1
|
Xia S, Gu X, Wang G, Zhong Y, Ma F, Liu Q, Xie J. Regulated Cell Death of Alveolar Macrophages in Acute Lung Inflammation: Current Knowledge and Perspectives. J Inflamm Res 2024; 17:11419-11436. [PMID: 39722732 PMCID: PMC11669335 DOI: 10.2147/jir.s497775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a common and serious clinical lung disease characterized by extensive alveolar damage and inflammation leading to impaired gas exchange. Alveolar macrophages (AMs) maintain homeostatic properties and immune defenses in lung tissues. Several studies have reported that AMs are involved in and regulate ALI/ARDS onset and progression via different regulated cell death (RCD) programs, such as pyroptosis, apoptosis, autophagic cell death, and necroptosis. Notably, the effects of RCD in AMs in disease are complex and variable depending on the environment and stimuli. In this review, we provide a comprehensive perspective on how regulated AMs death impacts on ALI/ARDS and assess its potential in new therapeutic development. Additionally, we describe the crosstalk between different RCD types in ALI, and provide new perspectives for the treatment of ALI/ARDS and other severe lung diseases.
Collapse
Affiliation(s)
- Siwei Xia
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Xiaoyan Gu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Gaojian Wang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yizhi Zhong
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Fengjie Ma
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Qinxue Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Junran Xie
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| |
Collapse
|
2
|
Song M, Lim KM, Song K, Kang GH, Kim SJ, Lee Y, Yu S, Jeong KH, Cho SG. Efficient Treatment of Psoriasis Using Conditioned Media from Mesenchymal Stem Cell Spheroids Cultured to Produce Transforming Growth Factor- β1-Enriched Small-Sized Extracellular Vesicles. Int J Stem Cells 2024; 17:407-417. [PMID: 39396918 PMCID: PMC11612221 DOI: 10.15283/ijsc24089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 10/15/2024] Open
Abstract
Psoriasis is a common chronic inflammatory disease in which keratinocytes proliferate abnormally due to excessive immune action. Psoriasis can be associated with various comorbidities and has a significant impact on health-related quality of life. Although many systemic treatments, including biologic agents, have been developed, topical treatment remains the main option for psoriasis management. Consequently, there is an urgent need to develop topical treatments with minimal side effects and high efficacy. Mesenchymal stem cells (MSCs) exhibit excellent immune regulation, anti-inflammatory activities, and therapeutic effects, and MSC-derived extracellular vesicles (EVs) can serve as crucial mediators of functional transfer from MSCs. Therefore, this study aimed to develop a safe and easy-to-use emulsion cream for treating psoriasis using MSC conditioned media (CM) containing EVs. We developed an enhanced Wharton's jelly MSC (WJ-MSC) culture method through a three-dimensional (3D) culture containing exogenous transforming growth factor-β3. Using the 3D culture system, we obtained CM from WJ-MSCs, which yielded a higher EV production compared to that of conventional WJ-MSC culture methods, and investigated the effect of EV-enriched 3D-WJ-MSC-CM cream on psoriasis-related inflammation. Administration of the EV-enriched 3D-WJ-MSC-CM cream significantly reduced erythema, thickness, and scaling of skin lesions, alleviated imiquimod-induced psoriasiform lesions in mice, and ameliorated histopathological changes in mouse skin. The upregulated mRNA expression of inflammatory cytokines, including IL-17a, IL-22, IL-23, and IL-36, decreased in the lesions. In conclusion, we present here a new topical treatment for psoriasis using an MSC EV-enriched cream.
Collapse
Affiliation(s)
- Myeongjin Song
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Kwonwoo Song
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Geun-Ho Kang
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Se Jong Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| | - Youngseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Sujin Yu
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
| | - Ki-Heon Jeong
- Department of Dermatology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul, Korea
- R&D Team, StemExOne Co., Ltd., Seoul, Korea
| |
Collapse
|
3
|
Lin S, Luo Y, Mao X, He W, Xu C, Zeng M. Homeobox B4 optimizes the therapeutic effect of bone marrow mesenchymal stem cells on endotoxin-associated acute lung injury in rats. Am J Med Sci 2024; 368:242-252. [PMID: 38795966 DOI: 10.1016/j.amjms.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 04/05/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Alveolar capillary endothelial cell (EC) injury has a pivotal role in driving acute respiratory distress syndrome (ARDS) progression and maintaining endothelial homeostasis. A previous ex vivo study revealed that overexpression of homeobox B4 (HOXB4) in bone marrow mesenchymal stem cells (BMSCs) enhanced protection against lipopolysaccharide (LPS)-induced EC injury by activating the Wnt/β-catenin pathway. This in vivo study was performed to verify whether BMSCs overexpressing HOXB4 exert similar protective effects on LPS-induced acute lung injury (ALI) in an animal model. METHODS The ALI rat model was established by intraperitoneal injection of LPS. Wildtype BMSCs or BMSCs overexpressing HOXB4 were then injected via the tail vein. The lung characteristics of rats were visualized by computed tomography. Lung histopathological characteristics and collagen deposition were assessed by hematoxylin-eosin and Masson's staining, respectively, which were combined with the lung wet/dry ratio and proinflammatory factor levels in bronchoalveolar lavage fluid to further evaluate therapeutic effects. Expression of β-catenin and VE-cadherin was assessed by western blotting and immunofluorescence. RESULTS Compared with wildtype BMSCs, overexpression of HOXB4 optimized the therapeutic effects of BMSCs, which manifested as improvements in lung exudation and histopathological features, reduced lung collagen deposition, amelioration of lung permeability, attenuation of lung inflammation, and enhanced expression of β-catenin and VE-cadherin proteins. CONCLUSIONS HOXB4-overexpressing BMSCs optimized the protective effect against LPS-induced ALI by partially activating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Shan Lin
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, PR China
| | - Yuling Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China
| | - Xueyan Mao
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China
| | - Caixia Xu
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China.
| |
Collapse
|
4
|
Margiana R, Pilehvar Y, Amalia FL, Lestari SW, Supardi S, I'tishom R. Mesenchymal stem cell secretome: A promising therapeutic strategy for erectile dysfunction? Asian J Urol 2024; 11:391-405. [PMID: 39139521 PMCID: PMC11318444 DOI: 10.1016/j.ajur.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/06/2023] [Indexed: 08/15/2024] Open
Abstract
Objective The secretome, comprising bioactive chemicals released by mesenchymal stem cells (MSCs), holds therapeutic promise in regenerative medicine. This review aimed to explore the therapeutic potential of the MSC secretome in regenerative urology, particularly for treating erectile dysfunction (ED), and to provide an overview of preclinical and clinical research on MSCs in ED treatment and subsequently to highlight the rationales, mechanisms, preclinical investigations, and therapeutic potential of the MSC secretome in this context. Methods The review incorporated an analysis of preclinical and clinical research involving MSCs in the treatment of ED. Subsequently, it delved into the existing knowledge regarding the MSC secretome, exploring its therapeutic potential. The methods included a comprehensive examination of relevant literature to discern the processes underlying the therapeutic efficacy of the MSC secretome. Results Preclinical research indicated the effectiveness of the MSC secretome in treating various models of ED. However, the precise mechanisms of its therapeutic efficacy remain unknown. The review provided insights into the anti-inflammatory, pro-angiogenic, and trophic properties of the MSC secretome. It also discussed potential advantages, such as avoiding issues related to cellular therapy, including immunogenicity, neoplastic transformation, and cost. Conclusion This review underscores the significant therapeutic potential of the MSC secretome in regenerative urology, particularly for ED treatment. While preclinical studies demonstrate promising outcomes, further research is essential to elucidate the specific mechanisms underlying the therapeutic efficacy before clinical application. The review concludes by discussing future perspectives and highlighting the challenges associated with the clinical translation of the MSC secretome in regenerative urology.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Indonesia General Academic Hospital, Depok, Indonesia
- Ciptomangunkusumo General Academic Hospital, Jakarta, Indonesia
| | - Younes Pilehvar
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Science, Urmia, Iran
| | - Fatkhurrohmah L. Amalia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Dr. Kariadi Hospital, Semarang, Indonesia
| | - Silvia W. Lestari
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Indonesia General Academic Hospital, Depok, Indonesia
- Ciptomangunkusumo General Academic Hospital, Jakarta, Indonesia
- Department of Medical Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Supardi Supardi
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Reny I'tishom
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga Surabaya, Indonesia
| |
Collapse
|
5
|
Fernandes R, Barbosa-Matos C, Borges-Pereira C, de Carvalho ALRT, Costa S. Glycogen Synthase Kinase-3 Inhibition by CHIR99021 Promotes Alveolar Epithelial Cell Proliferation and Lung Regeneration in the Lipopolysaccharide-Induced Acute Lung Injury Mouse Model. Int J Mol Sci 2024; 25:1279. [PMID: 38279281 PMCID: PMC10816825 DOI: 10.3390/ijms25021279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 01/28/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury that currently lacks effective clinical treatments. Evidence highlights the potential role of glycogen synthase kinase-3 (GSK-3) inhibition in mitigating severe inflammation. The inhibition of GSK-3α/β by CHIR99021 promoted fetal lung progenitor proliferation and maturation of alveolar epithelial cells (AECs). The precise impact of CHIR99021 in lung repair and regeneration during acute lung injury (ALI) remains unexplored. This study intends to elucidate the influence of CHIR99021 on AEC behaviour during the peak of the inflammatory phase of ALI and, after its attenuation, during the repair and regeneration stage. Furthermore, a long-term evaluation was conducted post CHIR99021 treatment at a late phase of the disease. Our results disclosed the role of GSK-3α/β inhibition in promoting AECI and AECII proliferation. Later administration of CHIR99021 during ALI progression contributed to the transdifferentiation of AECII into AECI and an AECI/AECII increase, suggesting its contribution to the renewal of the alveolar epithelial population and lung regeneration. This effect was confirmed to be maintained histologically in the long term. These findings underscore the potential of targeted therapies that modulate GSK-3α/β inhibition, offering innovative approaches for managing acute lung diseases, mostly in later stages where no treatment is available.
Collapse
Affiliation(s)
- Raquel Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (R.F.); (C.B.-M.); (C.B.-P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Braga, Portugal
| | - Catarina Barbosa-Matos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (R.F.); (C.B.-M.); (C.B.-P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Braga, Portugal
| | - Caroline Borges-Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (R.F.); (C.B.-M.); (C.B.-P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Braga, Portugal
| | - Ana Luísa Rodrigues Toste de Carvalho
- Department of Internal Medicine, São João Universitary Hospital Center, 4200-319 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Sandra Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (R.F.); (C.B.-M.); (C.B.-P.)
