51
|
Jariyasakulroj S, Shu Y, Lin Z, Chen J, Chang Q, Ko PF, Chen JF. Mapping cell diversity and dynamics in inflammatory temporomandibular joint osteoarthritis with pain at single-cell resolution. JCI Insight 2025; 10:e184379. [PMID: 39927459 PMCID: PMC11948589 DOI: 10.1172/jci.insight.184379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/13/2024] [Indexed: 02/11/2025] Open
Abstract
Temporomandibular joint (TMJ) osteoarthritis with pain is a highly prevalent disorder affecting patients' quality of life. A comprehensive understanding of cell type diversity and its dynamics in painful TMJ osteoarthritis (TMJOA) is lacking. Here, we utilized an inflammatory TMJOA mouse model via intra-articular injection of CFA. TMJOA mice exhibited cartilage remodeling, bone loss, synovitis, increased osteoarthritis (OA) score, and orofacial pain, recapitulating hallmark symptoms in patients. Single-cell transcriptomic profiling of the TMJ was performed in conjunction with mouse genetic labeling, tissue clearing, light sheet and confocal 3D imaging, multiplex RNAscope, and immunodetection. We visualized, reconstructed, and analyzed the distribution and density of nociceptive innervation of TMJ at single-axon levels. We systematically mapped the heterogeneity and anatomical position of blood endothelial cells, synovial fibroblasts, and immune cells, including Cx3cr1-positive barrier macrophages. Importantly, TMJOA mice exhibited enhanced neurovascular coupling, sublining fibroblast hyperplasia, inflammatory immune cell expansion, disrupted signaling-dependent cell-cell interaction, and a breakdown of the sandwich-like organization consisting of synovial barrier macrophages and fibroblasts. By utilizing a mouse model with combined TMJ pain history and OA, we reveal the cellular diversity, anatomical structure, and cell dynamics of the TMJ at single-cell resolution, which facilitate our understanding and potential targeting of TMJOA.
Collapse
Affiliation(s)
- Supawadee Jariyasakulroj
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
- Department of Masticatory Science, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Yang Shu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Ziying Lin
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Jingyi Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Qing Chang
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Pao-Fen Ko
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
52
|
Lou J, Zhang B, Cai J, Zhang L, Zhao Y, Zhao Z. Diabetes exacerbates periodontitis by disrupting IL-33-mediated interaction between periodontal ligament fibroblasts and macrophages. Int Immunopharmacol 2025; 147:113896. [PMID: 39740505 DOI: 10.1016/j.intimp.2024.113896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Tissue-resident fibroblasts with immunomodulatory properties have recently been identified as key players in inflammation. However, their roles within the periodontal niche in diabetes-associated periodontitis remain unclear. Interleukin (IL)-33, known as an "alarmin" in inflammatory responses, has recently emerged as a potential contributor to periodontitis. Herein, we show that IL-33 levels are reduced in periodontal ligament fibroblasts (PDLFs) in the in vivo models of diabetes-associated periodontitis and in vitro models of diabetic inflammation. In the in vitro co-culture model, overexpression of IL-33 in PDLFs promotes M2 macrophage polarization, while knockdown of IL-33 in PDLFs instigates M1 macrophage polarization. Notably, supplementation with IL-33 in vivosignificantly alleviates periodontal tissue destruction and enhances M2 macrophage infiltration, whereas targeting the IL-33/ST2 axis exacerbates tissue damage and promotes M1 macrophage polarization in diabetes-associated periodontitis. Additionally, theCUT&RUN assay confirms the direct regulation of IL-33 by Yes-associated protein (YAP). These findings demonstrate that IL-33 deficiency in PDLFs favors M1 macrophage polarization, thereby exacerbating the pathogenesis of diabetes-associated periodontitis. Our study underscores the essential immunomodulatory role of PDLFs in creating an inflammatory niche and unveils a novel interaction axis between PDLFs and macrophages in diabetes-associated periodontitis.
Collapse
Affiliation(s)
- Jingyang Lou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jingyi Cai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Linli Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yifan Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
53
|
Elghali M, Yosr B, Syrine D, Mahbouba J, Nabil S, Sonia H. Isolated anti-ribosomal P antibodies are associated with reduced risk of renal and articular involvement in systemic lupus erythematosus patients. An observational study from one center. Reumatologia 2025; 63:27-34. [PMID: 40206226 PMCID: PMC11977507 DOI: 10.5114/reum/197390] [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: 10/12/2024] [Accepted: 12/15/2024] [Indexed: 04/11/2025] Open
Abstract
Introduction The aim of the study was to compare the specific clinical manifestations of systemic lupus erythematosus (SLE) or laboratory findings between patients with and without anti-ribosomal P (anti-P) antibodies and to investigate possible associations between isolated anti-P antibodies and these features. Material and methods Seventy-five SLE patients were enrolled in this study. They were recruited from the Department of Internal Medicine and Department of Rheumatology at the University Hospital of Monastir, Tunisia (January 2008 - December 2022). All patients met at least four American College of Rheumatology criteria or Systemic Lupus Erythematosus International Collaborating Clinics criteria at the time of disease diagnosis. Antibody typing was performed using a commercial line blot technique. Statistical analysis was performed using the χ2 test, Fisher's test when appropriate, Student's t-test, or Mann-Whitney U test according to normality of the data distribution. Results Thirty patients (40%) were positive for anti-P (anti-P+). The anti-P+ had higher frequency of skin features (26/49 [53.1%] vs. 4/26 [15.4%], p = 0.003) and central nervous system (CNS) involvement (10/15 [66.7%] vs. 20/60 [33.3%], p = 0.018) than patients without anti-P. Interestingly, anti-P+ showed a lower frequency of SLE/rheumatoid arthritis overlap syndrome (1/11 [9.1%] vs. 29/64 [45.3%], p = 0.042). The comparison between groups of patients according to the presence of anti-P, anti-dsDNA, and anti-Sm showed that the group with anti-P lacking anti-dsDNA and anti-Sm had the highest frequency of neuropsychiatric SLE (75%, p = 0.034), and the lowest frequency of lupus nephritis (0%, p = 0.029) and arthritis (12.5%, p = 0.039). Conclusions This study supports the association of anti-P antibodies with CNS and cutaneous manifestations. To the best of our knowledge, this is the first study to report a negative association between isolated anti-P antibodies and renal and articular involvement in SLE.
Collapse
Affiliation(s)
- Mourad Elghali
- Laboratory of Immunology, University Hospital F.B., University of Monastir, Tunisia
| | - Boussoukaya Yosr
- Department of Internal Medicine, University Hospital F.B., University of Monastir, Tunisia
| | - Daadaa Syrine
- Department of Internal Medicine, University Hospital F.B., University of Monastir, Tunisia
| | - Jguirim Mahbouba
- Department of Rheumatology, University Hospital F.B., University of Monastir, Tunisia
| | - Sakly Nabil
- Laboratory of Immunology, University Hospital F.B., University of Monastir, Tunisia
- Laboratory of Medical and Molecular Parasitology-Mycology LR12ES08, Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Hammami Sonia
- Department of Internal Medicine, University Hospital F.B., University of Monastir, Tunisia
- Research Laboratory LR12ES05, Lab-NAFS – Nutrition-Functional Food and Health, Faculty of Medicine, University of Monastir, Tunisia
| |
Collapse
|
54
|
Zheng L, Gu M, Li X, Hu X, Chen C, Kang Y, Pan B, Chen W, Xian G, Wu X, Li C, Wang C, Li Z, Guan M, Zhou G, Mobasheri A, Song W, Peng S, Sheng P, Zhang Z. ITGA5 + synovial fibroblasts orchestrate proinflammatory niche formation by remodelling the local immune microenvironment in rheumatoid arthritis. Ann Rheum Dis 2025; 84:232-252. [PMID: 39919897 DOI: 10.1136/ard-2024-225778] [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: 03/08/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVES To investigate the phenotypic heterogeneity of tissue-resident synovial fibroblasts and their role in inflammatory response in rheumatoid arthritis (RA). METHODS We used single-cell and spatial transcriptomics to profile synovial cells and spatial gene expressions of synovial tissues to identify phenotypic changes in patients with osteoarthritis, RA in sustained remission and active state. Immunohistology, multiplex immunofluorescence and flow cytometry were used to identify synovial fibroblasts subsets. Deconvolution methods further validated our findings in two cohorts (PEAC and R4RA) with treatment response. Cell coculture was used to access the potential cell-cell interactions. Adoptive transfer of synovial cells in collagen-induced arthritis (CIA) mice and bulk RNA sequencing of synovial joints further validate the cellular functions. RESULTS We identified a novel tissue-remodelling CD45-CD31-PDPN+ITGA5+ synovial fibroblast population with unique transcriptome of POSTN, COL3A1, CCL5 and TGFB1, and enriched in immunoregulatory pathways. This subset was upregulated in active and lympho-myeloid type of RA, associated with an increased risk of multidrug resistance. Transforming growth factor (TGF)-β1 might participate in the differentiation of this subset. Moreover, ITGA5+ synovial fibroblasts might occur in early stage of inflammation and induce the differentiation of CXCL13hiPD-1hi peripheral helper T cells (TPHs) from naïve CD4+ T cells, by secreting TGF-β1. Intra-articular injection of ITGA5+ synovial fibroblasts exacerbates RA development and upregulates TPHs in CIA mice. CONCLUSIONS We demonstrate that ITGA5+ synovial fibroblasts might regulate the RA progression by inducing the differentiation of CXCL13hiPD-1hi TPHs and remodelling the proinflammatory microenvironments. Therapeutic modulation of this subpopulation could therefore be a potential treatment strategy for RA.
Collapse
Affiliation(s)
- Linli Zheng
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Minghui Gu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xuantao Hu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Spine Surgery, Sun Yat-sen University Third Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chen Chen
- Trauma Orthopedics, Foot and Ankle Surgery, Sun Yat-sen Memorial Hostpial, Guangzhou, Guangdong, China; Institute of Precision Medicine, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Yunze Kang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Baiqi Pan
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Weishen Chen
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | | | - Xiaoyu Wu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chengxin Li
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chao Wang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Zhiwen Li
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Mingqiang Guan
- Department of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Guanming Zhou
- Department of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Public Health Aspects of Musculoskeletal Health and Aging, World Health Organization Collaborating Centre, Liege, Belgium
| | - Weidong Song
- Trauma Orthopedics, Foot and Ankle Surgery, Sun Yat-sen Memorial Hostpial, Guangzhou, Guangdong, China
| | - Sui Peng
- Institute of Precision Medicine, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Clinical Trials Unit, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Gastroenterology and Hepatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| | - Puyi Sheng
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| | - Ziji Zhang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| |
Collapse
|
55
|
Kubo S, Miyazaki Y, Nishino T, Fujita Y, Kono M, Kawashima T, Ishigaki K, Kusaka K, Tanaka H, Ueno M, Satoh-Kanda Y, Inoue Y, Todoroki Y, Miyagawa I, Hanami K, Nakayamada S, Tanaka Y. Peripheral blood immunophenotypic diversity in patients with rheumatoid arthritis and its impact on therapeutic responsiveness. Ann Rheum Dis 2025; 84:210-220. [PMID: 39919895 DOI: 10.1136/ard-2024-226228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/20/2024] [Indexed: 10/06/2024]
Abstract
OBJECTIVE Considering the diverse aetiologies and immunodysregulatory statuses observed in each patient with rheumatoid arthritis (RA), stratification based on peripheral blood immunophenotyping holds the potential to enhance therapeutic responses to molecular targeted therapies, biological/targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs). METHODS Immunophenotype analysis was conducted on a cohort of over 500 b/tsDMARDs-naïve patients using flow cytometry. Patients with RA were stratified based on their immunophenotypes, and the treatment response to each targeted therapy was evaluated. Validation was performed using an additional cohort of 183 b/tsDMARDs-naïve patients with RA. RESULTS Patients with RA were stratified into five clusters, two of which exhibited distinct RA phenotypes compared with controls, characterised by significant increases in CD4+ effector memory T cells re-expressing CD45RA. Notably, the effectiveness of different b/tsDMARDs varied across clusters. The group using promising b/tsDMARDs was labelled as 'expected' whereas the 'non-expected' group comprised those using others. The expected group outperformed the non-expected group with higher 26-week remission rates (39.9% vs 24.6%, p=0.0004) and low disease activity achievement (80.8% vs 60.2%, p<0.0001). Trajectory analysis showed the non-expected group's 26-week disease activity was influenced by Clinical Disease Activity Index at baseline unlike the expected group. Additionally, different molecular targeted therapies influenced the proportions of each immune cell subset variably. To validate, immunophenotyping was performed on a validation cohort. When 183 cases were grouped based on their b/tsDMARDs usage into expected/non-expected groups, the expected group had a higher remission rate (p=0.0021), further confirming the observed trend. CONCLUSION Our findings offer valuable insights into the diversity of RA and potential therapeutic strategies grounded in the molecular underpinnings.
Collapse
Affiliation(s)
- Satoshi Kubo
- Department of Molecular Targeted Therapies, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yusuke Miyazaki
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Takahiro Nishino
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Department of Microbiology and Immunology, Keio University, Tokyo, Japan
| | - Yuya Fujita
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Michihiro Kono
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tsugumi Kawashima
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Department of Microbiology and Immunology, Keio University, Tokyo, Japan
| | - Katsuhide Kusaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Hiroaki Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Masanobu Ueno
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yurie Satoh-Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshino Inoue
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yasuyuki Todoroki
- Department of Molecular Targeted Therapies, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Ippei Miyagawa
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Kentaro Hanami
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan.
| |
Collapse
|
56
|
Yuan W, Liu J, Zhang Z, Ye C, Zhou X, Yi Y, Wu Y, Li Y, Zhang Q, Xiong X, Xiao H, Liu J, Wang J. Strontium-Alix interaction enhances exosomal miRNA selectively loading in synovial MSCs for temporomandibular joint osteoarthritis treatment. Int J Oral Sci 2025; 17:6. [PMID: 39890774 PMCID: PMC11785994 DOI: 10.1038/s41368-024-00329-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 02/03/2025] Open
Abstract
The ambiguity of etiology makes temporomandibular joint osteoarthritis (TMJOA) "difficult-to-treat". Emerging evidence underscores the therapeutic promise of exosomes in osteoarthritis management. Nonetheless, challenges such as low yields and insignificant efficacy of current exosome therapies necessitate significant advances. Addressing lower strontium (Sr) levels in arthritic synovial microenvironment, we studied the effect of Sr element on exosomes and miRNA selectively loading in synovial mesenchymal stem cells (SMSCs). Here, we developed an optimized system that boosts the yield of SMSC-derived exosomes (SMSC-EXOs) and improves their miRNA profiles with an elevated proportion of beneficial miRNAs, while reducing harmful ones by pretreating SMSCs with Sr. Compared to untreated SMSC-EXOs, Sr-pretreated SMSC-derived exosomes (Sr-SMSC-EXOs) demonstrated superior therapeutic efficacy by mitigating chondrocyte ferroptosis and reducing osteoclast-mediated joint pain in TMJOA. Our results illustrate Alix's crucial role in Sr-triggered miRNA loading, identifying miR-143-3p as a key anti-TMJOA exosomal component. Interestingly, this system is specifically oriented towards synovium-derived stem cells. The insight into trace element-driven, site-specific miRNA selectively loading in SMSC-EXOs proposes a promising therapeutic enhancement strategy for TMJOA.
