1
|
Jin J, Wang Z, Liu Y, Chen J, Jiang M, Lu L, Xu J, Gao F, Wang J, Zhang J, Xu GT, Jin C, Tian H, Zhao J, Ou Q. miR-143-3p boosts extracellular vesicles to improve the dermal fibrosis of localized scleroderma. J Autoimmun 2025; 153:103422. [PMID: 40273600 DOI: 10.1016/j.jaut.2025.103422] [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: 05/10/2024] [Revised: 03/15/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Localized scleroderma (LoSc) is an autoimmune disease that features extensive fibrosis of the skin. Due to its severity and limited understanding, no effective treatments have been developed to date. Bone marrow mesenchymal stem cells (BMSCs) derived extracellular vesicles (EVs) have been demonstrated promising therapeutic effects on the LoSc mouse model in our previous study. However, identifying the targets and underlying mechanisms of EVs remains a significant challenge for therapeutic applications. miR-143-3p, a critical and abundant factor in BMSC-EVs identified through miRNA sequencing, mediates antifibrotic effects in a LoSc mouse model and is significantly lacking in the dermis of LoSc patients. This microRNA inhibits myofibroblast formation and collagen synthesis, contributing to the therapeutic effects of BMSC-EVs in the LoSc mouse model. Moreover, miR-143-3p-reinforced BMSC-EVs demonstrated enhanced therapeutic efficacy compared to normal BMSC-EVs, reducing dermal thickening, collagen deposition, fibroblast differentiation into myofibroblasts, and promoting skin tissue remodeling. IGF1R, highly expressed in the skin of LoSc, was identified as a potential target of miR-143-3p and was inhibited by miR-143-3p-reinforced EVs, thereby modulating the IGF1/IGF1R-AKT/MAPK pathway. In conclusion, miR-143-3p-enriched EVs could be a more efficient candidate for treating dermal fibrosis in LoSc.
Collapse
Affiliation(s)
- Jiahui Jin
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Dermatology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Wang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Physiology, College of Basic Medical Sciences, Naval Medical University, Shanghai, China
| | - Yifan Liu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Chen
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Miao Jiang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lixia Lu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingying Xu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Furong Gao
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Juan Wang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jieping Zhang
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guo-Tong Xu
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Caixia Jin
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Haibin Tian
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Jingjun Zhao
- Department of Dermatology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qingjian Ou
- Department of Dermatology and Laboratory of Clinical and Visual Sciences, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
2
|
Bao N, Wang J, Yue Q, Cao F, Gu X, Wen K, Kong W, Gu M. Chrysophanol-mediated trx-1 activation attenuates renal fibrosis through inhibition of the JNK/Cx43 signaling pathway. Ren Fail 2024; 46:2398710. [PMID: 39238246 PMCID: PMC11382722 DOI: 10.1080/0886022x.2024.2398710] [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: 03/30/2024] [Revised: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
PURPOSE This study aimed to investigate the inhibitory effect of chrysophanol on renal fibrosis and its molecular mechanism. METHODS Initially, potential targets of chrysophanol were predicted through network pharmacology analysis, and a protein-protein interaction network of these targets was constructed using Venn diagrams and the STRING database. GO enrichment analysis predicted the biological process of chrysophanol in treating renal fibrosis. Subsequently, both in vivo and in vitro experiments were conducted using unilateral ureteral obstruction (UUO) induced CKD mouse model and HK-2 cell model, respectively. In the mouse model, different doses of chrysophanol were administered to assess its renal protective effects through biochemical indicators, histological examination, and immunofluorescence staining. In the cell model, the regulatory effect of chrysophanol on the Trx-1/JNK/Cx43 pathway was evaluated using western blotting and flow cytometry. RESULTS Chrysophanol treatment significantly ameliorated renal dysfunction and histopathological damage in the UUO mouse model, accompanied by a reduction in serum oxidative stress markers. Furthermore, chrysophanol markedly upregulated the expression of Trx-1 in renal tissues and inhibited the activation of the JNK/Cx43 signaling pathway. At the cellular level, chrysophanol enhanced the activity of Trx-1 and downregulated the JNK/Cx43 signaling pathway, thereby inhibiting TGF-β induced oxidative stress and cell apoptosis. CONCLUSION This study demonstrated a significant inhibitory effect of chrysophanol on renal fibrosis, mediated by the activation of Trx-1 to inhibit the JNK/Cx43 pathway. These findings provide experimental support for the potential use of chrysophanol as a therapeutic agent for renal fibrosis.