- ICVS/3B’s—PT Government Associate Laboratory, 4806-909 Braga, Portugal
| |
Collapse
|
6
|
Zhao J, Liang G, Yang J, Huang H, Dou Y, Gu Z, Liu J, Zeng L, Yang W. Liuwei Dihuang Pills Enhance Osteogenic Differentiation in MC3T3-E1 Cells through the Activation of the Wnt/β-Catenin Signaling Pathway. Pharmaceuticals (Basel) 2024; 17:99. [PMID: 38256932 PMCID: PMC10819701 DOI: 10.3390/ph17010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVE The therapeutic efficacy and molecular mechanisms of traditional Chinese medicines (TCMs), such as Liuwei Dihuang pills (LWDH pills), in treating osteoporosis (OP) remain an area of active research and interest in modern medicine. This study investigated the mechanistic underpinnings of LWDH pills in the treatment of OP based on network pharmacology, bioinformatics, and in vitro experiments. METHODS The active ingredients and targets of LWDH pills were retrieved through the TCMSP database. OP-related targets were identified using the CTD, GeneCards, and DisGeNET databases. The STRING platform was employed to construct a protein-protein interaction (PPI) network, and core targets for LWDH pills in treating OP were identified. The GO functional and KEGG pathway enrichment analyses for potential targets were performed using the R package "clusterProfiler". A "drug-target" network diagram was created using Cytoscape 3.7.1 software. The viability of MC3T3-E1 cells was evaluated using the CCK-8 method after treatment with various concentrations (1.25%, 2.5%, 5%, and 10%) of LWDH pill-medicated serum for 24, 48, and 72 h. Following a 48 h treatment of MC3T3-E1 cells with LWDH pill-medicated serum, the protein levels of collagen Ⅰ, RUNX2, Wnt3, and β-catenin were quantified using the Western blot analysis, and the activity of alkaline phosphatase (ALP) was measured. RESULTS A total of 197 putative targets for LWDH pills for OP treatment were pinpointed, from which 20 core targets were singled out, including TP53, JUN, TNF, CTNNB1 (β-catenin), and GSK3B. The putative targets were predominantly involved in signaling pathways such as the Wnt signaling pathway, the MAPK signaling pathway, and the PI3K-Akt signaling pathway. The intervention with LWDH pill-medicated serum for 24, 48, and 72 h did not result in any notable alterations in the cell viability of MC3T3-E1 cells relative to the control group (all p > 0.05). Significant upregulation in protein levels of collagen Ⅰ, RUNX2, Wnt3, and β-catenin in MC3T3-E1 cells was observed in response to the treatment with 2.5%, 5%, and 10% of LWDH pill-medicated serum in comparison to that with the 10% rabbit serum group (all p < 0.05). Furthermore, the intervention with LWDH pill-medicated serum resulted in the formation of red calcified nodules in MC3T3-E1 cells, as indicated by ARS staining. CONCLUSIONS LWDH pills may upregulate the Wnt/β-catenin signaling pathway to elevate the expression of osteogenic differentiation proteins, including collagen Ⅰ and RUNX2, and to increase the ALP activity in MC3T3-E1 cells for the treatment of OP.
Collapse
Affiliation(s)
- Jinlong Zhao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (G.L.); (Y.D.); (Z.G.)
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China;
- The Research Team on Bone and Joint Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China;
| | - Guihong Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (G.L.); (Y.D.); (Z.G.)
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China;
- The Research Team on Bone and Joint Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China;
| | - Junzheng Yang
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China;
| | - Hetao Huang
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China;
| | - Yaoxing Dou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (G.L.); (Y.D.); (Z.G.)
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China;
- The Research Team on Bone and Joint Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China;
| | - Zhuoxu Gu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (G.L.); (Y.D.); (Z.G.)
| | - Jun Liu
- The Research Team on Bone and Joint Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China;
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China;
- Guangdong Second Chinese Medicine Hospital (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou 510095, China
| | - Lingfeng Zeng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (G.L.); (Y.D.); (Z.G.)
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China;
- The Research Team on Bone and Joint Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou 510120, China;
| | - Weiyi Yang
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China;
| |
Collapse
|
7
|
Luo Y, Ge S, Chen Q, Lin S, He W, Zeng M. Overexpression of FoxM1 optimizes the therapeutic effect of bone marrow mesenchymal stem cells on acute respiratory distress syndrome. Stem Cell Res Ther 2023; 14:27. [PMID: 36788588 PMCID: PMC9926819 DOI: 10.1186/s13287-023-03240-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 01/17/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Injury of alveolar epithelial cells and capillary endothelial cells is crucial in the pathogenesis of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Mesenchymal stem cells (MSCs) are a promising cell source for ALI/ARDS treatment. Overexpression of Fork head box protein M1 (FoxM1) facilitates MSC differentiation into alveolar type II (AT II) cells in vitro. Moreover, FoxM1 has been shown to repair the endothelial barrier. Therefore, this study explored whether overexpression of FoxM1 promotes the therapeutic effect of bone marrow-derived MSCs (BMSCs) on ARDS by differentiation of BMSCs into AT II cells or a paracrine mechanism. METHODS A septic ALI model was established in mice by intraperitoneal administration of lipopolysaccharide. The protective effect of BMSCs-FoxM1 on ALI was explored by detecting pathological variations in the lung, total protein concentration in bronchoalveolar lavage fluid (BALF), wet/dry (W/D) lung weight ratio, oxidative stress levels, cytokine levels, and retention of BMSCs in the lung. In addition, we assessed whether FoxM1 overexpression promoted the therapeutic effect of BMSCs on ALI/ARDS by differentiating into AT II cells using SPC-/- mice. Furthermore, the protective effect of BMSCs-FoxM1 on lipopolysaccharide-induced endothelial cell (EC) injury was explored by detecting EC proliferation, apoptosis, scratch wounds, tube formation, permeability, and oxidative stress, and analyzing whether the Wnt/β-catenin pathway contributes to the regulatory mechanism in vitro using a pathway inhibitor. RESULTS Compared with BMSCs-Vector, treatment with BMSCs-FoxM1 significantly decreased the W/D lung weight ratio, total BALF protein level, lung injury score, oxidative stress, and cytokine levels. With the detected track of BMSCs-FoxM1, we observed a low residency rate and short duration of residency in the lung. Notably, SPC was not expressed in SPC-/- mice injected with BMSCs-FoxM1. Furthermore, BMSCs-FoxM1 enhanced EC proliferation, migration, and tube formation; inhibited EC apoptosis and inflammation; and maintained vascular integrity through activation of the Wnt/β-catenin pathway, which was partially reversed by XAV-939. CONCLUSION Overexpression of FoxM1 enhanced the therapeutic effect of BMSCs on ARDS, possibly through a paracrine mechanism rather than by promoting BMSC differentiation into AT II cells in vivo, and prevented LPS-induced EC barrier disruption partially through activating the Wnt/β-catenin signaling pathway in vitro.
Collapse
Affiliation(s)
- Yuling Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Shanhui Ge
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Qingui Chen
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Shan Lin
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
8
|
Pourhadi M, Zali H, Ghasemi R, Vafaei-Nezhad S. Promising Role of Oral Cavity Mesenchymal Stem Cell-Derived Extracellular Vesicles in Neurodegenerative Diseases. Mol Neurobiol 2022; 59:6125-6140. [PMID: 35867205 DOI: 10.1007/s12035-022-02951-y] [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: 09/21/2021] [Accepted: 06/28/2022] [Indexed: 10/17/2022]
Abstract
Mesenchymal stem cells (MSCs) and mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been regarded as the beneficial and available tools to treat various hereditary, multifactorial, acute, and chronic diseases. Mesenchymal stem cells can be extracted from numerous sources for clinical purposes while oral cavity-derived mesenchymal stem cells seem to be more effective in neuroregeneration than other sources due to their similar embryonic origins to neuronal tissues. In various studies and different neurodegenerative diseases (NDs), oral cavity mesenchymal stem cells have been applied to prove their promising capacities in disease improvement. Moreover, oral cavity mesenchymal stem cells' secretion is regarded as a novel and practical approach to neuroregeneration; hence, extracellular vesicles (EVs), especially exosomes, may provide promising results to improve CNS defects. This review article focuses on how oral cavity-derived stem cells and their extracellular vesicles can improve neurodegenerative conditions and tries to show which molecules are involved in the recovery process.
Collapse
Affiliation(s)
- Masoumeh Pourhadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Rasoul Ghasemi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Vafaei-Nezhad
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
9
|
Human Placental Mesenchymal Stem Cells for the Treatment of ARDS in Rat. Stem Cells Int 2022; 2022:8418509. [PMID: 35756754 PMCID: PMC9226970 DOI: 10.1155/2022/8418509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/21/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022] Open
Abstract
The acute respiratory distress syndrome (ARDS) is one of the main causes of high mortality in patients with coronavirus (COVID-19). In recent years, due to the coronavirus pandemic, the number of patients with ARDS has increased significantly. Unfortunately, until now, there are no effective treatments for ARDS caused by COVID-19. Many drugs are either ineffective or have a low effect. Currently, there have been reports of efficient use of mesenchymal stem cells (MSCs) for the treatment of ARDS caused by COVID-19. We investigated the influence of freeze-dried human placenta-derived mesenchymal stem cells (HPMSCs) in ARDS rat model. All animals have received intratracheal injection of 6 mg/kg of lipopolysaccharide (LPS). The rats were randomly divided into five groups: I: LPS, II: LPS+dexamethasone, III: LPS+HPMSCs, IV: HPMSC, and V: saline. ARDS observation time was short-term and amounted to 168 hours. The study has shown that HPMSCs are able to migrate and attach to damaged lung tissue, contributing to the resolution of pathology, restoration of function, and tissue repair in the alveolar space. Studies have also shown that the administration of HPMSCs in animals with ARDS model significantly reduced the levels of key cytokines such as IL-1β, IL-6, and TNF-α. Freeze-dried placental stem cell is a very promising biomaterial for the treatment of ARDS. The human placenta can be easily obtained because it is considered as a medical waste. At the same time, a huge number of MSCs can be obtained from the placental tissue, and there is no ethical controversy around their use. The freeze-dried MSCs from human placental tissue can be stored sterile at room temperature for a long time before use.
Collapse
|
10
|
Shen M, Wang L, Feng L, Gao Y, Li S, Wu Y, Xu C, Pei G. bFGF-Loaded Mesoporous Silica Nanoparticles Promote Bone Regeneration Through the Wnt/β-Catenin Signalling Pathway. Int J Nanomedicine 2022; 17:2593-2608. [PMID: 35698561 PMCID: PMC9188412 DOI: 10.2147/ijn.s366926] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/01/2022] [Indexed: 12/22/2022] Open
Abstract
Background Bone defects remain an unsolved clinical problem due to the lack of effective osteogenic induction protocols. Nanomaterials play an important role in bone defect repair by stimulating osteogenesis. However, constructing an effective bioactive nanomaterial remains a substantial challenge. Methods In this study, mesoporous silica nanoparticles (MSNs) were prepared and used as nanocarriers for basic fibroblast growth factor (bFGF). The characteristics and biological properties of the synthetic bFGF@MSNs were tested. The osteogenic effects of the particles on the behavior of MC3T3-E1 cells were investigated in vitro. In addition, the differentially expressed genes during induction of osteogenesis were analyzed by transcriptomic sequencing. Radiological and histological observations were carried out to determine bone regeneration capability in a distal femur defect model. Results Achieving bFGF sustained release, bFGF@MSNs had uniform spherical morphology and good biocompatibility. In vitro osteogenesis induction experiments showed that bFGF@MSNs exhibited excellent osteogenesis performance, with upregulation of osteogenesis-related genes (RUNX2, OCN, Osterix, ALP). Transcriptomic sequencing revealed that the Wnt/β-catenin signalling pathway could be activated in regulation of biological processes. In vivo, bone defect repair experiments showed enhanced bone regeneration, as indicated by radiological and histological analysis, after the application of bFGF@MSNs. Conclusion bFGF@MSNs can promote bone regeneration by activating the Wnt/β-catenin signalling pathway. These particles are expected to become a potential therapeutic bioactive material for clinical application in repairing bone defects in the future.
Collapse
Affiliation(s)
- Mingkui Shen
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Lulu Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Li Feng
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Yi Gao
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Sijing Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Yulan Wu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| | - Chuangye Xu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
- Correspondence: Chuangye Xu; Guoxian Pei, Email ;
| | - Guoxian Pei
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People’s Republic of China
| |
Collapse
|
11
|
In-vitro analysis on the potential use of dental pulp mesenchymal stem cells on arecoline-induced oral epithelial cells. Med Oncol 2022; 39:77. [PMID: 35195802 DOI: 10.1007/s12032-022-01673-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/30/2022] [Indexed: 10/19/2022]
Abstract
To assess the protective role of the secretome of dental pulp mesenchymal stem cells on arecoline-induced epithelial-mesenchymal transition and senescence on epithelial cells of the oral mucosa. Effect of varying concentrations of arecoline extract and dental pulp mesenchymal stem cell condition media (DPSC-CM) were noted on oral mucosal epithelial cells. MTT assay, Annexin V-FITC/PI assay, and the quantitative gene expressions of BCL2, PUMA, BAD, BAX, CASP3, CASP9, CASP12, TGFB1, CST3, COL1A2, COL3A1, TIMP1, TIMP2, CDH1, and CDH2 were assessed. Oral mucosal epithelial cells exposed only to the arecoline were the control. 50% and 100% DPSC-CM decreased apoptosis-related gene expression in the cells exposed with 25 μM arecoline compared to the control. 50% DPSC-CM attenuated the expression of all fibrotic genes and EMT-related genes. 20% and 100% DPSC-CM showed differential effects on fibrotic and EMT-related genes. DPSC-CM inhibited apoptosis, and attenuated expression of fibrotic and EMT-related genes on arecoline treated human oral epithelial cells.
