1
|
武 志, 胡 明, 赵 巧, 吕 凤, 张 晶, 张 伟, 王 永, 孙 晓, 王 慧. [Immunomodulatory mechanism of umbilical cord mesenchymal stem cells modified by miR-125b-5p in systemic lupus erythematosus]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2024; 56:860-867. [PMID: 39397466 PMCID: PMC11480562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To investigate the mechanism of immunomodulatory effects of umbilical cord mesenchymal stem cells (UC-MSCs) modified by miR-125b-5p on systemic lupus erythematosus (SLE). METHODS The expression level of miR-125b-5p was detected by real-time fluorescence quantitative PCR in UC-MSCs and peripheral blood mononuclear cells (PBMCs) from SLE patients and health checkers. Annexin V-FITC/PI apoptosis detection kit was used to detect the effect of miR-125b-5p on apoptosis of UC-MSCs. MRL/lpr mice in each group were injected with UC-MSCs via tail vein, and T-lymphocyte subsets in the spleen of the MRL/lpr mice were detected by flow cytometry after 5 weeks. The expression levels of interleukin (IL)-4 and IL-17A in serum of MRL/lpr mice were detected by ELISA. Hematoxylin-eosin staining was used to observe the pathological manifestations of the lungs and kidneys of the MRL/lpr mice. RESULTS miR-125b-5p was significantly down-regulated in PBMCs of SLE patients compared with healthy controls (P < 0.01). Compared with the UC-MSCs group, the expression of miR- 125b-5p in UC-MSCs modified by miR-125b-5p group was increased (P < 0.01). The survival rate of UC-MSCs was significantly increased by miR-125b-5p (P < 0.01). Compared with the untreated group of MRL/lpr mice, the expression level of IL-4 in serum was increased (P < 0.05); the expression level of IL-17A was decreased (P < 0.05); the proportion of Th17 cells in the spleen of MRL/lpr mice was decreased (P < 0.05); the inflammatory cells infiltration and micro-thrombosis of lungs and kidneys of MRL/lpr mice were significantly reduced in the UC-MSCs modified by miR-125b-5p treatment group. CONCLUSION UC-MSCs modified by miR-125b-5p have immunomodulatory effects on systemic lupus erythematosus.
Collapse
Affiliation(s)
- 志慧 武
- 包头医学院第一附属医院中心实验室(内蒙古自治区自体免疫学重点实验室),内蒙古自治区包头 014010Central Laboratory, First Affiliated Hospital of Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunology), Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 明智 胡
- 包头医学院第一附属医院中心实验室(内蒙古自治区自体免疫学重点实验室),内蒙古自治区包头 014010Central Laboratory, First Affiliated Hospital of Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunology), Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 巧英 赵
- 包头医学院第一附属医院中心实验室(内蒙古自治区自体免疫学重点实验室),内蒙古自治区包头 014010Central Laboratory, First Affiliated Hospital of Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunology), Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 凤凤 吕
- 包头医学院第一附属医院中心实验室(内蒙古自治区自体免疫学重点实验室),内蒙古自治区包头 014010Central Laboratory, First Affiliated Hospital of Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunology), Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 晶莹 张
- 包头医学院第一附属医院风湿免疫科,内蒙古自治区包头 014010Department of Rheumatism and Immunology, First Affiliated Hospital of Baotou Medical College, Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 伟 张
- 包头医学院第一附属医院中心实验室(内蒙古自治区自体免疫学重点实验室),内蒙古自治区包头 014010Central Laboratory, First Affiliated Hospital of Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunology), Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 永福 王
- 包头医学院第一附属医院风湿免疫科,内蒙古自治区包头 014010Department of Rheumatism and Immunology, First Affiliated Hospital of Baotou Medical College, Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 晓林 孙
- 包头医学院第一附属医院中心实验室(内蒙古自治区自体免疫学重点实验室),内蒙古自治区包头 014010Central Laboratory, First Affiliated Hospital of Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunology), Baotou 014010, Inner Mongolia Autonomous Region, China
| | - 慧 王
- 包头医学院第一附属医院风湿免疫科,内蒙古自治区包头 014010Department of Rheumatism and Immunology, First Affiliated Hospital of Baotou Medical College, Baotou 014010, Inner Mongolia Autonomous Region, China
| |
Collapse
|
2
|
Carstens M, Trujillo J, Dolmus Y, Rivera C, Calderwood S, Lejarza J, López C, Bertram K. Adipose-derived stromal vascular fraction cells to treat long-term pulmonary sequelae of coronavirus disease 2019: 12-month follow-up. Cytotherapy 2024; 26:1076-1083. [PMID: 38639670 DOI: 10.1016/j.jcyt.2024.03.491] [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: 02/06/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND AIMS Long coronavirus disease (COVID) is estimated to occur in up to 20% of patients with coronavirus disease 2019 (COVID-19) infections, with many having persistent pulmonary symptoms. Mesenchymal stromal cells (MSCs) have been shown to have powerful immunomodulatory and anti-fibrotic properties. Autologous adipose-derived (AD) stromal vascular fraction (SVF) contains MSC and other healing cell components and can be obtained by small-volume lipoaspiration and administered on the same day. This study was designed to study the safety of AD SVF infused intravenously to treat the pulmonary symptoms of long COVID. METHODS Five subjects with persistent cough and dyspnea after hospitalization and subsequent discharge for COVID-19 pneumonia were treated with 40 million intravenous autologous AD SVF cells and followed for 12 months, to include with pulmonary function tests and computed tomography scans of the lung. RESULTS SVF infusion was safe, with no significant adverse events related to the infusion out to 12 months. Four subjects had improvements in pulmonary symptoms, pulmonary function tests, and computed tomography scans, with some improvement noted as soon as 1 month after SVF treatment. CONCLUSIONS It is not possible to distinguish between naturally occurring improvement or improvement caused by SVF treatment in this small, uncontrolled study. However, the results support further study of autologous AD SVF as a treatment for long COVID.
Collapse
Affiliation(s)
- Michael Carstens
- Department of Surgery, Hospital Escuela Oscar Danilo Rosale Argüello, León, Nicaragua; Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, USA.
| | - Jessy Trujillo
- Department of Medicine, Hospital Monte España, Managua, Nicaragua
| | - Yanury Dolmus
- Department of Pediatrics, Hospital Escuela Cesar Amador Molina, Matagalpa, Nicaragua
| | - Carlos Rivera
- Department of Radiology, Hospital Escuela Cesar Amador Molina, Matagalpa, Nicaragua
| | - Santos Calderwood
- Department of Surgery, Hospital Escuela Cesar Amador Molina, Matagalpa, Nicaragua
| | - Judith Lejarza
- Department of Surgery, Hospital Escuela Oscar Danilo Rosale Argüello, León, Nicaragua
| | - Carlos López
- Department of Medicine, Hospital Escuela Oscar Danilo Rosales Argüello, León, Nicaragua
| | - Kenneth Bertram
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
3
|
Aouabdi S, Aboalola D, Zakari S, Alwafi S, Nedjadi T, Alsiary R. Protective potential of mesenchymal stem cells against COVID-19 during pregnancy. Future Sci OA 2024; 10:FSO924. [PMID: 38836262 PMCID: PMC11149780 DOI: 10.2144/fsoa-2023-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/17/2023] [Indexed: 06/06/2024] Open
Abstract
SARS-CoV-2 causes COVID-19. COVID-19 has led to severe clinical illnesses and an unprecedented death toll. The virus induces immune inflammatory responses specifically cytokine storm in lungs. Several published reports indicated that pregnant females are less likely to develop severe symptoms compared with non-pregnant. Putative protective role of maternal blood circulating fetal mesenchymal stem cells (MSCs) has emerged and have been put forward as an explanation to alleviated symptoms. MSCs with immune-modulatory, anti-inflammatory and anti-viral roles, hold great potential for the treatment of COVID-19. MSCs could be an alternative to treat infections resulting from the SARS-CoV-2 and potential future outbreaks. This review focuses on the MSCs putative protective roles against COVID-19 in pregnant females.
Collapse
Affiliation(s)
- Sihem Aouabdi
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Doaa Aboalola
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Samer Zakari
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Suliman Alwafi
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Taoufik Nedjadi
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| | - Rawiah Alsiary
- King Abdullah International Medical Research Center, Jeddah, 21423, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, 21423, Saudi Arabia
| |
Collapse
|
4
|
Wufuer M, Choi TH, Najmiddinov B, Kim J, Choi J, Kim T, Park Y, Kim JH, Jeon H, Kim BJ. Improving Facial Fat Graft Survival Using Stromal Vascular Fraction-Enriched Lipotransfer: A Multicenter Randomized Controlled Study. Plast Reconstr Surg 2024; 153:690e-700e. [PMID: 37141448 DOI: 10.1097/prs.0000000000010625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
BACKGROUND Although previous clinical studies have reported that cell-assisted lipotransfer increases the fat survival rate in facial fat transplants, most were case studies without quantitative evaluation. A multicenter randomized controlled study was performed to evaluate the safety and efficacy of the stromal vascular fraction (SVF) in facial fat grafts. METHODS Twenty-three participants were enrolled for autologous fat transfer in the face, and assigned randomly to the experimental ( n = 11) or control ( n = 12) group. Fat survival was assessed using magnetic resonance imaging at 6 and 24 weeks postoperatively. Subjective evaluations were performed by the patients and surgeons. To address safety concerns, results of an SVF culture and the postoperative complications were recorded. RESULTS The overall fat survival rate was significantly higher in the experimental group than in the control group (6 weeks, 74.5% ± 9.99% versus 66.55% ± 13.77%, P < 0.025; 24 weeks, 71.27% ± 10.43% versus 61.98% ± 13.46%, P < 0.012). Specifically, graft survival in the forehead was 12.82% higher in the experimental group when compared with that in the control group at 6 weeks ( P < 0.023). Furthermore, graft survival in the forehead ( P < 0.021) and cheeks ( P < 0.035) was superior in the experimental group at 24 weeks. At 24 weeks, the aesthetic scores given by the surgeons were higher in the experimental group than in the control group ( P < 0.03); however, no significant intergroup differences were noted in the patient-evaluated scores. Neither bacterial growth from SVF cultures nor postoperative complications were noted. CONCLUSION SVF enrichment for autologous fat grafting can be a safe and effective technique for increasing the fat retention rate. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, II.
Collapse
Affiliation(s)
| | | | - Bakhtiyor Najmiddinov
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine
| | - Junhyung Kim
- Department of Plastic and Reconstructive Surgery, Keimyung University School of Medicine
| | - Jaehoon Choi
- Department of Plastic and Reconstructive Surgery, Keimyung University School of Medicine
| | | | | | - Ji-Hoon Kim
- Radiology, Seoul National University Hospital, Seoul National University College of Medicine
| | - Heejung Jeon
- From the Departments of Plastic and Reconstructive Surgery
| | - Byung Jun Kim
- From the Departments of Plastic and Reconstructive Surgery
| |
Collapse
|
5
|
Li J, Wu Z, Zhao L, Liu Y, Su Y, Gong X, Liu F, Zhang L. The heterogeneity of mesenchymal stem cells: an important issue to be addressed in cell therapy. Stem Cell Res Ther 2023; 14:381. [PMID: 38124129 PMCID: PMC10734083 DOI: 10.1186/s13287-023-03587-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
With the continuous improvement of human technology, the medical field has gradually moved from molecular therapy to cellular therapy. As a safe and effective therapeutic tool, cell therapy has successfully created a research boom in the modern medical field. Mesenchymal stem cells (MSCs) are derived from early mesoderm and have high self-renewal and multidirectional differentiation ability, and have become one of the important cores of cell therapy research by virtue of their immunomodulatory and tissue repair capabilities. In recent years, the application of MSCs in various diseases has received widespread attention, but there are still various problems in the treatment of MSCs, among which the heterogeneity of MSCs may be one of the causes of the problem. In this paper, we review the correlation of MSCs heterogeneity to provide a basis for further reduction of MSCs heterogeneity and standardization of MSCs and hope to provide a reference for cell therapy.
Collapse
Affiliation(s)
- Jingxuan Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Zewen Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Li Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030600, China
| | - Yang Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yazhen Su
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Xueyan Gong
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Fancheng Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
6
|
Wang G, Wang Z, Zhang J, Shen Y, Hou X, Su L, Chen W, Chen J, Guo X, Song H. Treatment of androgenetic alopecia by exosomes secreted from hair papilla cells and the intervention effect of LTF. J Cosmet Dermatol 2023; 22:2996-3007. [PMID: 37553912 DOI: 10.1111/jocd.15890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/25/2023] [Accepted: 06/14/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Androgenetic alopecia (AGA) is the most common cause of chronic progressive hair loss in men, and AGA has a severe negative impact on the quality of life and physical and mental health of patients. METHODS Four female C57BL/6 mice were isolated from DP cells in culture (≤4 generations) after stimulation of DPC proliferation by herbal concentrations obtained by the CCK-8 method, and exosomes were isolated by differential centrifugation at low temperature. Testosterone propionate and topical hair removal treatments were used together to establish the C57BL/6 mouse AGA model, which was treated with LTF, 5% minoxidil, and LTF-DPC-EXO, respectively. ELISA was used to detect serum hormone levels, in vivo tracing was used to observe dynamic changes in exosomes, H&E staining showed changes in mouse hair follicle tissue, and (q) RT-PCR and WB were used to detect dorsal skin VEGF, AKT1, and CASP3 expression in dorsal skin tissues. RESULTS Hair regeneration was significant in the LTF group, minoxidil group, and LTF-DPC-EXO group mice, and the hair growth was only seen in the local skin in the model group. The hormone T in all treatment groups was lower than that in the model group, and e2 was higher than that in the model group. (q) RT-PCR and western blot showed that VEGF and AKT1 expressions were upregulated and Caspase3 expression was downregulated in the skin sections of mice in the treatment groups. CONCLUSION DPC-EXO obtained through LTF may activate AKT1 and VEGF in the PI3K/AKT signaling pathway to inhibit CASP3, thereby protecting DPC to restore the hair growth.