Collapse
Affiliation(s)
- Neng Bao
- Department of Nephrology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing city, Jiangsu, China
| | - Jin Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Qiyu Yue
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu, China
| | - Fang Cao
- Department of Nephrology, Changshu Hospital affiliated to Nanjing University of Chinese Medicine, Changshu city, Jiangsu, China
| | - Xuejing Gu
- Department of Nephrology, Changshu Hospital affiliated to Nanjing University of Chinese Medicine, Changshu city, Jiangsu, China
| | - Kejian Wen
- Department of Nephrology, Changshu Hospital affiliated to Nanjing University of Chinese Medicine, Changshu city, Jiangsu, China
| | - Wei Kong
- Department of Nephrology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing city, Jiangsu, China
| | - Mingjia Gu
- Department of Nephrology, Changshu Hospital affiliated to Nanjing University of Chinese Medicine, Changshu city, Jiangsu, China
| |
Collapse
|
3
|
Yang H, Cheong S, He Y, Lu F. Mesenchymal stem cell-based therapy for autoimmune-related fibrotic skin diseases-systemic sclerosis and sclerodermatous graft-versus-host disease. Stem Cell Res Ther 2023; 14:372. [PMID: 38111001 PMCID: PMC10729330 DOI: 10.1186/s13287-023-03543-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/23/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) and sclerodermatous graft-versus-host disease (Scl-GVHD)-characterized by similar developmental fibrosis, vascular abnormalities, and innate and adaptive immune response, resulting in severe skin fibrosis at the late stage-are chronic autoimmune diseases of connective tissue. The significant immune system dysfunction, distinguishing autoimmune-related fibrosis from mere skin fibrosis, should be a particular focus of treating autoimmune-related fibrosis. Recent research shows that innovative mesenchymal stem cell (MSC)-based therapy, with the capacities of immune regulation, inflammation suppression, oxidation inhibition, and fibrosis restraint, shows great promise in overcoming the disease. MAIN BODY This review of recent studies aims to summarize the therapeutic effect and theoretical mechanisms of MSC-based therapy in treating autoimmune-related fibrotic skin diseases, SSc and Scl-GVHD, providing novel insights and references for further clinical applications. It is noteworthy that the efficacy of MSCs is not reliant on their migration into the skin. Working on the immune system, MSCs can inhibit the chemotaxis and infiltration of immune cells to the skin by down-regulating the expression of skin chemokines and chemokine receptors and reducing the inflammatory and pro-fibrotic mediators. Furthermore, to reduce levels of oxidative stress, MSCs may improve vascular abnormalities, and enhance the antioxidant defenses through inducible nitric oxide synthase, thioredoxin 1, as well as other mediators. The oxidative stress environment does not weaken MSCs and may even strengthen certain functions. Regarding fibrosis, MSCs primarily target the transforming growth factor-β signaling pathway to inhibit fibroblast activation. Here, miRNAs may play a critical role in ECM remodeling. Clinical studies have demonstrated the safety of these approaches, though outcomes have varied, possibly owing to the heterogeneity of MSCs, the disorders themselves, and other factors. Nevertheless, the research clearly reveals the immense potential of MSCs in treating autoimmune-related fibrotic skin diseases. CONCLUSION The application of MSCs presents a promising approach for treating autoimmune-related fibrotic skin diseases: SSc and Scl-GVHD. Therapies involving MSCs and MSC extracellular vesicles have been found to operate through three primary mechanisms: rebalancing the immune and inflammatory disorders, resisting oxidant stress, and inhibiting overactivated fibrosis (including fibroblast activation and ECM remodeling). However, the effectiveness of these interventions requires further validation through extensive clinical investigations, particularly randomized control trials and phase III/IV clinical trials. Additionally, the hypothetical mechanism underlying these therapies could be elucidated through further research.
Collapse
Affiliation(s)
- Han Yang
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Sousan Cheong
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yunfan He
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| | - Feng Lu
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
4
|
Xie L, Long X, Mo M, Jiang J, Zhang Q, Long M, Li M. Bone marrow mesenchymal stem cell-derived exosomes alleviate skin fibrosis in systemic sclerosis by inhibiting the IL-33/ST2 axis via the delivery of microRNA-214. Mol Immunol 2023; 157:146-157. [PMID: 37028129 DOI: 10.1016/j.molimm.2023.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/24/2023] [Accepted: 03/23/2023] [Indexed: 04/09/2023]
Abstract
Interleukin (IL)- 33 is a tissue-derive proinflammatory cytokine that promotes fibrosis in systemic sclerosis (SSc). microRNA (miR)- 214 expression has been elaborated to be downregulated in SSc patients and exert anti-fibrotic and anti-inflammatory effects. This study elucidates the role of bone marrow mesenchymal stem cell-derived exosome (BMSC-Exos)-delivered miR-214 in SSc and the relationship between this miR and IL-33/ST2 axis. SSc clinical samples were obtained to evaluate levels of miR-214, IL-33, and ST2. Primary fibroblasts and BMSC-Exos were extracted, followed by the co-culture of PKH6-labeled BMSC-Exos and fibroblasts. Subsequently, Exos extracted from miR-214 inhibitor-transfected BMSCs were co-cultured with TGF-β1-stimulated fibroblasts, after which the expression of fibrotic markers, miR-214, IL-33, and ST2, as well as fibroblast proliferation and migration, was determined. A skin fibrosis mouse model was induced with bleomycin (BLM) and treated with BMSC-Exos. Collagen fiber accumulation, collagen content, α-SMA expression, and IL-33 and ST2 levels were examined in BLM-treated or IL-33-knockout mice. IL-33 and ST2 were upregulated and miR-214 was downregulated in SSc patients. Mechanistically, miR-214 targeted IL-33 and blocked the IL-33/ST2 axis. BMSC-Exos delivering miR-214 inhibitor augmented proliferation, migration, and fibrotic gene expression in TGF-β1-stimulated fibroblasts. Similarly, IL-33 induced migration, proliferation, and fibrotic gene expression in fibroblasts via ST2. In BLM-treated mice, IL-33 knockout suppressed skin fibrosis, and BMSC-Exos delivered miR-214 to suppress the IL-33/ST2 axis, thus mitigating skin fibrosis. Conclusively, BMSC-Exos alleviate skin fibrosis through the blockade of the IL-33/ST2 axis by delivering miR-214.
