractionation on 10 kDa molecular cut-off FASP (Filter Aided Sample Preparation) device and LC-MS analysis. The filter retained protein fraction underwent trypsin digestion, while the unretained was collected unchanged for intact small proteins and peptides analysis. This combined approach (C-FASP) collects in a single step two complementary fractions, advantageously saving sample volume and time of analysis. The bottom-up analysis of the C-FASP proteins fraction >10 kDa confirmed our previous findings, establishing a set of proteins consistently characterizing the hAMSC secretome. The analysis of the fraction <10 kDa, never been investigated to our knowledge, identified peptide fragments of thymosin beta 4 and beta 10, collagen alpha 1 chains I and III, alpha-enolase, and glyceraldehyde-3-phosphate dehydrogenase, involved in wound healing, anti-inflammatory response, tissue repair and regeneration, key biological activities of the secretome. C-FASP provided a comprehensive molecular profile of the hAMSC secretome offering new insights for enhanced therapeutic applications in regenerative medicine. SIGNIFICANCE: In this investigation we originally present the comprehensive proteomic investigation of the human amniotic mesenchymal stromal cell secretome by combining the analysis of the proteome and of the peptidome following sample pretreatment and fractionation by Filter Aided Sample Preparation (FASP) with 10 kDa molecular cut-off in coupling with LC-MS analysis. The proteome fraction retained by FASP filter was analyzed after enzymatic digestion, while the unretained fraction, below 10 kDa molecular mass, was analyzed unchanged in its intact form. This dual approach provides novel insights, previously unexplored, into the molecular components potentially responsible for the immunomodulatory and anti-inflammatory properties of the hAMSC secretome. These findings could significantly enhance the therapeutic potential of hAMSCs in regenerative medicine.
Collapse
Affiliation(s)
- Alexandra Muntiu
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy
| | - Federica Vincenzoni
- Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diana Valeria Rossetti
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Pietro Romele
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Antonietta Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Rome, Italy
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy.
| |
Collapse
|
3
|
Jiang X, Luo X, Cai C, Bai Y, Ding H, Yue H, Li Y, Yang Z, Zhang H, Liang Y, Peng C, Huang H, Liu M, Li Z, Shi Y, Han S, Li X, Zhang B. Umbilical cord mesenchymal stem cells in ulcerative colitis treatment: efficacy and possible mechanisms. Stem Cell Res Ther 2024; 15:272. [PMID: 39218946 PMCID: PMC11368034 DOI: 10.1186/s13287-024-03878-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) possess powerful immunomodulatory ability. This study aimed to assess the efficacy and safety of human umbilical cord-derived mesenchymal stem cells (UMSCs) in patients with ulcerative colitis (UC) and to explore the potential mechanisms. METHODS This prospective, self-controlled clinical study was conducted at Henan Provincial People's Hospital. Patients with moderate-to-severe active UC, unresponsive to traditional drugs were continuously enrolled from September 2018 to March 2023. UMSCs were administered intravenously monthly for two months at a cell dosage of 1 × 106 per kg. The primary outcome was a clinical response at 2 months. The levels of cytokines and progerin in the plasma of the patients were analyzed using enzyme-linked immunosorbent assay kits, and longitudinal data was analyzed using generalized estimation equation. RESULTS Forty-one patients were enrolled and received UMSC therapy. At 2 months, 73.2% (30/41) of patients achieved a clinical response, and 41.5% (17/41) achieved a clinical remission. At 6 months, 2 patients were lost to follow-up; the corresponding figures were 70.0% (25/41) and 34.2% (14/41), respectively. After UMSC therapy, the Mayo score, Mayo endoscopy score, mean and maximum values of Ulcerative Colitis Endoscopic Index of Severity and Nancy index were significantly reduced compared with baseline values. Additionally, the levels of progerin and inflammatory markers, such as interleukin (IL)-1β, IL-6, IL-8, IL-12, and IL-17 A decreased, while hemoglobin, albumin, and IL-10/IL-17 A ratio increased, particularly in the response group. Multiple stepwise logistic regression analysis showed age was an independent risk factor affecting efficacy (odds ratio, 0.875 (95% confidence interval (0.787, 0.972)); the area under the receiver operating characteristic curve for age was 0.79. No serious adverse events were observed during or after UMSC therapy. CONCLUSION UMSCs are safe and effective for patients with UC, with age being an independent risk factor affecting efficacy. Mechanistically, UMSC treatment may ameliorate cell senescence and suppress the secretion of pro-inflammatory cytokines. TRIAL REGISTRATION The study was retrospectively registered at www.chictr.org.cn/ (ChiCTR1900026035) on September 18, 2019.
Collapse
Affiliation(s)
- Xiaoke Jiang
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Xiaoying Luo
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
- Microbiome Laboratory, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Conghui Cai
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
- Microbiome Laboratory, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Yangqiu Bai
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Hui Ding
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Han Yue
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Yalong Li
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Zhiyu Yang
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
- Microbiome Laboratory, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Huimin Zhang
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Yuan Liang
- Department of Pulmonary and Critical Care Medicine, Xinyang Central Hospital, No.1, Siyi Road, Xinyang, Henan Province, 464000, China
| | - Cong Peng
- Department of Gastroenterology, Yunfu People's Hospital, No. 120, Huanshi East Road, Yunfu, Guangdong Province, 527300, China
| | - Huanrong Huang
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
- Microbiome Laboratory, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Min Liu
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
- Microbiome Laboratory, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Zhenjuan Li
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Yujie Shi
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
- Microbiome Laboratory, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
- Department of Pathology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China
| | - Shuangyin Han
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China.
| | - Xiuling Li
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China.
| | - Bingyong Zhang
- Department of Gastroenterology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, School of Clinical Medicine, Henan University, No.7 Weiwu Road, Jinshui District, Zhengzhou, Henan Province, 450003, China.
| |
Collapse
|
4
|
Burdzinska A, Szopa IM, Majchrzak-Kuligowska K, Roszczyk A, Zielniok K, Zep P, Dąbrowski FA, Bhale T, Galanty M, Paczek L. The Comparison of Immunomodulatory Properties of Canine and Human Wharton Jelly-Derived Mesenchymal Stromal Cells. Int J Mol Sci 2024; 25:8926. [PMID: 39201612 PMCID: PMC11354339 DOI: 10.3390/ijms25168926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Although therapies based on mesenchymal stromal cells (MSCs) are being implemented in clinical settings, many aspects regarding these procedures require further optimization. Domestic dogs suffer from numerous immune-mediated diseases similar to those found in humans. This study aimed to assess the immunomodulatory activity of canine (c) Wharton jelly (WJ)-derived MSCs and refer them to human (h) MSCs isolated from the same tissue. Canine MSC(WJ)s appeared to be more prone to in vitro aging than their human counterparts. Both canine and human MSC(WJ)s significantly inhibited the activation as well as proliferation of CD4+ and CD8+ T cells. The treatment with IFNγ significantly upregulated indoleamine-2,3-dioxygenase 1 (IDO1) synthesis in human and canine MSC(WJ)s, and the addition of poly(I:C), TLR3 ligand, synergized this effect in cells from both species. Unstimulated human and canine MSC(WJ)s released TGFβ at the same level (p > 0.05). IFNγ significantly increased the secretion of TGFβ in cells from both species (p < 0.05); however, this response was significantly stronger in human cells than in canine cells. Although the properties of canine and human MSC(WJ)s differ in detail, cells from both species inhibit the proliferation of activated T cells to a very similar degree and respond to pro-inflammatory stimulation by enhancing their anti-inflammatory activity. These results suggest that testing MSC transplantation in naturally occurring immune-mediated diseases in dogs may have high translational value for human clinical trials.
Collapse
Affiliation(s)
- Anna Burdzinska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska Str. 159, 02-776 Warsaw, Poland; (I.M.S.); (K.M.-K.); (T.B.)
| | - Iwona Monika Szopa
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska Str. 159, 02-776 Warsaw, Poland; (I.M.S.); (K.M.-K.); (T.B.)
| | - Kinga Majchrzak-Kuligowska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska Str. 159, 02-776 Warsaw, Poland; (I.M.S.); (K.M.-K.); (T.B.)
| | - Aleksander Roszczyk
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka Str. 59, 02-006 Warsaw, Poland (L.P.)
| | - Katarzyna Zielniok
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, Banacha Str. 1B, 02-097 Warsaw, Poland;
| | - Paweł Zep
- Veterinary Clinic “ochWET”, Pruszkowska Str. 19/21, 02-119 Warsaw, Poland
| | - Filip Andrzej Dąbrowski
- Department of Gynecology and Gynecological Oncology, Medical Centre of Postgraduate Education CMKP, Marymoncka Str. 99/103, 00-416 Warsaw, Poland;
| | - Tanushree Bhale
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska Str. 159, 02-776 Warsaw, Poland; (I.M.S.); (K.M.-K.); (T.B.)
| | - Marek Galanty
- Department of Small Animal Diseases and Clinic, Warsaw University of Life Sciences—SGGW, Nowoursynowska 159c, 02-776 Warsaw, Poland;
| | - Leszek Paczek
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka Str. 59, 02-006 Warsaw, Poland (L.P.)
| |
Collapse
|
5
|
Guo X, Zhang W, Lu J, Zhu Y, Sun H, Xu D, Xian S, Yao Y, Qian W, Lu B, Shi J, Ding X, Li Y, Tong X, Xiao S, Huang R, Ji S. Amniotic miracle: Investigating the unique development and applications of amniotic membrane in wound healing. Skin Res Technol 2024; 30:e13860. [PMID: 39073182 DOI: 10.1111/srt.13860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The perfect repair of damaged skin has always been a constant goal for scientists; however, the repair and reconstruction of skin is still a major problem and challenge in injury and burns medicine. Human amniotic membrane (hAM), with its good mechanical properties and anti-inflammatory, antioxidant and antimicrobial benefits, containing growth factors that promote wound healing, has evolved over the last few decades from simple skin sheets to high-tech dressings, such as being made into nanocomposites, hydrogels, powders, and electrostatically spun scaffolds. This paper aims to explore the historical development, applications, trends, and research hotspots of hAM in wound healing. METHODS We examined 2660 publications indexed in the Web of Science Core Collection (WoSCC) from January 1, 1975 to July 12, 2023. Utilizing bibliometric methods, we employed VOSviewer, CiteSpace, and R-bibliometrix to characterize general information, identify development trends, and highlight research hotspots. Subsequently, we identified a collection of high-quality English articles focusing on the roles of human amniotic epithelial stem cells (hAESCs), human amniotic mesenchymal stem cells (hAMSCs), and amniotic membrane (AM) scaffolds in regenerative medicine and tissue engineering. RESULTS Bibliometric analysis identified Udice-French Research Universities as the most productive affiliation and Tseng S.C.G. as the most prolific author. Keyword analysis, historical direct quotations network, and thematic analysis helped us review the historical and major themes in this field. Our examination included the knowledge structure, global status, trends, and research hotspots regarding the application of hAM in wound healing. Our findings indicate that contemporary research emphasizes the preparation and application of products derived from hAM. Notably, both hAM and the cells isolated from it - hADSCs and hAESCs are prominent and promising areas of research in regenerative medicine and tissue engineering. CONCLUSION This research delivers a comprehensive understanding of the knowledge frameworks, global dynamics, emerging patterns, and primary research foci in the realm of hAM applications for wound healing. The field is rapidly evolving, and our findings offer valuable insights for researchers. Future research outcomes are anticipated to be applied in clinical practice, enhancing methods for disease prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Xinya Guo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei Zhang
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jianyu Lu
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yushu Zhu
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Hanlin Sun
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Dayuan Xu
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shuyuan Xian
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yuntao Yao
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijin Qian
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingnan Lu
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaying Shi
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyi Ding
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yixu Li
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xirui Tong
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shichu Xiao
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Runzhi Huang
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shizhao Ji
- Department of Burn Surgery, Research Unit of key techniques for treatment of burns and combined burns and trauma injury, The First Affiliated Hospital of Naval Medical University, Shanghai, People's Republic of China, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
6
|
Yang Y, Li S, Peng Q, Guo Y, Gao Y, Gong Y, Lu J, Zhang Y, Shi X. Human umbilical cord mesenchymal stem cells promoted tumor cell growth associated with increased interleukin-18 in hepatocellular carcinoma. Mol Biol Rep 2024; 51:762. [PMID: 38874690 DOI: 10.1007/s11033-024-09688-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is experiencing a concerning rise in both incidence and mortality rates. Current therapeutic strategies are limited in their effectiveness, largely due to the complex causes of the disease and significant levels of drug resistance. Given the latest developments in human umbilical cord mesenchymal stem cells (hUC-MSCs) research, there is a debate over the continued use of stem cell transplantation for treating tumors. Consequently, this study seeks to explore the role of hUC-MSCs in the management of HCC. METHODS AND RESULTS HUC-MSCs increased the number (10.75 ± 1.50) in the DEN/TCPOBOP-induced mice hepatoma model, compared with DMSO group (7.25 ± 1.71). Moreover, the liver index in hUC-MSCs group (0.21 ± 0.06) was greater than that in DMSO group (0.09 ± 0.01). Immunohistochemical (IHC) analysis revealed that while hUC-MSCs did not alter Foxp3 expression, they significantly stimulated Ki67 expression, indicative of increased tumor cellular proliferation. Additionally, immunofluorescence (IF) studies showed that hUC-MSCs increased CD8+ T cell counts without affecting macrophage numbers. Notably, granzyme B expression remained nearly undetectable. We observed that serum IL-18 levels were higher in the hUC-MSCs group (109.66 ± 0.38 pg/ml) compared to the DMSO group (91.14 ± 4.37 pg/ml). Conversely, IL-1β levels decreased in the hUC-MSCs group (63.00 ± 0.53 pg/ml) relative to the DMSO group (97.38 ± 9.08 pg/ml). CONCLUSIONS According to this study, hUC-MSCs promoted the growth of liver tumors. Therefore, we proposed that hUC-MSCs are not suitable for treating HCC, as they exhibit clinically prohibited abnormalities.