Collapse
Affiliation(s)
- Wenxiu Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jiaqi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chengxinyue Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xueman Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yating Yi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yange Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yijun Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qinlanhui Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Liu
- Laboratory of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| | - Jun Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
57
|
Yin C, Fu L, Guo S, Liang Y, Shu T, Shao W, Xia H, Xia T, Wang M. Senescent Fibroblasts Drive FAP/OLN Imbalance Through mTOR Signaling to Exacerbate Inflammation and Bone Resorption in Periodontitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409398. [PMID: 39716898 PMCID: PMC11831441 DOI: 10.1002/advs.202409398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/02/2024] [Indexed: 12/25/2024]
Abstract
Fibroblast activation protein (FAP), predominantly expressed in activated fibroblasts, plays a key role in inflammatory bone diseases, but its role in periodontitis remains unclear. Accordingly, this study identified a positive association between FAP levels and periodontitis susceptibility using Mendelian randomization analysis. Human and mouse periodontitis tissues show elevated FAP and reduced osteolectin (OLN), an endogenous FAP inhibitor, indicating a FAP/OLN imbalance. Single-cell RNA sequencing revealed gingival fibroblasts (GFs) as the primary FAP and OLN source, with periodontitis-associated GFs showing increased reactive oxygen species, cellular senescence, and mTOR pathway activation. Rapamycin treatment restored the FAP/OLN balance in GFs. Recombinant FAP increased pro-inflammatory cytokine secretion and osteoclast differentiation in macrophages, exacerbating periodontal damage, whereas FAP inhibition reduced macrophage inflammation, collagen degradation, and bone resorption in experimental periodontitis. Therefore, senescent fibroblasts drive the FAP/OLN imbalance through mTOR activation, contributing to periodontitis progression. Consequently, targeting FAP may offer a promising therapeutic strategy for periodontitis.
Collapse
Affiliation(s)
- Chenghu Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Liangliang Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Shuling Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Youde Liang
- Department of Stomatology CenterThe People's Hospital of Baoan Shenzhen (The Second Affiliated Hospital of Shenzhen University)ShenzhenGuangdong518081P. R. China
| | - Taizhi Shu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Wenjun Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Haibin Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Ting Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Min Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationKey Laboratory of Oral Biomedicine Ministry of EducationHubei Key Laboratory of StomatologySchool & Hospital of StomatologyWuhan UniversityWuhan430079China
| |
Collapse
|
58
|
Cyndari KI, Scorza BM, Zacharias ZR, Pessôa-Pereira D, Strand L, Mahachi K, Oviedo JM, Gibbs L, Butler KL, Ausdal G, Hendricks D, Yahashiri R, Elkins JM, Gulbrandsen T, Peterson AR, Willey MC, Fairfax KC, Petersen CA. Resident synovial macrophages in synovial fluid: Implications for immunoregulation. Clin Immunol 2025; 271:110422. [PMID: 39701169 DOI: 10.1016/j.clim.2024.110422] [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: 09/20/2024] [Revised: 11/08/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Resident synovial macrophages (RSMs) are anti-inflammatory, self-renewing macrophages that provide physical immune sequestration of the joint space from the peripheral immune system. Increased permeability of this structure is associated with peripheral immune cells in the synovial fluid (SF). Direct measures of synovial barrier integrity are possible with tissue histology, but after barrier breakdown, if these cells perpetuate or initiate chronic inflammation in SF remains unknown. We sought to identify RSM in human SF as an indirect measure of synovial barrier integrity. To validate findings, we created a novel ex vivo explant model using human synovium. scRNA-seq revealed these SF RSMs upregulated pro-fibrotic and pro-osteoclastic pathways in inflammatory arthritis, but not septic arthritis. Increased frequencies of RSMs in SF was associated with increased sRANKL regardless of underlying pathology. These findings suggest the frequency of RSMs in SF may correlate with synovial barrier damage and in turn, potential damage to joint structures.
Collapse
Affiliation(s)
- Karen I Cyndari
- Department of Emergency Medicine, University of Iowa, Iowa City, IA, United States of America; Center for Emerging Infectious Diseases, United States of America.
| | - Breanna M Scorza
- Center for Emerging Infectious Diseases, United States of America; Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
| | - Zeb R Zacharias
- Human Immunology Core, University of Iowa, Iowa City, IA, United States of America; Holden Comprehensive Cancer Center, Iowa City, IA, United States of America
| | - Danielle Pessôa-Pereira
- Center for Emerging Infectious Diseases, United States of America; Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
| | - Leela Strand
- Harvard University, Cambridge, MA, United States of America
| | - Kurayi Mahachi
- Center for Emerging Infectious Diseases, United States of America; Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
| | - Juan Marcos Oviedo
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
| | - Lisa Gibbs
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
| | - Katherine L Butler
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Graham Ausdal
- Center for Emerging Infectious Diseases, United States of America; Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
| | - Dylan Hendricks
- Center for Emerging Infectious Diseases, United States of America; Department of Epidemiology, University of Iowa, Iowa City, IA, United States of America
| | - Rika Yahashiri
- Williams College, Williamstown, MA, United States of America
| | - Jacob M Elkins
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States of America
| | - Trevor Gulbrandsen
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States of America
| | - Andrew R Peterson
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States of America
| | - Michael C Willey
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States of America
| | - Keke C Fairfax
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
| | - Christine A Petersen
- College of Veterinary Medicine, Ohio State University, OH, United States of America
| |
Collapse
|
59
|
Swarnkar G, Naaz M, Mims D, Gupta P, Peterson T, Christopher MJ, Singamaneni S, Mbalaviele G, Abu-Amer Y. IĸBζ as a Central Modulator of Inflammatory Arthritis Pathogenesis. Arthritis Rheumatol 2025; 77:124-139. [PMID: 39279148 PMCID: PMC11785494 DOI: 10.1002/art.42990] [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: 09/17/2023] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE Current therapies targeting individual factors in inflammatory arthritis show variable efficacy, often requiring treatment with combinations of drugs, and are associated with undesirable side effects. NF-ĸB is critical for the production and function of most inflammatory cytokines. However, given its essential role in physiologic processes, targeting NF-ĸB is precarious. Hence, identifying pathways downstream of NF-ĸB that selectively govern the expression of inflammatory cytokines in inflammatory arthritis would be advantageous. We have previously identified IĸBζ as a unique inflammatory signature of NF-ĸB that controls the transcription of inflammatory cytokines only under pathologic conditions while sparing physiologic NF-ĸB signals. METHODS We generated mice harboring myeloid, lymphoid, and global deletion of Nfkbiz (the gene encoding IĸBζ). These models were subjected to serum transfer-induced arthritis. Additionally, pharmacologic inhibitors of IĸBζ were injected intraperitonially. Joint swelling, microcomputed tomography, immunohistochemistry, flow cytometry, and cytokine measurements were conducted using synovial tissue samples. RESULTS Global deletion of Nfkbiz or depletion of neutrophils (vastly IĸBζ+ cells) reduced inflammatory synovial cells and increased anti-inflammatory and regenerative synovial cells, plummeted expression of inflammatory factors and ameliorated experimental mouse inflammatory arthritis. Further, expression of immune responsive gene-1, the enzyme responsible for itaconate production, was increased in synovial cells. Accordingly, the itaconate derivative dimethyl itaconate (DI) inhibited IĸBζ-mediated inflammatory factors. Further, in silico screen identified 8-hydroxyquinoline (HQ) as a putative inhibitor of IĸBζ not affecting physiologic NF-ĸB activity. Congruently, systemic administration of either DI or HQ inhibited joint swelling and damage. CONCLUSION Our study positions IĸBζ as an inflammation-specific target for therapeutic consideration in rheumatoid arthritis because its inhibition spares the beneficial functions of NF-ĸB.
Collapse
Affiliation(s)
- Gaurav Swarnkar
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Musarrat Naaz
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Dorothy Mims
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Prashant Gupta
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, Missouri 63130
| | - Timothy Peterson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
- HealthSpan Technologies, Inc. St. Louis, Missouri 63110
- Bioio, Inc. St. Louis, Missouri 63110
| | - Matthew J. Christopher
- Division of Oncology, Cellular Therapy Section, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, Missouri 63130
| | - Gabriel Mbalaviele
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Yousef Abu-Amer
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri 63110
- Shriners Hospital for Children, St. Louis, Missouri 63110
| |
Collapse
|
60
|
Zhang R, Lin X, Lin R, Chen Z, Miao C, Wang Y, Deng X, Lin J, Lin S, Weng S, Chen M. Effectively alleviate rheumatoid arthritis via maintaining redox balance, inducing macrophage repolarization and restoring homeostasis of fibroblast-like synoviocytes by metformin-derived carbon dots. J Nanobiotechnology 2025; 23:58. [PMID: 39881361 PMCID: PMC11776225 DOI: 10.1186/s12951-025-03159-7] [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: 09/17/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Overproduction of reactive oxygen species (ROS), elevated synovial inflammation, synovial hyperplasia and fibrosis are the main characteristic of microenvironment in rheumatoid arthritis (RA). Macrophages and fibroblast-like synoviocytes (FLSs) play crucial roles in the progression of RA. Hence, synergistic combination of ROS scavenging, macrophage polarization from pro-inflammatory M1 phenotype towards M2 anti-inflammatory phenotype, and restoring homeostasis of FLSs will provide a promising therapeutic strategy for RA. In this study, we successfully synthesized metformin-derived carbon dots (MCDs), and investigated the antirheumatic effect in vivo and in vitro. Designed MCDs could target inflamed cells and accumulate at the inflammatory joints of collagen-induced arthritis (CIA) rats. In vivo therapeutic investigation suggested that MCDs reduced synovial inflammation and hyperplasia, ultimately prevented cartilage destruction, bone erosion, and synovial fibrosis in CIA rats. In addition, MCDs eliminated the cellular ROS in M1 phenotype macrophages in RA microenvironment through the enzyme-like catalytic activity as well as inhibiting NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome signaling pathway, effectively polarizing them into the M2 phenotype to realize the anti-inflammatory effect. Furthermore, MCDs could inhibit the proliferation, migration, and fibrosis of inflamed FLSs. Mechanistically, MCDs restored the homeostasis of FLSs while reducing the level of synovial inflammation by blocking IL-6/gp130 signaling pathway. Combined with preferable biocompatibility, MCDs offer a prospective treatment approach for RA.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xingyu Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Zhenbin Chen
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Chenfang Miao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Yao Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Xiaoqin Deng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Jianlong Lin
- Department of Orthopedic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Shishui Lin
- Department of Orthopedic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| | - Shaohuang Weng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Min Chen
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
61
|
Miyahara J, Omata Y, Chijimatsu R, Okada H, Ishikura H, Higuchi J, Tachibana N, Nagata K, Tani S, Kono K, Kawaguchi K, Yamagami R, Inui H, Taketomi S, Iwanaga Y, Terashima A, Yano F, Seki M, Suzuki Y, Baron R, Tanaka S, Saito T. CD34hi subset of synovial fibroblasts contributes to fibrotic phenotype of human knee osteoarthritis. JCI Insight 2025; 10:e183690. [PMID: 39846253 PMCID: PMC11790023 DOI: 10.1172/jci.insight.183690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Abstract
Osteoarthritis (OA) shows various clinical manifestations depending on the status of its joint components. We aimed to identify the synovial cell subsets responsible for OA pathophysiology by comprehensive analyses of human synovium samples in single-cell resolution. Two distinct OA synovial tissue groups were classified by gene expression profiles in RNA-Seq: inflammatory and fibrotic. The inflammatory group exhibited high expression of inflammatory cytokines, histologically inflammatory infiltrate, and a more severe pain score. The fibrotic group showed higher expression of fibroblast growth factor (FGFs) and bone morphogenetic proteins (BMPs), showed histologically perivascular fibrosis, and showed a lower pain score. In single-cell RNA-Seq (scRNA-Seq) of synovial cells, MERTKloCD206lo macrophages and CD34hi fibroblasts were associated with the inflammatory and fibrotic groups, respectively. Among the 3 fibroblast subsets, CD34loTHY1lo and CD34loTHY1hi fibroblasts were influenced by synovial immune cells, whereas CD34hi fibroblasts were influenced by mural and endothelial cells. Particularly, in CD34hi fibroblast subsets, CD34hiCD70hi fibroblasts promoted proliferation of Tregs, potentially suppressing synovitis and protecting articular cartilage. Elucidation of the mechanisms underlying the regulation of these synovial cell subsets may lead to novel strategies for OA therapeutics.
Collapse
Affiliation(s)
| | - Yasunori Omata
- Sensory & Motor System Medicine
- Bone and Cartilage Regenerative Medicine
| | | | - Hiroyuki Okada
- Sensory & Motor System Medicine
- Center for Disease Biology and Integrative Medicine, and
| | | | | | | | | | - Shoichiro Tani
- Sensory & Motor System Medicine
- Center for Disease Biology and Integrative Medicine, and
| | | | | | | | | | | | - Yasuhide Iwanaga
- Sensory & Motor System Medicine
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | | | | | - Masahide Seki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yutaka Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, Japan
| | - Roland Baron
- Department of Medicine, Harvard Medical School and Endocrine Unit, MGH, Boston, Massachusetts, USA
| | | | | |
Collapse
|
62
|
Neofotistou-Themeli E, Goutakoli P, Chanis T, Semitekolou M, Sevdali E, Sidiropoulos P. Fibroblasts in rheumatoid arthritis: novel roles in joint inflammation and beyond. Front Med (Lausanne) 2025; 11:1376925. [PMID: 39906351 PMCID: PMC11790453 DOI: 10.3389/fmed.2024.1376925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025] Open
Abstract
High-throughput technologies in human and animal studies have revealed novel molecular and cellular pathways involved in tissue inflammation of rheumatoid arthritis (RA). Fibroblasts have been in the forefront of research for several decades. Subpopulations with specific phenotypic and functional properties have been characterized both in mouse models and human disease. Data supporting the active involvement of fibroblasts in immune responses and tissue remodeling processes, as well as their central role in promoting clinical relapses and contributing to treatment resistance, have clearly reshaped their role in disease evolution. The lung is an important non-synovial component of RA both from a clinical and an immunopathogenic aspect. Interstitial lung disease (ILD) is a significant contributor to disease burden affecting morbidity and mortality. Although our knowledge of ILD has progressed, significant gaps in both basic and clinical science remain, posing hurdles to efficient diagnosis, prediction of disease course and its effective treatment. The specific role and contribution of fibroblasts to this process has not been clearly defined. The focus of this review is on fibroblasts and their contribution to RA and RA-ILD, presenting data on genetics and immune responses associated with RA-ILD in humans and animal models.