Collapse
Affiliation(s)
- Lihu Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiaoping Long
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Meili Mo
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Jinmei Jiang
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Qingxiu Zhang
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Mei Long
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Mei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
5
|
Shen YW, Cheng YA, Li Y, Li Z, Yang BY, Li X. Sambucus williamsii Hance maintains bone homeostasis in hyperglycemia-induced osteopenia by reversing oxidative stress via cGMP/PKG signal transduction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154607. [PMID: 36610352 DOI: 10.1016/j.phymed.2022.154607] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 11/30/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Sambucus williamsii Hance (SWH) has effectively been adopted to treat joint and bone disorders. Diabetes-induced osteopenia (DOP) is caused primarily by impaired bone formation as a result of hyperglycemia. We had previously demonstrated that SWH extract accelerated fracture healing and promoted osteoblastic MC3T3-E1 cell proliferation and osteogenic differentiation. This study assessed the impacts of SWH extract on diabetes-induced bone loss and explored the mechanisms underlying its osteoprotective effects. METHODS This work employed MC3T3-E1 cell line for evaluating how SWH extract affected osteogenesis, oxidative stress (OS), and the underlying mechanism in vitro. Streptozotocin-induced osteopenia mouse model was applied with the purpose of assessing SWH extract's osteoprotection on bone homeostasis in vivo. RESULTS The increased OS of MC3T3-E1 cells exposed to high glucose (HG) was largely because of the upregulation of pro-oxidant genes and the downregulation of antioxidant genes, whereas SWH extract reduced the OS by modulating NADPH oxidase-4 and thioredoxin-related genes by activating cyclic guanosine monophosphate (cGMP) production and increasing the level of cGMP-mediated protein kinase G type-2 (PKG2). The oral administration of SWH extract maintained bone homeostasis in type 1 diabetes mellitus (T1DM) mice by enhancing osteogenesis while decreasing OS. In bones from hyperglycemia-induced osteopenia mice and HG-treated MC3T3-E1 cells, the SWH extract achieved the osteoprotective effects through activating the cGMP/PKG2 signaling pathway, upregulating the level of antioxidant genes, as well as downregulating the level of pro-oxidant genes. CONCLUSION SWH extract exerts osteoprotective effects on hyperglycemia-induced osteopenia by reversing OS via cGMP/PKG signal transduction and is a potential therapy for DOP.
Collapse
Affiliation(s)
- Yi-Wei Shen
- Ningbo Hospital of Traditional Chinese Medicine (Ningbo Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medicine University), Ningbo, Zhejiang, 315010, China; The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, Heilongjiang 150040, China; Key Laboratory of Northern Medicine Base and Application under Ministry of d Education, Harbin, Heilongjiang 150040, China; Key Laboratory of Chinese Materia Medica, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Yang-Ang Cheng
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, Heilongjiang 150040, China; Key Laboratory of Northern Medicine Base and Application under Ministry of d Education, Harbin, Heilongjiang 150040, China
| | - Yi Li
- College of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Zuo Li
- College of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Bing-You Yang
- College of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, China
| | - Xue Li
- Ningbo Hospital of Traditional Chinese Medicine (Ningbo Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medicine University), Ningbo, Zhejiang, 315010, China; The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin, Heilongjiang 150040, China.
| |
Collapse
|
6
|
Abstract
Abstract
The pathogenesis of connective tissue diseases (CTDs), represented by systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), primary Sjögren’s syndrome (pSS), and idiopathic inflammatory myopathies (IIM), includes various immune cells involved in both innate and adaptive immunity. The mesenchymal stem cells (MSCs) are unique due to their regulatory effect on immunity. This makes them a promising therapeutic approach for patients with immune-mediated disorders such as CTD. The safety and clinical efficacy of MSC treatment in CTD have been tested in a growing number of preclinical and clinical studies. Administration of MSCs has consistently shown benefits with both symptomatic and histologic improvement in CTD animal models. MSC therapies in severe and drug-resistant CTD patients have shown promise in a number of the pilot studies, cohort studies, and randomized controlled trials in SLE, RA, and SSc, but some problems still need to be resolved in the transition from the bench to the bedside. The relevant studies in pSS and IIM are still in their infancy, but have displayed encouraging outcomes. Considerable efficacy variations have been observed in terms of the route of delivery, time of MSC injection, origin of the MSCs and dosage. Furthermore, the optimization of conventional drugs combined with MSC therapies and the applications of novel cell engineering approaches requires additional research. In this review, we summarize the current evidence about the immunoregulatory mechanism of MSCs, as well as the preclinical and clinical studies of MSC-based therapy for the treatment of CTDs.