Collapse
Affiliation(s)
- Yanguang Yang
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shenghao Li
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yinglin Guo
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Junlan Lu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yuman Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xinli Shi
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China.
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China.
| |
Collapse
|
7
|
Liu J, Yang Y, Qi Y. Efficacy of mesenchymal stromal cells in the treatment of type 1 diabetes: a systematic review. Cell Tissue Bank 2024; 25:663-676. [PMID: 38383908 PMCID: PMC11143029 DOI: 10.1007/s10561-024-10128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024]
Abstract
To investigate the efficacy of mesenchymal stromal cells in the treatment of type 1 diabetes. Articles about the effects of mesenchymal stromal cells for T1D were retrieved in PubMed, Web of Science, Embase, and the Cochrane Library databases up to July 2023. Additional relevant studies were manually searched through citations. HbA1c, FBG, PBG, insulin requirement and C-peptide were assessed. The risk of bias was evaluated with the ROB 2.0 and ROBINS-I tools. Six RCTs and eight nRCTs were included. Of the 14 studies included, two evaluated BM-MSCs, three evaluated UC-MSCs, five evaluated AHSCT, two evaluated CB-SCs, and two evaluated UC-SCs plus aBM-MNCs. At the end of follow-up, ten studies found that mesenchymal stromal cells improved glycemic outcomes in T1D, while the remaining four studies showed no significant improvement. Findings support the positive impacts observed from utilizing mesenchymal stromal cells in individuals with T1D. However, the overall methodological quality of the identified studies and findings is heterogeneous, limiting the interpretation of the therapeutic benefits of mesenchymal stromal cells in T1D. Methodically rigorous research is needed to further increase credibility.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Endocrinology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430034, China
| | - Yang Yang
- Department of Endocrinology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430034, China.
| | - Yun Qi
- Department of Endocrinology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430034, China.
| |
Collapse
|
8
|
Yang LF, He JD, Jiang WQ, Wang XD, Yang XC, Liang Z, Zhou YK. Interferon-gamma Treatment of Human Umbilical Cord Mesenchymal Stem Cells can Significantly Reduce Damage Associated with Diabetic Peripheral Neuropathy in Mice. Curr Stem Cell Res Ther 2024; 19:1129-1141. [PMID: 37644749 DOI: 10.2174/1574888x19666230829155046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Diabetic peripheral neuropathy causes significant pain to patients. Umbilical cord mesenchymal stem cells have been shown to be useful in the treatment of diabetes and its complications. The aim of this study was to investigate whether human umbilical cord mesenchymal stem cells treated with interferon-gamma can ameliorate nerve injury associated with diabetes better than human umbilical cord mesenchymal stem cells without interferon-gamma treatment. METHODS Human umbilical cord mesenchymal stem cells were assessed for adipogenic differentiation, osteogenic differentiation, and proliferation ability. Vonfry and a hot disc pain tester were used to evaluate tactile sensation and thermal pain sensation in mice. Hematoxylin-eosin and TUNEL staining were performed to visualize sciatic nerve fiber lesions and Schwann cell apoptosis in diabetic mice. Western blotting was used to detect expression of the apoptosis-related proteins Bax, B-cell lymphoma-2, and caspase-3 in mouse sciatic nerve fibers and Schwann cells. Real-Time Quantitative PCR was used to detect mRNA levels of the C-X-C motif chemokine ligand 1, C-X-C motif chemokine ligand 2, C-X-C motif chemokine ligand 9, and C-X-C motif chemokine ligand 10 in mouse sciatic nerve fibers and Schwann cells. Enzyme-linked immunosorbent assay was used to detect levels of the inflammatory cytokines, interleukin- 1β, interleukin-6, and tumor necrosis factor-α in serum and Schwann cells. RESULTS The adipogenic differentiation capacity, osteogenic differentiation capacity, and proliferation ability of human umbilical cord mesenchymal stem cells were enhanced after interferon-gamma treatment. Real-Time Quantitative PCR revealed that interferon-gamma promoted expression of the adipogenic markers, PPAR-γ and CEBP-α, as well as of the osteogenic markers secreted phosphoprotein 1, bone gamma-carboxyglutamate protein, collagen type I alpha1 chain, and Runt-related transcription factor 2. The results of hematoxylin-eosin and TUNEL staining showed that pathological nerve fiber damage and Schwann cell apoptosis were reduced after the injection of interferon-gamma-treated human umbilical cord mesenchymal stem cells. Expression of the apoptosis-related proteins, caspase-3 and Bax, was significantly reduced, while expression of the anti-apoptotic protein B-cell lymphoma-2 was significantly increased. mRNA levels of the cell chemokines, C-X-C motif chemokine ligand 1, C-X-C motif chemokine ligand 2, C-X-C motif chemokine ligand 9, and C-X-C motif chemokine ligand 10, were significantly reduced, and levels of the inflammatory cytokines, interleukin-1β, interleukin-6, and tumor necrosis factor-α, were decreased. Tactile and thermal pain sensations were improved in diabetic mice. CONCLUSION Interferon-gamma treatment of umbilical cord mesenchymal stem cells enhanced osteogenic differentiation, adipogenic differentiation, and proliferative potential. It can enhance the ability of human umbilical cord mesenchymal stem cells to alleviate damage to diabetic nerve fibers and Schwann cells, in addition to improving the neurological function of diabetic mice.
Collapse
Affiliation(s)
- Li-Fen Yang
- Department of Endocrinology and Metabolism, First People's Hospital of Yunnan Province, China
| | - Jun-Dong He
- Department of Endocrinology and Metabolism, First People's Hospital of Yunnan Province, China
- Medical School, Kunming University of Science and Technology, Kunming, 650300, Yunnan Province, People's Republic of China
| | - Wei-Qi Jiang
- The First Clinical Medical College, Kunming Medical University, Kunming, 650050, People's Republic of China
| | - Xiao-Dan Wang
- Kunming Yan'an Hospital Kunming, 650051, People's Republic of China
| | - Xiao-Chun Yang
- Department of Ophthalmology First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, People's Republic of China
| | - Zhi Liang
- Department of Information Center, First People's Hospital of Yunnan Province, China
| | - Yi-Kun Zhou
- Department of Endocrinology and Metabolism, First People's Hospital of Yunnan Province, China
| |
Collapse
|
9
|
Pérez I, Galindo S, López-Miguel A, Nieto-Miguel T, de la Mata A, López-Paniagua M, Alberca M, Herreras JM, Calonge M. In Vivo Confocal Microscopy in Limbal Stem Cell Deficiency After Mesenchymal Stem Cell Transplantation: A Sub-analysis from a Phase I-II Clinical Trial. Ophthalmol Ther 2023; 12:3251-3262. [PMID: 37773479 PMCID: PMC10640524 DOI: 10.1007/s40123-023-00809-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/31/2023] [Indexed: 10/01/2023] Open
Abstract
INTRODUCTION The aim of this work is to evaluate the effect of mesenchymal stem cell transplantation (MSCT) and cultivated limbal epithelial transplantation (CLET) therapies on the limbus of patients suffering from limbal stem cell deficiency (LSCD). METHODS A sub-analysis of a phase I-II randomized, controlled, and double-masked clinical trial was performed to assess the changes in the anatomical structures of the limbus. In vivo confocal microscopy (IVCM) analysis was carried out in LSCD eyes before and 12 months after allogeneic MSCT or CLET. Epithelial phenotype of the central cornea, as well as the presence of transition zones and palisades of Vogt in the limbus, were assessed using Wilcoxon test. RESULTS Twenty-three LSCD (14 MSCT and nine CLET) eyes were included. The epithelial phenotype of the central cornea improved significantly (p < 0.001) from 15 (eight MSCT, seven CLET) and eight (six MSCT, two CLET) LSCD eyes showing conjunctival and mixed phenotypes, respectively, to eight (five MSCT, three CLET), five (two MSCT, three CLET), and ten (seven MSCT, three CLET) eyes showing conjunctival, mixed, and corneal phenotypes, respectively. Transition areas and palisades of Vogt were observed in at least one quadrant in nine (five MSCT, four CLET) and 16 (nine MSCT, seven CLET), and in four (two MSCT, two CLET) and six (three MSCT, three CLET) LSCD eyes before and after surgery, respectively. Changes in the transition zones and palisades were solely significant (p = 0.046) for the nasal and inferior quadrants, respectively. CONCLUSIONS MSCT and CLET improved the central corneal epithelial phenotype despite only minor changes in the anatomical structures of the limbus, as detected by IVCM technology. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT01562002.
Collapse
Affiliation(s)
- Inmaculada Pérez
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain
| | - Sara Galindo
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain
- Department of Cell Biology, Genetics, Histology and Pharmacology, Universidad de Valladolid, Valladolid, Spain
| | - Alberto López-Miguel
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain.
- Departamento de Cirugía, Oftalmología, Otorrinolaringología y Fisioterapia, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.
| | - Teresa Nieto-Miguel
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain
- Department of Cell Biology, Genetics, Histology and Pharmacology, Universidad de Valladolid, Valladolid, Spain
| | - Ana de la Mata
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain
| | - Marina López-Paniagua
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain
- Department of Cell Biology, Genetics, Histology and Pharmacology, Universidad de Valladolid, Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
| | - Mercedes Alberca
- IBGM (Institute of Molecular Biology and Genetics) and University Scientific Park, Universidad de Valladolid, Valladolid, Spain
| | - José M Herreras
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain
- Departamento de Cirugía, Oftalmología, Otorrinolaringología y Fisioterapia, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Margarita Calonge
- IOBA (Institute of Applied Ophthalmobiology), Universidad de Valladolid, Campus Miguel Delibes, Paseo Belén, 17, 47011, Valladolid, Spain
- CIBER-BBN (Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine), Carlos III National Institute of Health, Valladolid, Spain
- Departamento de Cirugía, Oftalmología, Otorrinolaringología y Fisioterapia, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
10
|
Sababathy M, Ramanathan G, Abd Rahaman NY, Ramasamy R, Biau FJ, Qi Hao DL, Hamid NFS. A 'one stone, two birds' approach with mesenchymal stem cells for acute respiratory distress syndrome and Type II diabetes mellitus. Regen Med 2023; 18:913-934. [PMID: 38111999 DOI: 10.2217/rme-2023-0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023] Open
Abstract
This review explores the intricate relationship between acute respiratory distress syndrome (ARDS) and Type II diabetes mellitus (T2DM). It covers ARDS epidemiology, etiology and pathophysiology, along with current treatment trends and challenges. The lipopolysaccharides (LPS) role in ARDS and its association between non-communicable diseases and COVID-19 are discussed. The review highlights the therapeutic potential of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) for ARDS and T2DM, emphasizing their immunomodulatory effects. This review also underlines how T2DM exacerbates ARDS pathophysiology and discusses the potential of hUC-MSCs in modulating immune responses. In conclusion, the review highlights the multidisciplinary approach to managing ARDS and T2DM, focusing on inflammation, oxidative stress and potential therapy of hUC-MSCs in the future.
Collapse
Affiliation(s)
- Mogesh Sababathy
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Ghayathri Ramanathan
- Faculty of Computer Science & Information Technology, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nor Yasmin Abd Rahaman
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Foo Jhi Biau
- Centre for Drug Discovery & Molecular Pharmacology (CDDMP), Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
| | - Daniel Looi Qi Hao
- My Cytohealth Sdn. Bhd., 18-2, Jalan Radin Bagus 1, Bandar Seri Petaling, Kuala Lumpur, 57000, Malaysia
| | - Nur-Fazila Saulol Hamid
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
11
|
Huang L, Zhang S, Wu J, Guo B, Gao T, Shah SZA, Huang B, Li Y, Zhu B, Fan J, Wang L, Xiao Y, Liu W, Tian Y, Fang Z, Lv Y, Xie L, Yao S, Ke G, Huang X, Huang Y, Li Y, Jia Y, Li Z, Feng G, Huo Y, Li W, Zhou Q, Hao J, Hu B, Chen H. Immunity-and-matrix-regulatory cells enhance cartilage regeneration for meniscus injuries: a phase I dose-escalation trial. Signal Transduct Target Ther 2023; 8:417. [PMID: 37907503 PMCID: PMC10618459 DOI: 10.1038/s41392-023-01670-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 09/12/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023] Open
Abstract
Immunity-and-matrix-regulatory cells (IMRCs) derived from human embryonic stem cells have unique abilities in modulating immunity and regulating the extracellular matrix, which could be mass-produced with stable biological properties. Despite resemblance to mesenchymal stem cells (MSCs) in terms of self-renew and tri-lineage differentiation, the ability of IMRCs to repair the meniscus and the underlying mechanism remains undetermined. Here, we showed that IMRCs demonstrated stronger immunomodulatory and pro-regenerative potential than umbilical cord MSCs when stimulated by synovial fluid from patients with meniscus injury. Following injection into the knees of rabbits with meniscal injury, IMRCs enhanced endogenous fibrocartilage regeneration. In the dose-escalating phase I clinical trial (NCT03839238) with eighteen patients recruited, we found that intra-articular IMRCs injection in patients was safe over 12 months post-grafting. Furthermore, the effective results of magnetic resonance imaging (MRI) of meniscus repair and knee functional scores suggested that 5 × 107 cells are optimal for meniscus injury treatment. In summary, we present the first report of a phase I clinical trial using IMRCs to treat meniscus injury. Our results demonstrated that intra-articular injection of IMRCs is a safe and effective therapy by providing a permissive niche for cartilage regeneration.