Collapse
Affiliation(s)
- Elpida Neofotistou-Themeli
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| | - Panagiota Goutakoli
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| | - Theodoros Chanis
- Division of Immunology and Allergy, Department of Medicine, Karolinska Institute, Solna, Sweden
- Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Maria Semitekolou
- Dendritic Cells and Adaptive Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- Developmental Biology and Stem Cells, UMR3738 – National Center for Scientific Research (CNRS), Pasteur Institute, Paris, France
| | - Eirini Sevdali
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| | - Prodromos Sidiropoulos
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete, Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece
| |
Collapse
|
63
|
Bahatibieke A, Zhao J, Fan D, Zhou Z, Li J, Wang X, Zhao H, Wang T, Fang Z, Xie Y, Huang C, Xiao C, Zheng Y. Sea-Island Micelle Structured Hydrogel Scaffold: A Dual-Action Approach to Combat Cartilage Damage under RA Conditions. ACS APPLIED MATERIALS & INTERFACES 2025; 17:2911-2923. [PMID: 39746184 DOI: 10.1021/acsami.4c16005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Rheumatoid arthritis (RA) is a common autoimmune joint disease characterized by persistent synovial inflammation and cartilage damage. The current clinical treatments primarily utilize drugs such as triptolide (TP) to address inflammation, yet they are unable to directly repair damaged cartilage. Furthermore, the persistent inflammation often undermines the effectiveness of traditional cartilage repair strategies, preventing them from achieving optimal outcomes. To tackle this challenge, this study successfully developed a drug-loaded polyurethane hydrogel-oriented porous scaffold, designed to address persistent inflammation and facilitate cartilage repair under RA conditions. A drug-loaded hydrogel was formed via solvent-induced polyurethane-gelatin, resulting in the scaffold TP@GSPU. The sea-island micelle structure of TP@GSPU enables efficient loading of TP. The release of TP in the in vivo environment regulates the expression of inflammatory factors in macrophages, thereby improving the inflammatory microenvironment within the joint cavity. Additionally, the gelatin component of the scaffold provides robust support for cartilage regeneration. The efficacy of the TP@GSPU in regulating the inflammatory microenvironment and facilitating cartilage repair under RA conditions, which was demonstrated through cartilage damage repair experiments conducted in a rat collagen-induced arthritis (CIA) model. The design scheme of this material offers a potential approach to cartilage repair in the conditions of RA.
Collapse
Affiliation(s)
- Abudureheman Bahatibieke
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Jianming Zhao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Danping Fan
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100070, China
| | - Zixiang Zhou
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Junfei Li
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xing Wang
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hongyan Zhao
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100070, China
| | - Tianyang Wang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Ziyuan Fang
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yajie Xie
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Cheng Huang
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing 100029, China
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yudong Zheng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
64
|
Alivernini S, Masserdotti A, Magatti M, Cargnoni A, Papait A, Silini AR, Romoli J, Ficai S, Di Mario C, Gremese E, Tolusso B, Parolini O. Exploring perinatal mesenchymal stromal cells as a potential therapeutic strategy for rheumatoid arthritis. Heliyon 2025; 11:e41438. [PMID: 39811302 PMCID: PMC11732555 DOI: 10.1016/j.heliyon.2024.e41438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by inflammation in the synovial tissue, driven by aberrant activation of both the innate and adaptive immune systems, which can lead to irreversible disability. Despite the increasing therapeutic approaches for RA, only a low percentage of patients achieve sustained disease remission, and the persistence of immune dysregulation is likely responsible for disease recurrence once remission is attained. Cell therapy is an attractive, wide-spectrum strategy to modulate inflammation, and mesenchymal stromal cells (MSC) derived from perinatal tissues provide valuable tools for their use in regenerative medicine, mainly due to their immunomodulatory properties. Several in vitro studies have shown that perinatal MSC modulate the proliferation, maturation, and cytokine secretion profile of both innate and adaptive immune cells. Moreover, different beneficial effects have also been described when perinatal MSC were used to treat animal models of diseases associated with inflammatory conditions and degenerative processes. Specifically, in experimental models of RA, treatment with perinatal MSC resulted in a strong reduction of articular damage, which was associated with the modulation of both inflammation and activation of stromal resident cells in the synovial tissue. Here, we present in vitro and in vivo evidence supporting the use of perinatal MSC in RA. We also highlight the promising results from the few published clinical trials, which demonstrate the safety of perinatal MSC.
Collapse
Affiliation(s)
- Stefano Alivernini
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alice Masserdotti
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Marta Magatti
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Andrea Papait
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| | - Antonietta R. Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Jacopo Romoli
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Sara Ficai
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Clara Di Mario
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Elisa Gremese
- Rheumatology and Clinical Immunology Unit, Humanitas Research Hospital, Milan, Italy
| | - Barbara Tolusso
- Immunology Research Core Facility, Gemelli Science and Technology Park (GSTeP), Fondazione Policlinico Universitario A, Gemelli IRCCS, Rome, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
| |
Collapse
|
65
|
Qin Y, Liu L, Zhang Y, Chen X, Zhang J, Ling S, Wang J, Yang X. Citrullinated IGF2BP1 promotes rheumatoid synovial aggression via increasing the mRNA stability of SEMA3D. Commun Biol 2025; 8:50. [PMID: 39809921 PMCID: PMC11732996 DOI: 10.1038/s42003-025-07492-3] [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: 09/13/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Protein citrullination modification plays a pivotal role in the pathogenesis of rheumatoid arthritis (RA), and anti-citrullinated protein antibodies (ACPAs) are extensively employed for clinical diagnosis of RA. However, there remains limited understanding regarding specific citrullinated proteins and their implications in the progression of RA. In this study, we screen and verify insulin-like growth factor-2 mRNA binding protein 1 (IGF2BP1) as a novel citrullinated protein with significantly elevated citrullinated level in RA. Autoantibodies against citrullinated IGF2BP1 are further detected in serum and synovial fluid samples from RA patients, which are positively correlated with erythrocyte sedimentation rate (ESR) and disease activity score 28 (DAS28). Transcriptomic sequencing and functional verification show that citrullination at the R167 site of IGF2BP1 promotes the proliferation, migration, and invasion of RA fibroblast-like synoviocytes (RA-FLSs) by improving the mRNA stability of Semaphorin 3D (SEMA3D). Experiments in collagen-induced arthritis (CIA) mice, the classical animal model of RA, show that IGF2BP1 R176K point mutation (Igf2bp1R167K/R167K) mice exert reduced inflammatory response, clinical scores, and joint destruction. At a molecular level, citrullination of IGF2BP1 promotes the stability of SEMA3D mRNA by promoting the interaction between IGF2BP1 and its cofactor ELAV-like protein 1 (ELAVL1), thereby promoting the invasiveness of RA-FLSs. In this study, a new citrullinated protein of IGF2BP1 is discovered, and the molecular mechanism of its citrullinated modification promoting the progression of RA disease is elucidated, which provides theoretical basis for the diagnosis and treatment of RA.
Collapse
Affiliation(s)
- Yang Qin
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanwen Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaowei Chen
- Department of Immunology and Rheumatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiani Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunwang Ling
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianguang Wang
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Xinyu Yang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
66
|
Liu J, Li B, Zhou X, Liu G, Li C, Hu Z, Peng R. Uncovering the mechanisms of Zhubi decoction against rheumatoid arthritis through an integrated study of network pharmacology, metabolomics, and intestinal flora. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118736. [PMID: 39186991 DOI: 10.1016/j.jep.2024.118736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhubi Decoction (ZBD) is a modified formulation derived from the classic traditional Chinese medicine prescription "Er-Xian Decoction" documented in the esteemed "Clinical Manual of Chinese Medical Prescription". While the utilization of ZBD has exhibited promising clinical outcomes in treating rheumatoid arthritis (RA), the precise bioactive chemical constituents and the underlying mechanisms involved in its therapeutic efficacy remain to be comprehensively determined. AIM OF THE STUDY This study aims to systematically examine ZBD's pharmacological effects and molecular mechanisms for RA alleviation. MATERIALS AND METHODS Utilizing the collagen-induced arthritis (CIA) rat model, we comprehensively evaluated the anti-rheumatoid arthritis effects of ZBD in vivo through various indices, such as paw edema, arthritis index, ankle diameter, inflammatory cytokine levels, pathological conditions, and micro-CT analysis. The UPLC-MS/MS technique was utilized to analyze the compounds of ZBD. The potential therapeutic targets and signaling pathways of ZBD in the management of RA were predicted using network pharmacology. To analyze comprehensive metabolic profiles and identify underlying metabolic pathways, we conducted a serum-based widely targeted metabolomics analysis utilizing LC-MS technology. Key targets and predicted pathways were further validated using immunofluorescent staining, which integrated findings from serum metabolomics and network pharmacology analysis. Additionally, we analyzed the gut microbiota composition in rats employing 16 S rDNA sequencing and investigated the effects of ZBD on the microbiota of CIA rats through bioinformatics and statistical methods. RESULTS ZBD exhibited remarkable efficacy in alleviating RA symptoms in CIA rats without notable side effects. This included reduced paw redness and swelling, minimized joint damage, improved the histopathology of cartilage and synovium, mitigated the inflammatory state, and lowered serum concentrations of cytokines TNF-α, IL-1β and IL-6. Notably, the effectiveness of ZBD was comparable to MTX. Network pharmacology analysis revealed inflammation and immunity-related signaling pathways, such as PI3K/AKT, MAPK, IL-17, and TNF signaling pathways, as vital mediators in the effectual mechanisms of ZBD. Immunofluorescence analysis validated ZBD's ability to inhibit PI3K/AKT pathway proteins. Serum metabolomics studies revealed that ZBD modulates 170 differential metabolites, partially restored disrupted metabolic profiles in CIA rats. With a notable impact on amino acids and their metabolites, and lipids and lipid-like molecules. Integrated analysis of metabolomics and network pharmacology identified 6 pivotal metabolite pathways and 3 crucial targets: PTGS2, GSTP1, and ALDH2. Additionally, 16 S rDNA sequencing illuminated that ZBD mitigated gut microbiota dysbiosis in the CIA group, highlighting key genera such as Ligilactobacillus, Prevotella_9, unclassified_Bacilli, and unclassified_rumen_bacterium_JW32. Correlation analysis disclosed a significant link between 47 distinct metabolites and specific bacterial species. CONCLUSION ZBD is a safe and efficacious TCM formulation, demonstrates efficacy in treating RA through its multi-component, multi-target, and multi-pathway mechanisms. The regulation of inflammation and immunity-related signaling pathways constitutes a crucial mechanism of ZBD's efficacy. Furthermore, ZBD modulates host metabolism and intestinal flora. The integrated analysis presents experimental evidence of ZBD for the management of RA.
Collapse
Affiliation(s)
- Jing Liu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Bocun Li
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Xiaohong Zhou
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Guangya Liu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Chao Li
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Zhaoduan Hu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| | - Rui Peng
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430061, China.
| |
Collapse
|
67
|
Periyakoil PK, Smith MH, Kshirsagar M, Ramirez D, DiCarlo EF, Goodman SM, Rudensky AY, Donlin LT, Leslie CS. Deep topic modeling of spatial transcriptomics in the rheumatoid arthritis synovium identifies distinct classes of ectopic lymphoid structures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631928. [PMID: 39829741 PMCID: PMC11741433 DOI: 10.1101/2025.01.08.631928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Single-cell RNA sequencing studies have revealed the heterogeneity of cell states present in the rheumatoid arthritis (RA) synovium. However, it remains unclear how these cell types interact with one another in situ and how synovial microenvironments shape observed cell states. Here, we use spatial transcriptomics (ST) to define stable microenvironments across eight synovial tissue samples from six RA patients and characterize the cellular composition of ectopic lymphoid structures (ELS). To identify disease-relevant cellular communities, we developed DeepTopics, a scalable reference-free deconvolution method based on a Dirichlet variational autoencoder architecture. DeepTopics identified 22 topics across tissue samples that were defined by specific cell types, activation states, and/or biological processes. Some topics were defined by multiple colocalizing cell types, such as CD34+ fibroblasts and LYVE1+ macrophages, suggesting functional interactions. Within ELS, we discovered two divergent cellular patterns that were stable across ELS in each patient and typified by the presence or absence of a "germinal-center-like" topic. DeepTopics is a versatile and computationally efficient method for identifying disease-relevant microenvironments from ST data, and our results highlight divergent cellular architectures in histologically similar RA synovial samples that have implications for disease pathogenesis.
Collapse
Affiliation(s)
- Preethi K Periyakoil
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Weill Cornell Medical College, New York, NY 10021, USA
| | - Melanie H Smith
- Weill Cornell Medical College, New York, NY 10021, USA
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY 10021, USA
| | | | - Daniel Ramirez
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Edward F DiCarlo
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Susan M Goodman
- Weill Cornell Medical College, New York, NY 10021, USA
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY 10021, USA
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program at Sloan Kettering Institute, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY,10065, USA
| | - Laura T Donlin
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY 10021, USA
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
68
|
Mueller AA, Zou AE, Marsh LJ, Kemble S, Nayar S, Watts GFM, Murphy CL, Taylor E, Major T, Gardner D, Buckley CD, Wei K, Raychaudhuri S, Korsunsky I, Filer A, Croft AP, Brenner MB. Wnt signaling drives stromal inflammation in inflammatory arthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631510. [PMID: 39829745 PMCID: PMC11741264 DOI: 10.1101/2025.01.06.631510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The concept that fibroblasts are critical mediators of inflammation is an emerging paradigm. In rheumatoid arthritis (RA), they are the main producers of IL-6 as well as a host of other cytokines and chemokines. Their pathologic activation also directly causes cartilage and bone degradation. Yet, therapeutic agents specifically targeting fibroblasts are not available. Here, we find that Wnt receptors and modulators are predominantly expressed in stromal populations in the synovium. Importantly, non-canonical Wnt activation induces robust inflammatory gene expression including an abundance of cytokines and chemokines in synovial fibroblasts in vitro . Strikingly, the addition of Wnt ligands or inhibition of Wnt secretion exacerbates or reduces arthritis severity, respectively, in vivo in a murine model of inflammatory arthritis. These observations are relevant in human disease, as Wnt activation signatures are enhanced in fibroblasts derived from inflamed RA synovial tissue as well as fibroblasts across other inflammatory diseases. Together, these findings implicate Wnt signaling as a major driver of fibroblast-mediated inflammation and joint pathology. They further suggest that targeting the Wnt pathway is a therapeutically relevant approach to rheumatoid arthritis, particularly in patients who do not respond to conventional treatments and who often express fibroblast-predominant synovial phenotypes.