Collapse
|
7
|
Supramolecular Hydrogel-Wrapped Gingival Mesenchymal Stem Cells in Cutaneous Radiation Injury. Cells 2022; 11:cells11193089. [PMID: 36231051 PMCID: PMC9564043 DOI: 10.3390/cells11193089] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 12/12/2022] Open
Abstract
Radiation-induced skin wound/dermatitis is one of the common side effects of radiotherapy or interventional radiobiology. Gingiva-derived mesenchymal stem cells (GMSCs) were indicated to have therapeutic potentials in skin diseases. However, stem cells are prone to spread and difficult to stay in the skin for a long time, limiting their curative effects and application. This study investigated the therapeutic efficacy of Nap-GDFDFpDY (pY-Gel) self-assembled peptide hydrogel-encapsulated GMSCs to treat 137Cs γ-radiation-induced skin wounds in mice. The effects were evaluated by skin damage score, hind limb extension measurement and histological and immunohistochemical analysis. In vivo studies showed that pY-Gel self-assembled peptide hydrogel-encapsulated GMSCs could effectively improve wound healing in irradiated skin tissues. In addition, it was found that GMSCs conditioned medium (CM) could promote the proliferation, migration and DNA damage repair ability of skin cells after irradiation in human keratinocyte cell line HaCaT and normal human dermal fibroblasts (HFF). Mechanistically, GMSCs-CM can promote the expression of epidermal growth factor receptor (EGFR), signal transducers and activators of transcription 3 (STAT3) and matrix metalloproteinases (MMPs), suggesting that activation of the EGFR/STAT3 signaling pathway may be involved in the repair of skin cells after exposure to radiations. In conclusion, pY-Gel self-assembled peptide hydrogel-encapsulated GMSCs have a beneficial therapeutic effect on radiation-induced cutaneous injury and may serve as a basis of novel cells therapeutic approach.
Collapse
|
8
|
Assar S, Khazaei H, Naseri M, El-Senduny F, Momtaz S, Farzaei MH, Echeverría J. Natural Formulations: Novel Viewpoint for Scleroderma Adjunct Treatment. J Immunol Res 2021; 2021:9920416. [PMID: 34258301 PMCID: PMC8253639 DOI: 10.1155/2021/9920416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/27/2021] [Accepted: 06/08/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Scleroderma is a complex disease involving autoimmune, vascular, and connective tissues, with unknown etiology that can progress through any organ systems. OBJECTIVE Yet, no cure is available; the thorough treatment of scleroderma and current treatments are based on controlling inflammation. Nowadays, medicinal plants/natural-based formulations are emerging as important regulators of many diseases, including autoimmune diseases. Here, we provided an overview of scleroderma, also focused on recent studies on medicinal plants/natural-based formulations that are beneficial in scleroderma treatment/prevention. METHODS This study is the result of a search in PubMed, Scopus, and Cochrane Library with "scleroderma", "systemic sclerosis", "plant", "herb", and "phytochemical" keywords. Finally, 22 articles were selected from a total of 1513 results entered in this study. RESULTS Natural products can modulate the inflammatory and/or oxidative mediators, regulate the production or function of the immune cells, and control the collagen synthesis, thereby attenuating the experimental and clinical manifestation of the disease. CONCLUSION Natural compounds can be considered an adjunct treatment for scleroderma to improve the quality of life of patients suffering from this disease.
Collapse
Affiliation(s)
- Shirin Assar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hosna Khazaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Maryam Naseri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Fardous El-Senduny
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
- Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Javier Echeverría
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
9
|
Jin J, Ou Q, Wang Z, Tian H, Xu JY, Gao F, Hu S, Chen J, Wang J, Zhang J, Lu L, Jin C, Xu GT, Zhao J. BMSC-derived extracellular vesicles intervened the pathogenic changes of scleroderma in mice through miRNAs. Stem Cell Res Ther 2021; 12:327. [PMID: 34090522 PMCID: PMC8179710 DOI: 10.1186/s13287-021-02400-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is a disease that features severe fibrosis of the skin and lacks effective therapy. Bone marrow mesenchymal stem cell (BMSC)-derived extracellular vesicles (EVs) are potential stem cell-based tools for the treatment of SSc. METHODS BMSCs were isolated from the bone marrow of mice and identified with surface markers according to multilineage differentiation. EVs were isolated from the BMSC culture medium by ultracentrifugation and identified with a Nanosight NS300 particle size analyzer, transmission electron microscopy (TEM), and western blot. The microRNAs (miRNAs) of BMSC-derived EVs (BMSC-EVs) were studied via miRNA sequencing (miRNA-seq) and bioinformatic analysis. An SSc mouse model was established via subcutaneous bleomycin (BLM) injection, and the mice were treated with BMSCs or BMSC-derived EVs. Skin tissues were dissociated and analyzed with H&E staining, RNA sequencing (RNA-seq), western blot, and immunohistochemical staining. RESULTS Evident pathological changes, like fibrosis and inflammation, were induced in the skin of BLM-treated mice. BMSCs and BMSC-EVs effectively intervened such pathological manifestations and disease processes in a very similar way. The effects of the BMSC-EVs were found to be caused by the miRNAs they carried, which were proven to be involved in regulating the proliferation and differentiation of multiple cell types and in multiple EV-related biological processes. Furthermore, TGF-β1-positive cells and α-SMA-positive myofibroblasts were significantly increased in the scleroderma skin of BLM-treated mice but evidently reduced in the scleroderma skin of the EV-treated SSc group. In addition, the numbers of mast cells and infiltrating macrophages and lymphocytes were evidently increased in the skin of BLM-treated mice but significantly reduced by EV treatment. In line with these observations, there were significantly higher mRNA levels of the inflammatory cytokines Il6, Il10, and Tnf-α in SSc mice than in control mice, but the levels decreased following EV treatment. Through bioinformatics analysis, the TGFβ and WNT signaling pathways were revealed to be closely involved in the pathogenic changes seen in mouse SSc, and these pathways could be therapeutic targets for treating the disease. CONCLUSIONS BMSC-derived EVs could be developed as a potential therapy for treating skin dysfunction in SSc, especially considering that they show similar efficacy to BMSCs but have fewer developmental regulatory requirements than cell therapy. The effects of EVs are generated by the miRNAs they carry, which alleviate SSc pathogenic changes by regulating the WNT and TGFβ signaling pathways.