Collapse
|
12
|
Ebadi Zavieh S, Safari F. The Antitumor Activity of hAMSCs Secretome in HT-29 Colon Cancer Cells Through Downregulation of EGFR/c-Src/IRTKS Expression and p38/ERK1/2 Phosphorylation. Cell Biochem Biophys 2022; 80:395-402. [PMID: 35150389 DOI: 10.1007/s12013-022-01066-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2022] [Indexed: 11/03/2022]
Abstract
Colon cancer is considered as one of the main causes of mortality worldwide. Identifying a novel and more effective platform with fewer side effects is still progress. In various cancer types, Epidermal growth factor receptor (EGFR) and c-Src (a key mediator in EGFR signaling pathway) are the key targets for cancer therapy. Moreover, insulin receptor tyrosine kinase substrate (IRTKS or BAI1-associated protein 2-like 1: BAIAP2L1) is a member of the subfamily of inverse BAR (I-BAR) domain proteins, which mediates cell morphology and movement through regulation of actin polymerization. In this study, we employed a co-culture system using Transwell six-well plates. After 72 h, hAMSCs-treated HT-29 cells, EGFR, c-Src, IRTKS, p38, and ERK1/2 expression were analyzed using quantitative real time PCR (qRT-PCR) and western blot methods. The significant reduction in tumor cell growth and motility through downregulation of EGFR/c-Src/IRTKS expression and p38/ERK1/2 phosphorylation in HT-29 cells was demonstrated based on 2D and 3D cell culture models. The induction of cellular apoptosis was also found. Our results support the idea that the hAMSCS secretome has therapeutic effects on cancer cells. However, further experiments will be required to identify the exact molecular mechanisms.
Collapse
Affiliation(s)
- Shamin Ebadi Zavieh
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran.
| |
Collapse
|
13
|
Lee S, Chae DS, Song BW, Lim S, Kim SW, Kim IK, Hwang KC. ADSC-Based Cell Therapies for Musculoskeletal Disorders: A Review of Recent Clinical Trials. Int J Mol Sci 2021; 22:ijms221910586. [PMID: 34638927 PMCID: PMC8508846 DOI: 10.3390/ijms221910586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 01/04/2023] Open
Abstract
Recently published clinical trials involving the use of adipose-derived stem cells (ADSCs) indicated that approximately one-third of the studies were conducted on musculoskeletal disorders (MSD). MSD refers to a wide range of degenerative conditions of joints, bones, and muscles, and these conditions are the most common causes of chronic disability worldwide, being a major burden to the society. Conventional treatment modalities for MSD are not sufficient to correct the underlying structural abnormalities. Hence, ADSC-based cell therapies are being tested as a form of alternative, yet more effective, therapies in the management of MSDs. Therefore, in this review, MSDs subjected to the ADSC-based therapy were further categorized as arthritis, craniomaxillofacial defects, tendon/ligament related disorders, and spine disorders, and their brief characterization as well as the corresponding conventional therapeutic approaches with possible mechanisms with which ADSCs produce regenerative effects in disease-specific microenvironments were discussed to provide an overview of under which circumstances and on what bases the ADSC-based cell therapy was implemented. Providing an overview of the current status of ADSC-based cell therapy on MSDs can help to develop better and optimized strategies of ADSC-based therapeutics for MSDs as well as help to find novel clinical applications of ADSCs in the near future.
Collapse
Affiliation(s)
- Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, Catholic Kwandong University, Gangneung 210-701, Korea;
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
| | - Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
- Correspondence: (I.-K.K.); (K.-C.H.); Fax: +82-32-290-2774 (K.-C.H.)
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung 210-701, Korea; (S.L.); (B.-W.S.); (S.L.); (S.W.K.)
- Correspondence: (I.-K.K.); (K.-C.H.); Fax: +82-32-290-2774 (K.-C.H.)
| |
Collapse
|
14
|
Mesenchymal Stem Cells in the Treatment of COVID-19, a Promising Future. Cells 2021; 10:cells10102588. [PMID: 34685567 PMCID: PMC8533906 DOI: 10.3390/cells10102588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have a potent self-renewal capacity and can differentiate into multiple cell types. They also affect the ambient tissue by the paracrine secretion of numerous factors in vivo, including the induction of other stem cells’ differentiation. In vitro, the culture media supernatant is named secretome and contains soluble molecules and extracellular vesicles that retain potent biological function in tissue regeneration. MSCs are considered safe for human treatment; their use does not involve ethical issues, as embryonic stem cells do not require genetic manipulation as induced pluripotent stem cells, and after intravenous injection, they are mainly found in the lugs. Therefore, these cells are currently being tested in various preclinical and clinical trials for several diseases, including COVID-19. Several affected COVID-19 patients develop induced acute respiratory distress syndrome (ARDS) associated with an uncontrolled inflammatory response. This condition causes extensive damage to the lungs and may leave serious post-COVID-19 sequelae. As the disease may cause systemic alterations, such as thromboembolism and compromised renal and cardiac function, the intravenous injection of MSCs may be a therapeutic alternative against multiple pathological manifestations. In this work, we reviewed the literature about MSCs biology, focusing on their function in pulmonary regeneration and their use in COVID-19 treatment.
Collapse
|
15
|
Wang L, Li Y, Xu M, Deng Z, Zhao Y, Yang M, Liu Y, Yuan R, Sun Y, Zhang H, Wang H, Qian Z, Kang H. Regulation of Inflammatory Cytokine Storms by Mesenchymal Stem Cells. Front Immunol 2021; 12:726909. [PMID: 34394132 PMCID: PMC8358430 DOI: 10.3389/fimmu.2021.726909] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/14/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely used in preclinical and clinical trials for various diseases and have shown great potential in the treatment of sepsis and coronavirus disease (COVID-19). Inflammatory factors play vital roles in the pathogenesis of diseases. The interaction between inflammatory factors is extremely complex. Once the dynamics of inflammatory factors are unbalanced, inflammatory responses and cytokine storm syndrome develop, leading to disease exacerbation and even death. Stem cells have become ideal candidates for the treatment of such diseases due to their immunosuppressive and anti-inflammatory properties. However, the mechanisms by which stem cells affect inflammation and immune regulation are still unclear. This article discusses the therapeutic mechanism and potential value of MSCs in the treatment of sepsis and the novel COVID-19, outlines how MSCs mediate innate and acquired immunity at both the cellular and molecular levels, and described the anti-inflammatory mechanisms and related molecular pathways. Finally, we review the safety and efficacy of stem cell therapy in these two diseases at the preclinical and clinical levels.
Collapse
Affiliation(s)
- Lu Wang
- Medical School of Chinese PLA, Beijing, China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yun Li
- Medical School of Chinese PLA, Beijing, China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Moyan Xu
- Health Care Office, Chinese PLA General Hospital, Beijing, China
| | - Zihui Deng
- Department of Basic Medicine, Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Yan Zhao
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mengmeng Yang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuyan Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Rui Yuan
- Medical School of Chinese PLA, Beijing, China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Sun
- School of Public Health, Capital Medical University, Beijing, China
| | - Hao Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Heming Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhirong Qian
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hongjun Kang
- Medical School of Chinese PLA, Beijing, China.,Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Lin S, Chen Q, Zhang L, Ge S, Luo Y, He W, Xu C, Zeng M. Overexpression of HOXB4 Promotes Protection of Bone Marrow Mesenchymal Stem Cells Against Lipopolysaccharide-Induced Acute Lung Injury Partially Through the Activation of Wnt/β-Catenin Signaling. J Inflamm Res 2021; 14:3637-3649. [PMID: 34349541 PMCID: PMC8326777 DOI: 10.2147/jir.s319416] [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: 05/09/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Pulmonary vascular endothelial cell (EC) injury is recognized as one of the pathological factors of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Bone marrow mesenchymal stem cell (BMSC)-based cytotherapy has attracted substantial attention over recent years as a promising therapeutic approach for ALI/ARDS; however, its use remains limited due to inconsistent efficacy. Currently, gene modification techniques are widely applied to MSCs. In the present study, we aimed to investigate the effect of BMSCs overexpressing Homeobox B4 (HOXB4) on lipopolysaccharide (LPS)-induced EC injury. Methods We used LPS to induce EC injury and established EC-BMSC coculture system using transwell chambers. The effect of BMSCs on ECs was explored by detecting EC proliferation, apoptosis, migration, tube formation, and permeability, and determining whether the Wnt/β-catenin pathway is involved in the regulatory mechanism using XAV-939, inhibitor of Wnt/ β-catenin. Results As compared to BMSCWT, BMSCHOXB4 coculture promoted EC proliferation, migration, and tube formation after LPS stimulation and attenuated LPS-induced EC apoptosis and vascular permeability. Mechanistically, BMSCHOXB4 coculture prevented LPS-induced EC injury by activating the Wnt/β-catenin pathway, which is partially reversible by XAV-939. When cocultured with BMSCHOXB4, pro-inflammatory factors were dramatically decreased and anti-inflammatory factors were greatly increased in the EC medium compared to those in the LPS group (P<0.05). Additionally, when compared to BMSCWT coculture, the BMSCHOXB4 coculture showed an enhanced modulation of IL-6, TNF-α, and IL-10, but there was no statistically significant effect on IL-1β and IL-4. Conclusion Coculturing of BMSCHOXB4 prevented LPS-induced EC injury by reversing the inactivation of the Wnt/β-catenin signaling pathway. An in vivo study remains warranted to ascertain whether engraftment of BMSCHOXB4 can be an attractive strategy for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Shan Lin
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Qingui Chen
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Lishan Zhang
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shanhui Ge
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yuling Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Caixia Xu
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
17
|
Sharma A, Chakraborty A, Jaganathan BG. Review of the potential of mesenchymal stem cells for the treatment of infectious diseases. World J Stem Cells 2021; 13:568-593. [PMID: 34249228 PMCID: PMC8246252 DOI: 10.4252/wjsc.v13.i6.568] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The therapeutic value of mesenchymal stem cells (MSCs) for the treatment of infectious diseases and the repair of disease-induced tissue damage has been explored extensively. MSCs inhibit inflammation, reduce pathogen load and tissue damage encountered during infectious diseases through the secretion of antimicrobial factors for pathogen clearance and they phagocytose certain bacteria themselves. MSCs dampen tissue damage during infection by downregulating the levels of pro-inflammatory cytokines, and inhibiting the excessive recruitment of neutrophils and proliferation of T cells at the site of injury. MSCs aid in the regeneration of damaged tissue by differentiating into the damaged cell types or by releasing paracrine factors that direct tissue regeneration, differentiation, and wound healing. In this review, we discuss in detail the various mechanisms by which MSCs help combat pathogens, tissue damage associated with infectious diseases, and challenges in utilizing MSCs for therapy.