Collapse
Affiliation(s)
- Guiyue Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhili Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaqi Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Shen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Hou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Su
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wen Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiahao Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang Guo
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hong Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Wang L, Qiao S, Xia R, Liu Y, Hu Y, Wu Y, Zhou J, Liang G, Tian T, Cao L. Mesenchymal stromal cell-derived magnetic nanovesicles for enhanced skin retention and hair follicle growth. Cytotherapy 2023; 25:1176-1185. [PMID: 37516947 DOI: 10.1016/j.jcyt.2023.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND AIMS Extracellular vesicles and exosome-mimetic nanovesicles (NVs) derived from mesenchymal stromal cells (MSCs) have emerged as promising to promote hair growth. However, short local skin retention after subcutaneous administration hinders their clinical applications. METHODS In this study, we prepared magnetic nanovesicles (MNVs) from iron oxide nanoparticle-incorporated MSCs. MNVs contained more therapeutic growth factors than NVs derived from naive MSCs, and their localization and internalization were manipulated by external magnetic field. RESULTS Following the subcutaneous injection of MNVs into a mouse model of depilation-induced hair regeneration, the magnetic attraction increased their skin retention. Then, the cellular proliferation and β-catenin signaling in hair follicles (HF) were markedly enhanced by MNV injection and magnetic field application. Furthermore, an acceleration of HF growth was revealed by histological analysis. CONCLUSIONS The proposed strategy can enhance the therapeutic potential of MSC-derived NVs for hair regeneration and other dermatological diseases.
Collapse
Affiliation(s)
- Lei Wang
- Department of Dermatology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Shuya Qiao
- Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rushan Xia
- Department of Dermatology, Jiangnan University Medical Center, Wuxi, Jiangsu, China
| | - Yiwen Liu
- Department of Dermatology, Jiangnan University Medical Center, Wuxi, Jiangsu, China
| | - Yifei Hu
- Department of Dermatology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yajuan Wu
- Department of Dermatology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Junhao Zhou
- Department of Dermatology, Jiangnan University Medical Center, Wuxi, Jiangsu, China
| | - Gaofeng Liang
- School of Basic Medical Sciences, Henan University of Science & Technology, Luoyang, Henan, China
| | - Tian Tian
- Department of Neurobiology, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Lei Cao
- Department of Dermatology, Jiangnan University Medical Center, Wuxi, Jiangsu, China.
| |
Collapse
|
8
|
Yan C, Hu M, Dai R. Safety and efficacy of mesenchymal stem cells in COVID-19 patients: A systematic review and meta-analysis. Immun Inflamm Dis 2023; 11:e1000. [PMID: 37773722 PMCID: PMC10515507 DOI: 10.1002/iid3.1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/03/2023] [Accepted: 08/22/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Coronavirus disease-19 (COVID-19) is a zoonotic disease that has become a global pandemic. The fast evolution of the COVID-19 pandemic and persist problems make COVID-19 highly infectious; publicly accessible literature and other sources of information continue to expand in volume. The mesenchymal stem cells (MSCs) therapy efficacy for COVID-19 is debatable. OBJECTIVE This systematic review and meta-analysis (SRMA) aimed to evaluate the usefulness of MSCs in treating COVID-19. METHODS Relevant publications were retrieved from databases up to April 30, 2022. In the case of dichotomous data, the 95% confidence intervals (CIs) and pooled risk ratio (RR) were estimated with a random effects model (REM) or fixed effects model (FEM). The pooled mean difference (MD) and 95% CIs were calculated with REM or FEM in continuous data. In the outcomes, studies with insufficient or unusable data were reported descriptively. RESULTS A total of eight randomized controlled trials (RCTs) with 464 people were chosen for this SRMA. Relative to the control group, mortality was significantly lower in the MSCs group (RR: 0.66, 95% CI: 0.44, 0.99, Z = 2.01, p = .04); other secondary outcomes, such as the clinical symptom improvement rate improved in the MSCs group (RR: 1.44, 95% CI: 1.05, 1.99, Z = 2.24, p = .03), clinical symptom improvement time (MD: -4.01, 95% CI: -6.33, -1.68, Z = 3.38, p = .0007), C-reactive protein (CRP) (MD: -39.16, 95% CI: -44.39, -33.94, Z = 14.70, p < .00001) and days to hospital discharge (MD: -3.83, 95% CI: -6.19, -1.48, Z = 3.19, p = .001) reduced significantly in MSCs group. However, the adverse reaction incidence did not change significantly. CONCLUSIONS MSCs are a viable therapy option for COVID-19 because of their safety and potential efficacy. With no significant adverse effects, MSCs can reduce mortality, clinical symptom improvement time, and days to hospital discharge, improve clinical symptoms, and reduce inflammatory cytokines CRP in COVID-19. However, further high-quality clinical studies are required to confirm these results.
Collapse
Affiliation(s)
- Cai Yan
- Xiangtan Central HospitalDepartment of Infectious diseasesXiangtanHunan provincePeople's Republic of China
| | - Minjie Hu
- The First Affiliated Hospital, Department of Cardiothoracic Surgery, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan provincePeople's Republic of China
| | - Rongjuan Dai
- The First Affiliated Hospital, Department of Infectious Diseases, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan provincePeople's Republic of China
| |
Collapse
|
9
|
Armstrong BBS, Pedroso JCM, Conceição Carvalho JD, Ferreira LM. Mesenchymal stem cells in lung diseases and their potential use in COVID-19 ARDS: A systematized review. Clinics (Sao Paulo) 2023; 78:100237. [PMID: 37454534 PMCID: PMC10368758 DOI: 10.1016/j.clinsp.2023.100237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/23/2023] [Accepted: 04/24/2023] [Indexed: 07/18/2023] Open
Abstract
COVID-19 can converge with the pro-inflammatory immunoregulatory mechanisms of chronic lung diseases. Given the disorders inherent to lung transplantation and the inexistence of other definitive therapeutic alternatives, Adipose tissue-derived Stem Cells (ASCs) presented themselves as a therapeutic hope. The purpose of this review is to assess the basis for the potential use of ASCs in lung diseases unresponsive to conventional therapy, relating to their possible use in COVID-19 ARDS. 35 studies comprised this review, 14 being narrative reviews, 19 preclinical trials and two proofs of concept. COVID-19 can converge with the pro-inflammatory immunoregulatory mechanisms of chronic lung diseases. In view of the disorders inherent to lung transplantation and the inexistence of definitive therapeutic alternatives, Adipose tissue-derived Stem Cells (ASCs) presented themselves as a therapeutic hope. Its detailed reading indicated the absence of serious adverse effects and toxicity to the administration of ASCs and suggested possible effectiveness in reducing lung damage, in addition to promoting the recovery of leukocytes and lymphocytes with its immunomodulatory and anti-apoptotic effects. The revised clinical data suggests optimism in the applicability of ASCs in other immunoinflammatory diseases and in severe COVID-19 ARDS. However, further studies are needed to develop a consensus on the methods of collection of ASCs, the ideal dosage schedule, the most effective time and route of administration, as well as on the definition of indications for the administration of ASCs in cases of COVID-19 for conducting clinical trials in near future.
Collapse
Affiliation(s)
| | | | | | - Lydia Masako Ferreira
- Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
10
|
Li H, Dai H, Li J. Immunomodulatory properties of mesenchymal stromal/stem cells: The link with metabolism. J Adv Res 2023; 45:15-29. [PMID: 35659923 PMCID: PMC10006530 DOI: 10.1016/j.jare.2022.05.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/17/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) are the most promising stem cells for the treatment of multiple inflammatory and immune diseases due to their easy acquisition and potent immuno-regulatory capacities. These immune functions mainly depend on the MSC secretion of soluble factors. Recent studies have shown that the metabolism of MSCs plays critical roles in immunomodulation, which not only provides energy and building blocks for macromolecule synthesis but is also involved in the signaling pathway regulation. AIM OF REVIEW A thorough understanding of metabolic regulation in MSC immunomodulatory properties can provide new sights to the enhancement of MSC-based therapy. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC immune regulation can be affected by cellular metabolism (glucose, adenosine triphosphate, lipid and amino acid metabolism), which further mediates MSC therapy efficiency in inflammatory and immune diseases. The enhancement of glycolysis of MSCs, such as signaling molecule activation, inflammatory cytokines priming, or environmental control can promote MSC immune functions and therapeutic potential. Besides glucose metabolism, inflammatory stimuli also alter the lipid molecular profile of MSCs, but the direct link with immunomodulatory properties remains to be further explored. Arginine metabolism, glutamine-glutamate metabolism and tryptophan-kynurenine via indoleamine 2,3-dioxygenase (IDO) metabolism all contribute to the immune regulation of MSCs. In addition to the metabolism dictating the MSC immune functions, MSCs also influence the metabolism of immune cells and thus determine their behaviors. However, more direct evidence of the metabolism in MSC immune abilities as well as the underlying mechanism requires to be uncovered.
Collapse
Affiliation(s)
- Hanyue Li
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Hongwei Dai
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| |
Collapse
|
11
|
Luo D, Peng S, Li Q, Rao P, Tao G, Wang L, Xiao J. Methyltransferase-like 3 modulates osteogenic differentiation of adipose-derived stem cells in osteoporotic rats. J Gene Med 2023; 25:e3481. [PMID: 36782035 DOI: 10.1002/jgm.3481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/20/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Osteoporosis (OP) is a metabolic bone disease involving reduced bone mass. Adipose-derived stem cells (ASCs) play an important role in bone regeneration. Emerging evidence suggests that methyltransferase-like 3 (METTL3) plays a significant role in bone development and metabolism. Therefore, this study investigates changes to METTL3 in the osteogenic differentiation of adipose stem cells in osteoporotic rats (OP-ASCs) and explores ways to enhance their osteogenic ability. METHODS An animal model of osteoporosis was established by removing both ovaries in rats. Real-time PCR and western blotting were performed to detect the expression of METTL3 and bone-related molecules, including runt-related transcription factor 2 (Runx2) and osteopontin (Opn). Furthermore, alkaline phosphatase staining was used to confirm the osteogenic potential of stem cells. Mettl3 small interfering RNA and Mettl3 overexpression lentivirus were used to assess the role of METTL3 in osteogenic differentiation of ASCs and OP-ASCs. RESULTS The osteogenic differentiation capacity and Mettl3 expression significantly decreased in OP-ASCs. Moreover, Mettl3 silencing down-regulated the osteogenic ability of ASCs, and overexpression of Mettl3 recovered the impaired osteogenic capacity in OP-ASCs in vitro. CONCLUSION The Mettl3 expression levels and osteogenic potential of OP-ASCs decreased. However, overexpression of METTL3 rescued the osteogenic ability of OP-ASCs, providing a new target for treatment of osteoporotic bone defects.
Collapse
Affiliation(s)
- Daowen Luo
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Shuanglin Peng
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qing Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Pengcheng Rao
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Tao
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Lang Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China.,Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| | - Jingang Xiao
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China.,Department of Oral Implantology, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China.,Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Öner Ç, Irmak F, Eken G, Öner BB, Karsıdağ SH. The effect of stromal vascular fraction in an experimental frostbite injury model. Burns 2023; 49:149-161. [PMID: 35241296 DOI: 10.1016/j.burns.2022.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/29/2022] [Accepted: 02/14/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Despite current treatment modalities, frostbite remains an injury with a poor prognosis which may cause functional morbidities. Several experimental and clinical studies have demonstrated that stromal vascular fraction is an autologous mixture, which can improve wound healing and vasculogenesis. The aim of this study was to show the beneficial effects of stromal vascular fraction on experimental frostbite healing. MATERIAL AND METHODS Stromal vascular fraction (SVF) was harvested from 5 rats after excision of the inguinal fat pads. Another 20 rats were separated into 2 groups of 10 as the SVF group and the control group. A frostbite injury was created on each rat using a cryoprobe frozen with liquid nitrogen (-196 °C). SVF was applied to the SVF group and phosphate-buffered saline to the control group. All injections were performed subcutaneously within the frostbite injury area. Biopsies were performed on days 5 and 14 for histopathological and immunochemical evaluations. The tissue perfusion rates of both groups were assessed on day 14 using indocyanine green angiography (SPY system). RESULTS The increase in mean tissue perfusion was 373.3% ( ± 32.1) in the SVF group and 123.8% ( ± 16.3) in the control group (p < 0.001). The macroscopic wound reduction rates of the SVF and control groups were 25.5% ( ± 19.1) and 18.0% ( ± 5.9), respectively on day 5%, and 78.2% ( ± 9.2) and 57.3% ( ± 16.7) on day 14 (p = 0.007; p = 0.003). Acute inflammation and the fibrosis gradient were significantly decreased in the SVF group compared to the control group (p = 0.004, p = 0.054 respectively on day 14). Granulation tissue amount, re-epithelialization score and neovascularization were significantly increased in the SVF group (p = 0.006, p = 0.010 and p = 0.021, respectively on day 14). CONCLUSIONS The study results demonstrated that SVF increases frostbite wound healing by increasing tissue perfusion rate, neovascularization and re-epithelialization, and modulating acute inflammation and fibrosis.