Collapse
Affiliation(s)
- Liangjiang Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Zhang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Wu
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Baojie Guo
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Tingting Gao
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Sayed Zulfiqar Ali Shah
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Huang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yajie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaqi Fan
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Liu Wang
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yani Xiao
- Beijing Key Lab for Pre-clinical Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing, China
| | - Wenjing Liu
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Yao Tian
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Zhengyu Fang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Lv
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingfeng Xie
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Yao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaotan Ke
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolin Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Beijing Key Lab for Pre-clinical Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing, China
| | - Yujuan Li
- Beijing Zephyrm Biotechnologies Co., Ltd., Beijing, China
| | - Yi Jia
- Beijing Zephyrm Biotechnologies Co., Ltd., Beijing, China
| | - Zhongwen Li
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Guihai Feng
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan Huo
- Beijing Key Lab for Pre-clinical Safety Evaluation of Drugs, National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing, China
| | - Wei Li
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Zhou
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Hao
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Baoyang Hu
- National Stem Cell Resource Center, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Wang S, Yao Z, Chen L, Li J, Chen S, Fan C. Preclinical assessment of IL-1β primed human umbilical cord mesenchymal stem cells for tendon functional repair through TGF-β/IL-10 signaling. Heliyon 2023; 9:e21411. [PMID: 37954299 PMCID: PMC10638607 DOI: 10.1016/j.heliyon.2023.e21411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023] Open
Abstract
Background Inadequate repair capacity and disturbed immune compartments are the main pathological causes of tendinopathy. Transplantation of mesenchymal stem cells (MSCs) become an effective clinic option to alleviate tendinopathy. Interleukin-1β (IL-1β) could confer on MSCs enhanced immunoregulatory capability to remodel the repair microenvironment favoring tissue repair. Therefore, IL-1β activated UC-MSCs (1βUC-MSCs) may exert favorable efficacy in promoting tendon repair in a preclinical tendinopathy rat model. Methods Tendon-derived stem cells (TDSCs) were isolated and characterized. In vitro, the levels of immunoregulatory-related cytokines such as IL-1β, IL-6, IL-10, and TGF-β secreted by 1βUC-MSCs and unprimed UC-MSCs was measured. And tendon-specific markers expressed by TDSCs cultured with primed cultured medium (CM) or unprimed CM were detected. In vivo, Achilles tendinopathy was induced by 30 μL collagenase I injection in Sprague Dawley rats. One week later, the rats were randomly injected with UC-MSCs primed with IL-1β (106 cells per tendon), UC-MSCs, or PBS. After rats were sacrificed, histological evaluation, electron microscopy, biomechanical tests, gait performance were conducted to evaluate the structural and functional recovery of Achilles tendons. The inflammation and metabolic state of the extracellular matrix, and the potential mechanism were assessed by immunohistochemical staining and Western blot. Results UC-MSCs were activated by IL-1β to secrete higher levels of IL-10 and TGF-β while the secretion levels of IL-6 and IL-1β were not changed significantly, promoting a higher expression level of COL I and TNMD in TDSCs under proinflammatory environment. In vivo, the transplanted 1βUC-MSCs could survive up to 5 weeks after injection with tenogenic differentiation and improved tendon healing histologically semi-quantified by modified Bonar scores. This structural regeneration was further confirmed by observation of ultrastructural morphology, and led to good functional recovery including improved biomechanical properties and gait performance. During this process, the inflammatory response and metabolism of the extracellular matrix was improved through TGF-β/IL-10 pathway. Conclusion This study demonstrated that the transplantation of UC-MSCs activated by IL-1β exhibited satisfactory ability for promoting tendon functional repair in a tendinopathy rat model. During this process, the balance of inflammatory response and extracellular matrix metabolism was remodeled, and the TGF-β/Smad2/3 and IL-10 signaling pathways were activated simultaneously. We cautiously conclude that the IL-1β primed UC-MSCs could be a promising strategy for enhancing the ability of MSCs to treat tendinopathy.
Collapse
Affiliation(s)
- Shikun Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Zhixiao Yao
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Lei Chen
- Department of Orthopedics, Tongji Hospital, School of Medicine Tongji University, Shanghai, China
| | - Juehong Li
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Shuai Chen
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| | - Cunyi Fan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, China
| |
Collapse
|
13
|
Smolinská V, Boháč M, Danišovič Ľ. Current status of the applications of conditioned media derived from mesenchymal stem cells for regenerative medicine. Physiol Res 2023; 72:S233-S245. [PMID: 37888967 PMCID: PMC10669946 DOI: 10.33549/physiolres.935186] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 12/01/2023] Open
Abstract
Recently published studies suggest that the paracrine substances released by mesenchymal stem cells (MSCs) are the primary motive behind the therapeutic action reported in these cells. Pre-clinical and clinical research on MSCs has produced promising outcomes. Furthermore, these cells are generally safe for therapeutic use and may be extracted from a variety of anatomical regions. Recent research has indicated, however, that transplanted cells do not live long and that the advantages of MSC treatment may be attributable to the large diversity of bioactive substances they create, which play a crucial role in the control of essential physiological processes. Secretome derivatives, such as conditioned media or exosomes, may provide significant benefits over cells in terms of manufacture, preservation, handling, longevity of the product, and potential as a ready-to-use biologic product. Despite their immunophenotypic similarities, the secretome of MSCs appears to vary greatly depending on the host's age and the niches in which the cells live. The secretome's effect on multiple biological processes such as angiogenesis, neurogenesis, tissue repair, immunomodulation, wound healing, anti-fibrotic, and anti-tumor for tissue maintenance and regeneration has been discovered. Defining the secretome of cultured cultivated MSC populations by conditioned media analysis will allow us to assess its potential as a novel treatment approach. This review will concentrate on accumulating data from pre-clinical and clinical trials pointing to the therapeutic value of the conditioned medium. At last, the necessity of characterizing the conditioned medium for determining its potential for cell-free treatment therapy will be emphasized in this study.
Collapse
|
14
|
El-Naseery NI, Elewa YHA, El-Behery EI, Dessouky AA. Human umbilical cord blood-derived mesenchymal stem cells restored hematopoiesis by improving radiation induced bone marrow niche remodeling in rats. Ann Anat 2023; 250:152131. [PMID: 37460043 DOI: 10.1016/j.aanat.2023.152131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/07/2023] [Accepted: 06/20/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Functional hematopoiesis is governed by the bone marrow (BM) niche, which is compromised by radiotherapy, leading to radiation induced BM failure. The aim of this study was to demonstrate the radiation induced pathological remodeling of the niche and the efficacy of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) in restoring hematopoiesis via improvement of the niche. METHODS Thirty male Wistar rats were equally assigned to three groups: control (CON), irradiated (IR), and IR+hUCB-MSCs. Biochemical, histopathological and immunohistochemical analyses were performed to detect collagen type III and IV, Aquaporin 1+ sinusoidal endothelial cells and immature hematopoietic cells, CD11c+ dendritic cells, Iba1+ macrophages, CD9+ megakaryocytes, Sca-1+, cKit+, CD133 and N-cadherin+ hematopoietic stem and progenitor cells, CD20+, Gr1+ mature hematopoietic cells, in addition to ki67+ proliferation, Bcl-2+ anti-apoptotic, caspase-3+ apoptotic, TNF-α+ inflammatory cells. Histoplanimetry data were statistically analyzed using the one-way analysis of variance followed by the post hoc Duncan's test. Moreover, Pearson's correlation was used to assess the correlation between various parameters. RESULTS In comparison to the IR group, the IR+hUCB-MSCs group showed restored cell populations and extracellular collagen components of the BM niche with significant increase in hematopoietic stem, progenitor, mature and proliferating cells, and a considerable decrease in apoptotic and inflammatory cells. Furthermore, highly significant correlations between BM niche and blood biochemical, histopathological, and immunohistochemical parameters were observed. CONCLUSION hUCB-MSCs restored functional hematopoiesis through amelioration of the BM niche components via reduction of oxidative stress, DNA damage, inflammation, and apoptosis with upregulation of cellular proliferation.
Collapse
Affiliation(s)
- Nesma I El-Naseery
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, P.O. Box, 44511, Zagazig, Egypt.
| | - Yaser H A Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, P.O. Box, 44511, Zagazig, Egypt; Laboratory of Anatomy, Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-818, Japan
| | - Eman I El-Behery
- Anatomy & Embryology Department, Faculty of Veterinary Medicine, Zagazig University, P.O. Box, 44511 Zagazig, Egypt
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, P.O. Box, 44519 Zagazig, Egypt
| |
Collapse
|
15
|
Couto PS, Al-Arawe N, Filgueiras IS, Fonseca DLM, Hinterseher I, Catar RA, Chinnadurai R, Bersenev A, Cabral-Marques O, Moll G, Verter F. Systematic review and meta-analysis of cell therapy for COVID-19: global clinical trial landscape, published safety/efficacy outcomes, cell product manufacturing and clinical delivery. Front Immunol 2023; 14:1200180. [PMID: 37415976 PMCID: PMC10321603 DOI: 10.3389/fimmu.2023.1200180] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/24/2023] [Indexed: 07/08/2023] Open
Abstract
During the pandemic of severe respiratory distress syndrome coronavirus 2 (SARS-CoV2), many novel therapeutic modalities to treat Coronavirus 2019 induced disease (COVID-19) were explored. This study summarizes 195 clinical trials of advanced cell therapies targeting COVID-19 that were registered over the two years between January 2020 to December 2021. In addition, this work also analyzed the cell manufacturing and clinical delivery experience of 26 trials that published their outcomes by July 2022. Our demographic analysis found the highest number of cell therapy trials for COVID-19 was in United States, China, and Iran (N=53, 43, and 19, respectively), with the highest number per capita in Israel, Spain, Iran, Australia, and Sweden (N=0.641, 0.232, 0,223, 0.194, and 0.192 trials per million inhabitants). The leading cell types were multipotent mesenchymal stromal/stem cells (MSCs), natural killer (NK) cells, and mononuclear cells (MNCs), accounting for 72%, 9%, and 6% of the studies, respectively. There were 24 published clinical trials that reported on infusions of MSCs. A pooled analysis of these MSC studies found that MSCs provide a relative risk reduction for all-cause COVID-19 mortality of RR=0.63 (95% CI 0.46 to 0.85). This result corroborates previously published smaller meta-analyses, which suggested that MSC therapy demonstrated a clinical benefit for COVID-19 patients. The sources of the MSCs used in these studies and their manufacturing and clinical delivery methods were remarkably heterogeneous, with some predominance of perinatal tissue-derived products. Our results highlight the important role that cell therapy products may play as an adjunct therapy in the management of COVID-19 and its related complications, as well as the importance of controlling key manufacturing parameters to ensure comparability between studies. Thus, we support ongoing calls for a global registry of clinical studies with MSC products that could better link cell product manufacturing and delivery methods to clinical outcomes. Although advanced cell therapies may provide an important adjunct treatment for patients affected by COVID-19 in the near future, preventing pathology through vaccination still remains the best protection to date. We conducted a systematic review and meta-analysis of advanced cell therapy clinical trials as potential novel treatment for COVID-19 (resulting from SARS-CoV-2 coronavirus infection), including analysis of the global clinical trial landscape, published safety/efficacy outcomes (RR/OR), and details on cell product manufacturing and clinical delivery. This study had a 2-year observation interval from start of January 2020 to end of December 2021, including a follow-up period until end of July to identify published outcomes, which covers the most vivid period of clinical trial activity, and is also the longest observation period studied until today. In total, we identified 195 registered advanced cell therapy studies for COVID-19, employing 204 individual cell products. Leading registered trial activity was attributed to the USA, China, and Iran. Through the end of July 2022, 26 clinical trials were published, with 24 out of 26 articles employing intravenous infusions (IV) of mesenchymal stromal/stem cell (MSC) products. Most of the published trials were attributed to China and Iran. The cumulative results from the 24 published studies employing infusions of MSCs indicated an improved survival (RR=0.63 with 95% Confidence Interval 0.46 to 0.85). Our study is the most comprehensive systematic review and meta-analysis on cell therapy trials for COVID-19 conducted to date, clearly identifying the USA, China, and Iran as leading advanced cell therapy trial countries for COVID-19, with further strong contributions from Israel, Spain, Australia and Sweden. Although advanced cell therapies may provide an important adjunct treatment for patients affected by COVID-19 in the future, preventing pathology through vaccination remains the best protection.
Collapse
Affiliation(s)
- Pedro S. Couto
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, United Kingdom
- CellTrials.org and Parent’s Guide to Cord Blood Foundation, a non-profit organization headquartered in Brookeville, Brookeville, MD, United States
| | - Nada Al-Arawe
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
- Vascular Surgery Clinic, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Igor S. Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Dennyson L. M. Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo, SP, Brazil
| | - Irene Hinterseher
- Vascular Surgery Clinic, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Brandenburg Theodor Fontane, Neuruppin, Germany
- Fakultät der Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburg Technischen Universität (BTU) Cottbus-Senftenberg, Potsdam, Germany
| | - Rusan A. Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
| | - Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
| | - Alexey Bersenev
- Advanced Cell Therapy (ACT) Laboratory, Yale School of Medicine, New Haven, CT, United States
| | - Otávio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo, SP, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, SP, Brazil
- Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
- Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frances Verter
- CellTrials.org and Parent’s Guide to Cord Blood Foundation, a non-profit organization headquartered in Brookeville, Brookeville, MD, United States
| |
Collapse
|
16
|
Abatay-Sel F, Erol A, Suleymanoglu M, Demirayak G, Kekik-Cinar C, Kuruca DS, Savran-Oguz F. The in vitro treatment of mesenchymal stem cells for colorectal cancer cells. Med Oncol 2023; 40:103. [PMID: 36811793 DOI: 10.1007/s12032-023-01972-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/03/2023] [Indexed: 02/24/2023]
Abstract
Colorectal cancer is the most common tumor of the gastrointestinal system. The conventional treatment options for colorectal cancer are troublesome for both patients and clinicians. Recently, mesenchymal stem cells (MSCs) have been the novel focus for cell therapy due to their migration to tumor sites. In this study, the apoptotic effect of MSCs on colorectal cancer cell lines has been aimed. HCT-116 and HT-29 were selected as the colorectal cancer cell lines. Human umbilical cord blood and Wharton's jelly were used as mesenchymal stem cell sources. To discriminate against the apoptotic effect of MSC on cancer, we also used peripheral blood mononuclear cells (PBMC) as a healthy control group. Cord blood-MSC and PBMC were obtained by ficoll-paque density gradient, and Wharton's jelly-MSC by explant method. Transwell co-culture systems were used as cancer cells or PBMC/MSCs at ratios of 1/5 and 1/10, with incubation times of 24 h and 72 h. The Annexin V/PI-FITC-based apoptosis assay was performed by flow cytometry. Caspase-3 and HTRA2/Omi proteins were measured by ELISA. For both ratios in both cancer cells, it was found that the apoptotic effect of Wharton's jelly-MSC was significantly higher in 72-h incubations (p < 0.006), whereas the effect of cord blood mesenchymal stem cell in 24-h incubations were higher (p < 0.007). In this study, we showed that human cord blood and tissue-derived MSCs treatment led to colorectal cancers to apoptosis. We anticipate that further in vivo studies may shed light on the apoptotic effect of MSC.