Collapse
Affiliation(s)
- Amit Sharma
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Anuja Chakraborty
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Bithiah Grace Jaganathan
- Stem Cell and Cancer Biology Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| |
Collapse
|
18
|
Safari F, Shakery T, Sayadamin N. Evaluating the effect of secretome of human amniotic mesenchymal stromal cells on apoptosis induction and epithelial-mesenchymal transition inhibition in LNCaP prostate cancer cells based on 2D and 3D cell culture models. Cell Biochem Funct 2021; 39:813-820. [PMID: 34128234 DOI: 10.1002/cbf.3654] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Prostate cancer (PCa) is the second most prevalent cancer in men worldwide. Most cases of death from PCa are due to metastasis. Early stages of metastasis are mediated by epithelial-mesenchymal transition (EMT) process through which cancer cells acquire motility and invasive characteristics. Thus, more potent and novel therapeutic strategies must be designed based on the inhibition of EMT or metastasis. Herein, we employ a co-culture system to evaluate the anti-EMT effects of human amniotic mesenchymal stromal cells (hAMSCs) on LNCaP PCa cells. The RNA of treated (sample) and untreated cancer cells (control) and whole-cell lysates of related cells were prepared and analysed through quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, respectively. Based on the results, the expression of vimentin, Snail and Zeb1 in LNCaP cells decreased and the expression of E-cadherin increased after treatment with hAMSCs. Furthermore, induction of the cellular apoptosis in LNCaP cells was detected. The anti-cancer activity of conditioned medium from hAMSCs was shown using hanging drop technique (a 3D cell culture model). Our findings support the idea that stem cells can be considered as a novel therapeutic approach to inhibit prostate cancer cells. SIGNIFICANCE OF THE STUDY: The anti-tumour activity of hAMSCs on LNCaP prostate cancer cells using 2D and 3D cell culture models via induction of apoptosis, suppression of EMT process and down-regulation of EGFR was shown. The results of the present study support this idea that hAMSCs may be a potent therapeutic tool to suppress tumour growth in LNCaP prostate cancer cells.
Collapse
Affiliation(s)
- Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Termeh Shakery
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Nadiya Sayadamin
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| |
Collapse
|
19
|
Fuloria S, Subramaniyan V, Dahiya R, Dahiya S, Sudhakar K, Kumari U, Sathasivam K, Meenakshi DU, Wu YS, Sekar M, Malviya R, Singh A, Fuloria NK. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Regenerative Potential and Challenges. BIOLOGY 2021; 10:172. [PMID: 33668707 PMCID: PMC7996168 DOI: 10.3390/biology10030172] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Evidence suggests that stem cells exert regenerative potential via the release of extracellular vesicles. Mesenchymal stem cell extracellular vesicles (MSCEVs) offer therapeutic benefits for various pathophysiological ailments by restoring tissues. Facts suggest that MSCEV action can be potentiated by modifying the mesenchymal stem cells culturing methodology and bioengineering EVs. Limited clinical trials of MSCEVs have questioned their superiority, culturing quality, production scale-up and isolation, and administration format. Translation of preclinically successful MSCEVs into a clinical platform requires paying attention to several critical matters, such as the production technique, quantification/characterization, pharmacokinetics/targeting/transfer to the target site, and the safety profile. Keeping these issues as a priority, the present review was designed to highlight the challenges in translating preclinical MSCEV research into clinical platforms and provide evidence for the regenerative potential of MSCEVs in various conditions of the liver, kidney, heart, nervous system, bone, muscle, cartilage, and other organs/tissues.
Collapse
Affiliation(s)
| | - Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Rajiv Dahiya
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago;
| | - Sunita Dahiya
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
| | - Kalvatala Sudhakar
- School of Pharmaceutical Sciences (LIT-Pharmacy), Lovely Professional University, Jalandhar 144411, India;
| | - Usha Kumari
- Faculty of Medicine, AIMST University, Kedah 08100, Malaysia;
| | | | | | - Yuan Seng Wu
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Kuala Lumpur 42610, Malaysia; (V.S.); (Y.S.W.)
| | - Mahendran Sekar
- Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh 30450, Malaysia;
| | - Rishabha Malviya
- Department of Pharmacy, SMAS, Galgotias University, Greater Noida 203201, India; (R.M.); (A.S.)
| | - Amit Singh
- Department of Pharmacy, SMAS, Galgotias University, Greater Noida 203201, India; (R.M.); (A.S.)
| | | |
Collapse
|
20
|
Patel S, Werstuck GH. Macrophage Function and the Role of GSK3. Int J Mol Sci 2021; 22:ijms22042206. [PMID: 33672232 PMCID: PMC7926541 DOI: 10.3390/ijms22042206] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
Macrophages are present in nearly all vertebrate tissues, where they respond to a complex variety of regulatory signals to coordinate immune functions involved in tissue development, metabolism, homeostasis, and repair. Glycogen synthase kinase 3 (GSK3) is a ubiquitously expressed protein kinase that plays important roles in multiple pathways involved in cell metabolism. Dysregulation of GSK3 has been implicated in several prevalent metabolic disorders, and recent findings have highlighted the importance of GSK3 activity in the regulation of macrophages, especially with respect to the initiation of specific pathologies. This makes GSK3 a potential therapeutic target for the development of novel drugs to modulate immunometabolic responses. Here, we summarize recent findings that have contributed to our understanding of how GSK3 regulates macrophage function, and we discuss the role of GSK3 in the development of metabolic disorders and diseases.
Collapse
Affiliation(s)
- Sarvatit Patel
- Thrombosis and Atherosclerosis Research Institute, 237 Barton Street E, Hamilton, ON L9L 2X2, Canada;
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Geoff H. Werstuck
- Thrombosis and Atherosclerosis Research Institute, 237 Barton Street E, Hamilton, ON L9L 2X2, Canada;
- Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
- Correspondence: ; Tel.: +1-905-521-2100 (ext. 40747)
| |
Collapse
|
21
|
Bone marrow-derived mesenchymal stem cells modulate autophagy in RAW264.7 macrophages via the phosphoinositide 3-kinase/protein kinase B/heme oxygenase-1 signaling pathway under oxygen-glucose deprivation/restoration conditions. Chin Med J (Engl) 2021; 134:699-707. [PMID: 33605598 PMCID: PMC7989993 DOI: 10.1097/cm9.0000000000001133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Autophagy of alveolar macrophages is a crucial process in ischemia/reperfusion injury-induced acute lung injury (ALI). Bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent cells with the potential for repairing injured sites and regulating autophagy. This study was to investigate the influence of BM-MSCs on autophagy of macrophages in the oxygen-glucose deprivation/restoration (OGD/R) microenvironment and to explore the potential mechanism. Methods We established a co-culture system of macrophages (RAW264.7) with BM-MSCs under OGD/R conditions in vitro. RAW264.7 cells were transfected with recombinant adenovirus (Ad-mCherry-GFP-LC3B) and autophagic status of RAW264.7 cells was observed under a fluorescence microscope. Autophagy-related proteins light chain 3 (LC3)-I, LC3-II, and p62 in RAW264.7 cells were detected by Western blotting. We used microarray expression analysis to identify the differently expressed genes between OGD/R treated macrophages and macrophages co-culture with BM-MSCs. We investigated the gene heme oxygenase-1 (HO-1), which is downstream of the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. Results The ratio of LC3-II/LC3-I of OGD/R treated RAW264.7 cells was increased (1.27 ± 0.20 vs. 0.44 ± 0.08, t = 6.67, P < 0.05), while the expression of p62 was decreased (0.77 ± 0.04 vs. 0.95 ± 0.10, t = 2.90, P < 0.05), and PI3K (0.40 ± 0.06 vs. 0.63 ± 0.10, t = 3.42, P < 0.05) and p-Akt/Akt ratio was also decreased (0.39 ± 0.02 vs. 0.58 ± 0.03, t = 9.13, P < 0.05). BM-MSCs reduced the LC3-II/LC3-I ratio of OGD/R treated RAW264.7 cells (0.68 ± 0.14 vs. 1.27 ± 0.20, t = 4.12, P < 0.05), up-regulated p62 expression (1.10 ± 0.20 vs. 0.77 ± 0.04, t = 2.80, P < 0.05), and up-regulated PI3K (0.54 ± 0.05 vs. 0.40 ± 0.06, t = 3.11, P < 0.05) and p-Akt/Akt ratios (0.52 ± 0.05 vs. 0.39 ± 0.02, t = 9.13, P < 0.05). A whole-genome microarray assay screened the differentially expressed gene HO-1, which is downstream of the PI3K/Akt signaling pathway, and the alteration of HO-1 mRNA and protein expression was consistent with the data on PI3K/Akt pathway. Conclusions Our results suggest the existence of the PI3K/Akt/HO-1 signaling pathway in RAW264.7 cells under OGD/R circumstances in vitro, revealing the mechanism underlying BM-MSC-mediated regulation of autophagy and enriching the understanding of potential therapeutic targets for the treatment of ALI.
Collapse
|
22
|
Rao J, Yang C, Yang S, Lu H, Hu Y, Lu L, Cheng F, Wang X. Deficiency of TGR5 exacerbates immune-mediated cholestatic hepatic injury by stabilizing the β-catenin destruction complex. Int Immunol 2020; 32:321-334. [PMID: 31930324 PMCID: PMC7206975 DOI: 10.1093/intimm/dxaa002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022] Open
Abstract
Intrahepatic cholestasis induced by drug toxicity may cause cholestatic hepatic injury
(CHI) leading to liver fibrosis and cirrhosis. The G protein-coupled bile acid receptor 1
(TGR5) is a membrane receptor with well-known roles in the regulation of glucose
metabolism and energy homeostasis. However, the role and mechanism of TGR5 in the context
of inflammation during CHI remains unclear. Wild-type (WT) and TGR5 knockout
(TGR5−/−) mice with CHI induced by bile duct ligation (BDL) were involved
in vivo, and WT and TGR5−/− bone marrow-derived macrophages
(BMDMs) were used in vitro. TGR5 deficiency significantly exacerbated
BDL-induced liver injury, inflammatory responses and hepatic fibrosis compared with WT
mice in vivo. TGR5−/− macrophages were more susceptible to
lipopolysaccharide (LPS) stimulation than WT macrophages. TGR5 activation by its ligand
suppressed LPS-induced pro-inflammatory responses in WT but not TGR5−/− BMDMs.
Notably, expression of β-catenin was effectively inhibited by TGR5 deficiency.
Furthermore, TGR5 directly interacted with Gsk3β to repress the interaction between Gsk3β
and β-catenin, thus disrupting the β-catenin destruction complex. The pro-inflammatory
nature of TGR5-knockout was almost abolished by lentivirus-mediated β-catenin
overexpression in BMDMs. BMDM migration in vitro was accelerated under
TGR5-deficient conditions or supernatant from LPS-stimulated TGR5−/− BMDMs.
From a therapeutic perspective, TGR5−/− BMDM administration aggravated
BDL-induced CHI, which was effectively rescued by β-catenin overexpression. Our findings
reveal that TGR5 plays a crucial role as a novel regulator of immune-mediated CHI by
destabilizing the β-catenin destruction complex, with therapeutic implications for the
management of human CHI.