Collapse
Affiliation(s)
- Çağatay Öner
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey; Department of Plastic, Reconstructive and Aesthetic Surgery, Sirnak State Hospital, Sirnak, Turkey.
| | - Fatih Irmak
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey.
| | - Gülçin Eken
- Department of Clinical Pathology, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey.
| | - Burcu Bitir Öner
- Department of Anesthesiology and Reanimation, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey.
| | - Semra Hacıkerim Karsıdağ
- Department of Plastic, Reconstructive and Aesthetic Surgery, University of Health Sciences, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey.
| |
Collapse
|
13
|
Bazdyrev E, Panova M, Zherebtsova V, Burdenkova A, Grishagin I, Novikov F, Nebolsin V. The Hidden Pandemic of COVID-19-Induced Organizing Pneumonia. Pharmaceuticals (Basel) 2022; 15:1574. [PMID: 36559025 PMCID: PMC9780828 DOI: 10.3390/ph15121574] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Since the beginning of the COVID-19 pandemic, clinical, radiological, and histopathological studies have provided evidence that organizing pneumonia is a possible consequence of the SARS-CoV2 infection. This post-COVID-19 organizing pneumonia (PCOP) causes persisting dyspnea, impaired pulmonary function, and produces radiological abnormalities for at least 5 weeks after onset of symptoms. While most patients with PCOP recover within a year after acute COVID-19, 5-25% of cases need specialized treatment. However, despite substantial resources allocated worldwide to finding a solution to this problem, there are no approved treatments for PCOP. Oral corticosteroids produce a therapeutic response in a majority of such PCOP patients, but their application is limited by the anticipated high-relapse frequency and the risk of severe adverse effects. Herein, we conduct a systematic comparison of the epidemiology, pathogenesis, and clinical presentation of the organizing pneumonias caused by COVID-19 as well as other viral infections. We also use the clinical efficacy of corticosteroids in other postinfection OPs (PIOPs) to predict the therapeutic response in the treatment of PCOP. Finally, we discuss the potential application of a candidate anti-inflammatory and antifibrotic therapy for the treatment of PCOP based on the analysis of the latest clinical trials data.
Collapse
Affiliation(s)
- Evgeny Bazdyrev
- Research Institute for Complex Issues of Cardiovascular Diseases, 6, Sosnoviy Blvd., 650002 Kemerovo, Russia
| | - Maria Panova
- Pharmenterprises LLC, Skolkovo Innovation Center, Bolshoi Blvd., 42(1), 143026 Moscow, Russia
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Avenue, 119991 Moscow, Russia
| | - Valeria Zherebtsova
- Pharmenterprises LLC, Skolkovo Innovation Center, Bolshoi Blvd., 42(1), 143026 Moscow, Russia
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Avenue, 119991 Moscow, Russia
- Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya sq. 9, 125047 Moscow, Russia
| | - Alexandra Burdenkova
- Pharmenterprises LLC, Skolkovo Innovation Center, Bolshoi Blvd., 42(1), 143026 Moscow, Russia
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Avenue, 119991 Moscow, Russia
| | - Ivan Grishagin
- Rancho BioSciences, 16955 Via Del Campo Suite 200, San Diego, CA 92127, USA
| | - Fedor Novikov
- Pharmenterprises LLC, Skolkovo Innovation Center, Bolshoi Blvd., 42(1), 143026 Moscow, Russia
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 47 Leninsky Avenue, 119991 Moscow, Russia
| | - Vladimir Nebolsin
- Pharmenterprises LLC, Skolkovo Innovation Center, Bolshoi Blvd., 42(1), 143026 Moscow, Russia
| |
Collapse
|
14
|
Yudintceva N, Mikhailova N, Fedorov V, Samochernych K, Vinogradova T, Muraviov A, Shevtsov M. Mesenchymal Stem Cells and MSCs-Derived Extracellular Vesicles in Infectious Diseases: From Basic Research to Clinical Practice. Bioengineering (Basel) 2022; 9:662. [PMID: 36354573 PMCID: PMC9687734 DOI: 10.3390/bioengineering9110662] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/30/2022] [Accepted: 11/04/2022] [Indexed: 08/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are attractive in various fields of regenerative medicine due to their therapeutic potential and complex unique properties. Basic stem cell research and the global COVID-19 pandemic have given impetus to the development of cell therapy for infectious diseases. The aim of this review was to systematize scientific data on the applications of mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (MSC-EVs) in the combined treatment of infectious diseases. Application of MSCs and MSC-EVs in the treatment of infectious diseases has immunomodulatory, anti-inflammatory, and antibacterial effects, and also promotes the restoration of the epithelium and stimulates tissue regeneration. The use of MSC-EVs is a promising cell-free treatment strategy that allows solving the problems associated with the safety of cell therapy and increasing its effectiveness. In this review, experimental data and clinical trials based on MSCs and MSC-EVs for the treatment of infectious diseases are presented. MSCs and MSC-EVs can be a promising tool for the treatment of various infectious diseases, particularly in combination with antiviral drugs. Employment of MSC-derived EVs represents a more promising strategy for cell-free treatment, demonstrating a high therapeutic potential in preclinical studies.
Collapse
Affiliation(s)
- Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Natalia Mikhailova
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
| | - Viacheslav Fedorov
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Konstantin Samochernych
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, St. Petersburg 191036, Russia
| | - Alexandr Muraviov
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Health of the Russian Federation, St. Petersburg 191036, Russia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| |
Collapse
|
15
|
Kerstan A, Dieter K, Niebergall-Roth E, Klingele S, Jünger M, Hasslacher C, Daeschlein G, Stemler L, Meyer-Pannwitt U, Schubert K, Klausmann G, Raab T, Goebeler M, Kraft K, Esterlechner J, Schröder HM, Sadeghi S, Ballikaya S, Gasser M, Waaga-Gasser AM, Murphy GF, Orgill DP, Frank NY, Ganss C, Scharffetter-Kochanek K, Frank MH, Kluth MA. Translational development of ABCB5 + dermal mesenchymal stem cells for therapeutic induction of angiogenesis in non-healing diabetic foot ulcers. Stem Cell Res Ther 2022; 13:455. [PMID: 36064604 PMCID: PMC9444095 DOI: 10.1186/s13287-022-03156-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/25/2022] [Indexed: 11/15/2022] Open
Abstract
Background While rapid healing of diabetic foot ulcers (DFUs) is highly desirable to avoid infections, amputations and life-threatening complications, DFUs often respond poorly to standard treatment. GMP-manufactured skin-derived ABCB5+ mesenchymal stem cells (MSCs) might provide a new adjunctive DFU treatment, based on their remarkable skin wound homing and engraftment potential, their ability to adaptively respond to inflammatory signals, and their wound healing-promoting efficacy in mouse wound models and human chronic venous ulcers. Methods The angiogenic potential of ABCB5+ MSCs was characterized with respect to angiogenic factor expression at the mRNA and protein level, in vitro endothelial trans-differentiation and tube formation potential, and perfusion-restoring capacity in a mouse hindlimb ischemia model. Finally, the efficacy and safety of ABCB5+ MSCs for topical adjunctive treatment of chronic, standard therapy-refractory, neuropathic plantar DFUs were assessed in an open-label single-arm clinical trial. Results Hypoxic incubation of ABCB5+ MSCs led to posttranslational stabilization of the hypoxia-inducible transcription factor 1α (HIF-1α) and upregulation of HIF-1α mRNA levels. HIF-1α pathway activation was accompanied by upregulation of vascular endothelial growth factor (VEGF) transcription and increase in VEGF protein secretion. Upon culture in growth factor-supplemented medium, ABCB5+ MSCs expressed the endothelial-lineage marker CD31, and after seeding on gel matrix, ABCB5+ MSCs demonstrated formation of capillary-like structures comparable with human umbilical vein endothelial cells. Intramuscularly injected ABCB5+ MSCs to mice with surgically induced hindlimb ischemia accelerated perfusion recovery as measured by laser Doppler blood perfusion imaging and enhanced capillary proliferation and vascularization in the ischemic muscles. Adjunctive topical application of ABCB5+ MSCs onto therapy-refractory DFUs elicited median wound surface area reductions from baseline of 59% (full analysis set, n = 23), 64% (per-protocol set, n = 20) and 67% (subgroup of responders, n = 17) at week 12, while no treatment-related adverse events were observed. Conclusions The present observations identify GMP-manufactured ABCB5+ dermal MSCs as a potential, safe candidate for adjunctive therapy of otherwise incurable DFUs and justify the conduct of a larger, randomized controlled trial to validate the clinical efficacy. Trial registration: ClinicalTrials.gov, NCT03267784, Registered 30 August 2017, https://clinicaltrials.gov/ct2/show/NCT03267784 Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03156-9.
Collapse
Affiliation(s)
- Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | - Sabrina Klingele
- TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Michael Jünger
- Department of Dermatology, University Hospital Greifswald, Greifswald, Germany
| | | | - Georg Daeschlein
- Department of Dermatology, University Hospital Greifswald, Greifswald, Germany.,Clinic of Dermatology, Immunology and Allergology, Medical University Brandenburg "Theodor Fontane" Medical Center Dessau, Dessau, Germany
| | - Lutz Stemler
- Diabetologikum DDG Ludwigshafen, Ludwigshafen, Germany
| | | | | | | | | | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | - Samar Sadeghi
- TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Seda Ballikaya
- TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | - Martin Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Ana M Waaga-Gasser
- Department of Surgery, University Hospital Würzburg, Würzburg, Germany.,Division of Renal (Kidney) Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - George F Murphy
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dennis P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Natasha Y Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA.,Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Christoph Ganss
- RHEACELL GmbH & Co. KG, Heidelberg, Germany.,TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany
| | | | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.,School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Mark A Kluth
- RHEACELL GmbH & Co. KG, Heidelberg, Germany. .,TICEBA GmbH, Im Neuenheimer Feld 517, 69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Yao W, Dong H, Qi J, Zhang Y, Shi L. Safety and efficacy of mesenchymal stem cells in severe/critical patients with COVID-19: A systematic review and meta-analysis. EClinicalMedicine 2022; 51:101545. [PMID: 35844767 PMCID: PMC9270852 DOI: 10.1016/j.eclinm.2022.101545] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The present study aims to better understand the efficacy and safety of mesenchymal stromal cells (MSCs) in treating severe/critical patients with COVID-19. METHODS PubMed, the Cochrane Library, and the Chinese electronic database CNKI were searched from inception up to Dec 19, 2021. Original comparative studies for MSC treatment + standard treatment for severe/critical patients with COVID-19, with placebo or standard treatment as the control group, were included. The primary outcomes were in-hospital mortality and adverse events (AEs). A meta-analysis was performed to compare the mortality rates between the two groups. Then, a subgroup analysis was performed according to the category of the disease (severe or critical) and MSC dose. Afterwards, a descriptive analysis was performed for AEs and secondary outcomes. The funnel plot and Egger's test were used for the publication bias assessment. FINDINGS Compared to placebo or standard care, MSCs provide significant benefit in the treatment of patients with severe/critical COVID-19, in terms of in-hospital mortality rate (odds ratio: 0.52, 95% CI 0.32-0.84), with very low heterogeneity (P=0.998 [Q test], I 2=0.0%) and less AEs. No significant difference was found in mortality rate due to the different disease categories or MSC doses. Furthermore, no publication bias was found. INTERPRETATION The present study demonstrates that MSCs are highly likely to reduce mortality and are safe to use for patients with severe or critical COVID-19, regardless of whether 1-3 doses are applied. However, due to the small sample size of the included studies, further high-quality, large-scale trials are needed to confirm this statement in the future. FUNDING The National Key Research and Development Program of China (No. 2020YFC0860900), the Science and Technology Project of Wuhan (No. 2020020602012112), the Tianjin Science and Technology Research Program (18PTSYJC00070 and 16PTWYHZ00030), Haihe Laboratory of Cell Ecosystem Innovation Fund (HH22KYZX0046), and the Tianjin Free Trade Zone Innovation Development Project (ZMCY-03-2021002-01) funded the study. We are also grateful for the support from the 3551 Talent Plan of China Optics Valley.
Collapse
Affiliation(s)
- Weiqi Yao
- Department of Hematology, Union Hospital, Tong Ji Medical College, Hua Zhong University of Science and Technology, Hubei, China
- School of Biological Engineering and Food, Hubei University of Technology, Wuhan, Hubei, China
- Wuhan Optics Valley Vcanbio Cell & Gene Technology Co., Ltd., Hubei, China
- Wuhan Optics Valley Zhongyuan Pharmaceutical Co., Ltd., Hubei, China
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, China
| | - Haibo Dong
- Wuhan Optics Valley Vcanbio Cell & Gene Technology Co., Ltd., Hubei, China
- Wuhan Optics Valley Zhongyuan Pharmaceutical Co., Ltd., Hubei, China
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, China
| | - Ji Qi
- Wuhan Optics Valley Zhongyuan Pharmaceutical Co., Ltd., Hubei, China
| | - Yu Zhang
- Wuhan Optics Valley Vcanbio Cell & Gene Technology Co., Ltd., Hubei, China
- Wuhan Optics Valley Zhongyuan Pharmaceutical Co., Ltd., Hubei, China
- Hubei Engineering Research Center for Human Stem Cell Preparation, Application and Resource Preservation, Wuhan, China
- VCANBIO Cell & Gene Engineering Corp., Ltd., No. 12 Meiyuan Road, Tianjin, China
- State Industrial Base for Stem Cell Engineering Products, Tianjin, China
- Corresponding authors.
| | - Lei Shi
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, No. 100 Xi Si Huan Middle Road, Fengtai District, Beijing, China
- Corresponding authors.
| |
Collapse
|
17
|
Gentile P. Hair Loss and Telogen Effluvium Related to COVID-19: The Potential Implication of Adipose-Derived Mesenchymal Stem Cells and Platelet-Rich Plasma as Regenerative Strategies. Int J Mol Sci 2022; 23:9116. [PMID: 36012383 PMCID: PMC9409133 DOI: 10.3390/ijms23169116] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
The diffusion of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) inducing coronavirus disease 2019 (COVID-19) has increased the incidence of several dermatological disorders, including hair loss (HL). This article aims to review the literature regarding the incidence of HL and telogen effluvium (TE) in COVID-19 patients and critically appraise the available evidence regarding the role of regenerative strategies like Platelet-Rich Plasma (PRP) and Human Follicle Stem Cells (HFSCs). A literature review regarding the correlation of HL and TE in COVID-19 patients analyzing the biomolecular pathway involved and the role of regenerative strategies was performed using PubMed, MEDLINE, Embase, PreMEDLINE, Scopus, and the Cochrane databases. Observational studies revealed an escalated incidence of pattern HL and TE in COVID-19 patients. Psychological stress, systemic inflammation, and oxidative stress are potential culprits. Proinflammatory cytokines and stress hormones negatively affect the normal metabolism of proteoglycans. Reduced anagenic expression of proteoglycans is a potential mediating mechanism that connects HL to COVID-19. Currently, only one study has been published on PRP against HL in COVID-19 patients. Further controlled trials are required to confirm PRP and HFSCs efficacy in COVID-19 patients.