Collapse
Affiliation(s)
- Figen Abatay-Sel
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey. .,Institute of Graduate Studies in Health Science, Istanbul University, Istanbul, Turkey.
| | - Ayse Erol
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mediha Suleymanoglu
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Gokhan Demirayak
- Department of Gynecologic Oncology, Bakırköy Sadi Konuk Education and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Cigdem Kekik-Cinar
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Durdane Serap Kuruca
- Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Fatma Savran-Oguz
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
17
|
Sousa A, Coelho P, Leite F, Teixeira C, Rocha AC, Santos I, Baylina P, Fernandes R, Soares R, Costa R, Gomes A. Impact of umbilical cord mesenchymal stromal/stem cell secretome and cord blood serum in prostate cancer progression. Hum Cell 2023; 36:1160-1172. [PMID: 36806993 PMCID: PMC10110723 DOI: 10.1007/s13577-023-00880-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023]
Abstract
Prostate cancer (PCa) is the second most common malignancy in men, and the fifth leading cause of death worldwide. Mesenchymal stromal/stem cells (MSC) have been identified in PCa, although contradictory effects in malignant transformation and tumor progression have been described. Since umbilical cord (UC) MSC and cord blood serum (CBS) are rich in numerous growth and anti-inflammatory factors, UC-MSC secretome and CBS are able to modulate tumor cell proliferation and survival as well as immunity and angiogenesis. In the present study, we address this relationship and investigate the influence of UC-MSC secretome and CBS on two human PCa cell lines (PC3 and LNCaP) and a normal epithelial prostate cell line (HPEpiC). Our results disclosed that upon exposure to UC-MSC-conditioned medium or CBS, both PC3 and LNCaP cells exhibited reduced viability, proliferation, and motility while non-malignant epithelial prostate cells were unaffected. These findings were corroborated by expression analysis of AKT/PI3K signaling pathway, p53 and interleukin genes. UC-MSC and CBS factors decreased the expression of growth-stimulating AKT and PI3K effectors and simultaneously up-regulated the expression of tumor-suppressor p53. Moreover, a more anti-inflammatory expression profile was found in both malignant PCa cell lines. Altogether, these results shed light into possible mechanisms by which UC-MSC and CBS reduce PCa progression, further reinforcing their potential use as novel therapeutic agents in PCa.
Collapse
Affiliation(s)
- André Sousa
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Pedro Coelho
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,School of Health (ESS), Polytechnic of Porto, Porto, Portugal
| | - Fernanda Leite
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Clinical Haematology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Catarina Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Ana Catarina Rocha
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Inês Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Pilar Baylina
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal.,School of Health (ESS), Polytechnic of Porto, Porto, Portugal
| | - Ruben Fernandes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal.,FCS - Faculty of Health Sciences, HE-UFP, Hospital Escola - Universidade Fernando Pessoa, Porto, Portugal
| | - Raquel Soares
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Raquel Costa
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,LaBMI - Laboratório de Biotecnologia Médica e Industrial, PORTIC - Porto Research, Technology, and Innovation Center, Polytechnic of Porto, Porto, Portugal
| | - Andreia Gomes
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, Universidade do Porto, Porto, Portugal. .,Bebé Vida, Ciências Para a Vida, S.A, Av. da França 476, 4050-367, Porto, Portugal.
| |
Collapse
|
18
|
Li S, Wu C, Lin S, Wen Z, Luo W, Li C, Wang X, Li X, Gao L, Ding Y. HUCMSC-derived Exosomes Suppress the Titanium Particles-induced Osteolysis in Mice through Inhibiting CCL2 and CCL3. Orthop Surg 2023; 15:888-898. [PMID: 36720704 PMCID: PMC9977603 DOI: 10.1111/os.13608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE Wear particles induce inflammation and the further osteolysis around the prosthesis, has been proven to be the main cause of aseptic hip joint loosening. In this research, we aimed to clarify whether human umbilical cord mesenchymal stem cells (HUCMSCs) could inhibit the titanium particles-induced osteolysis and shed light upon its mechanism. METHODS The expression of chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-C motif) ligand 3 (CCL3) and chemokine (C-C motif) ligand 5 (CCL5) were examinjed in clinical specimens of aseptic hip prosthesis loosening patients. Local injection of lentivirus that knocked down CCL2 or CCL3 in a cranial osteolysis mice model were used to exam the effect of CCL2 and CCL3 on titanium particles-induced osteolysis in vivo. Transwell assay was used to examine the effect of CCL2 and CCL3 on titanium particles-induced activation of macrophage in vitro. Furthermore, the therapeutic effect of HUCMSCs, and exosomes from HUCMSCs were also examed in vivo and vitro. Immunohistochemical and real-time PCR were used to examine the expression of relative pathways. Analysis of variance (ANOVA) and Student-Newman-Keuls post hoc t test were used to analyze the results and determine the statistical significance of the differences. RESULTS Results showed that titanium particles caused the osteolysis at the mice cranial in vivo and a large number of macrophages that migrated, while local injection of HUCMSCs and exosomes did inhibit the cranial osteolysis and migration. An exosome inhibitor GW4869 significantly increased the osteolysis area in the mice cranium osteolysis model, and increased the number of migrated macrophages. Immunohistochemical results suggested that the expression of CCL2, CCL3 and CD68 in the cranial in Titanium particles mice increased significantly, but was significantly reduced by HUCMSCs or exosomes. HUCMSC and exosomes down-regulate the expression of CCL3 in vitro and in vivo. CONCLUSION HUCMSCs and HUCMSC-derived exosomes could suppress the titanium particles-induced osteolysis in mice through inhibiting chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 3.
Collapse
Affiliation(s)
- Shixun Li
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Chuangran Wu
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Sipeng Lin
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Zhenkang Wen
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Wenqiang Luo
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Changchuan Li
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Xiaoyan Wang
- Guangzhou Saliai Stem Cell Science and Technology Co., LTDGuangzhouChina
| | - Xuejia Li
- Guangzhou Saliai Stem Cell Science and Technology Co., LTDGuangzhouChina
| | - Liangbin Gao
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yue Ding
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
19
|
Zhang Y, Lv P, Li Y, Zhang Y, Cheng C, Hao H, Yue H. Inflammatory Cytokine Interleukin-6 (IL-6) Promotes the Proangiogenic Ability of Adipose Stem Cells from Obese Subjects via the IL-6 Signaling Pathway. Curr Stem Cell Res Ther 2023; 18:93-104. [PMID: 36883256 DOI: 10.2174/1574888x17666220429103935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/05/2022] [Accepted: 03/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The prevalence of obesity, as well as obesity-induced chronic inflammatory diseases, is increasing worldwide. Chronic inflammation is related to the complex process of angiogenesis, and we found that adipose-derived stem cells from obese subjects (obADSCs) had proangiogenic features, including higher expression levels of interleukin-6 (IL-6), Notch ligands and receptors, and proangiogenic cytokines, than those from control subjects. We hypothesized that IL-6 and Notch signaling pathways are essential for regulating the proangiogenic characteristics of obADSCs. OBJECTIVE This study aimed to investigate whether the inflammatory cytokine interleukin 6 (IL-6) promotes the proangiogenic capacity of adipose stem cells in obese subjects via the IL-6 signaling pathway. METHODS We compared the phenotype analysis as well as cell doubling time, proliferation, migration, differentiation, and proangiogenic properties of ADSCs in vitro. Moreover, we used small interfering RNAs to inhibit the gene and protein expression of IL-6. RESULTS We found that ADSCs isolated from control individuals (chADSCs) and obADSCs had similar phenotypes and growth characteristics, and chADSCs had a stronger differentiation ability than obADSCs. However, obADSCs were more potent in promoting EA.hy926 cell migration and tube formation than chADSCs in vitro. We confirmed that IL-6 siRNA significantly reduced the transcriptional level of IL-6 in obADSCs, thereby reducing the expression of vascular endothelial growth factor (VEGF)- A, VEGF receptor 2, transforming growth factor β, and Notch ligands and receptors in obADSCs. CONCLUSION The finding suggests that inflammatory cytokine interleukin-6 (IL-6) promotes the proangiogenic ability of obADSCs via the IL-6 signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Pengju Lv
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Yalong Li
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Yonghui Zhang
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Chaofei Cheng
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Hongbo Hao
- Neuroscience Initiative, Advanced Science Research Center at the Graduate Center, City University of New York, New York, 10031, USA
| | - Han Yue
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.,People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| |
Collapse
|
20
|
Tsuji S, Mukai T, Tsuchiya H, Iwatani C, Nakamura A, Nagamura‐Inoue T, Murakami T. Impact of administering umbilical cord-derived mesenchymal stem cells to cynomolgus monkeys with endometriosis. Reprod Med Biol 2023; 22:e12540. [PMID: 37693240 PMCID: PMC10491929 DOI: 10.1002/rmb2.12540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023] Open
Abstract
Purpose This study aimed to explore whether umbilical cord-derived mesenchymal stem cells (UC-MSCs) could be used as a therapeutic resource for endometriosis. Methods Of seven cynomolgus monkeys with endometriosis, five were administered UC-MSCs (intervention group) and two were administered saline (control group). First, intravenous US-MSC treatment was administered for three months. Second, weekly intravenous US-MSC administration combined with monthly intraperitoneal US-MSC administration was conducted for 3 months. Finally, weekly intraperitoneal US-MSC administration was conducted for 3 months. The dose of UC-MSCs was set to 2 × 106 cells/kg for all administration routes. Laparoscopic findings and serum cancer antigen 125 (CA125) levels were also evaluated. The Revised American Society for Reproductive Medicine classification was used for laparoscopic evaluation. Results Laparoscopic findings showed exacerbation of endometriosis after intraperitoneal UC-MSC administration, although no changes were observed in the control group. Intravenous UC-MSC administration decreased the level of CA125 in all monkeys; however, the difference was not significant. Intraperitoneal UC-MSC administration significantly exacerbated endometriosis compared with intravenous administration (p = 0.02). Conclusions This study revealed that intraperitoneal UC-MSC administration exacerbates endometriosis in a nonhuman primate model of the disease.
Collapse
Affiliation(s)
- Shunichiro Tsuji
- Department of Obstetrics and GynecologyShiga University of Medical ScienceOtsuJapan
| | - Takeo Mukai
- Department of PediatricsThe University of Tokyo HospitalBunkyo‐ku, TokyoJapan
| | - Hideaki Tsuchiya
- Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Chizuru Iwatani
- Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Akiko Nakamura
- Department of Obstetrics and GynecologyShiga University of Medical ScienceOtsuJapan
| | - Tokiko Nagamura‐Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical ScienceThe University of TokyoMinato‐ku, TokyoJapan
| | - Takashi Murakami
- Department of Obstetrics and GynecologyShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
21
|
Santilli F, Fabrizi J, Pulcini F, Santacroce C, Sorice M, Delle Monache S, Mattei V. Gangliosides and Their Role in Multilineage Differentiation of Mesenchymal Stem Cells. Biomedicines 2022; 10:biomedicines10123112. [PMID: 36551867 PMCID: PMC9775755 DOI: 10.3390/biomedicines10123112] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/10/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Gangliosides (GGs) are a glycolipid class present on Mesenchymal Stem Cells (MSCs) surfaces with a critical appearance role in stem cell differentiation, even though their mechanistic role in signaling and differentiation remains largely unknown. This review aims to carry out a critical analysis of the predictive role of gangliosides as specific markers of the cellular state of undifferentiated and differentiated MSCs, towards the osteogenic, chondrogenic, neurogenic, and adipogenic lineage. For this reason, we analyzed the role of GGs during multilineage differentiation processes of several types of MSCs such as Umbilical Cord-derived MSCs (UC-MSCs), Bone Marrow-derived MSCs (BM-MSCs), Dental Pulp derived MSCs (DPSCs), and Adipose derived MSCs (ADSCs). Moreover, we examined the possible role of GGs as specific cell surface markers to identify or isolate specific stem cell isotypes and their potential use as additional markers for quality control of cell-based therapies.
Collapse
Affiliation(s)
- Francesca Santilli
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, Angelo Maria Ricci 35A, 02100 Rieti, Italy
| | - Jessica Fabrizi
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, Angelo Maria Ricci 35A, 02100 Rieti, Italy
- Department of Experimental Medicine, Sapienza University, Regina Elena 324, 00161 Rome, Italy
| | - Fanny Pulcini
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Vetoio, 67100 L’Aquila, Italy
| | - Costantino Santacroce
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, Angelo Maria Ricci 35A, 02100 Rieti, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, Sapienza University, Regina Elena 324, 00161 Rome, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Vetoio, 67100 L’Aquila, Italy
- Correspondence: (S.D.M.); (V.M.)