Collapse
Affiliation(s)
- Jianhua Rao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Chao Yang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Shikun Yang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Hao Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Yuanchang Hu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Ling Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Feng Cheng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| |
Collapse
|
23
|
Gorman E, Millar J, McAuley D, O'Kane C. Mesenchymal stromal cells for acute respiratory distress syndrome (ARDS), sepsis, and COVID-19 infection: optimizing the therapeutic potential. Expert Rev Respir Med 2020; 15:301-324. [PMID: 33172313 DOI: 10.1080/17476348.2021.1848555] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Mesenchymal stromal (stem) cell (MSC) therapies are emerging as a promising therapeutic intervention in patients with Acute Respiratory Distress Syndrome (ARDS) and sepsis due to their reparative, immunomodulatory, and antimicrobial properties.Areas covered: This review provides an overview of Mesenchymal stromal cells (MSCs) and their mechanisms of effect in ARDS and sepsis. The preclinical and clinical evidence to support MSC therapy in ARDS and sepsis is discussed. The potential for MSC therapy in COVID-19 ARDS is discussed with insights from respiratory viral models and early clinical reports of MSC therapy in COVID-19. Strategies to optimize the therapeutic potential of MSCs in ARDS and sepsis are considered including preconditioning, altered gene expression, and alternative cell-free MSC-derived products, such as extracellular vesicles and conditioned medium.Expert opinion: MSC products present considerable therapeutic promise for ARDS and sepsis. Preclinical investigations report significant benefits and early phase clinical studies have not highlighted safety concerns. Optimization of MSC function in preclinical models of ARDS and sepsis has enhanced their beneficial effects. MSC-derived products, as cell-free alternatives, may provide further advantages in this field. These strategies present opportunity for the clinical development of MSCs and MSC-derived products with enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Ellen Gorman
- School of Medicine Dentistry and Biomedical Science, Queen's University Belfast, UK
| | - Jonathan Millar
- Division of Functional Genetics and Development, Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Danny McAuley
- School of Medicine Dentistry and Biomedical Science, Queen's University Belfast, UK
| | - Cecilia O'Kane
- School of Medicine Dentistry and Biomedical Science, Queen's University Belfast, UK
| |
Collapse
|
24
|
Zhang H, Qin H, Zhou C, Feng Q, Yang Y, Sui J, Tang Y. Gene expression profile of lipopolysaccharide‑induced apoptosis of nucleus pulposus cells reversed by syringic acid. Mol Med Rep 2020; 22:5012-5022. [PMID: 33174055 PMCID: PMC7646953 DOI: 10.3892/mmr.2020.11632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/03/2020] [Indexed: 11/26/2022] Open
Abstract
Apoptosis of nucleus pulposus (NP) cells has an important role in the process of intervertebral disc degeneration (IDD), and the search for novel compounds to prevent apoptosis from occurring is urgently required. In the present study, syringic acid (SyrA) was found to exhibit no cytotoxicity on NP cells, and was able to reverse the cytotoxicity, as well as the abnormal expression of Bcl-2 and caspase-3, that were induced by lipopolysaccharide (LPS). The transcriptomes of each group were then analyzed using RNA-Seq. A total of 65 differentially expressed genes (DEGs) were identified in LPS-stimulated groups (LPS group vs. control group), 819 DEGs were identified in the SyrA-reversed groups (SyrA plus LPS group vs. LPS group), and a further 25 DEGs were identified in the SyrA plus LPS group compared with the control group. Reverse transcription-quantitative PCR validation indicated that the alterations in expression of uroplakin 3B-like 1 (UPK3BL1), voltage-dependent calcium channel subunit α-2/δ-1 (CACNA2D1) and polo-like kinase 4 (PLK4) were consistent with the corresponding results of RNA-Seq, and that these genes were involved in both LPS-stimulation and SyrA-reversion processes. Kyoto Encyclopedia of Genes and Genomes analyses indicated that the DEGs in SyrA-reversed groups were involved in, amongst other pathways, ‘Autophagy-other’ and ‘Apoptosis-multiple species’. In conclusion, the addition of SyrA to the NP cells co-incubated with LPS appeared to help prevent the abnormal expression of mRNAs and apoptosis that had been identified in NP cells incubated with LPS alone. The potential mechanism underlying the reversion of SyrA might be attributed to the regulation of CACNA2D1 and PLK4.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Rehabilitation Medicine, Langdong Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530028, P.R. China
| | - Hong Qin
- Department of Health Care, Nanning Maternal and Child Health Hospital, Nanning, Guangxi Zhuang Autonomous Region 530011, P.R. China
| | - Chengen Zhou
- Department of Rehabilitation Medicine, Langdong Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530028, P.R. China
| | - Qiang Feng
- Department of Rehabilitation Medicine, Langdong Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530028, P.R. China
| | - Yuan Yang
- Department of Rehabilitation Medicine, Langdong Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530028, P.R. China
| | - Jinhui Sui
- Department of Rehabilitation Medicine, Langdong Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530028, P.R. China
| | - Yucai Tang
- Department of Rehabilitation Medicine, Langdong Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530028, P.R. China
| |
Collapse
|
25
|
Qin H, Zhao A. Mesenchymal stem cell therapy for acute respiratory distress syndrome: from basic to clinics. Protein Cell 2020; 11:707-722. [PMID: 32519302 PMCID: PMC7282699 DOI: 10.1007/s13238-020-00738-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/12/2020] [Indexed: 01/08/2023] Open
Abstract
The 2019 novel coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has occurred in China and around the world. SARS-CoV-2-infected patients with severe pneumonia rapidly develop acute respiratory distress syndrome (ARDS) and die of multiple organ failure. Despite advances in supportive care approaches, ARDS is still associated with high mortality and morbidity. Mesenchymal stem cell (MSC)-based therapy may be an potential alternative strategy for treating ARDS by targeting the various pathophysiological events of ARDS. By releasing a variety of paracrine factors and extracellular vesicles, MSC can exert anti-inflammatory, anti-apoptotic, anti-microbial, and pro-angiogenic effects, promote bacterial and alveolar fluid clearance, disrupt the pulmonary endothelial and epithelial cell damage, eventually avoiding the lung and distal organ injuries to rescue patients with ARDS. An increasing number of experimental animal studies and early clinical studies verify the safety and efficacy of MSC therapy in ARDS. Since low cell engraftment and survival in lung limit MSC therapeutic potentials, several strategies have been developed to enhance their engraftment in the lung and their intrinsic, therapeutic properties. Here, we provide a comprehensive review of the mechanisms and optimization of MSC therapy in ARDS and highlighted the potentials and possible barriers of MSC therapy for COVID-19 patients with ARDS.
Collapse
Affiliation(s)
- Hua Qin
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, China.
| | - Andong Zhao
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, Beijing, 100853, China
- Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
26
|
Terraza-Aguirre C, Campos-Mora M, Elizondo-Vega R, Contreras-López RA, Luz-Crawford P, Jorgensen C, Djouad F. Mechanisms behind the Immunoregulatory Dialogue between Mesenchymal Stem Cells and Th17 Cells. Cells 2020; 9:cells9071660. [PMID: 32664207 PMCID: PMC7408034 DOI: 10.3390/cells9071660] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) exhibit potent immunoregulatory abilities by interacting with cells of the adaptive and innate immune system. In vitro, MSCs inhibit the differentiation of T cells into T helper 17 (Th17) cells and repress their proliferation. In vivo, the administration of MSCs to treat various experimental inflammatory and autoimmune diseases, such as rheumatoid arthritis, type 1 diabetes, multiple sclerosis, systemic lupus erythematosus, and bowel disease showed promising therapeutic results. These therapeutic properties mediated by MSCs are associated with an attenuated immune response characterized by a reduced frequency of Th17 cells and the generation of regulatory T cells. In this manuscript, we review how MSC and Th17 cells interact, communicate, and exchange information through different ways such as cell-to-cell contact, secretion of soluble factors, and organelle transfer. Moreover, we discuss the consequences of this dynamic dialogue between MSC and Th17 well described by their phenotypic and functional plasticity.
Collapse
Affiliation(s)
- Claudia Terraza-Aguirre
- IRMB, University of Montpellier, INSERM, F-34090 Montpellier, France; (C.T.-A.); (R.A.C.-L.)
| | | | - Roberto Elizondo-Vega
- Facultad de Ciencias Biológicas, Departamento de Biología Celular, Laboratorio de Biología Celular, Universidad de Concepción, Concepción 4030000, Chile;
| | | | - Patricia Luz-Crawford
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago 7620001, Chile;
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, F-34090 Montpellier, France; (C.T.-A.); (R.A.C.-L.)
- CHU Montpellier, F-34295 Montpellier, France
- Correspondence: (C.J.); (F.D.); Tel.: +33-(0)-4-67-33-77-96 (C.J.); +33-(0)-4-67-33-04-75 (F.D.)
| | - Farida Djouad
- IRMB, University of Montpellier, INSERM, F-34090 Montpellier, France; (C.T.-A.); (R.A.C.-L.)
- Correspondence: (C.J.); (F.D.); Tel.: +33-(0)-4-67-33-77-96 (C.J.); +33-(0)-4-67-33-04-75 (F.D.)
| |
Collapse
|
27
|
Yang X, Ma X, Don O, Song Y, Chen X, Liu J, Qu J, Feng Y. Mesenchymal stem cells combined with liraglutide relieve acute lung injury through apoptotic signaling restrained by PKA/β-catenin. Stem Cell Res Ther 2020; 11:182. [PMID: 32429994 PMCID: PMC7238586 DOI: 10.1186/s13287-020-01689-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/19/2020] [Accepted: 04/23/2020] [Indexed: 12/21/2022] Open
Abstract
Background ARDS and ALI are life-threatening diseases with extremely high mortality in patients. Different sources of MSCs could mitigate the symptoms of ALI from diverse mechanisms. Liraglutide is an activator of glucagon-like peptide-1 receptor (GLP-1R) that activates anti-apoptotic pathways and exerts anti-inflammatory effects. We mainly compared the effects of human chorionic villus-derived mesenchymal stem cells (hCMSCs), human bone marrow-derived mesenchymal stem cells (hBMSCs), and human adipose-derived mesenchymal stem cells (hAMSCs) on the treatment of ALI and explored the apoptosis mechanism of combination MSCs of liraglutide. Methods The proliferation of MSCs was detected by MTT assay. Western blot and RT-qPCR were used to detect the expression of GLP-1R, SPC, Ang-1, and KGF in MSCs stimulated by LPS and liraglutide. By using flow cytometry and TUNEL assay to compare the apoptosis of three MSCs under the action of LPS and liraglutide, we selected hCMSCs as the target cells to study the expression of apoptotic protein through the PKA/β-catenin pathway. In ALI animal models, we observed the effects of liraglutide alone, MSCs alone, and MSCs combined with liraglutide by H&E staining, cell counting, immunohistochemistry, and ELISA assay. Results We demonstrated that LPS attenuates the proliferation of the three MSCs and the expression of GLP-1R. Liraglutide could reverse the effects of LPS; increase the expression of SPC, Ang-1, and KGF; and can reduce the apoptosis of three MSCs through the PKA/β-catenin pathway. In the LPS-induced ALI model, MSCs combined with liraglutide showed a significant therapeutic effect, and hCMSCs combined with liraglutide have advantages in the treatment of ALI. Conclusions The therapeutic effect of combination MSCs of liraglutide on ALI was higher than that of MSCs alone or liraglutide alone, and liraglutide could alleviate the symptoms of ALI by reducing MSCs apoptosis.
Collapse
Affiliation(s)
- Xiaotong Yang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20025, China.,State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Xiaoying Ma
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Ocholi Don
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20025, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 20003, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20025, China
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Jieming Qu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20025, China. .,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20025, China.
| | - Yun Feng
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20025, China. .,Institute of Respiratory Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, 20025, China.
| |
Collapse
|
28
|
Extracellular Vesicles, Apoptotic Bodies and Mitochondria: Stem Cell Bioproducts for Organ Regeneration. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00282-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abstract
Purpose of Review
In the current work, we will present the characterization of the main different stem cell-derived vesicular bio-products with potential application in organ regeneration.
Recent Findings
The therapeutic effects of stem cell therapy in organ repair, specifically those utilizing mesenchymal stromal cells, are largely dependent on the cells’ release of different bio-products. Among these bio-products, extracellular vesicles (EVs) appear to play a major role due to their ability to carry and deliver bioactive material for modulation of cellular pathways in recipient cells. Concurrently, mitochondria transfer emerged as a new mechanism of cell communication, in which the bioenergetics of a damaged cell are restored. Finally, apoptotic bodies released by dying apoptotic stem cells contribute to stimulation of the tissue’s stem cells and modulation of the immune response.
Summary
Exploitation of isolated extracellular vesicles, mitochondria and apoptotic bodies in preclinical models of organ damage shows promising results. Here, we describe the results of the pre-clinical applications of stem cell vesicular products, as well as the first clinical trials approaching artificial administration of extracellular vesicles and mitochondria in human subjects and their possible benefits and limitations.