Collapse
Affiliation(s)
- Jiahui Jin
- Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065 China
| | - Qingjian Ou
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Zhe Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120 China
| | - Haibin Tian
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Jing-Ying Xu
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Furong Gao
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Shuqin Hu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120 China
| | - Jie Chen
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120 China
| | - Juan Wang
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Jieping Zhang
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Lixia Lu
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Caixia Jin
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Guo-Tong Xu
- Department of Ophthalmology of Shanghai Tenth People’s Hospital, and Laboratory of Clinical Visual Science of Tongji Eye Institute, School of Medicine, Tongji University, Shanghai, 200072 China
| | - Jingjun Zhao
- Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065 China
| |
Collapse
|
10
|
Baral H, Sekiguchi A, Uchiyama A, Nisaa Amalia S, Yamazaki S, Inoue Y, Yokoyama Y, Ogino S, Torii R, Hosoi M, Akai R, Iwawaki T, Ishikawa O, Motegi SI. Inhibition of skin fibrosis in systemic sclerosis by botulinum toxin B via the suppression of oxidative stress. J Dermatol 2021; 48:1052-1061. [PMID: 33840125 DOI: 10.1111/1346-8138.15888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022]
Abstract
Oxidative stress has been reported to play an important role in the pathogenesis of skin fibrosis in systemic sclerosis (SSc). We previously identified that botulinum toxin (BTX) injection suppresses pressure ulcer formation in a cutaneous ischemia-reperfusion injury mouse model by regulation of oxidative stress. However, the therapeutic possibility of BTX administration for preventing skin fibrosis in SSc is unclear. The objective of this study was to investigate the effect of BTX-B on skin fibrosis in a murine model of SSc and determine the underlying mechanism. We found that BTX-B injection significantly reduced dermal thickness and inflammatory cell infiltration in bleomycin-induced skin fibrosis lesion in mice. We also identified that the oxidative stress signal detected through bioluminescence in OKD48 mice after bleomycin injection in the skin was significantly decreased by BTX-B. Additionally, mRNA levels of oxidative stress associated factors (NOX2, HO-1, Trx2) were significantly decreased by BTX-B. Apoptotic cells in the lesional skin of bleomycin-treated mice were significantly reduced by BTX-B. Oxidant-induced intracellular accumulation of reactive oxygen species in SSc fibroblasts was also inhibited by BTX-B. In conclusion, BTX-B might improve bleomycin-induced skin fibrosis via the suppression of oxidative stress and inflammatory cells in the skin. BTX-B injection may have a therapeutic effect on skin fibrosis in SSc.
Collapse
Affiliation(s)
- Hritu Baral
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akiko Sekiguchi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihiko Uchiyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Syahla Nisaa Amalia
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sahori Yamazaki
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yuta Inoue
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoko Yokoyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sachiko Ogino
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Torii
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Mari Hosoi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Akai
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Osamu Ishikawa
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
11
|
Varrica C, Dias HS, Reis C, Carvalheiro M, Simões S. Targeted delivery in scleroderma fibrosis. Autoimmun Rev 2020; 20:102730. [PMID: 33338593 DOI: 10.1016/j.autrev.2020.102730] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/29/2022]
Abstract
Systemic sclerosis (SSc) is considered one of the most challenging and difficult to treat among rheumatic disorders, due to its severity, multiorgan manifestation and different outcomes. It manifests fibrosis in different organs, mostly in skin and lungs. The skin fibrosis expression is considered the first sign of the disease and usually it is followed by internal organ fibrosis. An aberrant immune system activation seems to relate to the expression of the disease, but even environmental influences and dysregulation of many molecules signalling pathways are involved in the development of the disease. Current therapies are limited and characterized by multiple side effects: systemic route is the elective administration route, which decreases patient adherence to the therapy, as they are often already bothered by pain and disfigurement. Treatments available are organ-based, originally indicated for other conditions and there is no therapy available to reduce the fibroblast population size within existing fibrotic lesions. Disease-modifying therapies or immunomodulatory agents that are highly effective in other rheumatic diseases have shown disappointing results in SSc. There are thus no standardized and effective treatments for this disease, and there are even unanswered questions related to the insurgence of the pathology and all the mechanisms involved. An ideal approach could be considered "targeted therapy" that will be an increasingly attainable objective insofar as our understanding of the disease improves. The advantages in identifying the molecule and the signalling pathways involved in the pathology have helped to find some novel compounds for the therapy of scleroderma fibrosis or following innovative uses for already-approved drugs, corroborated by many clinical studies.