Collapse
Affiliation(s)
- Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Surgical Science, "Tor Vergata" University, 00133 Rome, Italy
| |
Collapse
|
18
|
Hoang DM, Pham PT, Bach TQ, Ngo ATL, Nguyen QT, Phan TTK, Nguyen GH, Le PTT, Hoang VT, Forsyth NR, Heke M, Nguyen LT. Stem cell-based therapy for human diseases. Signal Transduct Target Ther 2022; 7:272. [PMID: 35933430 PMCID: PMC9357075 DOI: 10.1038/s41392-022-01134-4] [Citation(s) in RCA: 435] [Impact Index Per Article: 145.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023] Open
Abstract
Recent advancements in stem cell technology open a new door for patients suffering from diseases and disorders that have yet to be treated. Stem cell-based therapy, including human pluripotent stem cells (hPSCs) and multipotent mesenchymal stem cells (MSCs), has recently emerged as a key player in regenerative medicine. hPSCs are defined as self-renewable cell types conferring the ability to differentiate into various cellular phenotypes of the human body, including three germ layers. MSCs are multipotent progenitor cells possessing self-renewal ability (limited in vitro) and differentiation potential into mesenchymal lineages, according to the International Society for Cell and Gene Therapy (ISCT). This review provides an update on recent clinical applications using either hPSCs or MSCs derived from bone marrow (BM), adipose tissue (AT), or the umbilical cord (UC) for the treatment of human diseases, including neurological disorders, pulmonary dysfunctions, metabolic/endocrine-related diseases, reproductive disorders, skin burns, and cardiovascular conditions. Moreover, we discuss our own clinical trial experiences on targeted therapies using MSCs in a clinical setting, and we propose and discuss the MSC tissue origin concept and how MSC origin may contribute to the role of MSCs in downstream applications, with the ultimate objective of facilitating translational research in regenerative medicine into clinical applications. The mechanisms discussed here support the proposed hypothesis that BM-MSCs are potentially good candidates for brain and spinal cord injury treatment, AT-MSCs are potentially good candidates for reproductive disorder treatment and skin regeneration, and UC-MSCs are potentially good candidates for pulmonary disease and acute respiratory distress syndrome treatment.
Collapse
Affiliation(s)
- Duc M Hoang
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam.
| | - Phuong T Pham
- Department of Cellular Therapy, Vinmec High-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Trung Q Bach
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Anh T L Ngo
- Department of Cellular Therapy, Vinmec High-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Quyen T Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Trang T K Phan
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Giang H Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Phuong T T Le
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Van T Hoang
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Nicholas R Forsyth
- Institute for Science & Technology in Medicine, Keele University, Keele, UK
| | - Michael Heke
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Liem Thanh Nguyen
- Department of Research and Development, Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Vietnam
| |
Collapse
|
19
|
Wiercinska E, Bönig H. Zelltherapie in den Zeiten von SARS-CoV-2. TRANSFUSIONSMEDIZIN 2022. [DOI: 10.1055/a-1720-7975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
ZusammenfassungEin breites Spektrum von Disruptionen, aber auch blitzschnelle Innovationen, hat
die SARS-CoV-2 Pandemie gebracht. Dieser Übersichtsartikel betrachtet
die Pandemie aus der Warte der Zelltherapie; konkret werden vier Aspekte
untersucht: Wie unterscheiden sich die Risiken von Zelltherapie-Patienten mit
SARS-CoV-2 Infektion und COVID von denen der Allgemeinbevölkerung? Sind
Empfänger von Zelltherapien, hier speziell autologe und allogene
Stammzelltransplantationsempfänger sowie Empfänger von
CAR-T-Zell-Präparaten, klinisch relevant durch SARS-CoV-2 Vakzine
immunisierbar? Welche Auswirkungen hat die Pandemie mit Spenderausfallrisiko und
Zusammenbruch von Supply Chains auf die Versorgung mit Zelltherapeutika? Gibt es
Zelltherapeutika, die bei schwerem COVID therapeutisch nutzbringend eingesetzt
werden können? In aller Kürze, das erwartete massiv
erhöhte Risiko von Zelltherapie-Patienten, im Infektionsfall einen
schweren Verlauf zu erleiden oder zu sterben, wurde bestätigt. Die
Vakzine induziert jedoch bei vielen dieser Patienten humorale und
zelluläre Immunität, wenn auch weniger zuverlässig als
bei Gesunden. Dank kreativer Lösungen gelang es, die Versorgung mit
Zelltherapeutika im Wesentlichen uneingeschränkt aufrecht zu erhalten.
SARS-CoV-2-spezifische T-Zell-Präparate für den adoptiven
Immuntransfer wurden entwickelt, eine therapeutische Konstellation diese
anzuwenden ergab sich jedoch nicht. Therapiestudien mit mesenchymalen
Stromazellen beim schweren COVID laufen weltweit; die Frage der Wirksamkeit
bleibt zurzeit offen, bei jedoch substanziellem Optimismus in der Szene. Einige
der Erkenntnisse und Innovationen aus der SARS-CoV-2-Pandemie können
möglicherweise verallgemeinert werden und so auf die Zeit nach ihrem
Ende langfristig nachwirken.
Collapse
Affiliation(s)
- Eliza Wiercinska
- DRK-Blutspendedienst Baden-Württemberg-Hessen, Institut
Frankfurt, Frankfurt a.M
| | - Halvard Bönig
- DRK-Blutspendedienst Baden-Württemberg-Hessen, Institut
Frankfurt, Frankfurt a.M
- Goethe Universität, Institut für Transfusionsmedizin
und Immunhämatologie, Frankfurt a.M
- University of Washington, Seattle, WA
| |
Collapse
|
20
|
Ren Y, Liu J, Xu H, Wang S, Li S, Xiang M, Chen S. Knockout of integrin β1 in induced pluripotent stem cells accelerates skin-wound healing by promoting cell migration in extracellular matrix. Stem Cell Res Ther 2022; 13:389. [PMID: 35908001 PMCID: PMC9338467 DOI: 10.1186/s13287-022-03085-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/27/2021] [Indexed: 11/10/2022] Open
Abstract
Background Induced pluripotent stem cells (iPSCs) have the potential to promote wound healing; however, their adhesion to the extracellular matrix (ECM) might decrease iPSC migration, thereby limiting their therapeutic potential. Integrin β1 (Itgb1) is the major integrin subunit that mediates iPSC-ECM adhesion, suggesting that knocking out Itgb1 might be an effective method for enhancing the therapeutic efficacy of iPSCs. Methods We knocked out Itgb1 in mouse iPSCs and evaluated its effects on the therapeutic potential of topically applied iPSCs, as well as their underlying in vivo and in vitro mechanisms. Results The Itgb1-knockout (Itgb1-KO) did not change iPSC pluripotency, function, or survival in the absence of embedding in an ECM gel but did accelerate wound healing, angiogenesis, blood perfusion, and survival in skin-wound lesions. However, embedding in an ECM gel inhibited the in vivo effects of wild-type iPSCs but not those of Itgb1-knockout iPSCs. Additionally, in vitro results showed that Itgb1-knockout decreased iPSC-ECM adhesion while increasing ECM-crossing migration. Moreover, ECM coating on the culture surface did not change cell survival, regardless of Itgb1 status; however, the in vivo and in vitro functions of both Itgb1-knockout and wild-type iPSCs were not affected by the presence of agarose gel, which does not contain integrin-binding sites. Knockout of Integrin α4 (Itga4) did not change the above-mentioned cellular and therapeutic functions of iPSCs. Conclusions Itgb1-knockout increased iPSCs migration and the wound-healing-promoting effect of topically applied iPSCs. These findings suggest the inhibition of Itgb1 expression is a possible strategy for increasing the efficacy of iPSC therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03085-7.
Collapse
Affiliation(s)
- Yansong Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jinbo Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Huijun Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Shun Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Shirui Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Meng Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Sifeng Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
21
|
Behrangi E, Moradi S, Ghassemi M, Goodarzi A, Hanifnia A, Zare S, Nouri M, Dehghani A, Seifadini A, Nilforoushzadeh MA, Roohaninasab M. The investigation of the efficacy and safety of stromal vascular fraction in the treatment of nanofat-treated acne scar: a randomized blinded controlled clinical trial. Stem Cell Res Ther 2022; 13:298. [PMID: 35841057 PMCID: PMC9284502 DOI: 10.1186/s13287-022-02957-2] [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: 02/20/2022] [Accepted: 05/06/2022] [Indexed: 01/16/2023] Open
Abstract
Background Acne is the most common skin disorder which is known as a chronic inflammatory disease with psychological burden and reduced quality of life. Adipose tissue-derived stromal vascular fraction (SVF) is recognized as a source of regenerative cells and improves the quality of skin by increasing collagen content. To date, a few studies have been performed on the therapeutic role of SVF in the treatment of acne scars. Methods This randomized, single-blinded clinical trial was performed on 7 patients with acne scars. In all patients, the initial grade of acne (volume, area and depth) was evaluated and ultrasound of the relevant scar was performed to evaluate neocollagenesis. As a spilt face study, for treating the scars, we used nanofat subcutaneously on one side of the face (control group) and combination of nanofat subcutaneously and SVF intradermally on the opposite side (intervention group). The patients were evaluated for severity of acne by visioface after one month, also for thickness of epidermis and dermis by ultrasound after one month and three months. Results All of the apparent findings of scars improved in two groups after one month, but these changes were significant just for the group treated with SVF (p value < 0.05). Epidermal, dermal and complete thicknesses during the first month in both control and intervention groups were significantly increased (p value < 0.05) but between the first and third months, there was no significant difference in the variables (p value > 0.05). The findings showed that dermal and complete thicknesses of the skin in the first month were different between two groups significantly (p value: 0.042 and 0.040, respectively). Conclusion The use of SVF in the treatment of patients with acne scars accelerates the improvement of volume, area and depth of the scar by increasing collagen content and the dermal thickness, so it can be used as a potentially effective treatment for these patients.
Collapse
Affiliation(s)
- Elham Behrangi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Moradi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Ghassemi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Goodarzi
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Hanifnia
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Nouri
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Dehghani
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Seifadini
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran. .,Skin Repair Research Center, Jordan Dermatology and Hair Transplantation Center, Tehran, Iran.
| | - Masoumeh Roohaninasab
- Department of Dermatology, Rasool Akram Medical Complex Clinical Research Development Center (RCRDC), School of Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Karadag Sari EC, Ovali E. Factors affecting cell viability and the yield of adipose-derived stromal vascular fraction. J Plast Surg Hand Surg 2022; 56:249-254. [PMID: 35819816 DOI: 10.1080/2000656x.2022.2097250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The stromal vascular fraction (SVF) is isolated from adipose tissue and has tremendous regenerative potential for proliferation and differentiation. This study aimed to investigate the factors affecting the cell yield and viability of the SVF to improve the outcomes of its clinical applications and enhance its clinical usage. We performed a retrospective analysis with 121 patients who underwent liposuction to harvest adipose-derived SVF. We recorded patient demographic and clinical characteristics, including age, sex, body mass index (BMI), blood type, medical comorbidities, and smoking and alcohol consumption. As for operative variables, we noted the amount of lipoaspirate and the donor areas, including the lower and entire abdomen. The viability and the cell count of SVF were documented. Sex was a statistically significant factor for viability rate (p < 0.015) and cell count (p < 0.009). Men had higher viability, while women had higher cell counts. We found a statistically significant difference in the presence of hypertension (p = 0.024) and alcohol consumption (p = 0.024). There was a statistically significant relationship between cell count and age (p < 0.001), BMI (p = 0.006), and amount of lipoaspirate (p < 0.001). Sex had significant associations with cell count and viability, while age, BMI, and lipoaspirate amount were significantly associated with cell count. Hypertension and alcohol consumption significantly affected cell count, which is the first such report of this association. Surgeons could apply this knowledge to patient selection for optimal treatment outcomes. Additionally, understanding these factors can help manage patient expectations.
Collapse
Affiliation(s)
- E Cigdem Karadag Sari
- Plastic, Reconstructive and Aesthetic Surgery, Acibadem University, School of Medicine, Acibadem Altunizade Hospital, Istanbul, Turkey
| | - Ercument Ovali
- Hematology, Acibadem University, School of Medicine, Acibadem Labcell Cellular Therapy Facility, Istanbul, Turkey
| |
Collapse
|
23
|
Tammaro A, Adebanjo GAR, Parisella FR, Luzi F, Scarabello A. The hair and nail manifestations of COVID-19. J Cosmet Dermatol 2022; 21:1339-1346. [PMID: 35032337 DOI: 10.1111/jocd.14774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/12/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022]
Abstract
Emerging literature evidence shows that the manifestations of the coronavirus disease 2019 (COVID-19), which is the disease caused by SARS-CoV-2, encompass alterations of the pulmonary, cardiovascular, gastrointestinal and neurological system. Moreover, hematologic and dermatologic manifestations have been documented. The aim of this review is to summarize the dermatologic manifestations of COVID-19 involving the hair and nails in a narrative way. A total of 1136 patients have been reported to have de-novo hair loss following COVID-19. Notably, 958 patients experienced telogen effluvium (TE) (female/male ratio = 3,86:1), two female patients experienced anagen effluvium and 176 people had alopecia areata (female/male ratio of 19:3). Ten patients were reported to have ungual changes following the infection with the novel coronavirus: the individuals affected were 6 women and 4 men. COVID-19 can be associated to hair and ungual manifestations. This review summarizes the evidence regarding the hair and ungual manifestations of COVID-19, which could be harnessed to better understand the clinical implications and pathophysiology of this disease that has been burdening society globally since December 2019.