Collapse
|
29
|
Pokrovskaya LA, Zubareva EV, Nadezhdin SV, Lysenko AS, Litovkina TL. Biological activity of mesenchymal stem cells secretome as a basis for cell-free therapeutic approach. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.49413] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem (stromal) cells (MSCs) are self-renewing, cultured adult stem cells which secrete a complex set of multiple soluble biologically active molecules such as chemokines, and cytokines, cell adhesion molecules, lipid mediators, interleukins (IL), growth factors (GFs), hormones, micro RNAs (miRNAs), long non-coding RNAs (lncRNAs), messenger RNAs (mRNAs), exosomes, as well as microvesicles, the secretome. MSCs of various origin, including adipose-derived stem cells (ASCs), bone marrow derived mesenchymal stem cells (BM-MSCs), human uterine cervical stem cells (hUCESCs), may be good candidates for obtaining secretome-derived products. Different population of MSCs can secret different factors which could have anti-inflammatory, anti-apoptotic, anti-fibrotic activities, a neuroprotective effect, could improve bone, muscle, liver regeneration and wound healing. Therefore, the paracrine activity of conditioned medium obtained when cultivating MSCs, due to a plethora of bioactive factors, was assumed to have the most prominent cell-free therapeutic impact and can serve as a better option in the field of regenerative medicine in future.
Collapse
|
30
|
Xia J, Minamino S, Kuwabara K, Arai S. Stem cell secretome as a new booster for regenerative medicine. Biosci Trends 2020; 13:299-307. [PMID: 31527327 DOI: 10.5582/bst.2019.01226] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Stem cells are an undifferentiated cell population that has the ability to develop into many different cell types and also has the ability to repair damaged tissues in some cases. For a long time, the stem cell regenerative paradigm has been based on the assumption that progenitor cells play a critical role in tissue repair by means of their plasticity and differentiation potential. However, recent works suggest that the mechanism underlying the benefits of stem cell transplantation might relate to a paracrine modulatory effect rather than the replacement of affected cells at the site of injury. This paracrine modulatory effect derives from secretome which comprises a diverse host of growth factors, cytokines, chemokines, angiogenic factors, and exosomes which are extracellular vesicles that are produced in the endosomal compartment of most eukaryotic cells and are from about 30 to several hundred nanometers in diameter. The role of these factors is being increasingly recognized as key to the regulation of many physiological processes including leading endogenous and progenitor cells to sites of injury as well as mediating apoptosis, proliferation, migration, and angiogenesis. In reality, the immunomodulatory and paracrine role of these factors may mainly account for the therapeutic effects of stem cells and a number of in vitro and in vivo researches have proved limited stem cell engraftment at the site of injury. As a cell-free way for regenerative medicine therapies, stem cell secretome has shown great potential in a variety of clinical applications including prevention of cardiac disfunction, neurodegenerative disease, type 1 diabetes, hair loss, tumors, and joint osteoarthritis.
Collapse
Affiliation(s)
- Jufeng Xia
- Graduate School of Frontier Science, The University of Tokyo.,Department of stem cell and regenerative medicine, Arai Japan Medical Institute
| | - Shuichi Minamino
- Department of stem cell and regenerative medicine, Arai Japan Medical Institute
| | - Kazuma Kuwabara
- Department of stem cell and regenerative medicine, Arai Japan Medical Institute
| | - Shunichi Arai
- Department of stem cell and regenerative medicine, Arai Japan Medical Institute
| |
Collapse
|
31
|
Sun DZ, Abelson B, Babbar P, Damaser MS. Harnessing the mesenchymal stem cell secretome for regenerative urology. Nat Rev Urol 2020; 16:363-375. [PMID: 30923338 DOI: 10.1038/s41585-019-0169-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The extensive arsenal of bioactive molecules secreted by mesenchymal stem cells (MSCs), known as the secretome, has demonstrated considerable therapeutic benefit in regenerative medicine. Investigation into the therapeutic potential of the secretome has enabled researchers to replicate the anti-inflammatory, pro-angiogenic and trophic effects of stem cells without the need for the cells themselves. Furthermore, treatment with the MSC secretome could circumvent hurdles associated with cellular therapy, including oncogenic transformation, immunoreactivity and cost. Thus, a clear rationale exists for investigating the therapeutic potential of the MSC secretome in regenerative urology. Indeed, preclinical studies have demonstrated the therapeutic benefits of the MSC secretome in models of stress urinary incontinence, renal disease, bladder dysfunction and erectile dysfunction. However, the specific mechanisms underpinning therapeutic activity are unclear and require further research before clinical translation. Improvements in current proteomic methods used to characterize the secretome will be necessary to provide further insight into stem cells and their secretome in regenerative urology.
Collapse
Affiliation(s)
- Daniel Z Sun
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA. .,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA. .,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Benjamin Abelson
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA.,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Paurush Babbar
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA.,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Margot S Damaser
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA.,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| |
Collapse
|
32
|
Lopes-Pacheco M, Robba C, Rocco PRM, Pelosi P. Current understanding of the therapeutic benefits of mesenchymal stem cells in acute respiratory distress syndrome. Cell Biol Toxicol 2019; 36:83-102. [PMID: 31485828 PMCID: PMC7222160 DOI: 10.1007/s10565-019-09493-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/27/2019] [Indexed: 12/20/2022]
Abstract
The acute respiratory distress syndrome (ARDS) is a multifaceted lung disorder in which no specific therapeutic intervention is able to effectively improve clinical outcomes. Despite an improved understanding of molecular mechanisms and advances in supportive care strategies, ARDS remains associated with high mortality, and survivors usually face long-term morbidity. In recent years, preclinical studies have provided mounting evidence of the potential of mesenchymal stem cell (MSC)-based therapies in lung diseases and critical illnesses. In several models of ARDS, MSCs have been demonstrated to induce anti-inflammatory and anti-apoptotic effects, improve epithelial and endothelial cell recovery, and enhance microbial and alveolar fluid clearance, thus resulting in improved lung and distal organ function and survival. Early-stage clinical trials have also demonstrated the safety of MSC administration in patients with ARDS, but further, large-scale investigations are required to assess the safety and efficacy profile of these therapies. In this review, we summarize the main mechanisms whereby MSCs have been shown to exert therapeutic effects in experimental ARDS. We also highlight questions that need to be further elucidated and barriers that must be overcome in order to efficiently translate MSC research into clinical practice.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Chiara Robba
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Patricia Rieken Macêdo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy. .,Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy.
| |
Collapse
|
33
|
Khan RS, Newsome PN. A Comparison of Phenotypic and Functional Properties of Mesenchymal Stromal Cells and Multipotent Adult Progenitor Cells. Front Immunol 2019; 10:1952. [PMID: 31555259 PMCID: PMC6724467 DOI: 10.3389/fimmu.2019.01952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/02/2019] [Indexed: 12/15/2022] Open
Abstract
Both Multipotent Adult Progenitor Cells and Mesenchymal Stromal Cells are bone-marrow derived, non-haematopoietic adherent cells, that are well-known for having immunomodulatory and pro-angiogenic properties, whilst being relatively non-immunogenic. However, they are phenotypically and functionally distinct cell types, which has implications for their efficacy in different settings. In this review we compare the phenotypic and functional properties of these two cell types, to help in determining which would be the superior cell type for different applications.
Collapse
Affiliation(s)
- Reenam S Khan
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Philip N Newsome
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
34
|
Li X, Yuan Z, Chen J, Wang T, Shen Y, Chen L, Wen F. Microarray analysis reveals the changes of circular RNA expression and molecular mechanism in acute lung injury mouse model. J Cell Biochem 2019; 120:16658-16667. [PMID: 31106457 DOI: 10.1002/jcb.28924] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/19/2019] [Indexed: 02/05/2023]
Abstract
Acute lung injury (ALI) is a severe disease with sudden onset, rapid progression, poor treatment response, and high mortality. An increasing number of studies had found that circular RNAs (circRNAs) has significant functions in various diseases, while the role of circRNAs in ALI is not yet clear. The purpose of this study was to find circRNAs related to ALI and their mechanism of action. Expression profiles of lung circRNAs and messenger RNAs (mRNAs) were analyzed by microarray in the ALI mice models and healthy controlled mice. Differentially expressed RNAs were identified, function and pathways were analyzed by bioinformatics analysis. Moreover, the results of the microarray were verified by real-time PCR. We identified 2262 differentially expressed mRNAs and 581 circRNAs between ALI mice and control. Validation of candidate circRNAs by real-time PCR indicates that the majority of circRNAs identified by microarray are reliable and worthy of further study. ALI induced circRNAs primarily function in the metabolic regulatory process. Moreover, differentially expressed circRNAs were mainly involved in signaling pathways of mitogen-activated protein kinases, focal adhesion, FoxO, neurotrophin, and Wnt. In addition, a competitive endogenous RNA network was constructed to further interpret the molecular mechanism of ALI. This study observed significantly changed circRNAs profiles in LPS-induced mouse model and revealed a potential role of circRNAs in ALI.
Collapse
Affiliation(s)
- Xiaoou Li
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhicheng Yuan
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Jun Chen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Tao Wang
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Yongchun Shen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Lei Chen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Fuqiang Wen
- State Key Laboratory of Biotherapy, Division of Pulmonary Diseases, Department of Respiratory and Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
35
|
miRNA-1246 suppresses acute lung injury-induced inflammation and apoptosis via the NF-κB and Wnt/β-catenin signal pathways. Biomed Pharmacother 2018; 108:783-791. [DOI: 10.1016/j.biopha.2018.09.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/22/2018] [Accepted: 09/08/2018] [Indexed: 02/03/2023] Open
|
36
|
Zhang E, Yang Y, Chen S, Peng C, Lavin MF, Yeo AJ, Li C, Liu X, Guan Y, Du X, Du Z, Shao H. Bone marrow mesenchymal stromal cells attenuate silica-induced pulmonary fibrosis potentially by attenuating Wnt/β-catenin signaling in rats. Stem Cell Res Ther 2018; 9:311. [PMID: 30428918 PMCID: PMC6234553 DOI: 10.1186/s13287-018-1045-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/27/2018] [Accepted: 10/14/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pulmonary fibrosis induced by silica dust is an irreversible, chronic, and fibroproliferative lung disease with no effective treatment at present. Previous studies have shown that early intervention with bone marrow mesenchymal stem/stromal cells (BMSCs) has positive effect on anti-pulmonary fibrosis caused by silica dust. However, early intervention using BMSCs is not practical, and the therapeutic effects of BMSCs advanced intervention on pulmonary fibrosis have rarely been reported. In this study, we investigated the effects of advanced transplantation (on the 28th day after exposure to silica suspension) of BMSCs on an established rat model of pulmonary fibrosis. METHODS Sprague Dawley (SD) rats were randomly divided into four groups including (1) control group (n = 6) which were normally fed, (2) silica model group (n = 6) which were exposed to silica suspension (1 mL of 50 mg/mL/rat), (3) BMSC transplantation group (n = 6) which received 1 mL BMSC suspension (2 × 106 cells/mL) by tail vein injection on the 28th day after exposure to silica suspension, and (4) BMSC-CM (conditioned medium) transplantation group (n = 6) which received CM from the same cell number by tail vein injection on the 28th day after exposure to silica suspension. On the 56th day after exposure to silica suspension, we used computed tomography (CT), hematoxylin and eosin (H&E), and Masson's trichrome staining to evaluate the changes in lung tissue. We examined the expression of epithelial-mesenchymal transition (EMT) and Wnt/β-catenin pathway-related proteins in lung tissue using immunohistochemistry and western blotting. RESULTS Successful construction of a pulmonary fibrosis model was confirmed by H&E and Masson's trichrome staining on the 28th day after exposure to silica suspension. On the 56th day after exposure, pulmonary CT examination showed a relieving effect of BMSCs on silica-induced pulmonary fibrosis which was confirmed by H&E and Masson's trichrome staining. Treatment of BMSCs increased the expression of epithelial marker proteins including E-cadherin (E-cad) and cytokeratin19 (CK19) and reduced the expression of fibrosis marker proteins including Vimentin (Vim) and α-Smooth actin (α-SMA) after exposure to silica suspension. Furthermore, we found that Wnt/β-catenin signaling pathway is abnormally activated in silica-induced pulmonary fibrosis, and exogenous transplantation of BMSCs may attenuate their expression. CONCLUSIONS BMSC transplantation inhibits the EMT to alleviate silica-induced pulmonary fibrosis in rats and the anti-fibrotic effect potentially by attenuating Wnt/β-catenin signaling. ᅟ: ᅟ.