Collapse
|
69
|
Jenkins BH, Tracy I, Rodrigues MFSD, Smith MJL, Martinez BR, Edmond M, Mahadevan S, Rao A, Zong H, Liu K, Aggarwal A, Li L, Diehl L, King EV, Bates JG, Hanley CJ, Thomas GJ. Single cell and spatial analysis of immune-hot and immune-cold tumours identifies fibroblast subtypes associated with distinct immunological niches and positive immunotherapy response. Mol Cancer 2025; 24:3. [PMID: 39757146 DOI: 10.1186/s12943-024-02191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/30/2024] [Indexed: 01/07/2025] Open
Abstract
Cancer-associated Fibroblasts (CAFs) have emerged as critical regulators of anti-tumour immunity, with both beneficial and detrimental properties that remain poorly characterised. To investigate this, we performed single-cell and spatial transcriptomic analysis, comparing head & neck squamous cell carcinoma (HNSCC) subgroups, which although heterogenous, can be considered broadly immune-hot and immune-cold (human papillomavirus [HPV]+ve and HPV-ve tumours respectively). This identified six fibroblast subpopulations, including two with immunomodulatory gene expression profiles (IL-11 + inflammatory [i]CAF and CCL19 + fibroblastic reticular cell [FRC]-like). IL-11 + iCAF were spatially associated with inflammatory monocytes and regulated in vitro through synergistic activation of canonical NF-κB signalling by IL-1β and TNF-α. FRC-like were enriched in immune-hot HPV+ve tumours, associated with CD4 + T-cells and B-cells in tertiary lymphoid structures and regulated through non-canonical NF-κB signalling via lymphotoxin. Pan-cancer analysis revealed several 'iCAF' subgroups present in both normal and cancer tissues; IL11 + iCAF were found in cancers from the gastrointestinal (GI) tract and transcriptomically distinct from iCAFs previously described in pancreatic and breast cancers with greater inflammatory properties; FRC-like fibroblasts were present at low frequencies in all tumour types, and were associated with significantly better survival in patients receiving checkpoint immunotherapy. This work clarifies and expands current literature on immunomodulatory CAFs, highlighting links with important immunological niches.
Collapse
Affiliation(s)
- Benjamin H Jenkins
- School of Cancer Sciences, University of Southampton, Southampton, UK
- NIHR Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK
| | - Ian Tracy
- School of Cancer Sciences, University of Southampton, Southampton, UK
- NIHR Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK
| | - Maria Fernanda S D Rodrigues
- School of Cancer Sciences, University of Southampton, Southampton, UK
- NIHR Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK
- Postgraduate Program in Medicine-Biophotonics, Nove de Julho University, São Paulo, Brazil
| | - Melanie J L Smith
- School of Cancer Sciences, University of Southampton, Southampton, UK
- NIHR Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK
| | - Begoña R Martinez
- School of Cancer Sciences, University of Southampton, Southampton, UK
- NIHR Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK
| | - Mark Edmond
- School of Cancer Sciences, University of Southampton, Southampton, UK
- Dorset Cancer Centre, Poole Hospital NHS Foundation Trust, Poole, UK
| | | | - Anjali Rao
- Gilead Sciences Inc., Foster City, CA, US
| | | | - Kai Liu
- Gilead Sciences Inc., Foster City, CA, US
| | | | - Li Li
- Gilead Sciences Inc., Foster City, CA, US
| | | | - Emma V King
- School of Cancer Sciences, University of Southampton, Southampton, UK
- Dorset Cancer Centre, Poole Hospital NHS Foundation Trust, Poole, UK
| | | | - Christopher J Hanley
- School of Cancer Sciences, University of Southampton, Southampton, UK
- NIHR Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK
| | - Gareth J Thomas
- School of Cancer Sciences, University of Southampton, Southampton, UK.
- NIHR Experimental Cancer Medicine Centre, University of Southampton, Southampton, UK.
| |
Collapse
|
70
|
McCool JL, Sebastian A, Hum NR, Wilson SP, Davalos OA, Murugesh DK, Amiri B, Morfin C, Christiansen BA, Loots GG. CD206+ Trem2+ macrophage accumulation in the murine knee joint after injury is associated with protection against post-traumatic osteoarthritis in MRL/MpJ mice. PLoS One 2025; 20:e0312587. [PMID: 39752388 PMCID: PMC11698337 DOI: 10.1371/journal.pone.0312587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/09/2024] [Indexed: 01/06/2025] Open
Abstract
Post-traumatic osteoarthritis (PTOA) is a painful joint disease characterized by the degradation of bone, cartilage, and other connective tissues in the joint. PTOA is initiated by trauma to joint-stabilizing tissues, such as the anterior cruciate ligament, medial meniscus, or by intra-articular fractures. In humans, ~50% of joint injuries progress to PTOA, while the rest spontaneously resolve. To better understand molecular programs contributing to PTOA development or resolution, we examined injury-induced fluctuations in immune cell populations and transcriptional shifts by single-cell RNA sequencing of synovial joints in PTOA-susceptible C57BL/6J (B6) and PTOA-resistant MRL/MpJ (MRL) mice. We identified significant differences in monocyte and macrophage subpopulations between MRL and B6 joints. A potent myeloid-driven anti-inflammatory response was observed in MRL injured joints that significantly contrasted the pro-inflammatory signaling seen in B6 joints. Multiple CD206+ macrophage populations classically described as M2 were found enriched in MRL injured joints. These CD206+ macrophages also robustly expressed Trem2, a receptor involved in inflammation and myeloid cell activation. These data suggest that the PTOA resistant MRL mouse strain displays an enhanced capacity of clearing debris and apoptotic cells induced by inflammation after injury due to an increase in activated M2 macrophages within the synovial tissue and joint space.
Collapse
Affiliation(s)
- Jillian L. McCool
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
- School of Natural Sciences, University of California Merced, Merced, CA, United States of America
| | - Aimy Sebastian
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Stephen P. Wilson
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Oscar A. Davalos
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Beheshta Amiri
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Cesar Morfin
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States of America
| | - Blaine A. Christiansen
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States of America
| | - Gabriela G. Loots
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
- School of Natural Sciences, University of California Merced, Merced, CA, United States of America
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States of America
| |
Collapse
|
71
|
Nayar S, Turner JD, Asam S, Fennell E, Pugh M, Colafrancesco S, Berardicurti O, Smith CG, Flint J, Teodosio A, Iannizzotto V, Gardner DH, van Roon J, Korsunsky I, Howdle D, Frei AP, Lassen KG, Bowman SJ, Ng WF, Croft AP, Filer A, Fisher BA, Buckley CD, Barone F. Molecular and spatial analysis of tertiary lymphoid structures in Sjogren's syndrome. Nat Commun 2025; 16:5. [PMID: 39747819 PMCID: PMC11697438 DOI: 10.1038/s41467-024-54686-0] [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: 10/20/2022] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
Tertiary lymphoid structures play important roles in autoimmune and non-autoimmune conditions. While many of the molecular mechanisms involved in tertiary lymphoid structure formation have been identified, the cellular sources and temporal and spatial relationship remain unknown. Here we use combine single-cell RNA-sequencing, spatial transcriptomics and proteomics of minor salivary glands of patients with Sjogren's disease and Sicca Syndrome, with ex-vivo functional studies to construct a cellular and spatial map of key components involved in the formation and function of tertiary lymphoid structures. We confirm the presence of a fibroblast cell state and identify a pericyte/mural cell state with potential immunological functions. The identification of cellular properties associated with these structures and the molecular and functional interactions identified by this analysis may provide key therapeutic cues for tertiary lymphoid structures associated conditions in autoimmunity and cancer.
Collapse
Affiliation(s)
- Saba Nayar
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Jason D Turner
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Saba Asam
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- UCL Genomics, Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Eanna Fennell
- School of Medicine & HRI & Bernal Institute, University of Limerick, Limerick, Ireland
| | - Matthew Pugh
- Department of Immunology and Immunotherapy, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | | | - Onorina Berardicurti
- Rheumatology, Immunology and Clinical Medicine Unit, Department of Medicine, Università Campus Bio-Medico, Rome, and Immunorheumatology Unit, Fondazione Policlinico Universitario Campus Bio Medico, Rome, Italy
| | - Charlotte G Smith
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Joe Flint
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Ana Teodosio
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Valentina Iannizzotto
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - David H Gardner
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Joel van Roon
- Department of Rheumatology & Clinical Immunology/Laboratory of Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ilya Korsunsky
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
| | - Dawn Howdle
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Andreas P Frei
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Kara G Lassen
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Simon J Bowman
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Wan-Fai Ng
- HRB Clinical Research Facility, University College Cork, Cork, Ireland
| | - Adam P Croft
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Andrew Filer
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Benjamin A Fisher
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Christopher D Buckley
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Francesca Barone
- Rheumatology Research Group, Department of Inflammation and Ageing, College of Medicine & Health, University of Birmingham, Birmingham, UK.
- Candel Therapeutics, Needham, MA, USA.
| |
Collapse
|
72
|
Hofman ZLM, Roodenrijs NMT, Nikiphorou E, Kent AL, Nagy G, Welsing PMJ, van Laar JM. Difficult-to-treat rheumatoid arthritis: what have we learned and what do we still need to learn? Rheumatology (Oxford) 2025; 64:65-73. [PMID: 39383505 PMCID: PMC11701314 DOI: 10.1093/rheumatology/keae544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 10/11/2024] Open
Abstract
Difficult-to-treat RA (D2T RA) is an area of high unmet need. The prevalence reported in the first D2T RA cohort studies ranged from 5.5% to 27.5%. Key to the definition is a conviction by the patient and/or rheumatologist that disease management has become problematic and failure of at least two biological or targeted synthetic DMARDs. D2T RA is a multifactorial disease state which was reflected in data from D2T RA cohort studies: these pointed towards high prevalence of comorbidities and/or lower socioeconomic status in D2T RA subgroups, while others had persistent symptoms without these factors being present. A holistic approach is necessary to identify the root problems underlying D2T RA in individual patients. In this review, biological and non-biological drivers that should be considered to be optimized will be discussed in view of what we have learned from patient data emerging from the first D2T RA cohort studies.
Collapse
Affiliation(s)
- Zonne L M Hofman
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Nadia M T Roodenrijs
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, King’s College London, London, UK
- Rheumatology Department, King’s College Hospital, London, UK
| | | | - György Nagy
- Department of Rheumatology and Clinical Immunology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- National Institute of Locomotor Diseases and Disabilities, Budapest, Hungary
| | - Paco M J Welsing
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jaap M van Laar
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
73
|
Pearce‐Fisher D, Smith MH, Mehta BY, Spolaore E, DiCarlo E, Sun D, Goodman SM. Patient-Reported Fatigue Associated with Joint Histopathology in Rheumatoid Arthritis. ACR Open Rheumatol 2025; 7:e11772. [PMID: 39846130 PMCID: PMC11755064 DOI: 10.1002/acr2.11772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/21/2024] [Accepted: 11/12/2024] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVE Fatigue is important for patients with rheumatoid arthritis (RA) but is poorly understood. We sought to study associations of fatigue with clinical features, disease activity, and synovial histology. METHODS Patients meeting the American College of Rheumatology/EULAR 1987 and/or 2010 RA criteria were recruited before elective total joint replacement. Demographics, RA characteristics, tender and swollen joints, erythrocyte sedimentation rate (ESR) and C-reactive protein, and patient-reported fatigue, categorized as mild, moderate, or severe, were collected. Hematoxylin and eosin stains of sectioned synovium were systematically scored by a pathologist. Relationships between fatigue and studied variables were evaluated with Kendall's tau. A directed acyclic graph (DAG) was used to illustrate associations of exposures, outcome variables, mediators, and confounders. Multivariable ordered logistic regression was used to further study associations. RESULTS Of 160 included patients, 85.6% were women, with a median age of 63.5 (55.25-71.40) and mean disease activity scores in 28 joints using ESR (DAS28-ESR) of 3.91 (SD 1.3). There were no differences in comorbidities across fatigue categories. Fatigue correlated with DAS28-ESR, synovial lining hyperplasia (SLH), anxiety, depression, and pain. In the DAG, DAS28-ESR was associated with fatigue, full mediation by pain, partial mediation by depression and anxiety, and confounding by female sex. SLH was independently associated with fatigue but did not confound the relationship between DAS28-ESR and fatigue. SLH was affected by synovial lymphocytic inflammation. In multivariable models, female sex, DAS28-ESR, and SLH were all associated with higher fatigue. CONCLUSION Although fatigue is associated with DAS28-ESR, it is also associated with SLH independently of disease activity.
Collapse
Affiliation(s)
- Diyu Pearce‐Fisher
- Hospital for Special Surgery, New York City, Stony Brook UniversityStony BrookNew York
| | - Melanie H. Smith
- Hospital for Special Surgery and Weill Cornell MedicineNew York CityNew York
| | - Bella Y. Mehta
- Hospital for Special Surgery and Weill Cornell MedicineNew York CityNew York
| | | | - Edward DiCarlo
- Hospital for Special Surgery and Weill Cornell MedicineNew York CityNew York
| | - Dongmei Sun
- Hospital for Special SurgeryNew York CityNew York
| | - Susan M. Goodman
- Hospital for Special Surgery and Weill Cornell MedicineNew York CityNew York
| |
Collapse
|
74
|
Cheng L, Rong X. Emodin promotes the recovery of rheumatoid arthritis by regulating the crosstalk between macrophage subsets and synovial fibroblast subsets. Animal Model Exp Med 2025; 8:44-56. [PMID: 38369605 PMCID: PMC11798747 DOI: 10.1002/ame2.12387] [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: 09/25/2023] [Accepted: 01/05/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND To study the relationships among emodin, synovial fibroblasts (FLSs), and macrophages (STMs) to provide guidance for the use of emodin in rheumatoid arthritis (RA) treatment. METHODS RA clinical samples from patients with different pathological processes were collected, and the correlations between the subsets of FLSs and STMs and pathological processes were analyzed via flow cytometry. In vitro experimental methods such as enzyme linked immunosorbent assay (ELISA), Western blotting, Transwell assays, CCK-8 assays and cell coculture were used to assess cell proliferation, migration and secretion of inflammatory factors. A collagen-induced arthritis mouse model was constructed to investigate the therapeutic potential of emodin in RA by flow cytometry, micro-CT and staining. RESULTS Unique subsets of FLSs and STMs, namely, FAPα+THY1- FLSs, FAPα+THY1+ FLSs, and MerTKposTREM2high STMs, were identified in synovial tissues from RA patients. The number of MerTKposTREM2high STMs was negatively correlated with the degree of damage in RA, while the number of FAPα+THY1- FLSs was positively correlated with damage. On the one hand, emodin promoted the aggregation of MerTKposTREM2high STMs. Moreover, MerTKposTREM2high STM-mediated secretion of exosomes was promoted, which can inhibit the secretion of pro-inflammatory factors by FAPα+THY1+ FLSs and promote the secretion of anti-inflammatory factors by FAPα+THY1+ FLSs, thereby inhibiting FAPα+THY1-FLS proliferation and migration, improving the local immune microenvironment, and inhibiting RA damage. CONCLUSION Emodin was shown to regulate the aggregation of STM subsets and exosome secretion, affecting the secretion, proliferation and migration of inflammatory factors in FLS subsets, and ultimately achieving good therapeutic efficacy in RA patients, suggesting that it has important clinical value.