Collapse
Affiliation(s)
- Carla Varrica
- University of Pavia, Corso Strada Nuova, 65, 27100 Pavia, Italy
| | - Helena Sofia Dias
- Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Catarina Reis
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal; IBEB, Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1649-016 Lisboa, Portugal
| | - Manuela Carvalheiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal
| | - Sandra Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
12
|
Abedi M, Alavi-Moghadam S, Payab M, Goodarzi P, Mohamadi-jahani F, Sayahpour FA, Larijani B, Arjmand B. Mesenchymal stem cell as a novel approach to systemic sclerosis; current status and future perspectives. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:20. [PMID: 33258056 PMCID: PMC7704834 DOI: 10.1186/s13619-020-00058-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
Systemic sclerosis is a rare chronic autoimmune disease with extensive microvascular injury, damage of endothelial cells, activation of immune responses, and progression of tissue fibrosis in the skin and various internal organs. According to epidemiological data, women's populations are more susceptible to systemic sclerosis than men. Until now, various therapeutic options are employed to manage the symptoms of the disease. Since stem cell-based treatments have developed as a novel approach to rescue from several autoimmune diseases, it seems that stem cells, especially mesenchymal stem cells as a powerful regenerative tool can also be advantageous for systemic sclerosis treatment via their remarkable properties including immunomodulatory and anti-fibrotic effects. Accordingly, we discuss the contemporary status and future perspectives of mesenchymal stem cell transplantation for systemic sclerosis.
Collapse
Affiliation(s)
- Mina Abedi
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Goodarzi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Mohamadi-jahani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Wang C, Zhou J, Wang J, Li S, Fukunaga A, Yodoi J, Tian H. Progress in the mechanism and targeted drug therapy for COPD. Signal Transduct Target Ther 2020; 5:248. [PMID: 33110061 PMCID: PMC7588592 DOI: 10.1038/s41392-020-00345-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is emphysema and/or chronic bronchitis characterised by long-term breathing problems and poor airflow. The prevalence of COPD has increased over the last decade and the drugs most commonly used to treat it, such as glucocorticoids and bronchodilators, have significant therapeutic effects; however, they also cause side effects, including infection and immunosuppression. Here we reviewed the pathogenesis and progression of COPD and elaborated on the effects and mechanisms of newly developed molecular targeted COPD therapeutic drugs. Among these new drugs, we focussed on thioredoxin (Trx). Trx effectively prevents the progression of COPD by regulating redox status and protease/anti-protease balance, blocking the NF-κB and MAPK signalling pathways, suppressing the activation and migration of inflammatory cells and the production of cytokines, inhibiting the synthesis and the activation of adhesion factors and growth factors, and controlling the cAMP-PKA and PI3K/Akt signalling pathways. The mechanism by which Trx affects COPD is different from glucocorticoid-based mechanisms which regulate the inflammatory reaction in association with suppressing immune responses. In addition, Trx also improves the insensitivity of COPD to steroids by inhibiting the production and internalisation of macrophage migration inhibitory factor (MIF). Taken together, these findings suggest that Trx may be the ideal drug for treating COPD.
Collapse
Affiliation(s)
- Cuixue Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Jiedong Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Jinquan Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Shujing Li
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Atsushi Fukunaga
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Junji Yodoi
- Laboratory of Infection and Prevention, Department of Biological Response, Institute for Virus Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Hai Tian
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China.
- Jiaozhimei Biotechnology (Shaoxing) Co, Ltd, Shaoxing, 312000, China.
| |
Collapse
|
14
|
Abstract
Autoimmune diseases, such as rheumatoid arthritis, systematic lupus erythematosus and Sjögren's syndrome, are a group of diseases characterized by the activation of immune cells and excessive production of autoantibodies. Although the pathogenesis of these diseases is still not completely understood, studies have shown that multiple factors including genetics, environment and immune responses play important roles in the development and progression of the diseases. In China, there are great achievements in the mechanisms of autoimmune diseases during the last decades. These studies provide new insight to understand the diseases and also shed light on the development of novel therapy.