Collapse
Affiliation(s)
- Enguo Zhang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Ye Yang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Shangya Chen
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Cheng Peng
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, Queensland, Australia
| | - Martin F Lavin
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia
| | - Abrey J Yeo
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia
| | - Chao Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Xiaoshan Liu
- Department of Radiology, Shandong Tumor Hospital, Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Yingjun Guan
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Xinjing Du
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Zhongjun Du
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.
| | - Hua Shao
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.
| |
Collapse
|
37
|
Pak J, Lee JH, Pak N, Pak Y, Park KS, Jeon JH, Jeong BC, Lee SH. Cartilage Regeneration in Humans with Adipose Tissue-Derived Stem Cells and Adipose Stromal Vascular Fraction Cells: Updated Status. Int J Mol Sci 2018; 19:ijms19072146. [PMID: 30041472 PMCID: PMC6073159 DOI: 10.3390/ijms19072146] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/18/2018] [Accepted: 07/21/2018] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue-derived stem cells (ASCs) in the form of stromal vascular fraction (SVF) and cultured expansion have been applied in clinical settings in some countries to treat osteoarthritis (OA) of knees, one of the most common debilitating, incurable disorders. Since the first report of successful cartilage-like tissue regeneration with autologous adipose SVF containing ASCs, there has been a gradual increase in the number of publications confirming such results. Thus far, most of the reports have been limited to treatments of OA of knees. Recently, successful applications of adipose SVF in treating OA of ankles and hips have been reported. In addition, several groups have reported modified methods of applying adipose SVF, such as combining bone marrow stimulation with adipose SVF or adding additional extracellular matrix (ECM) in treating OA. Here, we present an updated, systematic review of clinical effectiveness and safety in treating OA of knees, ankles, and one hip since 2016 using ASCs in the form of adipose SVF or in cultured expansion, along with a description and suggestion of potential biological mechanisms of cartilage regeneration.
Collapse
Affiliation(s)
- Jaewoo Pak
- Mipro Medical Clinic, 32-3 Chungdamdong, Gangnamgu, Seoul 06068, Korea.
| | - Jung Hun Lee
- National Leading Research Laboratory, Department of Biological Sciences, Myongji University, 116 Myongjiro, Yongin, Gyeonggido 17058, Korea.
| | - Natalie Pak
- Mipro Medical Clinic, 32-3 Chungdamdong, Gangnamgu, Seoul 06068, Korea.
| | - Yoon Pak
- First Medical Center, 11841 South St., Cerritos, CA 90703, USA.
| | - Kwang Seung Park
- National Leading Research Laboratory, Department of Biological Sciences, Myongji University, 116 Myongjiro, Yongin, Gyeonggido 17058, Korea.
| | - Jeong Ho Jeon
- National Leading Research Laboratory, Department of Biological Sciences, Myongji University, 116 Myongjiro, Yongin, Gyeonggido 17058, Korea.
| | - Byeong Chul Jeong
- National Leading Research Laboratory, Department of Biological Sciences, Myongji University, 116 Myongjiro, Yongin, Gyeonggido 17058, Korea.
| | - Sang Hee Lee
- National Leading Research Laboratory, Department of Biological Sciences, Myongji University, 116 Myongjiro, Yongin, Gyeonggido 17058, Korea.
| |
Collapse
|
38
|
Yu Y, Wang D, Li H, Liu Y, Xiang Z, Wu J, Jing X. IPSC‑MSC inhibition assessment in Raw 264.7 cells following oxygen and glucose deprivation reveals a distinct function for cardiopulmonary resuscitation. Mol Med Rep 2018; 17:8212-8220. [PMID: 29658608 PMCID: PMC5983996 DOI: 10.3892/mmr.2018.8864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/02/2017] [Indexed: 01/01/2023] Open
Abstract
Hypoxia is a serious stress state. The nervous system is less tolerant to hypoxia, and cell death due to hypoxia is irreversible. With the incidence of cardiovascular disease gradually increasing, the sudden cardiac death rate is additionally increasing. Although cardiopulmonary resuscitation (CPR) is an important development, recovery is frequently poor. In a successful recovery population, ~40% of the population was in a vegetative state or subsequently succumbed to their condition, and ~20% had brain damage. Therefore, the recovery of the brain is of particular importance in CPR. Immune disorders are one of the major mechanisms of cerebral resuscitation following CPR. Studies have demonstrated that induced pluripotent stem cell-derived mesenchymal stem cells (IPSC-MSCs) have a strong immune regulatory effect during tissue repair and anti-inflammatory effects. IPSC-MSCs may inhibit the inflammatory response by means of the inflammatory reaction network to improve brain function following CPR, although the cellular and molecular mechanisms remain unclear. Macrophages are a bridge between innate immune and specific immune responses in the body; therefore, it was hypothesized that macrophages may be the important effector cell of the role of IPSC-MSCs in improving brain function following recovery of spontaneous respiration and circulation subsequent to cardiopulmonary resuscitation. In the present study, IPSC-MSCs were applied to the oxygen and glucose deprivation (OGD) model. It was observed that intervention with IPSC-MSCs was able to alter the polarization direction of macrophages. The difference in the proportions of M1 and M2 macrophages was statistically significant at 6, 12, 24 and 48 h (P=0.037, P<0.05) in the OGD + IPSC-MSCs group (M1, 33.48±5.6%; M2, 50.84±6.9%) and in the OGD group (M1, 83.55±7.3%; M2, 11.41±3.2%), and over time this trend was more obvious. The polarization direction of macrophages is associated with the neurogenic locus notch homolog protein 1 (Notch-1) signaling pathway. In conclusion, it was observed that IPSC-MSCs may be associated with altered macrophage polarization, which may be accomplished by inhibiting the Notch-1 signaling pathway.
Collapse
Affiliation(s)
- Yi Yu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Dongping Wang
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hui Li
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yujie Liu
- Department of Breast Surgery and Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat‑Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhao Xiang
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Junlin Wu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoli Jing
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
39
|
Xu M, Liu S, Wan R, Chen Y. Combined treatment with sinomenine and acupuncture on collagen-induced arthritis through the NF-κB and MAPK signaling pathway. Oncol Lett 2018; 15:8770-8776. [PMID: 29805616 DOI: 10.3892/ol.2018.8394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 09/22/2017] [Indexed: 01/13/2023] Open
Abstract
Sinomenine is a monomer extracted from the traditional Chinese medicine plant Sabia japonica, which possesses several pharmacological properties including prominent abirritation, mitigation, anti-inflammation, immune suppression, cough relief, stimulation of histamine release, decrease in blood pressure and antiarrhythmia. Sinomenine is clinically employed to treat rheumatic disease. To investigate the impact of combined sinomenine treatment with acupuncture on the progression of arthritis and explore the potential underlying molecular mechanisms, the present study analyzed a collagen-induced arthritis model. Results from the combined curative (CC) treatment group (combined treatment with sinomenine and acupuncture) demonstrated a decrease in volume changes and arthritis score changes within rat paws, and increased the overall body weight in arthritic rats. CC treatment significantly decreased tumor necrosis factor α, interleukin (IL)-6, IL-1β and IL-8 serum levels in arthritic rats. CC treatment significantly increased superoxide dismutase and inhibited malondialdehyde levels in arthritic rats. The protein expression of cyclooxygenase-2, inducible nitric oxide synthase, matrix metalloproteinase (MMP)2 and MMP9 in arthritic rats was suppressed owing to CC treatment. Finally, nuclear factor κB and phosphorylated p38 mitogen-activated protein kinase (MAPK) protein expression in arthritic rats were also suppressed following CC treatment. The results indicate that the combined treatment of sinomenine and acupuncture on collagen-induced arthritis takes effect through the nuclear factor κB and MAPK signaling pathway.
Collapse
Affiliation(s)
- Minmin Xu
- Department of Orthopedics, Chongqing TCM Hospital, Chongqing 400000, P.R. China
| | - Shaofan Liu
- Department of Orthopedics, Chongqing TCM Hospital, Chongqing 400000, P.R. China
| | - Ruijie Wan
- Department of Orthopedics, Chongqing TCM Hospital, Chongqing 400000, P.R. China
| | - Yu Chen
- Department of Orthopedics, Chongqing TCM Hospital, Chongqing 400000, P.R. China
| |
Collapse
|
40
|
Huang N, Li W, Wang X, Qi S. MicroRNA-17-5p aggravates lipopolysaccharide-induced injury in nasal epithelial cells by targeting Smad7. BMC Cell Biol 2018; 19:1. [PMID: 29433423 PMCID: PMC5809994 DOI: 10.1186/s12860-018-0152-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/16/2018] [Indexed: 01/07/2023] Open
Abstract
Background Globally, rhinitis is one of the most common chronic disorders. Despite availability of drugs to manage the symptomatology of rhinitis, researchers still focus on identification of novel molecular targets for better management. MicroRNAs are implicated in many biological and pathological processes. However, the role of miR-17-5p in rhinitis remains unexplored. This study aimed to explore the role of miR-17-5p in lipopolysaccharide (LPS)-induced injury of nasal epithelial RPMI2650 cells and to elucidate the possible underlying molecular mechanism. Results LPS damaged RPMI2650 cells by inhibiting cell proliferation, promoting apoptosis, and stimulating the release of inflammatory cytokines. miR-17-5p expression was significantly increased in RPMI2650 cells following treatment with LPS. Furthermore, it was found that overexpression of miR-17-5p led to aggravation of LPS-induced injury. miR-17-5p negatively regulated expression of Smad7; overexpression of Smad7 protected the RPMI2650 cells by inactivating NF-κB and Wnt/β catenin pathways and vice versa. Conclusions Overexpression of miR-17-5p aggravated LPS-induced damage of RPMI2650 cells. Expression of Smad7 was negatively regulated by miR-17-5p; Smad7 expression inactivated NF-κB and Wnt/β catenin pathways.
Collapse
Affiliation(s)
- Nan Huang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenjing Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaolong Wang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shanshan Qi
- Department of Allergy, Wuhan No.1 Hospital, No. 215, Zhongshan Avenue, Wuhan, 430022, China.
| |
Collapse
|
41
|
The promise of mesenchymal stem cell therapy for acute respiratory distress syndrome. J Trauma Acute Care Surg 2018; 84:183-191. [PMID: 29019797 DOI: 10.1097/ta.0000000000001713] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review describes the current state of the science on mesenchymal stem cell (MSC) treatment for acute lung injury (ALI). The general characteristics, regenerative potential, and mechanism of action of MSCs are first presented. Next, particular emphasis is placed on the application of MSCs for the treatment of acute respiratory distress syndrome (ARDS) in preclinical and clinical studies. Finally, we discuss current challenges and future directions in the field presented from a clinician-researcher perspective. The objective of this work is to provide the readership with a current review of the literature discussing the hurdles and overall promise of MSCs as therapeutic interventions for the treatment of ARDS.
Collapse
|
42
|
Wnt/β-catenin pathway promotes acute lung injury induced by LPS through driving the Th17 response in mice. Biochem Biophys Res Commun 2018; 495:1890-1895. [DOI: 10.1016/j.bbrc.2017.12.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 12/11/2017] [Indexed: 12/19/2022]
|
43
|
Abstract
Abstract
Sepsis is a life-threatening syndrome resulting in shock and organ dysfunction stemming from a microbial infection. Sepsis has a mortality of 40% and is implicated in half of all in-hospital deaths. The host immune response to microbial infection is critical, with early-phase sepsis characterized by a hyperinflammatory immune response, whereas the later phase of sepsis is often complicated by suppression. Sepsis has no treatment, and management remains supportive.
Stem cells constitute exciting potential therapeutic agents for sepsis. In this review, we examine the rationale for stem cells in sepsis, focusing on mesenchymal stem/stromal cells, which currently demonstrate the greatest therapeutic promise. We examine the preclinical evidence base and evaluate potential mechanisms of action of these cells that are important in the setting of sepsis. We discuss early-phase clinical trials and critically appraise translational barriers to the use of mesenchymal stem/stromal cells in patients with sepsis.