Collapse
Affiliation(s)
- Lianying Cheng
- Department of Integrated Traditional Chinese and Western MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaofeng Rong
- Department of Integrated Traditional Chinese and Western MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
75
|
Ibrahim HE, De Bari C. Giant cell arteritis: update on pathogenesis and clinical implications. Curr Opin Rheumatol 2025; 37:72-79. [PMID: 39600290 DOI: 10.1097/bor.0000000000001051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
PURPOSE OF REVIEW Giant cell arteritis (GCA) is an age-related autoimmune disease with a complex pathogenesis that involves several pathogenic mechanisms. This review provides recent critical insights into novel aspects of GCA pathogenesis. RECENT FINDINGS The use of novel approaches, including multiomic techniques, has uncovered notable findings that broaden the understanding of GCA pathogenesis. TCF1hiCD4+ T cells have been identified as stem-like T cells residing in tertiary lymphoid structures in the adventitia of GCA aortic tissues, which likely supply the pathogenic effector T cells present in vasculitic lesions. Studies have demonstrated that fibroblasts present in GCA-inflamed arteries are not innocent bystanders, but they contribute to arterial inflammation via maintenance of Th1 and Th17 polarisation, cytokine secretion (IL-6, IL-1B, IL-12, and IL-23) and antigen presentation. Additionally, deregulated cellular senescence programs are present in GCA as an accumulation of IL-6 and matrix metalloproteinase 9-producing senescent cells have been identified in vasculitic lesions. SUMMARY Recent studies have unravelled interesting findings with potentially significant clinical relevance. Stem-like T cells are likely key contributors to vascular disease persistence, and targeted depletion or modulation of these cells holds promise in GCA management. Fibroblast-targeting therapies and senotherapeutics are also exciting prospects in the treatment of GCA.
Collapse
Affiliation(s)
- Hafeez E Ibrahim
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
76
|
Li JX, Dang YM, Liu MC, Gao LQ, Lin H. Fibroblasts in heterotopic ossification: mechanisms and therapeutic targets. Int J Biol Sci 2025; 21:544-564. [PMID: 39781450 PMCID: PMC11705629 DOI: 10.7150/ijbs.102297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/07/2024] [Indexed: 01/12/2025] Open
Abstract
Heterotopic ossification (HO) refers to the abnormal formation of bone in non-skeletal tissues. Fibroblasts have traditionally been viewed as stationary cells primarily responsible for producing extracellular matrix during tissue repair and fibrosis. However, recent discoveries regarding their plasticity-encompassing roles in inflammation, extracellular matrix remodeling, and osteogenesis-highlight their potential as key contributors to the development of HO. In this review, we systematically summarize the diverse phenotypic and functional plasticity of fibroblasts in HO. Furthermore, we evaluate the possible interaction between fibroblasts and macrophages in pathophysiological processes and signaling pathways. Finally, we highlight the potential strategies for preventing and treating HO by targeting fibroblast activities.
Collapse
Affiliation(s)
- Jia-xin Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- First Clinical School, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yan-miao Dang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Meng-chao Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lin-qing Gao
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Hui Lin
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| |
Collapse
|
77
|
Basalova NA, Vigovskiy MA, Popov VS, Lagereva EA, Grigorieva OA, Efimenko AY. The Role of Activated Stromal Cells in Fibrotic Foci Formation and Reversion. Cells 2024; 13:2064. [PMID: 39768155 PMCID: PMC11674712 DOI: 10.3390/cells13242064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
Fibrotic focus is a pivotal morphofunctional unit in developing fibrosis in various tissues. For most fibrotic diseases, including progressive forms, the foci are considered unable to remodel and contribute to the worsening of prognosis. Unfortunately, the dynamics of the fibrotic focus formation and resolution remains understudied. A number of data suggest that the key cell type for focus formation are activated stromal cells marked by fibroblast activated protein alpha (FAPα) due to their high capacity for extracellular matrix (ECM) remodeling. We evaluated the dynamics of fibrotic focus formation and the contribution of the main cell types, including FAPα+ cells, in this process using a murine model of bleomycin-induced lung fibrosis. We revealed the very early appearance of FAPα+ cells in lungs after injury and assumed their important involvement to the myofibroblast pool formation. During the first month after bleomycin administration, FAPα+ cells colocalize with CD206+ M2 macrophages. Interestingly, during the reversion stage, we unexpectedly observed the specific structured foci formed by CD90+FAPα+ cells, which we suggested calling "remodeling foci". Our findings highlight the crucial role of activated stromal cells in fibrosis initiation, progression, and reversion and provide emerging issues regarding the novel targets for antifibrotic therapy.
Collapse
Affiliation(s)
- Nataliya Andreevna Basalova
- Centre for Regenerative Medicine, Medical Research and Educational Institute, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.V.); (V.S.P.); (E.A.L.); (O.A.G.); (A.Y.E.)
| | | | | | | | | | | |
Collapse
|
78
|
Shen C, Peng C, Zhang S, Li R, Liu S, Kuang Y, Su F, Liu Y, Liang L, Xiao Y, Xu H. Eukaryotic translation initiation factor 6-mediated ribosome biogenesis promotes synovial aggression and inflammation by increasing the translation of SP1 in rheumatoid arthritis. Int Immunopharmacol 2024; 142:113164. [PMID: 39288622 DOI: 10.1016/j.intimp.2024.113164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Fibroblast-like synoviocytes (FLSs) play critical roles in synovial inflammation and aggression in rheumatoid arthritis (RA). Here, we explored the role of eukaryotic translation initiation factor 6 (eIF6) in regulating the biological behaviors of FLSs from patients with RA. METHODS FLSs were isolated from the synovial tissues of RA patients. Gene expression was assessed via RT-qPCR, and protein expression was evaluated via Western blotting or immunohistochemistry. Proliferation and nascent peptide synthesis were evaluated via EdU incorporation and HPG labeling, respectively. Cell migration and invasion were observed via Transwell assays. Polysome profiling was conducted to analyze the distribution of ribosomes and combined mRNAs. The in vivo effect of eIF6 inhibition was evaluated in a collagen-induced arthritis (CIA) rat model. RESULTS We found that eIF6 expression was elevated in FLSs and synovial tissues from RA patients compared to those from healthy controls and osteoarthritis patients. Knockdown of eIF6 inhibited the migration, invasion, inflammation, and proliferation of FLSs from patients with RA. Mechanistically, eIF6 knockdown downregulated ribosome biogenesis in FLSs from with RA, leading to a decrease in the proportion of polysome-associated specificity protein 1 (SP1) mRNA and a subsequent reduction in the translation initiation efficiency of SP1 mRNA. Thus, eIF6 controls SP1 expression through translation-mediated mechanisms. Interestingly, intra-articular eIF6 siRNA treatment attenuated symptoms and histological manifestations in CIA rats. CONCLUSIONS Our findings suggest that an increase in synovial eIF6 might contribute to rheumatoid synovial inflammation and aggression and that targeting eIF6 may have therapeutic potential in RA patients.
Collapse
Affiliation(s)
- Chuyu Shen
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Chenxi Peng
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Shuoyang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Ruiru Li
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Suling Liu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Yu Kuang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Fan Su
- Department of Geriatrics, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Yingli Liu
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Liuqin Liang
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Youjun Xiao
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China
| | - Hanshi Xu
- Department of Rheumatology and Immunology, the First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan Er Road, Guangzhou 510080, Guangdong Province, PR China.
| |
Collapse
|
79
|
Nakajima S, Tsuchiya H, Fujio K. Unraveling immune cell heterogeneity in autoimmune arthritis: insights from single-cell RNA sequencing. Immunol Med 2024; 47:217-229. [PMID: 39120105 DOI: 10.1080/25785826.2024.2388343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed our understanding of immune-mediated arthritis, which comprises rheumatoid arthritis and spondyloarthritis. This review outlines the key findings and advancements in scRNA-seq studies focused on the pathogenesis of autoimmune arthritis and its clinical application. In rheumatoid arthritis, scRNA-seq has elucidated the heterogeneity among synovial fibroblasts and immune cell subsets in inflammatory sites, offering insights into disease mechanisms and the differences in treatment responses. Various studies have identified distinct synovial fibroblast subpopulations, such as THY1+ inflammatory and THY1- destructive fibroblasts. Furthermore, scRNA-seq has revealed diverse T cell profiles in the synovium, including peripheral helper T cells and clonally expanded CD8+ T cells, shedding light on potential therapeutic targets and predictive markers of treatment response. Similarly, in spondyloarthritis, particularly psoriatic arthritis and ankylosing spondylitis, scRNA-seq studies have identified distinct cellular profiles associated with disease pathology. Challenges such as cost and sample size limitations persist, but collaborative efforts and utilization of public databases hold promise for overcoming these obstacles. Overall, scRNA-seq emerges as a powerful tool for dissecting cellular heterogeneity and driving precision medicine in immune-mediated arthritis.
Collapse
Affiliation(s)
- Sotaro Nakajima
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
80
|
Hasegawa T, Lee CYC, Hotchen AJ, Fleming A, Singh R, Suzuki K, Yuzaki M, Watanabe M, Birch MA, McCaskie AW, Lénárt N, Tóth K, Dénes Á, Liu Z, Ginhoux F, Richoz N, Clatworthy MR. Macrophages and nociceptor neurons form a sentinel unit around fenestrated capillaries to defend the synovium from circulating immune challenge. Nat Immunol 2024; 25:2270-2283. [PMID: 39587345 PMCID: PMC11588661 DOI: 10.1038/s41590-024-02011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024]
Abstract
A wide variety of systemic pathologies, including infectious and autoimmune diseases, are accompanied by joint pain or inflammation, often mediated by circulating immune complexes (ICs). How such stimuli access joints and trigger inflammation is unclear. Whole-mount synovial imaging revealed PV1+ fenestrated capillaries at the periphery of the synovium in the lining-sublining interface. Circulating ICs extravasated from these PV1+ capillaries, and nociceptor neurons and three distinct macrophage subsets formed a sentinel unit around them. Macrophages showed subset-specific responses to systemic IC challenge; LYVE1+CX3CR1+ macrophages orchestrated neutrophil recruitment and activated calcitonin gene-related peptide+ (CGRP+) nociceptor neurons via interleukin-1β. In contrast, major histocompatibility complex class II+CD11c+ (MHCII+CD11c+) and MHCII+CD11c- interstitial macrophages formed tight clusters around PV1+ capillaries in response to systemic immune stimuli, a feature enhanced by nociceptor-derived CGRP. Altogether, we identify the anatomical location of synovial PV1+ capillaries and subset-specific macrophage-nociceptor cross-talk that forms a blood-joint barrier protecting the synovium from circulating immune challenges.
Collapse
Affiliation(s)
- Tetsuo Hasegawa
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK.
| | - Colin Y C Lee
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Andrew J Hotchen
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Aaron Fleming
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Rahul Singh
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Kunimichi Suzuki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Mark A Birch
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Andrew W McCaskie
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Krisztina Tóth
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Nathan Richoz
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Menna R Clatworthy
- Molecular Immunity Unit, University of Cambridge, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
81
|
Cheng X, Meng X, Chen R, Song Z, Li S, Wei S, Lv H, Zhang S, Tang H, Jiang Y, Zhang R. The molecular subtypes of autoimmune diseases. Comput Struct Biotechnol J 2024; 23:1348-1363. [PMID: 38596313 PMCID: PMC11001648 DOI: 10.1016/j.csbj.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by their complexity and a wide range of clinical differences. Despite patients presenting with similar symptoms and disease patterns, their reactions to treatments may vary. The current approach of personalized medicine, which relies on molecular data, is seen as an effective method to address the variability in these diseases. This review examined the pathologic classification of ADs, such as multiple sclerosis and lupus nephritis, over time. Acknowledging the limitations inherent in pathologic classification, the focus shifted to molecular classification to achieve a deeper insight into disease heterogeneity. The study outlined the established methods and findings from the molecular classification of ADs, categorizing systemic lupus erythematosus (SLE) into four subtypes, inflammatory bowel disease (IBD) into two, rheumatoid arthritis (RA) into three, and multiple sclerosis (MS) into a single subtype. It was observed that the high inflammation subtype of IBD, the RA inflammation subtype, and the MS "inflammation & EGF" subtype share similarities. These subtypes all display a consistent pattern of inflammation that is primarily driven by the activation of the JAK-STAT pathway, with the effective drugs being those that target this signaling pathway. Additionally, by identifying markers that are uniquely associated with the various subtypes within the same disease, the study was able to describe the differences between subtypes in detail. The findings are expected to contribute to the development of personalized treatment plans for patients and establish a strong basis for tailored approaches to treating autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Zerun Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuai Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuhao Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hao Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
82
|
Tokuhiro T, Matsumae G, Endo T, Ogawa Y, Ogawa T, Liyile C, Nishida Y, Alhasan H, Kobayashi H, Ebata T, Shimizu T, Takahashi D, Onodera T, Kadoya K, Terkawi MA, Iwasaki N. Cellular communication network factor 3 contributes to the pathological process of rheumatoid arthritis through promoting cell senescence and osteoclastogenesis in the joint. J Autoimmun 2024; 149:103334. [PMID: 39549484 DOI: 10.1016/j.jaut.2024.103334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/21/2024] [Accepted: 11/03/2024] [Indexed: 11/18/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic and autoimmune disease that primarily affects joints and causes pain, stiffness and swelling. The affected joints exhibit severe inflammation in the synovium and bone erosion, leading to joint deformity. Aging is an important factor facilitating the development of RA, as it is associated with an increase in the number of senescent cells and the production of the autoantibodies and proinflammatory cytokines in tissues. Given that CCN3 is highly expressed in RA joints and that its level is associated with the severity of the disease, we explored its molecular function in joints and therapeutic potential in RA. An analysis of public scRNA-seq data from the RA synovium revealed that CCN3 is expressed by an inflammatory fibroblast subset. Interestingly, stimulation with CCN3 resulted in the activation of the senescence pathway in synoviocytes and osteoclast differentiation in monocytes in vitro. Consistent with these results, the administration of CCN3 into the knee joint and onto the calvarial bone resulted in increased numbers of senescent synoviocytes in the joint and osteoclasts in the bone, respectively. Furthermore, the therapeutic potential of targeting CCN3 was evaluated in an experimental RA model. Administration of the CCN3 antibody significantly suppressed inflammation and osteoclast numbers in the joints of the RA model mice. Our findings suggest that CCN3 contributes to pathological processes in RA and represents a promising therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Taiki Tokuhiro
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Gen Matsumae
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Tsutomu Endo
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yuki Ogawa
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takuya Ogawa
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Chen Liyile
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yoshio Nishida
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hend Alhasan
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hideyuki Kobayashi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Taku Ebata
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Tomohiro Shimizu
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Daisuke Takahashi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Tomohiro Onodera
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Ken Kadoya
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - M Alaa Terkawi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
83
|
Schmidkonz C, Kuwert T, Götz TI, Ramming A, Atzinger A. Recent advances in nuclear medicine and their role in inflammatory arthritis: focus on the emerging role of FAPI PET/CT. Skeletal Radiol 2024:10.1007/s00256-024-04834-w. [PMID: 39586916 DOI: 10.1007/s00256-024-04834-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 11/27/2024]
Abstract
Imaging molecular processes associated with inflammatory disease has been revolutionized by hybrid imaging using positron emission tomography/computed tomography (PET/CT). PET/CT visualizes metabolic activity as well as protein expression and provides a comprehensive whole-body evaluation. It has the potential to reveal inflammation prior to detection of structural changes in inflammatory joint diseases. FAP is a type II transmembrane glycoprotein overexpressed not only in the stroma of tumors but also in the fibrotic processes of certain immune-mediated disorders. The recent introduction of fibroblast activation protein inhibitors (FAPI) labeled by positron emitters and thus suitable for PET/CT allows to investigate FAP expression in vivo. This review will focus on the use of FAPI-PET/CT for the diagnosis and evaluation of treatment response in inflammatory joint diseases.