Collapse
Affiliation(s)
- Antonella Tammaro
- NESMOS Dermatology Department, Sapienza University of Rome, Rome, Italy
| | | | | | - Fabiola Luzi
- NESMOS Dermatology Department, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
24
|
Al-Ghadban S, Artiles M, Bunnell BA. Adipose Stem Cells in Regenerative Medicine: Looking Forward. Front Bioeng Biotechnol 2022; 9:837464. [PMID: 35096804 PMCID: PMC8792599 DOI: 10.3389/fbioe.2021.837464] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
Over the last decade, stem cell-based regenerative medicine has progressed to clinical testing and therapeutic applications. The applications range from infusions of autologous and allogeneic stem cells to stem cell-derived products. Adult stem cells from adipose tissue (ASCs) show significant promise in treating autoimmune and neurodegenerative diseases, vascular and metabolic diseases, bone and cartilage regeneration and wound defects. The regenerative capabilities of ASCs in vivo are primarily orchestrated by their secretome of paracrine factors and cell-matrix interactions. More recent developments are focused on creating more complex structures such as 3D organoids, tissue elements and eventually fully functional tissues and organs to replace or repair diseased or damaged tissues. The current and future applications for ASCs in regenerative medicine are discussed here.
Collapse
Affiliation(s)
| | | | - Bruce A. Bunnell
- Department of Microbiology Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
25
|
Miguez-Rey E, Choi D, Kim S, Yoon S, Săndulescu O. Monoclonal antibody therapies in the management of SARS-CoV-2 infection. Expert Opin Investig Drugs 2022; 31:41-58. [PMID: 35164631 PMCID: PMC8862171 DOI: 10.1080/13543784.2022.2030310] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/13/2022] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Neutralizing antibodies (NAbs) that target key domains of the spike protein in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may have therapeutic value because of their specificity. Depending on the targeted epitope, single agents may be effective, but combined treatment involving multiple NAbs may be necessary to prevent the emergence of resistant variants. AREAS COVERED This article highlights the accelerated regulatory processes established to facilitate the review and approval of potential therapies. An overview of treatment approaches for SARS-CoV-2 infection, with detailed examination of the preclinical and clinical evidence supporting the use of NAbs, is provided. Finally, insights are offered into the potential benefits and challenges associated with the use of these agents. EXPERT OPINION NAbs offer an effective, evidence-based therapeutic intervention during the early stages of SARS-CoV-2 infection when viral replication is the primary factor driving disease progression. As the pandemic progresses, appropriate use of NAbs will be important to minimize the risk of escape variants. Ultimately, the availability of effective treatments for COVID-19 will allow the establishment of treatment algorithms for minimizing the substantial rates of hospitalization, morbidity (including long COVID) and mortality currently associated with the disease.
Collapse
Affiliation(s)
- Enrique Miguez-Rey
- Enfermedades Infecciosas, Hospital Universitario de A Coruña, A Coruña, Spain
| | - Dasom Choi
- Celltrion Healthcare Co., Ltd., Incheon, Republic of Korea
| | - Seungmin Kim
- Celltrion Healthcare Co., Ltd., Incheon, Republic of Korea
| | - Sangwook Yoon
- Celltrion Healthcare Co., Ltd., Incheon, Republic of Korea
| | - Oana Săndulescu
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, National Institute for Infectious Diseases 'Prof. Dr. Matei Balș', Bucharest, Romania
| |
Collapse
|
26
|
Soni N, Gupta S, Rawat S, Krishnakumar V, Mohanty S, Banerjee A. MicroRNA-Enriched Exosomes from Different Sources of Mesenchymal Stem Cells Can Differentially Modulate Functions of Immune Cells and Neurogenesis. Biomedicines 2021; 10:biomedicines10010069. [PMID: 35052749 PMCID: PMC8772751 DOI: 10.3390/biomedicines10010069] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 01/10/2023] Open
Abstract
Adult Mesenchymal stem cells-derived exosomes carry several biologically active molecules that play prominent roles in controlling disease manifestations. The content of these exosomes, their functions, and effect on the immune cells may differ depending on their tissue sources. Therefore, in this study, we purified the exosomes from three different sources and, using the RNA-Seq approach, highly abundant microRNAs were identified and compared between exosomes and parental cells. The effects of exosomes on different immune cells were studied in vitro by incubating exosomes with PBMC and neutrophils and assessing their functions. The expression levels of several miRNAs varied within the different MSCs and exosomes. Additionally, the expression profile of most of the miRNAs was not similar to that of their respective sources. Exosomes isolated from different sources had different abilities to induce the process of neurogenesis and angiogenesis. Moreover, these exosomes demonstrated their varying effect on PBMC proliferation, neutrophil survival, and NET formation, highlighting their versatility and broad interaction with immune cells. The knowledge gained from this study will improve our understanding of the miRNA landscape of exosomes from hMSCs and provide a resource for further improving our understanding of exosome cargo and their interaction with immune cells.
Collapse
Affiliation(s)
- Naina Soni
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, India; (N.S.); (S.R.)
| | - Suchi Gupta
- DBT-Centre of Excellence for Stem Cell Research, Stem Cell Facility, All India Institute of Medical Sciences, New Delhi 110029, India; (S.G.); (V.K.)
| | - Surender Rawat
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, India; (N.S.); (S.R.)
| | - Vishnu Krishnakumar
- DBT-Centre of Excellence for Stem Cell Research, Stem Cell Facility, All India Institute of Medical Sciences, New Delhi 110029, India; (S.G.); (V.K.)
| | - Sujata Mohanty
- DBT-Centre of Excellence for Stem Cell Research, Stem Cell Facility, All India Institute of Medical Sciences, New Delhi 110029, India; (S.G.); (V.K.)
- Correspondence: (S.M.); (A.B.)
| | - Arup Banerjee
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, India; (N.S.); (S.R.)
- Correspondence: (S.M.); (A.B.)
| |
Collapse
|
27
|
Costela Ruiz VJ, Melguizo Rodríguez L, Illescas Montes R, García Recio E, Arias Santiago S, Ruiz C, De Luna Bertos E. Human adipose tissue-derived mesenchymal stromal cells and their phagocytic capacity. J Cell Mol Med 2021; 26:178-185. [PMID: 34854223 PMCID: PMC8742185 DOI: 10.1111/jcmm.17070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 11/30/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have evidenced considerable therapeutic potential in numerous clinical fields, especially in tissue regeneration. The immunological characteristics of this cell population include the expression of Toll‐like receptors and mannose receptors, among others. The study objective was to determine whether MSCs have phagocytic capacity against different target particles. We isolated and characterized three human adipose tissue MSC (HAT‐MSC) lines from three patients and analysed their phagocytic capacity by flow cytometry, using fluorescent latex beads, and by transmission electron microscopy, using Escherichia coli, Staphylococcus aureus and Candida albicans as biological materials and latex beads as non‐biological material. The results demonstrate that HAT‐MSCs can phagocyte particles of different nature and size. The percentage of phagocytic cells ranged between 33.8% and 56.2% (mean of 44.37% ± 11.253) according to the cell line, and a high phagocytic index was observed. The high phagocytic capacity observed in MSCs, which have known regenerative potential, may offer an advance in the approach to certain local and systemic infections.
Collapse
Affiliation(s)
- Víctor J Costela Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Lucía Melguizo Rodríguez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Rebeca Illescas Montes
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Enrique García Recio
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Salvador Arias Santiago
- Biosanitary Research Institute, ibs Granada, Granada, Spain.,Surgical Medical Dermatology and Venereology Service, Department of Medicine, Virgen de las Nieves Hospital, Granada, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain.,Institute of Neuroscience, Centre for Medical Research (CIBM), Health Technology Park (PTS), University of Granada, Granada, Spain
| | - Elvira De Luna Bertos
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| |
Collapse
|
28
|
Gentile P, Alves R, Cole JP, Andjelkov K, Van Helmelryck T, Fernandez J, Trivisonno A, Guillaume L, Verpaele A, Tonnard P, Magalon J, Magalon G, Menkes S. AIRMESS - Academy of International Regenerative Medicine & Surgery Societies: recommendations in the use of platelet-rich plasma (PRP), autologous stem cell-based therapy (ASC-BT) in androgenetic alopecia and wound healing. Expert Opin Biol Ther 2021; 21:1443-1449. [PMID: 33769906 DOI: 10.1080/14712598.2021.1908995] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/23/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Many investigations showed that platelet-rich plasma (PRP), human follicles stem cells (HFSCs), and adipose-derived stem cells (ASCs), considered autologous stem cell-based therapy (ASC-BT), are effective for hair regrowth (HR) in patients affected by androgenetic alopecia and for wound healing (WH). The aim of this article is to analyze the in vitro and in vivo impact of different PRP, HFSCs, and ASCs preparation methods on HR and in WH. AREAS COVERED The analyzed data intended to clarify the molecular mechanism in which PRP, HFSCs, and ASCs are involved, the clinical use and related indications, fully respecting the European rules. Comparative studies between different systems of PRP, HFSCs, and ASCs preparation revealed differences in terms of HR and WH. EXPERT OPINION Despite a lack of standardized protocols, there is convincing evidence with objective measurement modalities that display positive outcomes of ASC-BT in HR and WH.
Collapse
Affiliation(s)
- Pietro Gentile
- Plastic and Reconstructive Surgery, Surgical Science Department, University of Rome "Tor Vergata", Rome, Italy
- Scientific Director of AIRMESS, Academy of International Regenerative Medicine & Surgery Societies, Geneva, Switzerland
| | - Rubina Alves
- Dermatology Department, Hospital Central Do Funchal, Funchal, Portugal
- Treasurer of AIRMESS, Academy of International Regenerative Medicine & Surgery Societies, Geneva, Switzerland
| | - John P Cole
- Cole Hair Transplant Group, Alpharetta, GA, USA
- Vice President of AIRMESS, Academy of International Regenerative Medicine & Surgery Societies, Geneva, Switzerland
| | - Katarina Andjelkov
- Research Associate, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Secretary of AIRMESS, Academy of International Regenerative Medicine & Surgery Societies, Geneva, Switzerland
| | | | - Jonathan Fernandez
- Plastic-Reconstructive and Hand Surgery Unit, University Hospital of Nice, Nice, France
| | - Angelo Trivisonno
- Department of Surgical Science, University of Rome "La Sapienza", Rome, Italy
| | | | - Alexis Verpaele
- Plastic Surgery Unit, Coupure Center for Plastic Surgery, Ghent, Belgium
| | - Patrick Tonnard
- Founder of Coupure Center for Plastic Surgery, Ghent, Belgium
| | - Jeremy Magalon
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Guy Magalon
- Plastic Surgeon, Plastic Surgery Department, Assistance Publique Hôpitaux De Marseille (APHM), Aix Marseille University, Marseille, France
- Honorary President of AIRMESS, Academy of International Regenerative Medicine & Surgery Societies, Geneva, Switzerland
| | - Sophie Menkes
- Forever Institute, Geneva, Switzerland
- President of AIRMESS, Academy of International Regenerative Medicine & Surgery Societies, Geneva, Switzerland
| |
Collapse
|
29
|
Kim EY, Kim HS, Hong KS, Chung HM, Park SP, Noh G. Mesenchymal stem/stromal cell therapy in atopic dermatitis and chronic urticaria: immunological and clinical viewpoints. Stem Cell Res Ther 2021; 12:539. [PMID: 34635172 PMCID: PMC8503727 DOI: 10.1186/s13287-021-02583-4] [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: 06/14/2021] [Accepted: 08/30/2021] [Indexed: 12/29/2022] Open
Abstract
Allergic diseases are immune-mediated diseases. Allergies share a common immunopathogenesis, with specific differences according to the specific disease. Mesenchymal stem/stromal cells (MSCs) have been applied to people suffering from allergic and many other diseases. In this review, the immunologic roles of MSCs are systemically reviewed according to disease immunopathogenesis from a clinical viewpoint. MSCs seem to be a promising therapeutic modality not only as symptomatic treatments but also as causative and even preventive treatments for allergic diseases, including atopic dermatitis and chronic urticaria.
Collapse
Affiliation(s)
| | - Hyuk Soon Kim
- Department of Biomedical Sciences, College of Natural Science, The Graduate School of Dong-A University, Busan, Korea.,Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
| | | | - Hyung-Min Chung
- Miraecellbio Co., Ltd., Seoul, Korea.,Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Se-Pill Park
- Miraecellbio Co., Ltd., Seoul, Korea. .,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, 63243, Korea.
| | - Geunwoong Noh
- Department of Allergy, Allergy and Clinical Immunology Center, Cheju Halla General Hospital, Doreongno 65, Jeju-si, 63127, Jeju Special Self-Governing Province, Korea.
| |
Collapse
|
30
|
Ligotti ME, Pojero F, Accardi G, Aiello A, Caruso C, Duro G, Candore G. Immunopathology and Immunosenescence, the Immunological Key Words of Severe COVID-19. Is There a Role for Stem Cell Transplantation? Front Cell Dev Biol 2021; 9:725606. [PMID: 34595175 PMCID: PMC8477205 DOI: 10.3389/fcell.2021.725606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/06/2021] [Indexed: 01/08/2023] Open
Abstract
The outcomes of Coronavirus disease-2019 (COVID-19) vary depending on the age, health status and sex of an individual, ranging from asymptomatic to lethal. From an immunologic viewpoint, the final severe lung damage observed in COVID-19 should be caused by cytokine storm, driven mainly by interleukin-6 and other pro-inflammatory cytokines. However, which immunopathogenic status precedes this "cytokine storm" and why the male older population is more severely affected, are currently unanswered questions. The aging of the immune system, i.e., immunosenescence, closely associated with a low-grade inflammatory status called "inflammageing," should play a key role. The remodeling of both innate and adaptive immune response observed with aging can partly explain the age gradient in severity and mortality of COVID-19. This review discusses how aging impacts the immune response to the virus, focusing on possible strategies to rejuvenate the immune system with stem cell-based therapies. Indeed, due to immunomodulatory and anti-inflammatory properties, multipotent mesenchymal stem cells (MSCs) are a worth-considering option against COVID-19 adverse outcomes.