Collapse
|
44
|
Fang Y, Kang Y, Zou H, Cheng X, Xie T, Shi L, Zhang H. β-elemene attenuates macrophage activation and proinflammatory factor production via crosstalk with Wnt/β-catenin signaling pathway. Fitoterapia 2017; 124:92-102. [PMID: 29066299 DOI: 10.1016/j.fitote.2017.10.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/10/2017] [Accepted: 10/20/2017] [Indexed: 11/27/2022]
Abstract
β-elemene, extracted from Rhizoma zedoariae, has been widely used as a traditional medicine for its antitumor activity against a broad range of cancers. However, the effect of β-elemene in inflammation disorders has yet to be determined. The present study was designed to investigate the anti-inflammatory effects and potential molecular mechanisms of β-elemene in lipopolysaccharide (LPS)-induced murine macrophage cells RAW264.7. We found that the production of pro-inflammatory mediators, including interleukin-6(IL-6), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), induced by LPS was significantly suppressed by β-elemene in a dose-dependent manner in RAW264.7 macrophage cell line. Also, β-elemene inhibited LPS-induced nitric oxide synthase (iNOS) and interleukin-10 (IL-10) expression by RAW264.7, which was related to the down-regulation of Wnt/β-catenin signaling pathway. Importantly, this study demonstrates that β-catenin was significantly inhibited by β-elemene, which appeared to be largely responsible for the down-regulation of Wnt/β-catenin signaling pathway. Accordingly, the deletion of β-catenin in primary macrophages reversed β-catenin-elicited inhibition of immune response. Furthermore, β-catenin expression and Wnt/β-catenin signaling pathway induced by LPS in RAW264.7 was also significantly inhibited by α-humulene, one isomeric sesquiterpene of β-elemene. α-humulene was also found to significantly inhibit LPS-induced production of proinflammatory cytokines. However, α-humulene showed more cytotoxic ability than β-elemene. Collectively, our data illustrated that β-elemene exerted a potent inhibitory effect on pro-inflammatory meditator and cytokines production via the inactivation of β-catenin, and also demonstrated the protective functions of β-elemene in endotoxin-induced inflammation. β-elemene may serve as potential nontoxic modulatory agents for the prevention and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Yangyi Fang
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yanhua Kang
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Department of Immunology, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zou
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaxuan Cheng
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Department of Biochemistry and Molecular Biology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Tian Xie
- Institute of Holistic Integrative Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Institute of Holistic Integrative Oncology, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Liyun Shi
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Department of Immunology, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Hang Zhang
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Institute of Holistic Integrative Oncology, Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|
45
|
Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int J Mol Sci 2017; 18:ijms18091852. [PMID: 28841158 PMCID: PMC5618501 DOI: 10.3390/ijms18091852] [Citation(s) in RCA: 816] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 02/07/2023] Open
Abstract
Earlier research primarily attributed the effects of mesenchymal stem cell (MSC) therapies to their capacity for local engrafting and differentiating into multiple tissue types. However, recent studies have revealed that implanted cells do not survive for long, and that the benefits of MSC therapy could be due to the vast array of bioactive factors they produce, which play an important role in the regulation of key biologic processes. Secretome derivatives, such as conditioned media or exosomes, may present considerable advantages over cells for manufacturing, storage, handling, product shelf life and their potential as a ready-to-go biologic product. Nevertheless, regulatory requirements for manufacturing and quality control will be necessary to establish the safety and efficacy profile of these products. Among MSCs, human uterine cervical stem cells (hUCESCs) may be a good candidate for obtaining secretome-derived products. hUCESCs are obtained by Pap cervical smear, which is a less invasive and painful method than those used for obtaining other MSCs (for example, from bone marrow or adipose tissue). Moreover, due to easy isolation and a high proliferative rate, it is possible to obtain large amounts of hUCESCs or secretome-derived products for research and clinical use.
Collapse
|
46
|
Cao Y, Wu BJ, Zheng WP, Yin ML, Liu T, Song HL. Effect of heme oxygenase-1 transduced bone marrow mesenchymal stem cells on damaged intestinal epithelial cells in vitro. Cell Biol Int 2017; 41:726-738. [PMID: 28206713 DOI: 10.1002/cbin.10749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/13/2017] [Indexed: 12/21/2022]
Abstract
In this study, we explored the effects of mesenchymal stem cells (MSCs) from bone marrow overexpressing heme oxygenase-1 (HO-1) on the damaged human intestinal epithelial barrier in vitro. Rat MSCs were isolated from bone marrow and transduced with rat HO-1 recombinant adenovirus (HO-MSCs) for stable expression of HO-1. Colorectal adenocarinoma 2 (Caco2) cells were treated with tumor necrosis factor-α (TNF-α) to establish a damaged colon epithelial model. Damaged Caco2 were cocultured with MSCs, Ad-MSCs, Ad-HO + MSCs or HO-MSCs. mRNA and protein expression of Zona occludens-1 (ZO-1) and human HO-1 and the release of cytokines were measured. ZO-1 and human HO-1 in Caco2 were significantly decreased after treatment with TNF-α; and this effect was reduced when coculture with MSCs from bone marrow. Expression of ZO-1 was not significantly affected by Caco2 treatment with TNF-α, Ad-HO, and MSCs. In contrast, ZO-1 and human HO-1 increased significantly when the damaged Caco2 was treated with HO-MSCs. HO-MSCs showed the strongest effect on the expression of ZO-1 in colon epithelial cells. Coculture with HO-MSCs showed the most significant effects on reducing the expression of IL-2, IL-6, IFN-γ and increasing the expression of IL-10. HO-MSCs protected the intestinal epithelial barrier, in which endogenous HO-1 was involved. HO-MSCs play an important role in the repair process by reducing the release of inflammatory cytokines and increasing the release of anti-inflammatory factors. These results suggested that HO-MSCs from bone marrow were more effective in repairing the damaged intestinal epithelial barrier, and the effectiveness of MSCs was improved by HO-1 gene transduction, which provides favorable support for the application of stem cell therapy in the intestinal diseases.
Collapse
Affiliation(s)
- Yi Cao
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, China
| | - Ben-Juan Wu
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, China
| | - Wei-Ping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, China
| | - Ming-Li Yin
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, China
| | - Tao Liu
- Key Laboratory of Emergency Care Medicine of Ministry of Health, Tianjin First Central Hospital, Tianjin, China
| | - Hong-Li Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin, China.,Tianjin Key Laboratory of Organ Transplantation, Tianjin, China
| |
Collapse
|
47
|
Gonzalez P, Rodríguez FJ. Analysis of the expression of the Wnt family of proteins and its modulatory role on cytokine expression in non activated and activated astroglial cells. Neurosci Res 2016; 114:16-29. [PMID: 27562517 DOI: 10.1016/j.neures.2016.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 12/23/2022]
Abstract
Despite the essential functions of astrocytes and the emerging relevance of the Wnt family of proteins in the CNS under physiological and pathological conditions, the astroglial expression of this family of proteins and its potential modulatory role on astroglial activation is almost unknown. Thus, we have evaluated the expression of all Wnt ligands, receptors and regulators, and the activation state of Wnt-related signaling pathways in non-activated and differentially activated astroglial cultures. We found that numerous Wnt ligands, receptors and regulators were expressed in non-activated astrocytes, while the Wnt-dependent pathways were constitutively active. Moreover, the expression of most detectable Wnt-related molecules and the activity of the Wnt-dependent pathways suffered post-activation variations which frequently depended on the activation system. Finally, the analysis of the effects exerted by Wnt1 and 5a on the astroglial expression of prototypical genes related to astroglial activation showed that both Wnt ligands increased the astroglial expression of interleukin 1β depending on the experimental context, while did not modulate tumor necrosis factor α, interleukin 6, transforming growth factor β1 and glial fibrillary acidic protein expression. These results strongly suggest that the Wnt family of proteins is involved in how astrocytes modulate and respond to the physiological and pathological CNS.
Collapse
Affiliation(s)
- Pau Gonzalez
- Laboratory of Molecular Neurology, National Hospital for Paraplegics, Finca la Peraleda s/n, 45071 Toledo, Spain.
| | - Francisco Javier Rodríguez
- Laboratory of Molecular Neurology, National Hospital for Paraplegics, Finca la Peraleda s/n, 45071 Toledo, Spain.
| |
Collapse
|
48
|
Cruz FF, Weiss DJ, Rocco PRM. Prospects and progress in cell therapy for acute respiratory distress syndrome. Expert Opin Biol Ther 2016; 16:1353-1360. [DOI: 10.1080/14712598.2016.1218845] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
49
|
Could stem cells be the future therapy for sepsis? Blood Rev 2016; 30:439-452. [PMID: 27297212 DOI: 10.1016/j.blre.2016.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/15/2022]
Abstract
The severity and threat of sepsis is well known, and despite several decades of research, the mortality continues to be high. Stem cells have great potential to be used in various clinical disorders. The innate ability of stem cells such as pluripotency, self-renewal makes them potential agents for therapeutic intervention. The pathophysiology of sepsis is a plethora of complex mechanisms which include the initial microbial infection, followed by "cytokine storm," endothelial dysfunction, coagulation cascade, and the late phase of apoptosis and immune paralysis which ultimately results in multiple organ dysfunction. Stem cells could potentially alter each step of this complex pathophysiology of sepsis. Multiple organ dysfunction associated with sepsis most often leads to death and stem cells have shown their ability to prevent the organ damage and improve the organ function. The possible mechanisms of therapeutic potential of stem cells in sepsis have been discussed in detail. The route of administration, dose level, and timing also play vital role in the overall effect of stem cells in sepsis.
Collapse
|
50
|
Wang J, Qin Y, Mi X. The protective effects of bone marrow-derived mesenchymal stem cell (BMSC) on LPS-induced acute lung injury via TLR3-mediated IFNs, MAPK and NF-κB signaling pathways. Biomed Pharmacother 2016; 79:176-87. [PMID: 27044826 DOI: 10.1016/j.biopha.2016.02.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 12/29/2022] Open
Abstract
The study attempted to clarify the protective role of bone marrow-derived mesenchymal stem cell (BMSC) transplantation on LPS-induced acute lung injury (ALI) of rats. BMSC were obtained from bone marrow of rat, cultured and proliferated in vitro. Rats of ALI were established through lipopolysaccharide (LPS) administration. Male rats were allocated to control group, ALI group and BMSC, transplantation group. Rats were sacrificed after BMSC injection after 12h, 24h and 48h. Here we investigated the role of BMSC in LPS-induced alveolar macrophages to further demonstrate the mechanism of BMSC to lung injury. TLR3, a member of Toll-like receptor family, has been found in macrophages and the cell surface. In our study, first BMSC successfully reversed LPS-induced lung injury by hematoxylin-eosin (H&E) staining, ameliorated apoptosis via TUNEL and flow cytometer analysis, as well as improved cell structure. And then, western blot, quantitative real-time PCR, immunohistochemistry and immunofluorescence analysis were used to confirm that TLR3 was significantly down-regulated for BMSC treatment. Subsequently, TRIF and RIP1, down-streaming signals of TLR3, were inhibited greatly, leading to TRAF3, MAPK as well as NF-κB inactivity. Our results indicated that BMSC transplantation group displayed inhibitory effects on interferon (IFNs) levels via TLR3 in LPS-induced ALI and preventive effects on inflammation response via TLR3-regualted MAPK and NF-κB signaling pathway in LPS-induced lung injury. The present study indicated that BMSC could display protective effects on LPS-induced ALI and provide an experimental basis for clinical therapy.
Collapse
Affiliation(s)
- Jingcai Wang
- Department of Pediatrics, People's Hospital of Liaocheng, Shandong 252000, China
| | - Ying Qin
- General Hospital of Jinan Iron and Steel Group Co., Ltd., Shandong 252000, China.
| | - Xiuju Mi
- Department of Pediatrics, People's Hospital of Liaocheng, Shandong 252000, China
| |
Collapse
|