Your browser doesn't support javascript.
loading
miRNA-148a-containing GMSC-derived EVs modulate Treg/Th17 balance via IKKB/NF-κB pathway and treat a rheumatoid arthritis model.
Chen, Jingrong; Shi, Xiaoyi; Deng, Yanan; Dang, Junlong; Liu, Yan; Zhao, Jun; Liang, Rongzhen; Zeng, Donglan; Wu, Wenbin; Xiong, Yiding; Yuan, Jia; Chen, Ye; Wang, Julie; Lin, Weidong; Chen, Xiangfang; Huang, Weishan; Olsen, Nancy; Pan, Yunfeng; Fu, Qingling; Zheng, Song Guo.
Affiliation
  • Chen J; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Shi X; Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
  • Deng Y; Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
  • Dang J; Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, China.
  • Liu Y; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Zhao J; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Liang R; Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
  • Zeng D; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Wu W; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Xiong Y; Department of Clinical Immunology.
  • Yuan J; Department of Spine Surgery, and.
  • Chen Y; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Wang J; Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
  • Lin W; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Chen X; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Huang W; Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
  • Olsen N; Department of Endocrinology, Second Affiliated Hospital of Naval Medical University, Shanghai, China.
  • Pan Y; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA.
  • Fu Q; Division of Rheumatology, Department of Medicine, The Penn State University Hershey Medical Center, Hershey, Pennsylvania, USA.
  • Zheng SG; Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
JCI Insight ; 9(10)2024 Apr 23.
Article in En | MEDLINE | ID: mdl-38652539
ABSTRACT
Mesenchymal stem cells (MSCs) have demonstrated potent immunomodulatory properties that have shown promise in the treatment of autoimmune diseases, including rheumatoid arthritis (RA). However, the inherent heterogeneity of MSCs triggered conflicting therapeutic outcomes, raising safety concerns and limiting their clinical application. This study aimed to investigate the potential of extracellular vesicles derived from human gingival mesenchymal stem cells (GMSC-EVs) as a therapeutic strategy for RA. Through in vivo experiments using an experimental RA model, our results demonstrate that GMSC-EVs selectively homed to inflamed joints and recovered Treg and Th17 cell balance, resulting in the reduction of arthritis progression. Our investigations also uncovered miR-148a-3p as a critical contributor to the Treg/Th17 balance modulation via IKKB/NF-κB signaling orchestrated by GMSC-EVs, which was subsequently validated in a model of human xenograft versus host disease (xGvHD). Furthermore, we successfully developed a humanized animal model by utilizing synovial fibroblasts obtained from patients with RA (RASFs). We found that GMSC-EVs impeded the invasiveness of RASFs and minimized cartilage destruction, indicating their potential therapeutic efficacy in the context of patients with RA. Overall, the unique characteristics - including reduced immunogenicity, simplified administration, and inherent ability to target inflamed tissues - position GMSC-EVs as a viable alternative for RA and other autoimmune diseases.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Arthritis, Rheumatoid / NF-kappa B / T-Lymphocytes, Regulatory / MicroRNAs / Th17 Cells / Mesenchymal Stem Cells / Extracellular Vesicles Limits: Animals / Humans / Male Language: En Journal: JCI Insight Year: 2024 Document type: Article Affiliation country: China

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Arthritis, Rheumatoid / NF-kappa B / T-Lymphocytes, Regulatory / MicroRNAs / Th17 Cells / Mesenchymal Stem Cells / Extracellular Vesicles Limits: Animals / Humans / Male Language: En Journal: JCI Insight Year: 2024 Document type: Article Affiliation country: China