Collapse
Affiliation(s)
- Christian Schmidkonz
- Department of Nuclear Medicine, Erlangen University Hospital, Erlangen, Germany.
- Institute for Medical Engineering, Ostbayerische Technische Hochschule Amberg-Weiden, Amberg, Germany.
| | - Torsten Kuwert
- Department of Nuclear Medicine, Erlangen University Hospital, Erlangen, Germany
| | - Theresa Ida Götz
- Institute for Medical Engineering, Ostbayerische Technische Hochschule Amberg-Weiden, Amberg, Germany
- Department of Internal Medicine 3, Rheumatology and Immunology, Erlangen University Hospital, Erlangen, Germany
| | - Andreas Ramming
- Department of Internal Medicine 3, Rheumatology and Immunology, Erlangen University Hospital, Erlangen, Germany
- Deutsches Zentrum Fuer Immuntherapie (DZI), Erlangen University Hospital, Erlangen, Germany
| | - Armin Atzinger
- Department of Nuclear Medicine, Erlangen University Hospital, Erlangen, Germany
| |
Collapse
|
84
|
Kim WS, Prasongyuenyong K, Ko A, Debnath R, Chen Z, Zhou JX, Shaaf E, Ko KI. ICAM1 + gingival fibroblasts modulate periodontal inflammation to mitigate bone loss. Front Immunol 2024; 15:1484483. [PMID: 39650645 PMCID: PMC11621011 DOI: 10.3389/fimmu.2024.1484483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Tissue-resident fibroblasts are heterogeneous and provide an endogenous source of cytokines that regulate immunologic events in many osteolytic diseases. Identifying distinct inflammatory fibroblast subsets and conducting mechanistic in vivo studies are critical for understanding disease pathogenesis and precision therapeutics, which is poorly explored in periodontitis. Here, we surveyed published single-cell datasets for fibroblast-specific analysis and show that Intercellular Adhesion Molecule-1 (ICAM1) expression selectively defines a fibroblast subset that exhibits an inflammatory transcriptional profile associated with nuclear factor-κB (NF-κB) pathway. ICAM1+ fibroblasts expand in both human periodontitis and murine ligature-induced periodontitis model, which have upregulated expression of CCL2 and CXCL1 compared to other fibroblast populations. Using a mouse model to selectively target gingival stromal cells, we further show that disruption of an inflammatory pathway by inhibiting transcriptional activity of NF-κB in these cells accelerated periodontal bone loss. Mechanistically, this was linked to a reduction of CCL2 expression by the ICAM1+ fibroblasts, leading to impaired macrophage recruitment and efferocytosis that was associated with persistent neutrophilic inflammation. These results may have a significant therapeutic implication as ICAM1+ gingival fibroblasts exert a protective response by regulating innate immune responses that are needed for the controlled inflammatory events in early stages of periodontitis.
Collapse
Affiliation(s)
- William S. Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kawintip Prasongyuenyong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Annette Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rahul Debnath
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhaoxu Chen
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jonathan X. Zhou
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emon Shaaf
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kang I. Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
85
|
Eiers AK, Vettorazzi S, Tuckermann JP. Journey through discovery of 75 years glucocorticoids: evolution of our knowledge of glucocorticoid receptor mechanisms in rheumatic diseases. Ann Rheum Dis 2024; 83:1603-1613. [PMID: 39107081 DOI: 10.1136/ard-2023-225371] [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: 05/05/2024] [Accepted: 07/20/2024] [Indexed: 08/09/2024]
Abstract
For three-quarters of a century, glucocorticoids (GCs) have been used to treat rheumatic and autoimmune diseases. Over these 75 years, our understanding of GCs binding to nuclear receptors, mainly the glucocorticoid receptor (GR) and their molecular mechanisms has changed dramatically. Initially, in the late 1950s, GCs were considered important regulators of energy metabolism. By the 1970s/1980s, they were characterised as ligands for hormone-inducible transcription factors that regulate many aspects of cell biology and physiology. More recently, their impact on cellular metabolism has been rediscovered. Our understanding of cell-type-specific GC actions and the crosstalk between various immune and stromal cells in arthritis models has evolved by investigating conditional GR mutant mice using the Cre/LoxP system. A major achievement in studying the complex, cell-type-specific interplay is the recent advent of omics technologies at single-cell resolution, which will provide further unprecedented insights into the cell types and factors mediating GC responses. Alongside gene-encoded factors, anti-inflammatory metabolites that participate in resolving inflammation by GCs during arthritis are just being uncovered. The translation of this knowledge into therapeutic concepts will help tackle inflammatory diseases and reduce side effects. In this review, we describe major milestones in preclinical research that led to our current understanding of GC and GR action 75 years after the first use of GCs in arthritis.
Collapse
Affiliation(s)
- Ann-Kathrin Eiers
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Baden-Württemberg, Germany
| | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Baden-Württemberg, Germany
| | - Jan P Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Baden-Württemberg, Germany
| |
Collapse
|
86
|
Faust HJ, Cheng TY, Korsunsky I, Watts GFM, Gal-Oz ST, Trim WV, Kongthong S, Jonsson AH, Simmons DP, Zhang F, Padera R, Chubinskaya S, Wei K, Raychaudhuri S, Lynch L, Moody DB, Brenner MB. Adipocyte associated glucocorticoid signaling regulates normal fibroblast function which is lost in inflammatory arthritis. Nat Commun 2024; 15:9859. [PMID: 39543086 PMCID: PMC11564742 DOI: 10.1038/s41467-024-52586-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/12/2024] [Indexed: 11/17/2024] Open
Abstract
Fibroblasts play critical roles in tissue homeostasis, but in pathologic states they can drive fibrosis, inflammation, and tissue destruction. Little is known about what regulates the homeostatic functions of fibroblasts. Here, we perform RNA sequencing and identify a gene expression program in healthy synovial fibroblasts characterized by enhanced fatty acid metabolism and lipid transport. We identify cortisol as the key driver of the healthy fibroblast phenotype and that depletion of adipocytes, which express high levels of Hsd11b1, results in loss of the healthy fibroblast phenotype in mouse synovium. Additionally, fibroblast-specific glucocorticoid receptor Nr3c1 deletion in vivo leads to worsened arthritis. Cortisol signaling in fibroblasts mitigates matrix remodeling induced by TNF and TGF-β1 in vitro, while stimulation with these cytokines represses cortisol signaling and adipogenesis. Together, these findings demonstrate the importance of adipocytes and cortisol signaling in driving the healthy synovial fibroblast state that is lost in disease.
Collapse
Affiliation(s)
- Heather J Faust
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tan-Yun Cheng
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ilya Korsunsky
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Gerald F M Watts
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shani T Gal-Oz
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - William V Trim
- Department of Endocrinology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suppawat Kongthong
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna Helena Jonsson
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Daimon P Simmons
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Fan Zhang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology and the Center for Health Artificial Intelligence, University of Colorado School of Medicine, Aurora, CO, USA
| | - Robert Padera
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- Department of Endocrinology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - D Branch Moody
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
87
|
Figueiredo ML. Applications of single-cell RNA sequencing in rheumatoid arthritis. Front Immunol 2024; 15:1491318. [PMID: 39600707 PMCID: PMC11588722 DOI: 10.3389/fimmu.2024.1491318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Single cell RNA sequencing (scRNA-seq) is a relatively new technology that provides an unprecedented, detailed view of cellular heterogeneity and function by delineating the transcriptomic difference among individual cells. This will allow for mapping of cell-type-specific signaling during physiological and pathological processes, to build highly specific models of cellular signaling networks between the many discrete clusters that are present. This technology therefore provides a powerful approach to dissecting the cellular and molecular mechanisms that contribute to autoimmune diseases, including rheumatoid arthritis (RA). scRNA-seq can offer valuable insights into RA unique cellular states and transitions, potentially enabling development of novel drug targets. However, some challenges that still limit its mainstream utilization and include higher costs, a lower sensitivity for low-abundance transcripts, and a relatively complex data analysis workflow relative to bulk or traditional RNA-seq. This minireview explores the emerging application of scRNA-seq in RA research, highlighting its role in producing important insights that can help pave the way for innovative and more effective therapeutic strategies.
Collapse
Affiliation(s)
- Marxa L. Figueiredo
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue
University, West Lafayette, IN, United States
| |
Collapse
|
88
|
Venken K, Jarlborg M, Stevenaert F, Malfait TLA, Vlieghe C, Abraham Y, Manuello T, Decruy T, Vanhee S, Wils H, Peeters PJ, Carron P, Van den Bosch F, Van Tendeloo V, Lambrecht BN, Wittoek R, Jacques P, Elewaut D. Shared lung and joint T cell repertoire in early rheumatoid arthritis driven by cigarette smoking. Ann Rheum Dis 2024:ard-2024-226284. [PMID: 39521450 DOI: 10.1136/ard-2024-226284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Smoking has been associated with an increased risk of developing rheumatoid arthritis (RA) in individuals carrying shared epitope (SE) HLA-DRB1 alleles. Yet, little is known about the regional and systemic T cell dynamics of smoking and a potential link to T cell infiltration in inflamed synovia. In this study, we, therefore, sought to study T cell features in lung and inflamed joints in smoking versus non-smoking patients. METHODS We set up a framework to monitor T cells in paired bronchoalveolar lavage fluid, blood and inflamed synovium tissue samples from 17 new-onset treatment naïve anticitrullinated protein antibody+RA patients. T cell receptor (TCR) repertoire of index-sorted tissue residing in T cells was determined by single-cell TCR sequencing coupled with deep immunophenotyping. RESULTS A significant enrichment of CD4+ and CD8+ T cells was seen in synovial samples from smoking versus non-smoking patients, along with an increase in expanded T cell clonotypes. This was particularly pronounced among SE+smokers, suggestive of a synergic gene-smoke effect. Strikingly, identical TCR clonalities were present in matched lung and joint samples of RA smokers, the majority being also detectable in circulation. This was mirrored by an increased clustering of lung and synovium TCRs across patients, suggesting a shared specificity by conserved motifs. The lung-joint shared T cell clonotypes showed a restricted TCR gene usage and exhibited a particular 4-1BB+CD57 hi effector profile within the inflamed synovium. CONCLUSION The data indicate a profound interplay between a strong MHC predisposition, smoking and induction of autoimmunity by shaping the TCR repertoire.
Collapse
Affiliation(s)
- Koen Venken
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | - Matthias Jarlborg
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | | | - Thomas L A Malfait
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Department of Respiratory Medicine, University Hospital Ghent, Gent, Belgium
| | - Carolien Vlieghe
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | - Yann Abraham
- Janssen Research and Development, Beerse, Belgium
| | - Teddy Manuello
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | - Tine Decruy
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | - Stijn Vanhee
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Zwijnaarde, Belgium
- Department of Head and Skin, Ghent University Hospital, Ghent, Belgium
| | - Hans Wils
- Janssen Research and Development, Beerse, Belgium
| | | | - Philippe Carron
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | - Filip Van den Bosch
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | | | - Bart N Lambrecht
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Department of Respiratory Medicine, University Hospital Ghent, Gent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Zwijnaarde, Belgium
| | - Ruth Wittoek
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | - Peggy Jacques
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| | - Dirk Elewaut
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics, Ghent University, Gent, Belgium
- Molecular Immunology and Inflammation Unit, VIB-UGent Center for Inflammation Research, Zwijnaarde, Belgium
| |
Collapse
|
89
|
Xu L, Wang H, Sun C, Zhao Q, Wang L, Yan Q, Wang J, Lin N, Liu C. GZMK Facilitates Experimental Rheumatoid Arthritis Progression by Interacting with CCL5 and Activating the ERK Signaling. Inflammation 2024:10.1007/s10753-024-02166-4. [PMID: 39489858 DOI: 10.1007/s10753-024-02166-4] [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: 06/13/2024] [Revised: 10/07/2024] [Accepted: 10/13/2024] [Indexed: 11/05/2024]
Abstract
Synovial over-proliferation is a key event in the progression of rheumatoid arthritis (RA) disease. Ferroptosis may be essential for maintaining the balance between synovial proliferation and death. This study aimed to investigate the molecular mechanisms mediating the activation and ferroptosis of collagen-induced arthritis (CIA)-synovial fibroblasts (SFs). Differentially expressed genes (DEGs) in the synovial tissues of CIA rats and normal rats were screened through sequencing. The GSE115662 dataset from the GEO database was analyzed and screened for DEGs. The viability, proliferation, migration, invasion, cell cycle, and apoptosis of CIA-SFs were analyzed by cell counting kit-8, 5-ethynyl-2'-deoxyuridine, flow cytometry, transwell migration, and invasion assays. The ferroptosis of CIA-SFs was assessed using matching reagent kits to detect indicators like reactive oxygen species, ferrous iron, malondialdehyde, glutathione, and superoxide dismutase. The interaction between Granzyme K (GZMK) and C-C motif chemokine 5 (CCL5) was determined by coimmunoprecipitation assay. We found abnormal GZMK expression in the GSE115662 database and mRNA sequencing data. GZMK was overexpressed in CIA-SFs, and GZMK promoted cell proliferation, migration, invasion, inflammation, and decreased cell apoptosis and ferroptosis in CIA-SFs. GZMK could interact with CCL5 to activate the ERK signaling. GZMK and CCL5 knockdown improved by reducing arthritis scores, redness and swelling of paws, and pathological changes in joint synovium of CIA rats. CCL5 overexpression reversed the effects of GZMK silencing on CIA-SFs cell proliferation, migration, invasion, apoptosis, and ferroptosis. We confirmed that GZMK accelerated experimental rheumatoid arthritis progression by interacting with CCL5 and activating the ERK signaling.