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, Angelo Maria Ricci 35A, 02100 Rieti, Italy
- Correspondence: (S.D.M.); (V.M.)
| |
Collapse
|
22
|
Baranovskii DS, Klabukov ID, Arguchinskaya NV, Yakimova AO, Kisel AA, Yatsenko EM, Ivanov SA, Shegay PV, Kaprin AD. Adverse events, side effects and complications in mesenchymal stromal cell-based therapies. Stem Cell Investig 2022; 9:7. [PMID: 36393919 PMCID: PMC9659480 DOI: 10.21037/sci-2022-025] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/28/2022] [Indexed: 07/22/2023]
Abstract
Numerous clinical studies have shown a wide clinical potential of mesenchymal stromal cells (MSCs) application. However, recent experience has accumulated numerous reports of adverse events and side effects associated with MSCs therapy. Furthermore, the strategies and methods of MSCs therapy did not change significantly in recent decades despite the clinical impact and awareness of potential complications. An extended understanding of limitations could lead to a wider clinical implementation of safe cell therapies and avoid harmful approaches. Therefore, our objective was to summarize the possible negative effects observed during MSCs-based therapies. We were also aimed to discuss the risks caused by weaknesses in cell processing, including isolation, culturing, and storage. Cell processing and cell culture could dramatically influence cell population profile, change protein expression and cell differentiation paving the way for future negative effects. Long-term cell culture led to accumulation of chromosomal abnormalities. Overdosed antibiotics in culture media enhanced the risk of mycoplasma contamination. Clinical trials reported thromboembolism and fibrosis as the most common adverse events of MSCs therapy. Their delayed manifestation generally depends on the patient's individual phenotype and requires specific awareness during the clinical trials with obligatory inclusion in the patient' informed consents. Finally we prepared the safety checklist, recommended for clinical specialists before administration or planning of MSCs therapy.
Collapse
Affiliation(s)
- Denis S. Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Ilya D. Klabukov
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering of the National Research Nuclear University MEPhI, Obninsk, Russia
| | - Nadezhda V. Arguchinskaya
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anna O. Yakimova
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anastas A. Kisel
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Elena M. Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Sergei A. Ivanov
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Peter V. Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
- Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
23
|
Zhao J, Meng H, Liao S, Su Y, Guo L, Wang A, Xu W, Zhou H, Peng J. Therapeutic effect of human umbilical cord mesenchymal stem cells in early traumatic osteonecrosis of the femoral head. J Orthop Translat 2022; 37:126-142. [PMID: 36313533 PMCID: PMC9582590 DOI: 10.1016/j.jot.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Background Osteonecrosis of the femoral head (ONFH) is a refractory disease due to its unclear pathomechanism. Therapies during the early stage of ONFH have not achieved satisfactory results. Therefore, this study aims to explore the available evidence for the therapeutic effect of human umbilical cord mesenchymal stem cells (HUCMSCs) on early-stage traumatic ONFH. Methods Early-stage traumatic ONFH was established. The femoral heads of rats were then locally administered HUCMSCs. Four weeks and eight weeks after surgery, bone repair of the necrotic area in the femoral head was analyzed to evaluate the therapeutic effect of HUCMSCs using micro-CT, histopathological staining, immunofluorescence staining, Luminex. Results HUCMSCs were still present in the femoral head four weeks later, and the morphological, micro-CT and histopathological outcomes in the 4-week HUCMSC-treated group were better than those in the model, NS and 8-week HUCMSC-treated groups. Local transplantation of HUCMSCs promoted bone repair and prevented bone loss in the necrotic area of the femoral head. Conclusions HUCMSCs can survive and positively affect the femoral head through local transplantation in early-stage traumatic ONFH. The conclusions of this study can provide a treatment option for patients who have ONFH and can serve as basic research on the advanced development of this disease. The Translational potential of this article The study indicated that the positive effect of exogenous HUCMSCs in the treatment of early-stage traumatic ONFH provides the solid basis and guidance for the clinical application of HUCMSCs.
Collapse
Affiliation(s)
- Jun Zhao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Haoye Meng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Sida Liao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yaoyu Su
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Li Guo
- The Eight Medical Center of PLA General Hospital, China
| | - Aiyuan Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Wenjing Xu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Hao Zhou
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China,Corresponding author.
| |
Collapse
|
24
|
Silini AR, Ramuta TŽ, Pires AS, Banerjee A, Dubus M, Gindraux F, Kerdjoudj H, Maciulatis J, Weidinger A, Wolbank S, Eissner G, Giebel B, Pozzobon M, Parolini O, Kreft ME. Methods and criteria for validating the multimodal functions of perinatal derivatives when used in oncological and antimicrobial applications. Front Bioeng Biotechnol 2022; 10:958669. [PMID: 36312547 PMCID: PMC9607958 DOI: 10.3389/fbioe.2022.958669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
Perinatal derivatives or PnDs refer to tissues, cells and secretomes from perinatal, or birth-associated tissues. In the past 2 decades PnDs have been highly investigated for their multimodal mechanisms of action that have been exploited in various disease settings, including in different cancers and infections. Indeed, there is growing evidence that PnDs possess anticancer and antimicrobial activities, but an urgent issue that needs to be addressed is the reproducible evaluation of efficacy, both in vitro and in vivo. Herein we present the most commonly used functional assays for the assessment of antitumor and antimicrobial properties of PnDs, and we discuss their advantages and disadvantages in assessing the functionality. This review is part of a quadrinomial series on functional assays for the validation of PnDs spanning biological functions such as immunomodulation, anticancer and antimicrobial, wound healing, and regeneration.
Collapse
Affiliation(s)
- Antonietta R. Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Taja Železnik Ramuta
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Ana Salomé Pires
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment, Genetics and Oncobiology (CIMAGO), Institute of Biophysics, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Asmita Banerjee
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Marie Dubus
- Université de Reims Champagne Ardenne, EA 4691 Biomatériaux et Inflammation en Site Osseux (BIOS), Reims, France
| | - Florelle Gindraux
- Service de Chirurgie Orthopédique, Traumatologique et Plastique, CHU Besançon and Laboratoire de Nanomédecine, Imagerie, Thérapeutique EA 4662, Université Bourgogne Franche-Comté, Besançon, France
| | - Halima Kerdjoudj
- Université de Reims Champagne Ardenne, EA 4691 Biomatériaux et Inflammation en Site Osseux (BIOS), Reims, France
| | - Justinas Maciulatis
- The Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Günther Eissner
- Systems Biology Ireland, UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Lab, Department of Women’s and Children’s Health, University of Padova, Fondazione Istituto di Ricerca Pediatrica Città Della Speranza, Padoa, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Mateja Erdani Kreft
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Mateja Erdani Kreft,
| |
Collapse
|
25
|
Hoang VT, Nguyen HP, Nguyen VN, Hoang DM, Nguyen TST, Nguyen Thanh L. “Adipose-derived mesenchymal stem cell therapy for the management of female sexual dysfunction: Literature reviews and study design of a clinical trial”. Front Cell Dev Biol 2022; 10:956274. [PMID: 36247008 PMCID: PMC9554747 DOI: 10.3389/fcell.2022.956274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Hormone imbalance and female sexual dysfunction immensely affect perimenopausal female health and quality of life. Hormone therapy can improve female hormone deficiency, but long-term use increases the risk of cardiovascular diseases and cancer. Therefore, it is necessary to develop a novel effective treatment to achieve long-term improvement in female general and sexual health. This study reviewed factors affecting syndromes of female sexual dysfunction and its current therapy options. Next, the authors introduced research data on mesenchymal stromal cell/mesenchymal stem cell (MSC) therapy to treat female reproductive diseases, including Asherman’s syndrome, premature ovarian failure/primary ovarian insufficiency, and vaginal atrophy. Among adult tissue-derived MSCs, adipose tissue-derived stem cells (ASCs) have emerged as the most potent therapeutic cell therapy due to their abundant presence in the stromal vascular fraction of fat, high proliferation capacity, superior immunomodulation, and strong secretion profile of regenerative factors. Potential mechanisms and side effects of ASCs for the treatment of female sexual dysfunction will be discussed. Our phase I clinical trial has demonstrated the safety of autologous ASC therapy for women and men with sexual hormone deficiency. We designed the first randomized controlled crossover phase II trial to investigate the safety and efficacy of autologous ASCs to treat female sexual dysfunction in perimenopausal women. Here, we introduce the rationale, trial design, and methodology of this clinical study. Because aging and metabolic diseases negatively impact the bioactivity of adult-derived MSCs, this study will use ASCs cultured in physiological oxygen tension (5%) to cope with these challenges. A total of 130 perimenopausal women with sexual dysfunction will receive two intravenous infusions of autologous ASCs in a crossover design. The aims of the proposed study are to evaluate 1) the safety of cell infusion based on the frequency and severity of adverse events/serious adverse events during infusion and follow-up and 2) improvements in female sexual function assessed by the Female Sexual Function Index (FSFI), the Utian Quality of Life Scale (UQOL), and the levels of follicle-stimulating hormone (FSH) and estradiol. In addition, cellular aging biomarkers, including plasminogen activator inhibitor-1 (PAI-1), p16 and p21 expression in T cells and the inflammatory cytokine profile, will also be characterized. Overall, this study will provide essential insights into the effects and potential mechanisms of ASC therapy for perimenopausal women with sexual dysfunction. It also suggests direction and design strategies for future research.
Collapse
Affiliation(s)
- Van T. Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hanoi, Vietnam
| | - Hoang-Phuong Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hanoi, Vietnam
| | - Viet Nhan Nguyen
- Vinmec International Hospital—Times City, Vinmec Health Care System, Hanoi, Vietnam
- College of Health Science, Vin University, Vinhomes Ocean Park, Hanoi, Vietnam
| | - Duc M. Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hanoi, Vietnam
| | - Tan-Sinh Thi Nguyen
- Vinmec International Hospital—Times City, Vinmec Health Care System, Hanoi, Vietnam
| | - Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hanoi, Vietnam
- Vinmec International Hospital—Times City, Vinmec Health Care System, Hanoi, Vietnam
- College of Health Science, Vin University, Vinhomes Ocean Park, Hanoi, Vietnam
- *Correspondence: Liem Nguyen Thanh,
| |
Collapse
|
26
|
Li T, Zhou L, Fan M, Chen Z, Yan L, Lu H, Jia M, Wu H, Shan L. Human Umbilical Cord-Derived Mesenchymal Stem Cells Ameliorate Skin Aging of Nude Mice Through Autophagy-Mediated Anti-Senescent Mechanism. Stem Cell Rev Rep 2022; 18:2088-2103. [PMID: 35864432 DOI: 10.1007/s12015-022-10418-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
Skin aging is a currently irreversible process, affected by increased oxidative stress, activated cellular senescence, and lacked regeneration of the dermal layer. Mesenchymal stem cells (MSCs), such as human umbilical cord-derived MSCs (hucMSCs), have pro-regeneration and anti-aging potencies. To explore whether hucMSCs can be used to treat skin aging, this study employed skin-aging model of nude mice to conduct in vivo assays, including biochemical analysis of superoxide dismutase (SOD) and malondialdehyde (MDA), gross observation, histopathological observation, and immunohistochemical analysis. To clarify how hucMSCs work on skin aging, this study employed skin-aging model of human dermal fibroblasts (HDFs) to conduct in vitro assays by applying conditional medium of hucMSCs (CMM), including wound healing assay, senescence staining, flow cytometric oxidative detection, real time PCR, and western blot analysis. The in vivo data demonstrated that hucMSCs dose-dependently removed wrinkles, smoothed skin texture, and increased dermal thickness and collagen production of aged skin by reversing SOD and MDA levels and up-regulating Col-1 and VEGF expressions, indicating anti-oxidative and pro-regenerative effects against skin aging. The in vitro data revealed that hucMSCs significantly reversed the senescence of HDFs by promoting cell migration, inhibiting ROS production, and restoring the overexpressions of oxidative and senescent markers through paracrine mode of action, and the paracrine mechanism was mediated by the inhibition of autophagy. This study provided novel knowledge regarding the anti-aging efficacy and paracrine mechanism of hucMSCs on skin, making hucMSCs-based therapy a promising regime for skin aging treatment.
Collapse
Affiliation(s)
- Ting Li
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengqiang Fan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China
| | - Haishan Lu
- Department of Dermatology, PLA 903 Hospital, Hangzhou, China
| | - Ming Jia
- Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiling Wu
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China. .,Department of Plastic and Aesthetic Center, The First Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China. .,Cell Resource Bank and Integrated Cell Preparation Center of Xiaoshan District, Hangzhou Regional Cell Preparation Center (Shangyu Biotechnology Co., Ltd), Hangzhou, China.
| |
Collapse
|
27
|
Moonshi SS, Adelnia H, Wu Y, Ta HT. Placenta‐Derived Mesenchymal Stem Cells for Treatment of Diseases: A Clinically Relevant Source. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202200054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Shehzahdi S. Moonshi
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hossein Adelnia
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| | - Yuao Wu
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
| | - Hang T. Ta
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland 4111 Australia
- Bioscience Discipline School of Environment and Science Griffith University Nathan Queensland 4111 Australia
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St Lucia Queensland 4072 Australia
| |
Collapse
|
28
|
Naeem A, Gupta N, Naeem U, Khan MJ, Elrayess MA, Cui W, Albanese C. A comparison of isolation and culture protocols for human amniotic mesenchymal stem cells. Cell Cycle 2022; 21:1543-1556. [PMID: 35412950 PMCID: PMC9291641 DOI: 10.1080/15384101.2022.2060641] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The successful translation of mesenchymal stem cells (MSCs) from bench to bedside is predicated upon their regenerative capabilities and immunomodulatory potential. Many challenges still exist in making MSCs a viable and cost-effective therapeutic option, due in part to the challenges of sourcing MSCs from adult tissues and inconsistencies in the characterization of MSCs. In many cases, adult MSC collection is an invasive procedure, and ethical concerns and age-related heterogeneity further complicate obtaining adult tissue derived MSCs at the scales needed for clinical applications. Alternative adult sources, such as post-partum associated tissues, offer distinct advantages to overcome these challenges. However, successful therapeutic applications rely on the efficient ex-vivo expansion of the stem cells while avoiding any culture-related phenotypic alterations, which requires optimized and standardized isolation, culture, and cell preservation methods. In this review, we have compared the isolation and culture methods for MSCs originating from the human amniotic membrane (hAMSCs) of the placenta to identify the elements that support the extended subculture potential of hAMSCs without compromising their immune-privileged, pluripotent regenerative potential.Abbreviations:AM: Human amniotic membrane; ASCs: Adipose tissue-derived stem cells; BM-MSCs: Bone marrow-mesenchymal stem cells; DMEM: Dulbecco's modified eagle medium; DT: Doubling time; EMEM: Eagle's modified essential medium; ESCM: Embryonic stem cell markers; ESCs: Embryonic stem cells; hAECs: Human amniotic epithelial cells; hAMSCs: Human amniotic mesenchymal stem cells; HLA: Human leukocyte antigen; HM: Hematopoietic markers; IM: Immunogenicity markers; MHC: Major histocompatibility complex; MSCs: Mesenchymal stem cells; MCSM: Mesenchymal cell surface markers; Nanog: NANOG homeobox; Oct: Octamer binding transcription factor 4; P: Passage; PM: Pluripotency markers; STRO-1: Stromal precursor antigen-1; SCP: Subculture potential; Sox-2: Sry-related HMG box gene 2; SSEA-4: Stage-specific embryonic antigen; TRA: Tumor rejection antigen.