Collapse
Affiliation(s)
- Ru Li
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China.
| | - Xing Sun
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xu Liu
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yue Yang
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Zhanguo Li
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
15
|
Sekiguchi A, Motegi SI, Fujiwara C, Yamazaki S, Inoue Y, Uchiyama A, Akai R, Iwawaki T, Ishikawa O. Inhibitory effect of kaempferol on skin fibrosis in systemic sclerosis by the suppression of oxidative stress. J Dermatol Sci 2019; 96:8-17. [DOI: 10.1016/j.jdermsci.2019.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/30/2019] [Accepted: 08/14/2019] [Indexed: 02/06/2023]
|
16
|
Zhang F, Peng W, Zhang J, Dong W, Yuan D, Zheng Y, Wang Z. New strategy of bone marrow mesenchymal stem cells against oxidative stress injury via Nrf2 pathway: oxidative stress preconditioning. J Cell Biochem 2019; 120:19902-19914. [PMID: 31347718 PMCID: PMC6852471 DOI: 10.1002/jcb.29298] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022]
Abstract
Clinically, bone marrow mesenchymal stem cells (BMSCs) have been used in treatment of many diseases, but the local oxidative stress (OS) of lesion severely limits the survival of BMSCs, which reduces the efficacy of BMSCs transplantation. Therefore, enhancing the anti‐OS stress ability of BMSCs is a key breakthrough point. Preconditioning is a common protective mechanism for cells or body. Here, the aim of this study was to investigate the effects of OS preconditioning on the anti‐OS ability of BMSCs and its mechanism. Fortunately, OS preconditioning can increase the expression of superoxide dismutase, catalase, NQO1, and heme oxygenase 1 through the nuclear factor erythroid 2‐related factor 2 pathway, thereby decreased the intracellular reactive oxygen species (ROS) levels, relieved the damage of ROS to mitochondria, DNA and cell membrane, enhanced the anti‐OS ability of BMSCs, and promoted the survival of BMSCs under OS.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Trauma orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,Trauma Teaching and Research Department, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wuxun Peng
- Department of Trauma orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,Trauma Teaching and Research Department, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jian Zhang
- Department of Trauma orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,Trauma Teaching and Research Department, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wentao Dong
- Department of Trauma orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,Trauma Teaching and Research Department, Guizhou Medical University, Guiyang, Guizhou, China
| | - Dajiang Yuan
- Trauma Teaching and Research Department, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yinggang Zheng
- Trauma Teaching and Research Department, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhenwen Wang
- Trauma Teaching and Research Department, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
17
|
Rozier P, Maria A, Goulabchand R, Jorgensen C, Guilpain P, Noël D. Mesenchymal Stem Cells in Systemic Sclerosis: Allogenic or Autologous Approaches for Therapeutic Use? Front Immunol 2018; 9:2938. [PMID: 30619298 PMCID: PMC6302042 DOI: 10.3389/fimmu.2018.02938] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/29/2018] [Indexed: 12/18/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare autoimmune disease, which is potentially lethal. The physiopathology of the disease is still incompletely elucidated although the role of fibroblasts, endothelial cells (ECs), immune cells. and the environment (i.e., oxidative stress) has been demonstrated. This is an intractable disease with an urgent need to provide better therapeutic options to patients. Mesenchymal stem cells (MSCs) represent a promising therapeutic approach thanks to the number of trophic and pleiotropic properties they exert. Among these, MSCs display anti-fibrotic, angiogenic, and immunomodulatory capacities that might be of interest in the treatment of SSc by acting on different processes that are dysregulated in the disease. In the recent years, the therapeutic effectiveness of MSCs has been demonstrated in different preclinical animal models and is being investigated in phase I clinical trials. Both allogenic and autologous transplantation of MSCs isolated from bone marrow or adipose tissue is being evaluated. The rationale for using allogenic MSCs in SSc, as well as in other autoimmune diseases, is based on the possibility that autologous MSCs might be altered in these diseases. In SSc, reports from the literature are controversial. Nevertheless, the role of the oxidative environment and of the crosstalk with neighboring cells (fibroblasts and ECs) on the functional properties of MSCs has been reported. Here, we review the preclinical and clinical data reporting the interest of MSC-based treatment in SSc and question the use of autologous or allogeneic MSCs in perspective of clinical applications.
Collapse
Affiliation(s)
- Pauline Rozier
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Alexandre Maria
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Radjiv Goulabchand
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Lapeyronie University Hospital, Montpellier, France
| | - Philippe Guilpain
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Department of Internal Medicine, Multiorganic Diseases, Saint-Eloi Hospital, Montpellier, France
| | - Danièle Noël
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Lapeyronie University Hospital, Montpellier, France
| |
Collapse
|
18
|
Peltzer J, Aletti M, Frescaline N, Busson E, Lataillade JJ, Martinaud C. Mesenchymal Stromal Cells Based Therapy in Systemic Sclerosis: Rational and Challenges. Front Immunol 2018; 9:2013. [PMID: 30271402 PMCID: PMC6146027 DOI: 10.3389/fimmu.2018.02013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
Systemic Sclerosis (SSc) is a rare chronic disease, related to autoimmune connective tissue diseases such as Systemic Lupus Erythematosus and Sjögren's Syndrome. Although its clinical heterogeneity, main features of the disease are: extensive tissue fibrosis with increase matrix deposition in skin and internal organ, microvascular alterations and activation of the immune system with autoantibodies against various cellular antigens. In the diffuse cutaneous scleroderma subtype, the disease is rapidly progressive with a poor prognosis, leading to failure of almost any internal organ, especially lung which is the leading cause of death. Primary trigger is unknown but may involve an immune process against mesenchymal cells in a genetically receptive host. Pathophysiology reveals a pivotal role of fibrosis and inflammation alterations implicating different cell subtypes, cytokines and growth factors, autoantibodies and reactive oxygen species. Despite improvement, the overall survival of SSc patients is still lower than that of other inflammatory diseases. Recommended drugs are agents capable of modulating fibrotic and inflammatory pathways. Cellular therapy has recently emerged as a credible option. Besides autologous hematopoietic stem cell transplantation which demonstrated remarkable improvement, mesenchymal stromal cells (MSCs) represent promising therapeutic candidates. Indeed, these cells possess anti-inflammatory, antiproliferative, antifibrotic, and immunomodulary properties especially by secreting a large panel of bioactive molecules, addressing the most important key points of the SSc. In addition, these cells are very sensitive to their environment and are able to modulate their activity according to the pathophysiological context in which they are located. Autologous or allogeneic MSCs from various sources have been tested in many trials in different auto-immune diseases such as multiple sclerosis, Crohn's disease or systemic lupus erythematosus. They are characterized by a broad availability and no or low acute toxicity. However, few randomized prospective clinical trials were published and their production under ATMP regulatory procedures is complex and time-consuming. Many aspects have still to be addressed to ascertain their potential as well as the potential of their derived products in the management of SSc, probably in association with other therapies.