Collapse
Affiliation(s)
- Liting Xu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Hui Wang
- Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China
| | - Congcong Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Qingyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Lili Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Qianqian Yan
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Jialin Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Na Lin
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| | - Chunfang Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| |
Collapse
|
90
|
Rutter-Locher Z, Kirkham BW, Bannister K, Bennett DL, Buckley CD, Taams LS, Denk F. An interdisciplinary perspective on peripheral drivers of pain in rheumatoid arthritis. Nat Rev Rheumatol 2024; 20:671-682. [PMID: 39242949 DOI: 10.1038/s41584-024-01155-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/09/2024]
Abstract
Pain is one of the most debilitating symptoms of rheumatoid arthritis (RA), and yet remains poorly understood, especially when pain occurs in the absence of synovitis. Without active inflammation, experts most often attribute joint pain to central nervous system dysfunction. However, advances in the past 5 years in both immunology and neuroscience research suggest that chronic pain in RA is also driven by a variety of abnormal interactions between peripheral neurons and mediators produced by resident cells in the local joint environment. In this Review, we discuss these novel insights from an interdisciplinary neuro-immune perspective. We outline a potential working model for the peripheral drivers of pain in RA, which includes autoantibodies, resident immune and mesenchymal cells and their interactions with different subtypes of peripheral sensory neurons. We also offer suggestions for how future collaborative research could be designed to accelerate analgesic drug development.
Collapse
Affiliation(s)
- Zoe Rutter-Locher
- Department of Rheumatology, Guy's Hospital, London, UK
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | | | - Kirsty Bannister
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London, London, UK
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Leonie S Taams
- Centre for Inflammation Biology & Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, London, UK.
| | - Franziska Denk
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London, London, UK.
| |
Collapse
|
91
|
Thomas T, Friedrich M, Rich-Griffin C, Pohin M, Agarwal D, Pakpoor J, Lee C, Tandon R, Rendek A, Aschenbrenner D, Jainarayanan A, Voda A, Siu JHY, Sanches-Peres R, Nee E, Sathananthan D, Kotliar D, Todd P, Kiourlappou M, Gartner L, Ilott N, Issa F, Hester J, Turner J, Nayar S, Mackerodt J, Zhang F, Jonsson A, Brenner M, Raychaudhuri S, Kulicke R, Ramsdell D, Stransky N, Pagliarini R, Bielecki P, Spies N, Marsden B, Taylor S, Wagner A, Klenerman P, Walsh A, Coles M, Jostins-Dean L, Powrie FM, Filer A, Travis S, Uhlig HH, Dendrou CA, Buckley CD. A longitudinal single-cell atlas of anti-tumour necrosis factor treatment in inflammatory bowel disease. Nat Immunol 2024; 25:2152-2165. [PMID: 39438660 PMCID: PMC11519010 DOI: 10.1038/s41590-024-01994-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Precision medicine in immune-mediated inflammatory diseases (IMIDs) requires a cellular understanding of treatment response. We describe a therapeutic atlas for Crohn's disease (CD) and ulcerative colitis (UC) following adalimumab, an anti-tumour necrosis factor (anti-TNF) treatment. We generated ~1 million single-cell transcriptomes, organised into 109 cell states, from 216 gut biopsies (41 subjects), revealing disease-specific differences. A systems biology-spatial analysis identified granuloma signatures in CD and interferon (IFN)-response signatures localising to T cell aggregates and epithelial damage in CD and UC. Pretreatment differences in epithelial and myeloid compartments were associated with remission outcomes in both diseases. Longitudinal comparisons demonstrated disease progression in nonremission: myeloid and T cell perturbations in CD and increased multi-cellular IFN signalling in UC. IFN signalling was also observed in rheumatoid arthritis (RA) synovium with a lymphoid pathotype. Our therapeutic atlas represents the largest cellular census of perturbation with the most common biologic treatment, anti-TNF, across multiple inflammatory diseases.
Collapse
Affiliation(s)
- Tom Thomas
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Centre for Human Genetics, University of Oxford, Oxford, UK
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Matthias Friedrich
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | | | - Mathilde Pohin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Devika Agarwal
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Julia Pakpoor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Centre for Human Genetics, University of Oxford, Oxford, UK
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Carl Lee
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Ruchi Tandon
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Aniko Rendek
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Dominik Aschenbrenner
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | | | - Alexandru Voda
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | | | - Eloise Nee
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Dharshan Sathananthan
- University of Adelaide, Adelaide, Australia
- Lyell McEwin Hospital, Adelaide, Australia
| | - Dylan Kotliar
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter Todd
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Lisa Gartner
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Nicholas Ilott
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Jason Turner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and NIHR Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Jonas Mackerodt
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fan Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Center for Health AI, University of Colorado Anschutz, Anschutz, CO, USA
| | - Anna Jonsson
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Brenner
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Soumya Raychaudhuri
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | - Noah Spies
- Celsius Therapeutics, Cambridge, MA, USA
| | - Brian Marsden
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Stephen Taylor
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Allon Wagner
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA
- The Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Paul Klenerman
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Alissa Walsh
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Mark Coles
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Fiona M Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Filer
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and NIHR Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Simon Travis
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Holm H Uhlig
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Calliope A Dendrou
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Centre for Human Genetics, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Christopher D Buckley
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK.
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
92
|
Rocha PS, Silva AA, Queiroz-Junior CM, Braga AD, Moreira TP, Teixeira MM, Amaral FA. Trained immunity of synovial macrophages is associated with exacerbated joint inflammation and damage after Staphylococcus aureus infection. Inflamm Res 2024; 73:1995-2008. [PMID: 39340660 DOI: 10.1007/s00011-024-01946-w] [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: 05/09/2024] [Revised: 08/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
OBJECTIVES Investigate whether and which synoviocytes would acquire trained immunity characteristics that could exacerbate joint inflammation following a secondary Staphylococcus aureus infection. METHODS Lipopolysaccharide (LPS) and S. aureus were separately or double injected (21 days of interval) into the tibiofemoral joint cavity of male C57BL/6 mice. At different time points after these stimulations, mechanical nociception was analyzed followed by the analysis of signs of inflammation and damage in the affected joints. The trained immunity markers, including the glycolytic and mTOR pathway, were analyzed in whole tissue or isolated synoviocytes. A group of mice was treated with Rapamycin, an mTOR inhibitor before LPS or S. aureus stimulation. RESULTS The double LPS - S. aureus hit promoted intense joint inflammation and damage compared to single joint stimulation, including markers in synoviocyte activation, production of proinflammatory cytokines, persistent nociception, and bone damage, despite not reducing the bacterial clearance. The double LPS - S. aureus hit joints increased the synovial macrophage population expressing CX3CR1 alongside triggering established epigenetic modifications associated with trained immunity events in these cells, such as the upregulation of the mTOR signaling pathway (p-mTOR and HIF1α) and the trimethylation of histone H3. Mice treated with Rapamycin presented reduced CX3CR1+ macrophage activation, joint inflammation, and bone damage. CONCLUSIONS There is a trained immunity phenotype in CX3CR1+ synovial macrophages that contributes to the exacerbation of joint inflammation and damage during septic arthritis caused by S. aureus.
Collapse
Affiliation(s)
- Peter Silva Rocha
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Adryan Aparecido Silva
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Amanda Dias Braga
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Thaiane Pinto Moreira
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil
| | - Flávio Almeida Amaral
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brazil.
| |
Collapse
|
93
|
Matsuoka T, Yashiro M. Molecular Mechanism for Malignant Progression of Gastric Cancer Within the Tumor Microenvironment. Int J Mol Sci 2024; 25:11735. [PMID: 39519285 PMCID: PMC11546171 DOI: 10.3390/ijms252111735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide. Most patients are diagnosed at the progressive stage of GC, and progress in the development of effective anti-GC drugs has been insufficient. The tumor microenvironment (TME) regulates various functions of tumor cells, and interactions between the cellular and molecular components of the TME-e.g., inflammatory cells, fibroblasts, vasculature cells, and innate and adaptive immune cells-promote the aggressiveness of cancer cells and dissemination to distant organs. This review summarizes the roles of various TME cells and molecules in regulating the malignant progression and metastasis of GC. We also address the important roles of signaling pathways in mediating the interaction between cancer cells and the different components of the GC TME. Finally, we discuss the implications of these molecular mechanisms for developing novel and effective therapies targeting molecular and cellular components of the GC TME to control the malignant progression of GC.
Collapse
Affiliation(s)
- Tasuku Matsuoka
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| |
Collapse
|
94
|
Rius Rigau A, Liang M, Devakumar V, Neelagar R, Matei AE, Györfi AH, Bergmann C, Filla T, Fedorchenko V, Schett G, Distler JHW, Li YN. Imaging mass cytometry-based characterisation of fibroblast subsets and their cellular niches in systemic sclerosis. Ann Rheum Dis 2024:ard-2024-226336. [PMID: 39442983 DOI: 10.1136/ard-2024-226336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES Transcriptomic data demonstrated that fibroblasts are heterogeneous with functionally diverse subpopulations. Although fibroblasts are key effector cells of fibrotic diseases such as systemic sclerosis (SSc), they have not yet been characterised spatially at the cellular level. Here, we aimed to investigate fibroblast subpopulations using imaging mass cytometry (IMC) as a proteomic-based, spatially resolved omics approach. METHODS We applied IMC to deconvolute the heterogeneity of 49 969 cells including 6501 fibroblasts at the single-cell level, to analyse their spatial distribution and to characterise their cellular niches in skin sections of patients with SSc and controls in situ. RESULTS We identified 13 different subpopulations of fibroblasts in SSc and control skin, the proportion increases in five fibroblast subpopulations (myofibroblasts, FAPhigh, S1PR+, Thy1+;ADAM12high;PU.1high and ADAM12+;GLI1+ fibroblasts) and decreases in three subpopulations (TFAMhigh, PI16+;FAP+ and Thy1+;ADAM12low fibroblasts). Several fibroblast subpopulations demonstrated spatial enrichment and altered cellular interactions in SSc. The proportion of S1PR+-fibroblast positively correlated with more extensive skin fibrosis, whereas high numbers of PI16+;FAP--fibroblasts were associated with milder skin fibrosis. The frequency of aberrant cellular interaction between S1PR+ and ADAM12+;GLI1+-fibroblasts also positively associated with the extent of skin fibrosis in SSc. CONCLUSION Using IMC, we demonstrated profound changes in composition and localisation of the majority of fibroblast subpopulations in SSc skin. These findings may provide a rationale for specific targeting of deregulated fibroblast subpopulations in SSc. Quantification of S1PR+-fibroblast and PI16+;FAP--fibroblasts may offer potential for patient stratification according to severity of skin fibrosis.
Collapse
Affiliation(s)
- Aleix Rius Rigau
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen University Hospital, Erlangen, Bayern, Germany
| | - Minrui Liang
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Huashan Hospital Fudan University, Shanghai, China
| | - Veda Devakumar
- Department of Rheumatology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
- Hiller Research Center, University Hospital of Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
| | - Ranjana Neelagar
- Department of Rheumatology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
- Hiller Research Center, University Hospital of Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
| | - Alexandru-Emil Matei
- Department of Rheumatology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
- Hiller Research Center, University Hospital of Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
| | - Andrea-Hermina Györfi
- Department of Rheumatology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
- Hiller Research Center, University Hospital of Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen University Hospital, Erlangen, Bayern, Germany
| | - Tim Filla
- Department of Rheumatology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
- Hiller Research Center, University Hospital of Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
| | - Vladyslav Fedorchenko
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen University Hospital, Erlangen, Bayern, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen University Hospital, Erlangen, Bayern, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Bayern, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen University Hospital, Erlangen, Bayern, Germany
- Department of Rheumatology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
- Hiller Research Center, University Hospital of Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
| | - Yi-Nan Li
- Department of Rheumatology, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
- Hiller Research Center, University Hospital of Düsseldorf, Düsseldorf, Nordrhein-Westfalen, Germany
| |
Collapse
|
95
|
Ammons DT, Chow L, Goodrich L, Bass L, Larson B, Williams ZJ, Stoneback JW, Dow S, Pezzanite LM. Characterization of the single cell landscape in normal and osteoarthritic equine joints. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:88. [PMID: 39507442 PMCID: PMC11534742 DOI: 10.21037/atm-24-40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/30/2024] [Indexed: 11/08/2024]
Abstract
Background Osteoarthritis (OA) is a major source of pain and disability worldwide. Understanding of disease progression is evolving, but OA is increasingly thought to be a multifactorial disease in which the innate immune system plays a role in regulating and perpetuating low-grade inflammation. The aim of this study was to enhance our understanding of OA immunopathogenesis through characterization of the transcriptomic responses in OA joints, with the goal to facilitate the development of targeted therapies. Methods Single-cell RNA sequencing (scRNA-seq) was completed on cells isolated from the synovial fluid of three normal and three OA equine joints. In addition to synovial fluid, scRNA-seq was also performed on synovium from one normal joint and one OA joint. Results Characterization of 28,639 cells isolated from normal and OA-affected equine synovial fluid revealed the composition to be entirely immune cells (CD45+) with 8 major populations and 26 subpopulations identified. In synovial fluid, we found myeloid cells (macrophage and dendritic cells) to be overrepresented and T cells (CD4 and CD8) to be underrepresented in OA relative to normal joints. Through subcluster and differential abundance analysis of T cells we further identified a relative overrepresentation of IL23R+ gamma-delta (γδ) T cells in OA-affected joints (a cell type we report to be enriched in gene signatures associated with T helper 17 mediated immunity). Analysis of an additional 17,690 cells (11 distinct cell type clusters) obtained from synovium of one horse led to the identification of an OA-associated reduction in the relative abundance of synovial macrophages, which contrasts with the increased relative abundance of macrophages in synovial fluid. Completion of cell-cell interaction analysis implicated myeloid cells in disease progression, suggesting that the myeloid-myeloid interactions were increased in OA-affected joints. Conclusions Overall, this work provides key insights into the composition of equine synovial fluid and synovium in health and OA. The data generated in this study provides equine-specific cell type gene signatures which can be applied to future investigations. Furthermore, our analysis highlights the potential role of macrophages and IL23R+ γδ T cells in OA immunopathogenesis.