Collapse
Affiliation(s)
- Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
- International Society on Aging and Disease, Fort Worth, TX, United States
| | - Giovanni Duro
- Institute for Biomedical Research and Innovation, National Research Council of Italy, Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
31
|
Abdelgawad M, Bakry NS, Farghali AA, Abdel-Latif A, Lotfy A. Mesenchymal stem cell-based therapy and exosomes in COVID-19: current trends and prospects. Stem Cell Res Ther 2021; 12:469. [PMID: 34419143 PMCID: PMC8379570 DOI: 10.1186/s13287-021-02542-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
Novel coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus-2. The virus causes an exaggerated immune response, resulting in a cytokine storm and acute respiratory distress syndrome, the leading cause of COVID-19-related mortality and morbidity. So far, no therapies have succeeded in circumventing the exacerbated immune response or cytokine storm associated with COVID-19. Mesenchymal stem cells (MSCs), through their immunomodulatory and regenerative activities, mostly mediated by their paracrine effect and extracellular vesicle production, have therapeutic potential in many autoimmune, inflammatory, and degenerative diseases. In this paper, we review clinical studies on the use of MSCs for COVID-19 treatment, including the salutary effects of MSCs on the pathophysiology of COVID-19 and the immunomodulation of the cytokine storm. Ongoing clinical trial designs, cell sources, dose and administration, and populations are summarized, and the paracrine mode of benefit is discussed. We also offer suggestions for optimizing MSC-based therapies, including genetic engineering, strategies for cell surface modification, nanotechnology applications, and combination therapies.
Collapse
Affiliation(s)
- Mai Abdelgawad
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt
| | - Nourhan Saied Bakry
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt
| | - Ahmed A Farghali
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt
| | - Ahmed Abdel-Latif
- Gill Heart Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA. .,College of Medicine, University of Kentucky, Lexington, KY, 40506-0046, USA.
| | - Ahmed Lotfy
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni Suef, 62511, Egypt.
| |
Collapse
|
32
|
Ebrahim N, Dessouky AA, Mostafa O, Hassouna A, Yousef MM, Seleem Y, El Gebaly EAEAM, Allam MM, Farid AS, Saffaf BA, Sabry D, Nawar A, Shoulah AA, Khalil AH, Abdalla SF, El-Sherbiny M, Elsherbiny NM, Salim RF. Adipose mesenchymal stem cells combined with platelet-rich plasma accelerate diabetic wound healing by modulating the Notch pathway. Stem Cell Res Ther 2021; 12:392. [PMID: 34256844 PMCID: PMC8276220 DOI: 10.1186/s13287-021-02454-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/12/2021] [Indexed: 02/08/2023] Open
Abstract
Background Diabetic foot ulceration is a serious chronic complication of diabetes mellitus characterized by high disability, mortality, and morbidity. Platelet-rich plasma (PRP) has been widely used for diabetic wound healing due to its high content of growth factors. However, its application is limited due to the rapid degradation of growth factors. The present study aimed to evaluate the efficacy of combined adipose-derived mesenchymal stem cells (ADSCs) and PRP therapy in promoting diabetic wound healing in relation to the Notch signaling pathway. Methods Albino rats were allocated into 6 groups [control (unwounded), sham (wounded but non-diabetic), diabetic, PRP-treated, ADSC-treated, and PRP+ADSCs-treated groups]. The effect of individual and combined therapy was evaluated by assessing wound closure rate, epidermal thickness, dermal collagen, and angiogenesis. Moreover, gene and protein expression of key elements of the Notch signaling pathway (Notch1, Delta-like canonical Notch ligand 4 (DLL4), Hairy Enhancer of Split-1 (Hes1), Hey1, Jagged-1), gene expression of angiogenic marker (vascular endothelial growth factor and stromal cell-derived factor 1) and epidermal stem cells (EPSCs) related gene (ß1 Integrin) were assessed. Results Our data showed better wound healing of PRP+ADSCs compared to their individual use after 7 and 14 days as the combined therapy caused reepithelialization and granulation tissue formation with a marked increase in area percentage of collagen, epidermal thickness, and angiogenesis. Moreover, Notch signaling was significantly downregulated, and EPSC proliferation and recruitment were enhanced compared to other treated groups and diabetic groups. Conclusions These data demonstrated that PRP and ADSCs combined therapy significantly accelerated healing of diabetic wounds induced experimentally in rats via modulating the Notch pathway, promoting angiogenesis and EPSC proliferation.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt.,Stem Cell Unit, Faculty of Medicine, Benha University, Benha, Egypt
| | - Arigue A Dessouky
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ola Mostafa
- Department of Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt
| | - Amira Hassouna
- School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland, New Zealand
| | - Mohamed M Yousef
- Department of Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt
| | - Yasmin Seleem
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | | | - Mona M Allam
- Department of Medical Physiology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Qalyubia, 13736, Egypt
| | - Bayan A Saffaf
- Department of Pharmacology, Faculty of Pharmacy, Future University, New Cairo, Egypt
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt.,Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University, Cairo, 11562, Egypt
| | - Ahmed Nawar
- Department of General Surgery, Faculty of Medicine, Benha University, Benha, Egypt
| | - Ahmed A Shoulah
- Department of General Surgery, Faculty of Medicine, Benha University, Benha, Egypt
| | - Ahmed H Khalil
- Department of Surgery, & Radiology Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Sami F Abdalla
- Clinical Department, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia.,Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nehal M Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt. .,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt.
| |
Collapse
|
33
|
Cava C, Bertoli G, Castiglioni I. Potential drugs against COVID-19 revealed by gene expression profile, molecular docking and molecular dynamic simulation. Future Virol 2021; 16:10.2217/fvl-2020-0392. [PMID: 34306168 PMCID: PMC8293696 DOI: 10.2217/fvl-2020-0392] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Aim: SARS-CoV-2, an emerging betacoronavirus, is the causative agent of COVID-19. Currently, there are few specific and selective antiviral drugs for the treatment and vaccines to prevent contagion. However, their long-term effects can be revealed after several years, and new drugs for COVID-19 should continue to be investigated. Materials & methods: In the first step of our study we identified, through a gene expression analysis, several drugs that could act on the biological pathways altered in COVID-19. In the second step, we performed a docking simulation to test the properties of the identified drugs to target SARS-CoV-2. Results: The drugs that showed a higher binding affinity are bardoxolone (-8.78 kcal/mol), irinotecan (-8.40 kcal/mol) and pyrotinib (-8.40 kcal/mol). Conclusion: We suggested some drugs that could be efficient in treating COVID-19.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging & Physiology, National Research Council (IBFM-CNR), Via F. Cervi 93, Segrate-Milan, Milan, 20090, Italy
| | - Gloria Bertoli
- Institute of Molecular Bioimaging & Physiology, National Research Council (IBFM-CNR), Via F. Cervi 93, Segrate-Milan, Milan, 20090, Italy
| | - Isabella Castiglioni
- Department of Physics “Giuseppe Occhialini”, University of Milan-Bicocca Piazza dell'Ateneo Nuovo, Milan, 20126, Italy
| |
Collapse
|
34
|
Shetty R, Murugeswari P, Chakrabarty K, Jayadev C, Matalia H, Ghosh A, Das D. Stem cell therapy in coronavirus disease 2019: current evidence and future potential. Cytotherapy 2021; 23:471-482. [PMID: 33257213 PMCID: PMC7649634 DOI: 10.1016/j.jcyt.2020.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023]
Abstract
The end of 2019 saw the beginning of the coronavirus disease 2019 (COVID-19) pandemic that soared in 2020, affecting 215 countries worldwide, with no signs of abating. In an effort to contain the spread of the disease and treat the infected, researchers are racing against several odds to find an effective solution. The unavailability of timely and affordable or definitive treatment has caused significant morbidity and mortality. Acute respiratory distress syndrome (ARDS) caused by an unregulated host inflammatory response toward the viral infection, followed by multi-organ dysfunction or failure, is one of the primary causes of death in severe cases of COVID-19 infection. Currently, empirical management of respiratory and hematological manifestations along with anti-viral agents is being used to treat the infection. The quest is on for both a vaccine and a more definitive management protocol to curtail the spread. Researchers and clinicians are also exploring the possibility of using cell therapy for severe cases of COVID-19 with ARDS. Mesenchymal stromal cells are known to have immunomodulatory properties and have previously been used to treat viral infections. This review explores the potential of mesenchymal stromal cells as cell therapy for ARDS.
Collapse
Affiliation(s)
- Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Institute, Bangalore, India
| | - Ponnalagu Murugeswari
- Stem Cell Research Laboratory, GROW Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | | | - Chaitra Jayadev
- Department of Vitreo-Retinal Surgery, Narayana Nethralaya Eye Institute, Bangalore, India
| | - Himanshu Matalia
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Institute, Bangalore, India
| | - Arkasubhra Ghosh
- GROW Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Debashish Das
- Stem Cell Research Laboratory, GROW Laboratory, Narayana Nethralaya Foundation, Bangalore, India.
| |
Collapse
|
35
|
Role of Fzd6 in Regulating the Osteogenic Differentiation of Adipose-derived Stem Cells in Osteoporotic Mice. Stem Cell Rev Rep 2021; 17:1889-1904. [PMID: 34041696 DOI: 10.1007/s12015-021-10182-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Although it has been demonstrated that adipose-derived stem cells (ASCs) from osteoporotic mice (OP-ASCs) exhibited impaired osteogenic differentiation potential, the molecular mechanism has not yet been elucidated. We found that Fzd6 was decreased in OP-ASCs compared with ASCs. This study investigates effects and underlying mechanisms of Fzd6 in the osteogenic potential of OP-ASCs, and explores methods to enhance osteogenic capacity of OP-ASCs. METHODS Fzd6 overexpression and silencing lentiviruses were used to evaluate the role of Fzd6 in the osteogenic differentiation of OP-ASCs. Real-time PCR (qPCR) and western blotting (WB) was performed to detect the expression of Fzd6 and bone-related molecules, including runt-related transcription factor 2 (Runx2) and osteopontin (Opn). Alizarin red staining and Alkaline phosphatase (ALP) staining were performed following osteogenic induction. Microscopic CT (Micro-CT), hematoxylin and eosin staining (HE) staining, and Masson staining were used to assess the role of Fzd6 in osteogenic differentiation of osteoporosis (OP) mice in vivo. RESULTS Expression of Fzd6 was decreased significantly in OP-ASCs. Fzd6 silencing down-regulated the osteogenic ability of OP-ASCs in vitro. Overexpression of Fzd6 rescued the impaired osteogenic capacity in OP-ASCs in vitro. We obtained similar results in vivo. CONCLUSIONS Fzd6 plays an important role in regulating the osteogenic ability of OP-ASCs both in vivo and in vitro. Overexpression of Fzd6 promotes the osteogenic ability of OP-ASCs, which provides new insights for the prevention and treatment of OP mice.
Collapse
|
36
|
Ngo BT, Marik P, Kory P, Shapiro L, Thomadsen R, Iglesias J, Ditmore S, Rendell M, Varon J, Dubé M, Nanda N, In G, Arkfeld D, Chaudhary P, Campese VM, Hanna DL, Sawcer DE, Ehresmann G, Peng D, Smogorewski M, Armstrong A, Dasgupta R, Sattler F, Brennan-Rieder D, Mussini C, Mitja O, Soriano V, Peschanski N, Hayem G, Confalonieri M, Piccirillo MC, Lobo-Ferreira A, Bello Rivero I, Turkia M, Vingevoll EH, Griffin D, Hung IF. The time to offer treatments for COVID-19. Expert Opin Investig Drugs 2021; 30:505-518. [PMID: 33721548 PMCID: PMC8074648 DOI: 10.1080/13543784.2021.1901883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 12/23/2022]
Abstract
Background: COVID-19 has several overlapping phases. Treatments to date have focused on the late stage of disease in hospital. Yet, the pandemic is by propagated by the viral phase in out-patients. The current public health strategy relies solely on vaccines to prevent disease.Methods: We searched the major national registries, pubmed.org, and the preprint servers for all ongoing, completed and published trial results.Results: As of 2/15/2021, we found 111 publications reporting findings on 14 classes of agents, and 9 vaccines. There were 62 randomized controlled studies, the rest retrospective observational analyses. Only 21 publications dealt with outpatient care. Remdesivir and high titer convalescent plasma have emergency use authorization for hospitalized patients in the U.S.A. There is also support for glucocorticoid treatment of the COVID-19 respiratory distress syndrome. Monoclonal antibodies are authorized for outpatients, but supply is inadequate to treat all at time of diagnosis. Favipiravir, ivermectin, and interferons are approved in certain countries.Expert Opinion: Vaccines and antibodies are highly antigen specific, and new SARS-Cov-2 variants are appearing. We call on public health authorities to authorize treatments with known low-risk and possible benefit for outpatients in parallel with universal vaccination.