Collapse
Affiliation(s)
- Juliette Peltzer
- Unité de Thérapie tissulaire et traumatologie de guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Marc Aletti
- Service de Médecine Interne, Hôpital d'Instruction des Armées Percy, Clamart, France
| | - Nadira Frescaline
- UMR7648 Laboratoire de physique des plasmas, École Polytechnique, Palaiseau, France
| | - Elodie Busson
- Unité de Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Jean-Jacques Lataillade
- Unité de Thérapie tissulaire et traumatologie de guerre, Institut de Recherche Biomédicale des Armées, Clamart, France
| | - Christophe Martinaud
- Unité de Médicaments de Thérapie Innovante, Centre de Transfusion Sanguine des Armées, Clamart, France
| |
Collapse
|
19
|
Vona R, Giovannetti A, Gambardella L, Malorni W, Pietraforte D, Straface E. Oxidative stress in the pathogenesis of systemic scleroderma: An overview. J Cell Mol Med 2018; 22:3308-3314. [PMID: 29664231 PMCID: PMC6010858 DOI: 10.1111/jcmm.13630] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 03/11/2018] [Indexed: 01/23/2023] Open
Abstract
Systemic sclerosis (SSc) is a rare disorder of the connective tissue characterized by fibrosis of the skin, skeletal muscles and visceral organs. Additional manifestations include activation of the immune system and vascular injury. SSc causes disability and death as the result of end‐stage organ failure. Two clinical subsets of the SSc are accepted: limited cutaneous SSc (lc‐SSc) and diffuse cutaneous SSc (dc‐SSc). At present, the aetiology and pathogenesis of SSc remain obscure, and consequently, disease outcome is unpredictable. Numerous studies suggest that reactive oxidizing species (ROS) play an important role in the pathogenesis of scleroderma. Over the years, several reports have supported this hypothesis for both lc‐SSc and dc‐SSc, although the specific role of oxidative stress in the pathogenesis of vascular injury and fibrosis remains to be clarified. The aim of the present review was to report and comment the recent findings regarding the involvement and role of oxidative stress in SSc pathogenesis. Biomarkers proving the link between ROS and the main pathological features of SSc have been summarized.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, Biomarkers Unit, Rome, Italy
| | | | | | - Walter Malorni
- Center for Gender-Specific Medicine, Biomarkers Unit, Rome, Italy
| | - Donatella Pietraforte
- Center for Gender-Specific Medicine, Biomarkers Unit, Rome, Italy.,Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
20
|
Marangoni RG, Lu TT. The roles of dermal white adipose tissue loss in scleroderma skin fibrosis. Curr Opin Rheumatol 2017; 29:585-590. [PMID: 28800024 DOI: 10.1097/bor.0000000000000437] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Dermal white adipose tissue (DWAT) is distinct from subcutaneous white adipose tissue and is lost in scleroderma skin fibrosis. The roles of DWAT loss in scleroderma skin fibrosis have not been well understood, and here we discuss recent findings that begin to provide insight into the multiple mechanisms involved. RECENT FINDINGS The DWAT loss in part reflects the direct contribution of DWAT cells to the fibrotic tissue, with the reprogramming of adipocytes to myofibroblasts. The DWAT contains reparative adipose-derived stromal cells and expresses antifibrotic cytokines such as adiponectin, and the loss of these skin-protective mechanisms with DWAT loss further contributes to skin fibrosis and injury. SUMMARY Potentially, halting or reversing the transdifferentiation of adipocytes to myofibroblasts along with improving survival of reparative adipose-derived stromal cells (ADSCs) and expression of antifibrotic cytokines may be effective therapeutic avenues.
Collapse
Affiliation(s)
- Roberta G Marangoni
- aDivision of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois bAutoimmunity and Inflammation Program and Pediatric Rheumatology, Hospital for Special Surgery cMicrobiology and Immunology Department, Weill Cornell Medical School, New York, New York, USA
| | | |
Collapse
|