Collapse
Affiliation(s)
- Aisha Naeem
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Health Research Governance Department, Ministry of Public Health, Qatar
| | - Nikita Gupta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Usra Naeem
- Department of Health Professional Technology, University of Lahore, Pakistan
| | | | - Mohamed A Elrayess
- Omics, Biomedical Research Center, Qatar University, Doha, Qatar.,Research and Graduate Studies, College of Pharmacy, Qu Health, Qatar University, Doha, Qatar
| | - Wanxing Cui
- Cell Therapy Manufacturing Facility, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Chris Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Department of Radiology, Georgetown University Medical Center, Washington, DC, USA.,Center for Translational Imaging, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
29
|
Xue J, Gao J, Gu Y, Wang A, Yu S, Li B, Yin Y, Wang J, Su W, Zhang H, Ren W, Gu W, Lv Z, Mu Y, Cheng Y. Human umbilical cord-derived mesenchymal stem cells alleviate insulin resistance in diet-induced obese mice via an interaction with splenocytes. Stem Cell Res Ther 2022; 13:109. [PMID: 35313972 PMCID: PMC8935757 DOI: 10.1186/s13287-022-02791-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/28/2022] [Indexed: 11/22/2022] Open
Abstract
Background Previous research has demonstrated that the spleen plays an important role in mesenchymal stem cell (MSC)-mediated alleviation of acute inflammation, as MSC infusion increases the spleen-derived anti-inflammatory cytokine interleukin 10 (IL-10) levels. However, studies on splenic involvement in MSC-induced protection against chronic inflammatory diseases are limited. Obesity is characterized by chronic low-grade inflammation, a key driver of insulin resistance. This study aims to evaluate the effects of MSCs on obesity-related insulin resistance and explore the underlying mechanism, particularly regarding splenic involvement.
Methods We induced obesity in mice by feeding them high-fat diets for 20 weeks. Human umbilical cord-derived MSCs (UC-MSCs) were systemically infused into the obese mice once per week for 6 weeks. Systemic glucose metabolic homeostasis and insulin sensitivity in epididymal adipose tissue (EAT) were evaluated. Then, we conducted in vivo blockade of IL-10 during UC-MSC infusion by intraperitoneally administrating an IL-10-neutralizing antibody twice per week. We also investigated the therapeutic effects of UC-MSCs on obese mice after removal of the spleen by splenectomy. Results UC-MSC infusions improved systemic metabolic homeostasis and alleviated insulin resistance in EAT but elicited no change in weight. Despite rare engraftment of UC-MSCs in EAT, UC-MSC infusions attenuated insulin resistance in EAT by polarizing macrophages into the M2 phenotype, coupled with elevated serum IL-10 levels. In vivo blockade of IL-10 blunted the effects of UC-MSCs on obese mice. Furthermore, UC-MSCs overwhelmingly homed to the spleen, and the ability of UC-MSCs to elevate serum IL-10 levels and alleviate insulin resistance was impaired in the absence of the spleen. Further in vivo and in vitro studies revealed that UC-MSCs promoted the capacity of regulatory T cells (Treg cells) to produce IL-10 in the spleen. Conclusions Our results demonstrated that UC-MSCs elevated serum IL-10 levels and subsequently promoted macrophage polarization, leading to alleviation of insulin resistance in EAT. The underlying mechanism was that UC-MSCs improved the capacity of Treg cells to produce IL-10 in the spleen. Our findings indicated that the spleen played a critical role in amplifying MSC-mediated immunomodulatory effects, which may contribute to maximizing MSC efficacy in clinical applications in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02791-6.
Collapse
Affiliation(s)
- Jing Xue
- Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,Department of Endocrinology, Diabetes Center of People's Liberation Army (PLA), PLA Strategic Support Force Characteristic Medical Center (The 306th Hospital of PLA), Beijing, China
| | - Jieqing Gao
- Department of Endocrinology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Yulin Gu
- Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Aihong Wang
- Department of Endocrinology, Diabetes Center of People's Liberation Army (PLA), PLA Strategic Support Force Characteristic Medical Center (The 306th Hospital of PLA), Beijing, China
| | - Songyan Yu
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bing Li
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jie Wang
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Wanlu Su
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Haixia Zhang
- Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Weizheng Ren
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Weijun Gu
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhaohui Lv
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yiming Mu
- Medical School of Chinese PLA, Beijing, China. .,Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Yu Cheng
- Department of Endocrinology, The First Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
30
|
Strategies to enhance immunomodulatory properties and reduce heterogeneity in mesenchymal stromal cells during ex vivo expansion. Cytotherapy 2022; 24:456-472. [DOI: 10.1016/j.jcyt.2021.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
|
31
|
Wang Z, Li H, Fang J, Wang X, Dai S, Cao W, Guo Y, Li Z, Zhu H. Comparative Analysis of the Therapeutic Effects of Amniotic Membrane and Umbilical Cord Derived Mesenchymal Stem Cells for the Treatment of Type 2 Diabetes. Stem Cell Rev Rep 2022; 18:1193-1206. [PMID: 35015214 PMCID: PMC8749914 DOI: 10.1007/s12015-021-10320-w] [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] [Accepted: 12/16/2021] [Indexed: 11/09/2022]
Abstract
Type 2 diabetes mellitus (T2DM), one of the most common carbohydrate metabolism disorders, is characterized by chronic hyperglycemia and insulin resistance (IR), and has become an urgent global health challenge. Mesenchymal stem cells (MSCs) originating from perinatal tissues such as umbilical cord (UC) and amniotic membrane (AM) serve as ideal candidates for the treatment of T2DM due to their great advantages in terms of abundant source, proliferation capacity, immunomodulation and plasticity for insulin-producing cell differentiation. However, the optimally perinatal MSC source to treat T2DM remains elusive. This study aims to compare the therapeutic efficacy of MSCs derived from AM and UC (AMMSCs and UCMSCs) of the same donor in the alleviation of T2DM symptoms and explore the underlying mechanisms. Our results showed that AMMSCs and UCMSCs displayed indistinguishable immunophenotype and multi-lineage differentiation potential, but UCMSCs had a much higher expansion capacity than AMMSCs. Moreover, we uncovered that single-dose intravenous injection of either AMMSCs or UCMSCs could comparably reduce hyperglycemia and improve IR in T2DM db/db mice. Mechanistic investigations revealed that either AMMSC or UCMSC infusion could greatly improve glycolipid metabolism in the liver of db/db mice, which was evidenced by decreased liver to body weight ratio, reduced lipid accumulation, upregulated glycogen synthesis, and increased Akt phosphorylation. Taken together, these data indicate that the same donor-derived AMMSCs and UCMSCs possessed comparable effects and shared a similar hepatoprotective mechanism on the alleviation of T2DM symptoms.
Collapse
Affiliation(s)
- Zhifeng Wang
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China. .,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Haisen Li
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China
| | - Jingmeng Fang
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China
| | - Xiaoyu Wang
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China
| | - Shuhang Dai
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China
| | - Wei Cao
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China
| | - Yinhong Guo
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China
| | - Zhe Li
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China
| | - Hao Zhu
- Sinoneural Cell Engineering Group Holdings Co., Ltd, Shanghai, 201100, China.
| |
Collapse
|
32
|
Dymowska M, Aksamit A, Zielniok K, Kniotek M, Kaleta B, Roszczyk A, Zych M, Dabrowski F, Paczek L, Burdzinska A. Interaction between Macrophages and Human Mesenchymal Stromal Cells Derived from Bone Marrow and Wharton's Jelly-A Comparative Study. Pharmaceutics 2021; 13:pharmaceutics13111822. [PMID: 34834238 PMCID: PMC8624657 DOI: 10.3390/pharmaceutics13111822] [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: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 01/15/2023] Open
Abstract
Despite intensive clinical research on the use of mesenchymal stromal cells (MSCs), further basic research in this field is still required. Herein, we compared human bone marrow MSCs (BM-MSCs, n = 6) and Wharton’s jelly MSCs (WJ-MSCs, n = 6) in their ability to interact with human primary macrophages. Evaluation of secretory potential revealed that under pro-inflammatory stimulation, WJ-MSCs secreted significantly more IL-6 than BM-MSCs (2-fold). This difference did not translate into the effect of MSCs on macrophages: both types of MSCs significantly directed M1-like macrophages toward the M2 phenotype (based on CD206 expression) to a similar extent. This observation was consistent both in flow cytometry analysis and immunocytochemical assessment. The effect of MSCs on macrophages was sustained when IL-6 signaling was blocked with Tocilizumab. Macrophages, regardless of polarization status, enhanced chemotaxis of both BM-MSCs and WJ-MSCs (p < 0.01; trans-well assay), with WJ-MSCs being significantly more responsive to M1-derived chemotactic signals than BM-MSCs. Furthermore, WJ-MSCs increased their motility (scratch assay) when exposed to macrophage-conditioned medium while BM-MSCs did not. These results indicate that although both BM-MSCs and WJ-MSCs have the ability to reciprocally interact with macrophages, the source of MSCs could slightly but significantly modify the response under clinical settings.
Collapse
Affiliation(s)
- Marta Dymowska
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.D.); (A.A.); (L.P.)
- Laboratory of Cell Research and Application, Medical University of Warsaw, Banacha 1B, 02-097 Warsaw, Poland
| | - Aleksandra Aksamit
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.D.); (A.A.); (L.P.)
| | - Katarzyna Zielniok
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.D.); (A.A.); (L.P.)
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.K.); (B.K.); (A.R.); (M.Z.)
- Correspondence: (K.Z.); (A.B.)
| | - Monika Kniotek
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.K.); (B.K.); (A.R.); (M.Z.)
| | - Beata Kaleta
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.K.); (B.K.); (A.R.); (M.Z.)
| | - Aleksander Roszczyk
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.K.); (B.K.); (A.R.); (M.Z.)
| | - Michal Zych
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.K.); (B.K.); (A.R.); (M.Z.)
| | - Filip Dabrowski
- Department of Gynecology and Obstetrics, Medical University of Silesia, Medykow 14, 40-752 Katowice, Poland;
| | - Leszek Paczek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.D.); (A.A.); (L.P.)
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106 Warsaw, Poland
| | - Anna Burdzinska
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.D.); (A.A.); (L.P.)
- Correspondence: (K.Z.); (A.B.)
| |
Collapse
|
33
|
Ahani-Nahayati M, Niazi V, Moradi A, Pourjabbar B, Roozafzoon R, Baradaran-Rafii A, Keshel SH. Umbilical cord mesenchymal stem/stromal cells potential to treat organ disorders; an emerging strategy. Curr Stem Cell Res Ther 2021; 17:126-146. [PMID: 34493190 DOI: 10.2174/1574888x16666210907164046] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022]
Abstract
Currently, mesenchymal stem/stromal cells (MSCs) have attracted growing attention in the context of cell-based therapy in regenerative medicine. Following the first successful procurement of human MSCs from bone marrow (BM), these cells isolation has been conducted from various origins, in particular, the umbilical cord (UC). Umbilical cord-derived mesenchymal stem/stromal cells (UC-MSCs) can be acquired by a non-invasive plan and simply cultured, and thereby signifies their superiority over MSCs derived from other sources for medical purposes. Due to their unique attributes, including self-renewal, multipotency, and accessibility concomitant with their immunosuppressive competence and lower ethical concerns, UC-MSCs therapy is described as encouraging therapeutic options in cell-based therapies. Regardless of their unique aptitude to adjust inflammatory response during tissue recovery and delivering solid milieu for tissue restoration, UC-MSCs can be differentiated into a diverse spectrum of adult cells (e.g., osteoblast, chondrocyte, type II alveolar, hepatocyte, and cardiomyocyte). Interestingly, they demonstrate a prolonged survival and longer telomeres compared with MSCs derived from other sources, suggesting that UC-MSCs are desired source to use in regenerative medicine. In the present review, we deliver a brief review of UC-MSCs isolation, expansion concomitantly with immunosuppressive activities, and try to collect and discuss recent pre-clinical and clinical researches based on the use of UC-MSCs in regenerative medicine, focusing on with special focus on in vivo researches.