Collapse
Affiliation(s)
- Binh T. Ngo
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
- The Rose Salter Medical Research Foundation, Newport Coast, USA
| | - Paul Marik
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Pierre Kory
- Pulmonary and Critical Care Medicine, Aurora St. Luke’s Medical Center, Milwaukee, USA
| | - Leland Shapiro
- Department of Internal Medicine, Rocky Mountain Regional Veterans Affairs Medical Center in Aurora, CO and University of Colorado Anschutz Medical Campus in Aurora, CO Supported by the Emily Foundation, Boston, USA
| | | | - Jose Iglesias
- Department of Internal Medicine, Jersey Shore University Medical Center, Hackensack Meridian School of Medicine at Seton Hall, Neptune, USA
| | | | - Marc Rendell
- The Rose Salter Medical Research Foundation, Newport Coast, USA
| | - Joseph Varon
- United Memorial Medical Center, University of Texas School of Medicine, Houston, USA
| | - Michael Dubé
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Neha Nanda
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Gino In
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Daniel Arkfeld
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Preet Chaudhary
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Vito M. Campese
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Diana L. Hanna
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - David E. Sawcer
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Glenn Ehresmann
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - David Peng
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Miroslaw Smogorewski
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - April Armstrong
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Rajkumar Dasgupta
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | - Fred Sattler
- Department of Internal Medicine, Eastern Virginia Medical School, Pulmonary and Critical Care Medicine, Norfolk, USA
| | | | - Cristina Mussini
- Department of Infectious Disease, University of Modena and Reggio Emilia, Modena, Italy
| | - Oriol Mitja
- Department of Internal Medicine, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Vicente Soriano
- Director, Centro Medico, UNIR Health Sciences School & Medical Center, Madrid, Spain
| | - Nicolas Peschanski
- Department of Emergency Medicine, UniversityHospital of Rennes, Rennes, France
| | - Gilles Hayem
- Department of Rheumatology, Hôpital Paris Saint-Joseph, Paris, France
| | - Marco Confalonieri
- Department of Respiratory Diseases, Azienda Ospedaliero-Universitaria Di Trieste, Trieste, Italia
| | | | - Antonio Lobo-Ferreira
- Unidade De Investigação Cardiovascular (Unic), Faculdade De Medicina, Da Universidade Do Porto, Centro Hospitalar Universitário De São João, Porto, and Hospital Rainha Santa Isabel, Marco De Canaveses, Portugal
| | - Iraldo Bello Rivero
- Department of Clinical Investigations, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | | | - Daniel Griffin
- Department of Internal Medicine, Rocky Mountain Regional Veterans Affairs Medical Center in Aurora, CO and University of Colorado Anschutz Medical Campus in Aurora, CO Supported by the Emily Foundation, Boston, USA
- Department of Internal Medicine and Department of Biochemistry and Molecular Biophysics, ProHEALTH, an OPTUM Company, Columbia University, College of Physicians and Surgeons, USA
| | - Ivan Fn Hung
- Department of Internal Medicine, Rocky Mountain Regional Veterans Affairs Medical Center in Aurora, CO and University of Colorado Anschutz Medical Campus in Aurora, CO Supported by the Emily Foundation, Boston, USA
| |
Collapse
|
37
|
Ni X, Shan X, Xu L, Yu W, Zhang M, Lei C, Xu N, Lin J, Wang B. Adipose-derived stem cells combined with platelet-rich plasma enhance wound healing in a rat model of full-thickness skin defects. Stem Cell Res Ther 2021; 12:226. [PMID: 33823915 PMCID: PMC8022317 DOI: 10.1186/s13287-021-02257-1] [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: 09/21/2020] [Accepted: 03/01/2021] [Indexed: 12/20/2022] Open
Abstract
Background Wound healing is impaired in patients with diabetes due to the multifactorial etiology of the disease, which limits the therapeutic efficacy of various approaches. This study hypothesizes that the combination of adipose-derived stem cells (ADSCs) and platelet-rich plasma (PRP) might achieve optimally efficient diabetic wound healing. Methods ADSCs were isolated from the adipose tissues of Sprague-Dawley (SD) rats. PRP was prepared by using a two-step centrifugation technique. A diabetic wound model was established on the backs of SD rats to evaluate the effect of ADSCs incorporated into PRP. Hematoxylin and eosin staining, immunofluorescence, and immunohistochemistry were performed to observe the changes in neovascularization. ELISA and Western blot were utilized to detect the angiogenesis-related protein expression levels. The proliferation of endothelial cells was assessed by the MTS assay. Results ADSCs incorporated into PRP induced a higher wound closure rate than ADSCs, PRP, and negative control. The expression levels of VEGF, p-STAT3, and SDF-1 in the ADSC+PRP group were higher than those in the other groups. Moreover, the proliferation of endothelial cells was strongly stimulated by treatment with the combination of ADSC-conditioned medium (ADSC-CM) and PRP. Conclusions PRP enhanced diabetic wound healing induced by ADSCs, and its promoting effect involved neovascularization.
Collapse
Affiliation(s)
- Xuejun Ni
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xiuying Shan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Lili Xu
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Wenjun Yu
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Mingliang Zhang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Chen Lei
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Nating Xu
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Junyu Lin
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Biao Wang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
38
|
Gentile P. SARS-CoV-2: the "Uncensored" Truth about Its Origin and Adipose-Derived Mesenchymal Stem Cells as New Potential Immune-Modulatory Weapon. Aging Dis 2021; 12:330-344. [PMID: 33815867 PMCID: PMC7990360 DOI: 10.14336/ad.2021.0121] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 01/21/2021] [Indexed: 12/16/2022] Open
Abstract
In this second return of the pandemic, January 2021, it appears to be clear that a Nano-sized organism, the SARS-CoV-2, has rendered the human race helpless, made the global health status decline, and drowned the world economy. However, it does not appear clear the real origin of the SARS-CoV-2 and the aim of this work is to report and discuss, maybe for the first time since the pandemic began, the scientific data published in this specific field, analyzing the potentially available weapons against the SARS-CoV-2. About this last point, a ray of hope comes from the potential of Mesenchymal Stem Cells (MSCs) that has already been established in Coronavirus Disease 2019 (COVID-19), and in particular from the Adipose-Derived Mesenchymal Stem Cells (AD-MSCs). However, cell-based therapy has its own limits, especially represented by the know-how in this field and by the rules of applications. It was suggested a biological therapy using AD-MSCs as a weapon against COVID-19, as they can be a game-changer owing to their immuno-modulatory nature, which combats the cytokine storm characterizing this disease, and their practical efficiency, which will realistically aid large access to therapy worldwide.
Collapse
Affiliation(s)
- Pietro Gentile
- Department of Surgical Science, Plastic and Reconstructive Surgery, “Tor Vergata” University, Rome, 00133, Italy.
- Founder and Scientific Director of Academy of International Regenerative Medicine & Surgery Societies (AIRMESS), 1201 Geneva, Switzerland.
| |
Collapse
|
39
|
Najafi-Ghalehlou N, Roudkenar MH, Langerodi HZ, Roushandeh AM. Taming of Covid-19: potential and emerging application of mesenchymal stem cells. Cytotechnology 2021; 73:253-298. [PMID: 33776206 PMCID: PMC7982879 DOI: 10.1007/s10616-021-00461-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 02/17/2021] [Indexed: 12/24/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has turned out to cause a pandemic, with a sky scraping mortality. The virus is thought to cause tissue injury by affecting the renin-angiotensin system. Also, the role of the over-activated immune system is noteworthy, leading to severe tissue injury via the cytokine storms. Thus it would be feasible to modulate the immune system response in order to attenuate the disease severity, as well as treating the patients. Today different medicines are being administered to the patients, but regardless of the efficacy of these treatments, adverse effects are pretty probable. Meanwhile, mesenchymal stem cells (MSCs) prove to be an effective candidate for treating the patients suffering from COVID-19 pneumonia, owing to their immunomodulatory and tissue-regenerative potentials. So far, several experiments have been conducted; transplanting MSCs and results are satisfying with no adverse effects being reported. This paper aims to review the recent findings regarding the novel coronavirus and the conducted experiments to treat patients suffering from COVID-19 pneumonia utilizing MSCs.
Collapse
Affiliation(s)
- Nima Najafi-Ghalehlou
- Department of Medical Laboratory Sciences, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehryar Habibi Roudkenar
- Burn and Regenerative Medicine Research Center, Velayat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Medical Biotechnology Department, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Habib Zayeni Langerodi
- Guilan Rheumatology Research Center (GRRC), Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Burn and Regenerative Medicine Research Center, Velayat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Anatomical Sciences Department, Medicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
40
|
Yan J, Liang J, Cao Y, El Akkawi MM, Liao X, Chen X, Li C, Li K, Xie G, Liu H. Efficacy of topical and systemic transplantation of mesenchymal stem cells in a rat model of diabetic ischemic wounds. Stem Cell Res Ther 2021; 12:220. [PMID: 33789742 PMCID: PMC8010295 DOI: 10.1186/s13287-021-02288-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) exert positive effects in chronic wounds. However, critical parameters, such as the most effective administration routes, remain unclear. Accordingly, the purpose of this study was to compare the effects of topical and systemic transplantation MSCs on diabetic ischemic wound healing and explored the underlying mechanisms. METHOD A diabetic ischemic wound model was created on the dorsal foot of type 2 diabetes mellitus (T2DM) rat. Bone marrow-derived mesenchymal stem cells (BM-MSCs) were administered via two routes: topical injection and intravenous (IV) infusion. Wound healing outcomes and blood glucose level were assessed dynamically. Meanwhile, blood flow recovery was evaluated in ischemic gastrocnemius muscles. The homing and transdifferentiation of mKate2-labeled BM-MSCs were assessed by fluorescence imaging and immunohistochemistry (IHC) analysis. RESULT Both topical and systemic treatments had a positive effect on the diabetic ischemic wound showing a significant reduction in wound area at day 14. Histological results showed an increase in the length of epithelial edges, collagen content, microvessel density in the wound bed, and a higher expression of vascular endothelial growth factor (VEGF). Meanwhile, systemic administration can ameliorate hyperglycemia and improve the blood perfusion of the ischemic hindlimb. BM-MSCs administered systemically were found distributed in wounded tissue and transdifferentiated into endothelial cells. Furthermore, BM-MSCs stimulated angiogenesis at wound sites by downregulating phosphatase and tensin homolog (PTEN) and activation of AKT signaling pathway. CONCLUSIONS The results demonstrated that both transplantation delivery method (topical and systemic) of BM-MSCs accelerated wound healing remarkably under pathological conditions. Nevertheless, systemic administration has the potential to ameliorate hyperglycemia and repair the damaged tissue.
Collapse
Affiliation(s)
- Jianxin Yan
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| | - Jiaji Liang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| | - Yingxuan Cao
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| | - Mariya M. El Akkawi
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| | - Xuan Liao
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| | - Xiaojia Chen
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632 People’s Republic of China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou, 510632 People’s Republic of China
- Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangzhou, 510632 People’s Republic of China
- National Engineering Research Center of Genetic Medicine, Guangzhou, 510632 People’s Republic of China
| | - Chengzhi Li
- Department of Interventional Radiology and Vascular Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
| | - Kecheng Li
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| | - Guanghui Xie
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| | - Hongwei Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630 People’s Republic of China
- Innovative Technology Research Institute of Plastic Surgery, Guangzhou, 510630 People’s Republic of China
- Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, 510632 People’s Republic of China
| |
Collapse
|
41
|
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: 0.5] [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
|
42
|
Song N, Wakimoto H, Rossignoli F, Bhere D, Ciccocioppo R, Chen KS, Khalsa JK, Mastrolia I, Samarelli AV, Dominici M, Shah K. Mesenchymal stem cell immunomodulation: In pursuit of controlling COVID-19 related cytokine storm. STEM CELLS (DAYTON, OHIO) 2021; 39:707-722. [PMID: 33586320 PMCID: PMC8014246 DOI: 10.1002/stem.3354] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/26/2021] [Indexed: 11/09/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has grown to be a global public health crisis with no safe and effective treatments available yet. Recent findings suggest that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the coronavirus pathogen that causes COVID-19, could elicit a cytokine storm that drives edema, dysfunction of the airway exchange, and acute respiratory distress syndrome in the lung, followed by acute cardiac injury and thromboembolic events leading to multiorgan failure and death. Mesenchymal stem cells (MSCs), owing to their powerful immunomodulatory abilities, have the potential to attenuate the cytokine storm and have therefore been proposed as a potential therapeutic approach for which several clinical trials are underway. Given that intravenous infusion of MSCs results in a significant trapping in the lung, MSC therapy could directly mitigate inflammation, protect alveolar epithelial cells, and reverse lung dysfunction by normalizing the pulmonary microenvironment and preventing pulmonary fibrosis. In this review, we present an overview and perspectives of the SARS-CoV-2 induced inflammatory dysfunction and the potential of MSC immunomodulation for the prevention and treatment of COVID-19 related pulmonary disease.
Collapse
Affiliation(s)
- Na Song
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroaki Wakimoto
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Filippo Rossignoli
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak Bhere
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Kok-Siong Chen
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jasneet Kaur Khalsa
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ilenia Mastrolia
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging (CSTI), Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
43
|
Richard SA, Kampo S, Sackey M, Hechavarria ME, Buunaaim ADB, Kuugbee ED, Anabah TW. Elucidating the Pivotal Role of Immune Players in the Management of COVID-19: Focus on Mesenchymal Stem Cells and Inflammation. Curr Stem Cell Res Ther 2021; 16:189-198. [PMID: 32628591 DOI: 10.2174/1574888x15666200705213751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 01/08/2023]
Abstract
The world is currently engulfed with a viral disease with no cure. Thus, far, millions of people are infected with the virus across the length and breadth of the world, with thousands losing their lives each passing day. The WHO in February 2020 classified the virus as a coronavirus and the name Coronavirus-19 (CoV-19) was offered to the virus. The disease caused by the virus was termed coronavirus disease-19 (COVID-19). The pathogenesis of COVID-19 is associated with elevation of several immune players as well as inflammatory factors which contribute to cytokine storms. Currently, the detection of CoV-19 RNA is through reverse transcriptase-polymerase chain reaction (RTPCR). Mesenchymal stem cells (MSCs) are capable of suppressing several kinds of cytokines via the paracrine secretion system. Therefore, MSCs therapy could be game changer in the treatment of the current COVID-19 pandemic. Moreover, intravenous IG may be capable of suppressing the high expression of IL-6 by the CoV-19 resulting in lessen disease burden. Anti-inflammatory medications like, corticosteroids, tocilizumab, glycyrrhetinic acid, as well as etoposide may be very advantageous in decreasing the COVID-19 burden because their mode of action targets the cytokine storms initiated by the CoV-19. It is important to indicate that, these medications do not target the virus itself. Therefore, potent CoV-19 anti-viral medications are needed to completely cure patients with COVID-19. Furthermore, a vaccine is urgently needed to stop the spread of the virus. This review, therefore, elucidates the immune players in the management of COVID-19; focusing principally on MSCs and inflammatory mediators.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Medicine, Princefield University, P. O. Box MA128, Ho, Ghana
| | - Sylvanus Kampo
- Department of Anesthesia and Critical care, School of Medicine, University of Health and Allied Sciences, Ho, Ghana
| | - Marian Sackey
- Department of Pharmacy, Ho Teaching Hospital, P.O. Box MA-374, Ho, Ghana
| | | | - Alexis D B Buunaaim
- Department of Surgery, School of Medicine and Health Science, University for Development Studies, Tamale, Ghana
| | - Eugene Dogkotenge Kuugbee
- Department of Clinical Microbiology, School of Medicine and Health Science, University for Development Studies, Tamale, Ghana
| | | |
Collapse
|
44
|
Engineered microtissues for the bystander therapy against cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111854. [PMID: 33579487 DOI: 10.1016/j.msec.2020.111854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022]
Abstract
Thymidine kinase expressing human adipose mesenchymal stem cells (TK-hAMSCs) in combination with ganciclovir (GCV) are an effective platform for antitumor bystander therapy in mice models. However, this strategy requires multiple TK-hAMSCs administrations and a substantial number of cells. Therefore, for clinical translation, it is necessary to find a biocompatible scaffold providing TK-hAMSCs retention in the implantation site against their rapid wash-out. We have developed a microtissue (MT) composed by TKhAMSCs and a scaffold made of polylactic acid microparticles and cell-derived extracellular matrix deposited by hAMSCs. The efficacy of these MTs as vehicles for TK-hAMSCs/GCV bystander therapy was evaluated in a rodent model of human prostate cancer. Subcutaneously implanted MTs were integrated in the surrounding tissue, allowing neovascularization and maintenance of TK-hAMSCs viability. Furthermore, MTs implanted beside tumors allowed TK-hAMSCs migration towards tumor cells and, after GCV administration, inhibited tumor growth. These results indicate that TK-hAMSCs-MTs are promising cell reservoirs for clinical use of therapeutic MSCs in bystander therapies.