Collapse
Affiliation(s)
- Dylan T Ammons
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Laurie Goodrich
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Luke Bass
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Blaine Larson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Zoë J Williams
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jason W Stoneback
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Steven Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Lynn M Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
96
|
Bergström B, Selldén T, Bollmann M, Svensson MND, Ekwall AKH. Methotrexate promotes the release of granulocyte-macrophage colony-stimulating factor from rheumatoid arthritis fibroblast-like synoviocytes via autocrine interleukin-1 signaling. Arthritis Res Ther 2024; 26:178. [PMID: 39394168 PMCID: PMC11468154 DOI: 10.1186/s13075-024-03406-6] [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: 05/01/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Activated fibroblast-like synoviocytes (FLS) are drivers of synovitis and structural joint damage in rheumatoid arthritis (RA). Despite the use of disease-modifying drugs, only about 50% of RA patients reach remission in real-world settings. We used an unbiased approach to investigate the effects of standard-of-care methotrexate (MTX) and a Janus kinase inhibitor, tofacitinib (TOFA), on gene expression in RA-FLS, in order to identify untargeted disease mediators. METHODS Primary RA-FLS were activated by stimulation with interleukin-1β (IL-1β) or platelet-derived growth factor + IL-1β in the presence or absence of MTX or TOFA, with or without additional inhibitors. Co-cultures of synovial cells were performed in direct and indirect systems. Cells were collected for RNA sequencing or qPCR, and supernatants were analyzed for protein concentrations. RESULTS Six thousand three hundred fifty genes were differentially expressed, the majority being upregulated, in MTX-treated activated RA-FLS and 970 genes, the majority being downregulated, in TOFA-treated samples. Pathway analysis showed that MTX had largest effects on 'Molecular mechanisms of cancer' and TOFA on 'Interferon signaling'. Targeted analysis of disease-associated genes revealed that MTX increased the expression of cell cycle-regulating genes but also of pro-inflammatory mediators like IL-1α (IL1A) and granulocyte-macrophage colony-stimulating factor, GM-CSF (CSF2). The MTX-promoted expression of CSF2 in activated RA-FLS peaked at 48 h, could be mediated via either NF-κB or AP-1 transcription factors, and was abrogated by IL-1 inhibitors (IRAK4 inhibitor and anakinra). In a co-culture setting, MTX-treatment of activated RA-FLS induced IL1B expression in macrophages. CONCLUSIONS MTX treatment induces secretion of IL-1 from activated RA-FLS which by autocrine signaling augments their release of GM-CSF. This unexpected effect of MTX might contribute to the persistence of synovitis.
Collapse
Affiliation(s)
- Beatrice Bergström
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tilia Selldén
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Miriam Bollmann
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Mattias N D Svensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Hultgård Ekwall
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Rheumatology, Division 3, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
97
|
Li R, Colombo M, Wang G, Rodriguez-Romera A, Benlabiod C, Jooss NJ, O’Sullivan J, Brierley CK, Clark SA, Pérez Sáez JM, Aragón Fernández P, Schoof EM, Porse B, Meng Y, Khan AO, Wen S, Dong P, Zhou W, Sousos N, Murphy L, Clarke M, Olijnik AA, C. Wong Z, Karali CS, Sirinukunwattana K, Ryou H, Norfo R, Cheng Q, Carrelha J, Ren Z, Thongjuea S, Rathinam VA, Krishnan A, Royston D, Rabinovich GA, Mead AJ, Psaila B. A proinflammatory stem cell niche drives myelofibrosis through a targetable galectin-1 axis. Sci Transl Med 2024; 16:eadj7552. [PMID: 39383242 PMCID: PMC7616771 DOI: 10.1126/scitranslmed.adj7552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024]
Abstract
Myeloproliferative neoplasms are stem cell-driven cancers associated with a large burden of morbidity and mortality. Most patients present with early-stage disease, but a substantial proportion progress to myelofibrosis or secondary leukemia, advanced cancers with a poor prognosis and high symptom burden. Currently, it remains difficult to predict progression, and therapies that reliably prevent or reverse fibrosis are lacking. A major bottleneck to the discovery of disease-modifying therapies has been an incomplete understanding of the interplay between perturbed cellular and molecular states. Several cell types have individually been implicated, but a comprehensive analysis of myelofibrotic bone marrow is lacking. We therefore mapped the cross-talk between bone marrow cell types in myelofibrotic bone marrow. We found that inflammation and fibrosis are orchestrated by a "quartet" of immune and stromal cell lineages, with basophils and mast cells creating a TNF signaling hub, communicating with megakaryocytes, mesenchymal stromal cells, and proinflammatory fibroblasts. We identified the β-galactoside-binding protein galectin-1 as a biomarker of progression to myelofibrosis and poor survival in multiple patient cohorts and as a promising therapeutic target, with reduced myeloproliferation and fibrosis in vitro and in vivo and improved survival after galectin-1 inhibition. In human bone marrow organoids, TNF increased galectin-1 expression, suggesting a feedback loop wherein the proinflammatory myeloproliferative neoplasm clone creates a self-reinforcing niche, fueling progression to advanced disease. This study provides a resource for studying hematopoietic cell-niche interactions, with relevance for cancer-associated inflammation and disorders of tissue fibrosis.
Collapse
Affiliation(s)
- Rong Li
- CAMS Oxford Institute; University of Oxford; Oxford, United Kingdom (UK)
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Michela Colombo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Human Technopole; Milan, Italy
| | - Guanlin Wang
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- MRC WIMM Centre for Computational Biology, University of Oxford; Oxford, United Kingdom
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
- Qizhi Institute, Shanghai, China
| | - Antonio Rodriguez-Romera
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Camelia Benlabiod
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Natalie J. Jooss
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Jennifer O’Sullivan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Charlotte K. Brierley
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Sally-Ann Clark
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Juan M. Pérez Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | | - Erwin M. Schoof
- Department of Biotechnology and Biomedicine, Technical University of Denmark; Denmark
| | - Bo Porse
- The Finsen Laboratory, Copenhagen University Hospital; Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Department of Clinical Medicine, University of Copenhagen; Copenhagen, Denmark
| | - Yiran Meng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Abdullah O. Khan
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Sean Wen
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Pengwei Dong
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Wenjiang Zhou
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology; Fudan University, Shanghai, China
| | - Nikolaos Sousos
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Lauren Murphy
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Matthew Clarke
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Aude-Anais Olijnik
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Zoë C. Wong
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Christina Simoglou Karali
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Korsuk Sirinukunwattana
- Oxford Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford; Oxford, UK
| | - Hosuk Ryou
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford; Oxford, UK
| | - Ruggiero Norfo
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Qian Cheng
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Joana Carrelha
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford; Oxford, UK
| | - Zemin Ren
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Supat Thongjuea
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
| | - Vijay A Rathinam
- Department of Immunology, University of Connecticut Health School of Medicine; Farmington, ConnecticutUSA
| | - Anandi Krishnan
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, California, USA
| | - Daniel Royston
- Biotech Research and Innovation Centre, Faculty of Health Sciences, University of Copenhagen; Denmark
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adam J Mead
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
| | - Bethan Psaila
- Medical Research Council Weatherall Institute of Molecular Medicine (MRC WIMM) and NIHR Biomedical Research Centre Hematology Theme; University of Oxford; Oxford, UK
- Oxford University Hospitals NHS Trust; Oxford, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| |
Collapse
|
98
|
Han Y, Liu C, Chen S, Sun H, Jia Z, Shi J, Wang L, Du K, Chang Y. Columbianadin ameliorates rheumatoid arthritis by attenuating synoviocyte hyperplasia through targeted vimentin to inhibit the VAV2/Rac-1 signaling pathway. J Adv Res 2024:S2090-1232(24)00432-6. [PMID: 39369957 DOI: 10.1016/j.jare.2024.09.030] [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: 07/24/2024] [Revised: 09/13/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is an autoimmune disease pathologically characterized by synovial inflammation. The abnormal activation of synoviocytes seems to accompany the progression of RA. The role and exact molecular mechanism in RA of columbianadin (CBN) which is a natural coumarin is still unclear. OBJECTIVES The present research aimed to investigate the effect of vimentin on the abnormal growth characteristics of RA synoviocytes and the targeted regulatory role of CBN. METHODS Cell migration and invasion were detected using the wound healing and transwell method. Mechanistically, the direct molecular targets of CBN were screened and identified by activity-based protein profiling. The expression of relevant proteins and mRNA in cells and mouse synovium was detected by western blotting and qRT-PCR. Changes in the degree of paw swelling and body weight of mice were recorded. H&E staining, toluidine blue staining, and micro-CT were used to visualize the degree of pathological damage in the ankle joints of mice. Small interfering RNA and plasmid overexpression of vimentin were used to observe their effects on MH7A cell proliferation, migration, apoptosis, and downstream molecular signaling. RESULTS The TNF-α-induced proliferation and migration of MH7A cells could be significantly repressed by CBN (25,50 μM), and the expression of apoptosis and autophagy-associated proteins could be modulated. Furthermore, CBN could directly bind to vimentin and inhibit its expression and function in synoviocytes, thereby ameliorating foot and paw swelling and joint damage in CIA mice. Silencing and overexpression of vimentin might be involved in developing RA synovial hyperplasia and invasive cartilage by activating VAV2 phosphorylation-mediated expression of Rac-1, which affects abnormal growth characteristics, such as synoviocyte invasion and migration. CONCLUSION CBN-targeted vimentin restrains the overactivation of RA synoviocytes thereby delaying the pathological process in CIA mice, which provides valuable targets and insights for understanding the pathological mechanisms of RA synovial hyperplasia.
Collapse
Affiliation(s)
- Yuli Han
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Changqing Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shujing Chen
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Huihui Sun
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Zhaoyu Jia
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jiaxin Shi
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lirong Wang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kunze Du
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yanxu Chang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
99
|
Huang D, Jiao X, Huang S, Liu J, Si H, Qi D, Pei X, Lu D, Wang Y, Li Z. Analysis of the heterogeneity and complexity of murine extraorbital lacrimal gland via single-cell RNA sequencing. Ocul Surf 2024; 34:60-95. [PMID: 38945476 DOI: 10.1016/j.jtos.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
PURPOSE The lacrimal gland is essential for maintaining ocular surface health and avoiding external damage by secreting an aqueous layer of the tear film. However, a healthy lacrimal gland's inventory of cell types and heterogeneity remains understudied. METHODS Here, 10X Genome-based single-cell RNA sequencing was used to generate an unbiased classification of cellular diversity in the extraorbital lacrimal gland (ELG) of C57BL/6J mice. From 43,850 high-quality cells, we produced an atlas of cell heterogeneity and defined cell types using classic marker genes. The possible functions of these cells were analyzed through bioinformatics analysis. Additionally, the CellChat was employed for a preliminary analysis of the cell-cell communication network in the ELG. RESULTS Over 37 subclasses of cells were identified, including seven types of glandular epithelial cells, three types of fibroblasts, ten types of myeloid-derived immune cells, at least eleven types of lymphoid-derived immune cells, and five types of vascular-associated cell subsets. The cell-cell communication network analysis revealed that fibroblasts and immune cells play a pivotal role in the dense intercellular communication network within the mouse ELG. CONCLUSIONS This study provides a comprehensive transcriptome atlas and related database of the mouse ELG.
Collapse
Affiliation(s)
- Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Xinwei Jiao
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Jiangman Liu
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China
| | - Yimian Wang
- Division of Medicine, Faculty of Medical Sciences, University College London, Gower Street, London, WC1E 6BT, UK
| | - Zhijie Li
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China; Henan Eye Institute, Henan Eye Hospital and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, 450000, China.
| |
Collapse
|
100
|
Rouse JR, Danner R, Wahhab A, Pereckas M, Nguyen C, McClune ME, Steere AC, Strle K, Jutras BL, Lochhead RB. HLA-DR-Expressing Fibroblast-Like Synoviocytes Are Inducible Antigen Presenting Cells That Present Autoantigens in Lyme Arthritis. ACR Open Rheumatol 2024; 6:678-689. [PMID: 39073021 PMCID: PMC11471949 DOI: 10.1002/acr2.11710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/09/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE HLA-DR-expressing fibroblast-like synoviocytes (FLS) are a prominent cell type in synovial tissue in chronic inflammatory forms of arthritis. FLS-derived extracellular matrix (ECM) proteins, including fibronectin-1 (FN1), contain immunogenic CD4+ T cell epitopes in patients with postinfectious Lyme arthritis (LA). However, the role of FLS in presentation of these T cell epitopes remains uncertain. METHODS Primary LA FLS and primary murine FLS stimulated with interferon gamma (IFNγ), Borrelia burgdorferi, and/or B burgdorferi peptidoglycan (PG) were assessed for properties associated with antigen presentation. HLA-DR-presented peptides from stimulated LA FLS were identified by immunopeptidomics analysis. OT-II T cells were co-cultured with stimulated murine FLS in the presence of cognate ovalbumin antigen to determine the potential of FLS to act as inducible antigen presenting cells (APCs). RESULTS FLS expressed HLA-DR molecules within inflamed synovial tissue and tendons from patients with postinfectious LA in situ. Major histocompatibility complex (MHC) class II and co-stimulatory molecules were expressed by FLS following in vitro stimulation with IFNγ and B burgdorferi and presented both foreign and self-MHC-II peptides, including an immunogenic T cell epitope derived from Lyme autoantigen FN1. Stimulated FLS induced proliferation of naive OT-II CD4+ T cells that were dependent on OT-II antigen and CD40. Stimulation with B burgdorferi PG enhanced FLS-mediated T cell activation. CONCLUSION MHC-II+ FLS are inducible APCs that can induce CD4+ T cell activation in an antigen- and CD40-dependent manner. Activated FLS can also present ECM-derived Lyme autoantigens, implicating FLS in amplifying tissue-localized autoimmunity in LA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Allen C. Steere
- Massachusetts General Hospital and Harvard Medical SchoolBoston
| | | | | | | |
Collapse
|