Collapse
Affiliation(s)
- Milad Ahani-Nahayati
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | - Alireza Moradi
- Department of Physiology, School of Medicine, Iran University of Medical Science, Tehran. Iran
| | - Bahareh Pourjabbar
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | - Reza Roozafzoon
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | | | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| |
Collapse
|
34
|
Labunets IF, Utko NA, Toporova OK. Effects of Multipotent Mesenchymal Stromal Cells of the Human Umbilical Cord and Their Combination with Melatonin in Adult and Aging Mice with a Toxic Cuprizone Model of Demyelination. ADVANCES IN GERONTOLOGY 2021. [DOI: 10.1134/s2079057021020077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
35
|
Human Umbilical Cord-Derived Mesenchymal Stem Cells Promote Corneal Epithelial Repair In Vitro. Cells 2021; 10:cells10051254. [PMID: 34069578 PMCID: PMC8160941 DOI: 10.3390/cells10051254] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/08/2021] [Accepted: 05/14/2021] [Indexed: 12/31/2022] Open
Abstract
Corneal injuries are among the leading causes of blindness and vision impairment. Trauma, infectious keratitis, thermal and chemical (acids and alkali burn) injuries may lead to irreversible corneal scarring, neovascularization, conjunctivalization, and limbal stem cell deficiency. Bilateral blindness constitutes 12% of total global blindness and corneal transplantation remains a stand-alone treatment modality for the majority of end-stage corneal diseases. However, global shortage of donor corneas, the potential risk of graft rejection, and severe side effects arising from long-term use of immunosuppressive medications, demands alternative therapeutic approaches. Umbilical cord-derived mesenchymal stem cells can be isolated in large numbers using a relatively less invasive procedure. However, their role in injury induced corneal repair is largely unexplored. Here, we isolated, cultured and characterized mesenchymal stem cells from human umbilical cord, and studied the expression of mesenchymal (CD73, CD90, CD105, and CD34), ocular surface and epithelial (PAX6, WNT7A, and CK-8/18) lineage markers through immunofluorescence. The cultured human limbal and corneal epithelial cells were used as controls. Scratch assay was used to study the corneal epithelial repair potential of umbilical cord-derived mesenchymal stem cells, in vitro. The in vitro cultured umbilical cord-derived mesenchymal stem cells were plastic adherent, showed trilineage differentiation and expressed: mesenchymal markers CD90, CD105, CD73; epithelial marker CK-8/18, and ocular lineage developmental markers PAX6 and WNT-7A. Our findings suggest that umbilical cord-derived mesenchymal stem cells promote repair of the injured corneal epithelium by stimulating the proliferation of corneal epithelial cells, in vitro. They may serve as a potential non-ocular source of stem cells for treating injury induced bilateral corneal diseases.
Collapse
|
36
|
Shang Y, Guan H, Zhou F. Biological Characteristics of Umbilical Cord Mesenchymal Stem Cells and Its Therapeutic Potential for Hematological Disorders. Front Cell Dev Biol 2021; 9:570179. [PMID: 34012958 PMCID: PMC8126649 DOI: 10.3389/fcell.2021.570179] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 04/08/2021] [Indexed: 01/14/2023] Open
Abstract
Umbilical cord mesenchymal stem cells (UC-MSCs) are a class of multifunctional stem cells isolated and cultured from umbilical cord. They possessed the characteristics of highly self-renewal, multi-directional differentiation potential and low immunogenicity. Its application in the field of tissue engineering and gene therapy has achieved a series of results. Recent studies have confirmed their characteristics of inhibiting tumor cell proliferation and migration to nest of cancer. The ability of UC-MSCs to support hematopoietic microenvironment and suppress immune system suggests that they can improve engraftment after hematopoietic stem cell transplantation, which shows great potential in treatment of hematologic diseases. This review will focus on the latest advances in biological characteristics and mechanism of UC-MSCs in treatment of hematological diseases.
Collapse
Affiliation(s)
- Yufeng Shang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Haotong Guan
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
37
|
Fang Z, Chen P, Tang S, Chen A, Zhang C, Peng G, Li M, Chen X. Will mesenchymal stem cells be future directions for treating radiation-induced skin injury? Stem Cell Res Ther 2021; 12:179. [PMID: 33712078 PMCID: PMC7952822 DOI: 10.1186/s13287-021-02261-5] [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: 01/20/2021] [Accepted: 03/01/2021] [Indexed: 01/09/2023] Open
Abstract
Radiation-induced skin injury (RISI) is one of the common serious side effects of radiotherapy (RT) for patients with malignant tumors. Mesenchymal stem cells (MSCs) are applied to RISI repair in some clinical cases series except some traditional options. Though direct replacement of damaged cells may be achieved through differentiation capacity of MSCs, more recent data indicate that various cytokines and chemokines secreted by MSCs are involved in synergetic therapy of RISI by anti-inflammatory, immunomodulation, antioxidant, revascularization, and anti-apoptotic activity. In this paper, we not only discussed different sources of MSCs on the treatment of RISI both in preclinical studies and clinical trials, but also summarized the applications and mechanisms of MSCs in other related regenerative fields.
Collapse
Affiliation(s)
- Zhuoqun Fang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Penghong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Shijie Tang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Aizhen Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Chaoyu Zhang
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Guohao Peng
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Ming Li
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xiaosong Chen
- Department of Plastic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, People's Republic of China.
| |
Collapse
|
38
|
Fu YX, Ji J, Shan F, Li J, Hu R. Human mesenchymal stem cell treatment of premature ovarian failure: new challenges and opportunities. Stem Cell Res Ther 2021; 12:161. [PMID: 33658073 PMCID: PMC7931610 DOI: 10.1186/s13287-021-02212-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Premature ovarian failure (POF) is one of the common disorders found in women leading to 1% female infertility. Clinical features of POF are hypoestrogenism or estrogen deficiency, increased gonadotropin level, and, most importantly, amenorrhea. With the development of regenerative medicine, human mesenchymal stem cell (hMSC) therapy brings new prospects for POF. This study aimed to describe the types of MSCs currently available for POF therapy, their biological characteristics, and their mechanism of action. It reviewed the latest findings on POF to provide the theoretical basis for further investigation and clinical therapy.
Collapse
Affiliation(s)
- Yun-Xing Fu
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jing Ji
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Fang Shan
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jialing Li
- Ningxia Medical University, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Rong Hu
- Reproductive Medicine Center, General Hospital of Ningxia Medical University, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
39
|
Mesenchymal Stem Cells for Mitigating Radiotherapy Side Effects. Cells 2021; 10:cells10020294. [PMID: 33535574 PMCID: PMC7912747 DOI: 10.3390/cells10020294] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/19/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy for cancers also damages healthy cells and causes side effects. Depending on the dosage and exposure region, radiotherapy may induce severe and irreversible injuries to various tissues or organs, especially the skin, intestine, brain, lung, liver, and heart. Therefore, promising treatment strategies to mitigate radiation injury is in pressing need. Recently, stem cell-based therapy generates great attention in clinical care. Among these, mesenchymal stem cells are extensively applied because it is easy to access and capable of mesodermal differentiation, immunomodulation, and paracrine secretion. Here, we summarize the current attempts and discuss the future perspectives about mesenchymal stem cells (MSCs) for mitigating radiotherapy side effects.
Collapse
|
40
|
Meng X, Gao X, Chen X, Yu J. Umbilical cord-derived mesenchymal stem cells exert anti-fibrotic action on hypertrophic scar-derived fibroblasts in co-culture by inhibiting the activation of the TGF β1/Smad3 pathway. Exp Ther Med 2021; 21:210. [PMID: 33574910 PMCID: PMC7818529 DOI: 10.3892/etm.2021.9642] [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: 01/30/2019] [Accepted: 08/23/2019] [Indexed: 12/27/2022] Open
Abstract
A hypertrophic scar (HS) is a severe fibrotic skin disease that causes disfigurement and deformity. It occurs after deep cutaneous injury and presents a major clinical challenge. The present study aimed to evaluate the effects of umbilical cord-derived mesenchymal stem cells (UCMSCs) on hypertrophic scar fibroblasts (HSFs), one of the main effector cells for HS formation, in a co-culture system and to investigate the potential underlying molecular mechanism. Cultured HSFs were divided into control and co-culture groups. The proliferation ability of HSFs was evaluated using cell counting kit-8 and the percentage of Ki67-positive fibroblasts was assessed by immunofluorescence. The apoptosis of HSFs was determined using a TUNEL assay and by assessing the expression of capase-3 via western blotting. A scratch wound healing assay was employed to examine the migration of HSFs. The expression levels of HS-associated genes (collagen type Iα 2 chain, collagen type IIIα 1 chain and actin α 2 smooth muscle) and proteins (collagen I, collagen III and α-smooth muscle actin) were measured by reverse transcription-quantitative PCR (RT-qPCR) and western blotting, respectively, to assess the pro-fibrotic phenotype of HSFs. The modulation of the transforming growth factor β1 (TGF β1)/Smad3 pathway in HSFs was evaluated by measuring the protein levels of TGF β1, Smad3 and phosphorylated Smad3 using western blotting, and the mRNA levels of TGFβ1 and several other target genes (cellular communication network factor 2, metalloproteinase inhibitor 1 and periostin) were measured by RT-qPCR. The proliferative and migratory ability of co-cultured HSFs was suppressed compared with controls, and no significant difference in apoptosis was observed between the two groups. The pro-fibrotic phenotype of co-cultured HSFs was inhibited due to a decline in expression levels of HS-associated genes and proteins. Furthermore, co-culture with UCMSCs inhibited the activation of the TGF β1/Smad3 pathway. In conclusion, the present study indicated that UCMSCs may exert an anti-fibrotic action on HSFs in co-culture through inhibition of the TGF β1/Smad3 pathway, which suggests a potential use for UCMSCs in HS therapy.
Collapse
Affiliation(s)
- Xianglong Meng
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Xinxin Gao
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Xinxin Chen
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Jiaao Yu
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
41
|
Galindo S, de la Mata A, López-Paniagua M, Herreras JM, Pérez I, Calonge M, Nieto-Miguel T. Subconjunctival injection of mesenchymal stem cells for corneal failure due to limbal stem cell deficiency: state of the art. Stem Cell Res Ther 2021; 12:60. [PMID: 33441175 PMCID: PMC7805216 DOI: 10.1186/s13287-020-02129-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have unique and beneficial properties and are currently used to treat a broad variety of diseases. These properties include the potential for differentiation into other cell types, secretion of different trophic factors that promote a regenerative microenvironment, anti-inflammatory actions, selective migration to damaged tissues, and non-immunogenicity. MSCs are effective for the treatment of ocular surface diseases such as dry eye, corneal burns, and limbal stem cell deficiency (LSCD), both in experimental models and in humans. LSCD is a pathological condition in which damage occurs to the limbal epithelial stem cells, or their niche, that are responsible for the continuous regeneration of the corneal epithelium. If LSCD is extensive and/or severe, it usually causes corneal epithelial defects, ulceration, and conjunctival overgrowth of the cornea. These changes can result in neovascularization and corneal opacity, severe inflammation, pain, and visual loss. The effectiveness of MSCs to reduce corneal opacity, neovascularization, and inflammation has been widely studied in different experimental models of LSCD and in some clinical trials; however, the methodological disparity used in the different studies makes it hard to compare outcomes among them. In this regard, the MSC route of administration used to treat LSCD and other ocular surface diseases is an important factor. It should be efficient, minimally invasive, and safe. So far, intravenous and intraperitoneal injections, topical administration, and MSC transplantation using carrier substrata like amniotic membrane (AM), fibrin, or synthetic biopolymers have been the most commonly used administration routes in experimental models. However, systemic administration carries the risk of potential side effects and transplantation requires surgical procedures that could complicate the process. Alternatively, subconjunctival injection is a minimally invasive and straightforward technique frequently used in ophthalmology. It enables performance of local treatments using high cell doses. In this review, we provide an overview of the current status of MSC administration by subconjunctival injection, analyzing the convenience, safety, and efficacy for treatment of corneal failure due to LSCD in different experimental models. We also provide a summary of the clinical trials that have been completed, are in progress, or being planned.
Collapse
Affiliation(s)
- Sara Galindo
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Edificio IOBA, Campus Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
| | - Ana de la Mata
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Edificio IOBA, Campus Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain. .,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.
| | - Marina López-Paniagua
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Edificio IOBA, Campus Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
| | - Jose M Herreras
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Edificio IOBA, Campus Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
| | - Inmaculada Pérez
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Edificio IOBA, Campus Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
| | - Margarita Calonge
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Edificio IOBA, Campus Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain
| | - Teresa Nieto-Miguel
- Instituto de Oftalmobiología Aplicada (IOBA), Universidad de Valladolid, Edificio IOBA, Campus Miguel Delibes, Paseo de Belén 17, 47011, Valladolid, Spain. .,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.
| |
Collapse
|
42
|
Xie Q, Liu R, Jiang J, Peng J, Yang C, Zhang W, Wang S, Song J. What is the impact of human umbilical cord mesenchymal stem cell transplantation on clinical treatment? Stem Cell Res Ther 2020; 11:519. [PMID: 33261658 PMCID: PMC7705855 DOI: 10.1186/s13287-020-02011-z] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Background Human umbilical cord mesenchymal stem cells (HUC-MSCs) present in the umbilical cord tissue are self-renewing and multipotent. They can renew themselves continuously and, under certain conditions, differentiate into one or more cell types constituting human tissues and organs. HUC-MSCs differentiate, among others, into osteoblasts, chondrocytes, and adipocytes and have the ability to secrete cytokines. The possibility of noninvasive harvesting and low immunogenicity of HUC-MSCs give them a unique advantage in clinical applications. In recent years, HUC-MSCs have been widely used in clinical practice, and some progress has been made in their use for therapeutic purposes. Main body This article describes two aspects of the clinical therapeutic effects of HUC-MSCs. On the one hand, it explains the benefits and mechanisms of HUC-MSC treatment in various diseases. On the other hand, it summarizes the results of basic research on HUC-MSCs related to clinical applications. The first part of this review highlights several functions of HUC-MSCs that are critical for their therapeutic properties: differentiation into terminal cells, immune regulation, paracrine effects, anti-inflammatory effects, anti-fibrotic effects, and regulating non-coding RNA. These characteristics of HUC-MSCs are discussed in the context of diabetes and its complications, liver disease, systemic lupus erythematosus, arthritis, brain injury and cerebrovascular diseases, heart diseases, spinal cord injury, respiratory diseases, viral infections, and other diseases. The second part emphasizes the need to establish an HUC-MSC cell bank, discusses tumorigenicity of HUC-MSCs and the characteristics of different in vitro generations of these cells in the treatment of diseases, and provides technical and theoretical support for the clinical applications of HUC-MSCs. Conclusion HUC-MSCs can treat a variety of diseases clinically and have achieved good therapeutic effects, and the development of HUC-MSC assistive technology has laid the foundation for its clinical application.