Collapse
|
45
|
Zuo Z, Wu T, Pan L, Zuo C, Hu Y, Luo X, Jiang L, Xia Z, Xiao X, Liu J, Ye M, Deng M. Modalities and Mechanisms of Treatment for Coronavirus Disease 2019. Front Pharmacol 2021; 11:583914. [PMID: 33643033 PMCID: PMC7908061 DOI: 10.3389/fphar.2020.583914] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is spreading rapidly throughout the world. Although COVID-19 has a relatively low case severity rate compared to SARS and Middle East Respiratory syndrome it is a major public concern because of its rapid spread and devastating impact on the global economy. Scientists and clinicians are urgently trying to identify drugs to combat the virus with hundreds of clinical trials underway. Current treatments could be divided into two major part: anti-viral agents and host system modulatory agents. On one hand, anti-viral agents focus on virus infection process. Umifenovir blocks virus recognizing host and entry. Remdesivir inhibits virus replication. Chloroquine and hydroxychloroquine involve preventing the whole infection process, including virus transcription and release. On the other hand, host system modulatory agents are associated with regulating the imbalanced inflammatory reaction and biased immune system. Corticosteroid is believed to be commonly used for repressing hyper-inflammation, which is one of the major pathologic mechanisms of COVID-19. Convalescent plasma and neutralizing antibodies provide essential elements for host immune system and create passive immunization. Thrombotic events are at high incidence in COVID-19 patients, thus anti-platelet and anti-coagulation are crucial, as well. Here, we summarized these current or reproposed agents to better understand the mechanisms of agents and give an update of present research situation.
Collapse
Affiliation(s)
- Zhihong Zuo
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ting Wu
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Department of Cardiovascular Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Liangyu Pan
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Chenzhe Zuo
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yingchuo Hu
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Xuan Luo
- Hunan Yuanpin Cell Biotechnology Co., Ltd., Changsha, China
| | - Liping Jiang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zanxian Xia
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Xiaojuan Xiao
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Jing Liu
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Meichun Deng
- Department of Biochemistry and Molecular Biology and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
46
|
Croci S, Bonacini M, Dolci G, Massari M, Facciolongo N, Pignatti E, Pisciotta A, Carnevale G, Negro A, Cassone G, Muratore F, Belloni L, Zerbini A, Salvarani C. Human Dental Pulp Stem Cells Modulate Cytokine Production in vitro by Peripheral Blood Mononuclear Cells From Coronavirus Disease 2019 Patients. Front Cell Dev Biol 2021; 8:609204. [PMID: 33634100 PMCID: PMC7901970 DOI: 10.3389/fcell.2020.609204] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022] Open
Abstract
A subset of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) developed a condition of hyper-inflammation, which can cause multi-organ damage and the more severe forms of coronavirus disease 2019 (COVID-19). Mesenchymal stem cells (MSCs) can promote tissue regeneration and modulate immune responses and, thus, have the rational requirements to be used to counteract SARS-CoV-2-induced pneumonia and hyper-inflammation. The aim of the present study was to gain insight into possible mechanisms of action of MSCs obtained from human dental pulp [dental pulp stem cells (DPSCs)] in COVID-19 patients. We investigated the concentrations of 18 cytokines in supernatants of peripheral blood mononuclear cells (PBMCs) obtained from COVID-19 patients cultured in vitro alone and in contact with DPSCs. The modulation of cytokines in PBMCs was confirmed by real-time PCR. IL-6 was the sole cytokine detected in supernatants of DPSCs. In resting conditions, co-culture increased IL-1β, IL-2, IL-5, IL-6, IL-10, IL-18, TNFα, and granulocyte macrophage colony-stimulating factor (GM-CSF) levels. When PBMCs were activated with anti-CD3/CD28 antibody-coated beads, co-culture increased IL-6 and GM-CSF, whereas it decreased IFNγ, TNFα, IL-2, IL-5, IL-9, IL-10, IL-12 (p70), IL-17A, IL-18, IL-21, IL-23, and IL-27 levels. Concentrations of IL-1β, IL-4, IL-13, and IL-22 were not affected. The comparison of cytokine concentrations in supernatants of PBMCs from COVID-19 patients vs. healthy subjects revealed lower concentrations of IL-10 and higher concentrations of IL-18 in supernatants of CD3/CD28-activated PBMCs from COVID-19 patients. Results are explorative but indicate that DPSCs can modulate the production of cytokines deregulated in COVID-19 patients, supporting their potential use in COVID-19.
Collapse
Affiliation(s)
- Stefania Croci
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Martina Bonacini
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giovanni Dolci
- Infectious Disease Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marco Massari
- Infectious Disease Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Nicola Facciolongo
- Pulmonology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisa Pignatti
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Pisciotta
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, University of Modena and Reggio Emilia, Modena, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, University of Modena and Reggio Emilia, Modena, Italy
| | - Aurelio Negro
- Internal Medicine and Secondary Hypertension Center, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Cassone
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy.,Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesco Muratore
- Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Lucia Belloni
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandro Zerbini
- Clinical Immunology, Allergy and Advanced Biotechnologies Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Salvarani
- Department of Surgery, Medicine Dentistry and Morphological Sciences with Interest in Transplant, University of Modena and Reggio Emilia, Modena, Italy.,Rheumatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
47
|
Mazini L, Ezzoubi M, Malka G. Overview of current adipose-derived stem cell (ADSCs) processing involved in therapeutic advancements: flow chart and regulation updates before and after COVID-19. Stem Cell Res Ther 2021; 12:1. [PMID: 33397467 PMCID: PMC7781178 DOI: 10.1186/s13287-020-02006-w] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/01/2020] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) have raised big interest in therapeutic applications in regenerative medicine and appear to fulfill the criteria for a successful cell therapy. Their low immunogenicity and their ability to self-renew, to differentiate into different tissue-specific progenitors, to migrate into damaged sites, and to act through autocrine and paracrine pathways have been altogether testified as the main mechanisms whereby cell repair and regeneration occur. The absence of standardization protocols in cell management within laboratories or facilities added to the new technologies improved at patient's bedside and the discrepancies in cell outcomes and engraftment increase the limitations on their widespread use by balancing their real benefit versus the patient safety and security. Also, comparisons across pooled patients are particularly difficult in the fact that multiple medical devices are used and there is absence of harmonized assessment assays despite meeting regulations agencies and efficient GMP protocols. Moreover, the emergence of the COVID-19 breakdown added to the complexity of implementing standardization. Cell- and tissue-based therapies are completely dependent on the biological manifestations and parameters associated to and induced by this virus where the scope is still unknown. The initial flow chart identified for stem cell therapies should be reformulated and updated to overcome patient infection and avoid significant variability, thus enabling more patient safety and therapeutic efficiency. The aim of this work is to highlight the major guidelines and differences in ADSC processing meeting the current good manufacturing practices (cGMP) and the cellular therapy-related policies. Specific insights on standardization of ADSCs proceeding at different check points are also presented as a setup for the cord blood and bone marrow.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Center of Biological and Medical Sciences CIAM, Mohammed VI Polytechnic University (UM6P), Lot 660, Hay Moulay Rachid, 43150 Ben Guerir, Morocco
| | - Mohamed Ezzoubi
- Centre des Brûlés et chirurgie réparatrice, Centre Hospitalier Universitaire Ibn Rochd Casablanca, Faculté de Médecine et de Pharmacie Casablanca, Casablanca, Morocco
| | - Gabriel Malka
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Center of Biological and Medical Sciences CIAM, Mohammed VI Polytechnic University (UM6P), Lot 660, Hay Moulay Rachid, 43150 Ben Guerir, Morocco
| |
Collapse
|
48
|
Abstract
The COVID-19 pandemic has led to a major setback in both the health and economic sectors across the globe. The scale of the problem is enormous because we still do not have any specific anti-SARS-CoV-2 antiviral agent or vaccine. The human immune system has never been exposed to this novel virus, so the viral interactions with the human immune system are completely naive. New approaches are being studied at various levels, including animal in vitro models and human-based studies, to contain the COVID-19 pandemic as soon as possible. Many drugs are being tested for repurposing, but so far only remdesivir has shown some positive benefits based on preliminary reports, but these results also need further confirmation via ongoing trials. Otherwise, no other agents have shown an impactful response against COVID-19. Recently, research exploring the therapeutic application of mesenchymal stem cells (MSCs) in critically ill patients suffering from COVID-19 has gained momentum. The patients belonging to this subset are most likely beyond the point where they could benefit from an antiviral therapy because most of their illness at this stage of disease is driven by inflammatory (over)response of the immune system. In this review, we discuss the potential of MSCs as a therapeutic option for patients with COVID-19, based on the encouraging results from the preliminary data showing improved outcomes in the progression of COVID-19 disease.
Collapse
Affiliation(s)
- Kamal Kant Sahu
- Department of Hematology and Oncology, Department of Internal Medicine, Saint Vincent Hospital, Worcester, Massachusetts
| | - Ahmad Daniyal Siddiqui
- Department of Hematology and Oncology, Department of Internal Medicine, Saint Vincent Hospital, Worcester, Massachusetts
| | - Jan Cerny
- Division of Hematology and Oncology, Department of Medicine, UMass Memorial Health Care, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
49
|
Rezakhani L, Kelishadrokhi AF, Soleimanizadeh A, Rahmati S. Mesenchymal stem cell (MSC)-derived exosomes as a cell-free therapy for patients Infected with COVID-19: Real opportunities and range of promises. Chem Phys Lipids 2021; 234:105009. [PMID: 33189639 PMCID: PMC7658620 DOI: 10.1016/j.chemphyslip.2020.105009] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
There are no commercially available effective antiviral medications or vaccines to deal with novel coronavirus disease (COVID-19). Hence there is a substantial unmet medical need for new and efficacious treatment options for COVID-19. Most COVID-19 deaths result from acute respiratory distress syndrome (ARDS). This virus induces excessive and aberrant inflammation so it is important to control the inflammation as soon as possible. To date, results of numerous studies have been shown that mesenchymal stem cells and their derivatives can suppress inflammation. Exosomes function as intercellular communication vehicles to transfer bioactive molecules (based on their origins), between cells. In this review, the recent exosome-based clinical trials for the treatment of COVID-19 are presented. Potential therapy may include the following items: First, using mesenchymal stem cells secretome. Second, incorporating specific miRNAs and mRNAs into exosomes and last, using exosomes as carriers to deliver drugs.
Collapse
Affiliation(s)
- Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Arghavan Soleimanizadeh
- International Graduate School in Molecular Medicine Ulm, Medical Faculty, University of Ulm, 89081 Ulm, Germany
| | - Shima Rahmati
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran; Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
50
|
Shyr D, Shyr Y. The design and analysis of non-randomized studies: a case study of off-label use of hydroxychloroquine in the COVID-19 pandemic. Expert Opin Investig Drugs 2020; 30:111-117. [PMID: 33353432 DOI: 10.1080/13543784.2021.1868435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: The COVID-19 pandemic has prompted researchers to conduct non-randomized studies in an effort to find an off-label drug that can effectively combat the virus and its effects. While these studies can expedite the drug approval process, researchers must carefully design and analyze such studies in order to perform rigorous science that is reproducible and credible. This article focuses on several key design and analysis considerations that can improve the scientific rigor of non-randomized studies of off-label drugs. Areas covered: The aim of this article is to provide an overview of best approaches that should be considered for non-randomized studies on off-label drugs. We discuss these approaches in detail and use a non-randomized study by Rivera et al. in Cancer Discovery as an example of methods that have been undertaken for COVID-19. Expert opinion: While non-randomized studies are inherently biased, they may be unavoidable in situations such as the COVID-19 pandemic, where researchers need to find an effective treatment quickly. We believe that a well-formed experimental design, high-quality data collection, and a well-thought-out statistical and data analysis plan are sufficient to produce rigorous and credible results for making an optimal decision.
Collapse
Affiliation(s)
- Derek Shyr
- Department of Biostatistics, Harvard T.H. Chan School of Public Health , Boston, MA, USA
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University School of Medicine , Nashville, TN, USA.,Center for Quantitative Sciences, Vanderbilt University School of Medicine , Nashville, TN, USA
| |
Collapse
|