Collapse
Affiliation(s)
- Qixin Xie
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Rui Liu
- Department of Medical Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Jia Jiang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Jing Peng
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Chunyan Yang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Wen Zhang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Sheng Wang
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China
| | - Jing Song
- Anhui Key Laboratory, Department of Pharmacy, Yijishan Hospital Affiliated to Wannan Medical College, Wuhu, China.
| |
Collapse
|
43
|
Chen S, He Z, Xu J. Application of adipose-derived stem cells in photoaging: basic science and literature review. Stem Cell Res Ther 2020; 11:491. [PMID: 33225962 PMCID: PMC7682102 DOI: 10.1186/s13287-020-01994-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Photoaging is mainly induced by continuous exposure to sun light, causing multiple unwanted skin characters and accelerating skin aging. Adipose-derived stem cells(ADSCs) are promising in supporting skin repair because of their significant antioxidant capacity and strong proliferation, differentiation, and migration ability, as well as their enriched secretome containing various growth factors and cytokines. The identification of the mechanisms by which ADSCs perform these functions for photoaging has great potential to explore therapeutic applications and combat skin aging. We also review the basic mechanisms of UV-induced skin aging and recent improvement in pre-clinical applications of ADSCs associated with photoaging. Results showed that ADSCs are potential to address photoaging problem and might treat skin cancer. Compared with ADSCs alone, the secretome-based approaches and different preconditionings of ADSCs are more promising to overcome the current limitations and enhance the anti-photoaging capacity.
Collapse
Affiliation(s)
- Shidie Chen
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Zhigang He
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China.
| | - Jinghong Xu
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
44
|
Human mesenchymal stem cells treatment improved hepatic lesions and reversed gut microbiome disorder in non-alcoholic steatohepatitis. Aging (Albany NY) 2020; 12:21660-21673. [PMID: 33168782 PMCID: PMC7695425 DOI: 10.18632/aging.103962] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/01/2020] [Indexed: 01/05/2023]
Abstract
Effective therapies for non-alcoholic steatohepatitis (NASH) are urgently needed. We investigated the effect of human mesenchymal stem cells (hMSCs) on the intestinal flora in NASH treatment. We isolated the hMSCs from the umbilical cords and divided male C57BL/6 mice into three groups, namely, chow, methionine-choline-deficient (MCD), and MCD+hMSCs. After collecting the feces and liver of the mice, we evaluated the histological changes in the liver and measured the inflammatory and fibrogenesis cytokines. Fecal microbiome and metabolome were analyzed using 16S rRNA gene sequencing analyses. The hMSCs treatment could alleviate hepatic steatosis, inflammation and fibrosis induced by MCD diet. It could also reverse the microbiome and metabolome disorders in the NASH model. Correlation analysis of the interaction among bacteria amplified the effects of the bacteria in host. In conclusion, hMSCs treatment could improve NASH-related lesions and reverse gut microbiome and metabolome disorder in NASH.
Collapse
|
45
|
Role of MSC in the Tumor Microenvironment. Cancers (Basel) 2020; 12:cancers12082107. [PMID: 32751163 PMCID: PMC7464647 DOI: 10.3390/cancers12082107] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/24/2022] Open
Abstract
The tumor microenvironment represents a dynamically composed matrix in which tissue-associated cancer cells are embedded together with a variety of further cell types to form a more or less separate organ-like structure. Constantly mutual interactions between cells of the tumor microenvironment promote continuous restructuring and growth in the tumor. A distinct organization of the tumor stroma also facilitates the formation of transient cancer stem cell niches, thereby contributing to progressive and dynamic tumor development. An important but heterogeneous mixture of cells that communicates among the cancer cells and the different tumor-associated cell types is represented by mesenchymal stroma-/stem-like cells (MSC). Following recruitment to tumor sites, MSC can change their functionalities, adapt to the tumor's metabolism, undergo differentiation and synergize with cancer cells. Vice versa, cancer cells can alter therapeutic sensitivities and change metastatic behavior depending on the type and intensity of this MSC crosstalk. Thus, close cellular interactions between MSC and cancer cells can eventually promote cell fusion by forming new cancer hybrid cells. Consequently, newly acquired cancer cell functions or new hybrid cancer populations enlarge the plasticity of the tumor and counteract successful interventional strategies. The present review article highlights some important features of MSC within the tumor stroma.
Collapse
|
46
|
Umbilical Cord-Derived Mesenchymal Stem Cells Are Able to Use bFGF Treatment and Represent a Superb Tool for Immunosuppressive Clinical Applications. Int J Mol Sci 2020; 21:ijms21155366. [PMID: 32731615 PMCID: PMC7432622 DOI: 10.3390/ijms21155366] [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: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have become a promising tool in cellular therapy for restoring immune system haemostasis; however, the success of clinical trials has been impaired by the lack of standardized manufacturing processes. This study aims to determine the suitability of source tissues and culture media for the production of MSC-based advanced therapy medicinal products (ATMPs) and to define parameters to extend the set of release criteria. MSCs were isolated from umbilical cord (UC), bone marrow and lipoaspirate and expanded in three different culture media. MSC phenotype, proliferation capacity and immunosuppressive parameters were evaluated in normal MSCs compared to primed MSCs treated with cytokines mimicking an inflammatory environment. Compared to bone marrow and lipoaspirate, UC-derived MSCs (UC-MSCs) showed the highest proliferative capacity, which was further enhanced by media supplemented with bFGF, while the cells maintained their immunosuppressive characteristics. Moreover, UC-MSCs expanded in the bFGF-enriched medium were the least sensitive to undesirable priming-induced changes in the MSC phenotype. Surface markers and secreted factors were identified to reflect the cell response to inflammatory priming and to be variable among MSCs from different source tissues. This study demonstrates that UC is a favorable cell source for manufacturing MSC-based ATMPs for immunosuppressive applications. UC-MSCs are able to use the bFGF-enriched medium for higher cell yields without the impairment of immunosuppressive parameters and undesirable phenotype changes after inflammatory preconditioning of MSCs before transplantation. Additionally, immunosuppressive parameters were identified to help finding predictors of clinically efficient MSCs in the following clinical trials.
Collapse
|
47
|
Thamm K, Möbus K, Towers R, Segeletz S, Wetzel R, Bornhäuser M, Zhang Y, Wobus M. A Novel Synthetic, Xeno‐Free Biomimetic Surface for Serum‐Free Expansion of Human Mesenchymal Stromal Cells. ACTA ACUST UNITED AC 2020; 4:e2000008. [DOI: 10.1002/adbi.202000008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/05/2020] [Indexed: 12/27/2022]
Affiliation(s)
| | - Kristin Möbus
- University Hospital Carl Gustav Carus der Technischen Universität Dresden Medizinische Klinik und Poliklinik 1 Fetscherstraße 74 Dresden 01307 Germany
| | - Russell Towers
- University Hospital Carl Gustav Carus der Technischen Universität Dresden Medizinische Klinik und Poliklinik 1 Fetscherstraße 74 Dresden 01307 Germany
| | | | | | - Martin Bornhäuser
- University Hospital Carl Gustav Carus der Technischen Universität Dresden Medizinische Klinik und Poliklinik 1 Fetscherstraße 74 Dresden 01307 Germany
| | - Yixin Zhang
- Technische Universität Dresden Tatzberg 41 Dresden 01307 Germany
| | - Manja Wobus
- University Hospital Carl Gustav Carus der Technischen Universität Dresden Medizinische Klinik und Poliklinik 1 Fetscherstraße 74 Dresden 01307 Germany
| |
Collapse
|
48
|
Wen H, Wang M, Gong S, Li X, Meng J, Wen J, Wang Y, Zhang S, Xin S. Human Umbilical Cord Mesenchymal Stem Cells Attenuate Abdominal Aortic Aneurysm Progression in Sprague-Dawley Rats: Implication of Vascular Smooth Muscle Cell Phenotypic Modulation. Stem Cells Dev 2020; 29:981-993. [PMID: 32486904 PMCID: PMC7410303 DOI: 10.1089/scd.2020.0058] [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] [Indexed: 02/06/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is life-threatening, for which efficient nonsurgical treatment strategy has not been available so far. Several previous studies investigating the therapeutic effect of mesenchymal stem cells (MSCs) in AAA indicated that MSCs could inhibit aneurysmal inflammatory responses and extracellular matrix destruction, and suppress aneurysm occurrence and expansion. Vascular smooth muscle cell (VSMC) phenotypic plasticity is reported to be predisposed in AAA initiation and progression. However, little is known about the effect of MSCs on VSMC phenotypic modulation in AAA. In this study, we investigate the therapeutic efficacy of umbilical cord mesenchymal stem cells (UC-MSCs) in elastase-induced AAA model and evaluate the effect of UC-MSC on VSMC phenotypic regulation. We demonstrate that the intravenous injection of UC-MSC attenuates elastase-induced aneurysmal expansion, reduces elastin degradation and fragmentation, inhibits MMPs and TNF-α expression, and preserves and/or restores VSMC contractile phenotype in AAA. Taken together, these results highlight the therapeutic and VSMC phenotypic modulation effects of UC-MSC in AAA progression, which further indicates the potential of applying UC-MSC as an alternative treatment candidate for AAA.
Collapse
Affiliation(s)
- Hao Wen
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China.,Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Mingjing Wang
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Shiqiang Gong
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Xintong Li
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China.,Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Jinze Meng
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Jie Wen
- Department of Ultrasonography, Inner Mongolia Baotou City Central Hospital, Baotou, China
| | - Yifei Wang
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Shuqing Zhang
- Department of Pharmacology, China Medical University, Shenyang, China
| | - Shijie Xin
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China.,Regenerative Medicine Research Center of China Medical University, Shenyang, China
| |
Collapse
|
49
|
Arrigoni C, D’Arrigo D, Rossella V, Candrian C, Albertini V, Moretti M. Umbilical Cord MSCs and Their Secretome in the Therapy of Arthritic Diseases: A Research and Industrial Perspective. Cells 2020; 9:cells9061343. [PMID: 32481562 PMCID: PMC7348802 DOI: 10.3390/cells9061343] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
The prevalence of arthritic diseases is increasing in developed countries, but effective treatments are currently lacking. The injection of mesenchymal stem cells (MSCs) represents a promising approach to counteract the degenerative and inflammatory environment characterizing those pathologies, such as osteoarthritis (OA). However, the majority of clinical approaches based on MSCs are used within an autologous paradigm, with important limitations. For this reason, allogeneic MSCs isolated from cord blood (cbMSCs) and Wharton’s jelly (wjMSCs) gained increasing interest, demonstrating promising results in this field. Moreover, recent evidences shows that MSCs beneficial effects can be related to their secretome rather than to the presence of cells themselves. Among the trophic factors secreted by MSCs, extracellular vesicles (EVs) are emerging as a promising candidate for the treatment of arthritic joints. In the present review, the application of umbilical cord MSCs and their secretome as innovative therapeutic approaches in the treatment of arthritic joints will be examined. With the prospective of routine clinical applications, umbilical cord MSCs and EVs will be discussed also within an industrial and regulatory perspective.
Collapse
Affiliation(s)
- Chiara Arrigoni
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900 Lugano, Switzerland; (C.A.); (D.D.)
| | - Daniele D’Arrigo
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900 Lugano, Switzerland; (C.A.); (D.D.)
| | - Valeria Rossella
- Swiss Stem Cells Biotech, Via Pizzamiglio 12, 6833 Vacallo, Switzerland; (V.R.); (V.A.)
| | - Christian Candrian
- Unità di Ortopedia e Traumatologia, Ospedale Regionale di Lugano, Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland
| | - Veronica Albertini
- Swiss Stem Cells Biotech, Via Pizzamiglio 12, 6833 Vacallo, Switzerland; (V.R.); (V.A.)
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900 Lugano, Switzerland; (C.A.); (D.D.)
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, via R. Galeazzi 4., 20161 Milano, Italy
- Correspondence: ; Tel.: +41-91-811-7076
| |
Collapse
|
50
|
Zhang Y, Lv P, Li Y, Zhang Y, Cheng C, Hao H, Yue H. Comparison of the biological characteristics of umbilical cord mesenchymal stem cells derived from the human heterosexual twins. Differentiation 2020; 114:1-12. [PMID: 32460139 DOI: 10.1016/j.diff.2020.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/17/2020] [Accepted: 05/11/2020] [Indexed: 12/24/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) are attracting more and more attention due to their tissue regenerative properties and immunomodulatory functions. MSCs may be the most acceptable, safe, and effective source for allogeneic cell therapy, and have been used in medical treatment. However, the similarities and differences between umbilical cord-derived MSCs (UC-MSCs) of heterosexual twins remain poorly understood. In this study, we compared the biological characteristics of UC-MSCs of heterosexual twins in vitro. We found that male fetal UC-MSCs and female fetal UC-MSCs share a similar phenotype and multi-lineage differentiation potential, and male fetal UC-MSCs show a significantly higher proliferation and adipogenic ability than female fetal UC-MSCs. UC-MSCs from heterosexual twins showed significant differences in the expression levels of NANOG, OCT4, TERT, and SOX2. In addition, male MSCs are more potent in the expression of inflammatory cytokines to lipopolysaccharide (LPS)-induced inflammation. In future clinical applications using MSCs for inflammation-related diseases, these biological characteristics differences with different genders will guide our clinical methods.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Pengju Lv
- Translational Medicine Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, 450007, China
| | - Yalong Li
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Yonghui Zhang
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Chaofei Cheng
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Hongbo Hao
- Neuroscience Initiative, Advanced Science Research Center at the Graduate Center, City University of New York, New York, 10031, USA
| | - Han Yue
- Stem Cell Research Center, Henan Key Laboratory of Stem Cell Differentiation and Modification Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China.
| |
Collapse
|