1
|
Li YY, Zhou LW, Qian FC, Fang QL, Yu ZM, Cui T, Dong FJ, Cai FH, Yu TT, Li LD, Wang QY, Zhu YB, Tang HF, Hu BY, Li CQ. scImmOmics: a manually curated resource of single-cell multi-omics immune data. Nucleic Acids Res 2025; 53:D1162-D1172. [PMID: 39494524 PMCID: PMC11701750 DOI: 10.1093/nar/gkae985] [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: 08/13/2024] [Revised: 09/30/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
Single-cell sequencing technology has enabled the discovery and characterization of subpopulations of immune cells with unique functions, which is critical for revealing immune responses under healthy or disease conditions. Efforts have been made to collect and curate single-cell RNA sequencing (scRNA-seq) data, yet an immune-specific single-cell multi-omics atlas with harmonized metadata is still lacking. Here, we present scImmOmics (https://bio.liclab.net/scImmOmics/home), a manually curated single-cell multi-omics immune database constructed based on high-quality immune cells with known immune cell labels. Currently, scImmOmics documents >2.9 million cell-type labeled immune cells derived from seven single-cell sequencing technologies, involving 131 immune cell types, 47 tissues and 4 species. To ensure data consistency, we standardized the nomenclature of immune cell types and presented them in a hierarchical tree structure to clearly describe the lineage relationships within the immune system. scImmOmics also provides comprehensive immune regulatory information, including T-cell/B-cell receptor sequencing clonotype information, cell-specific regulatory information (e.g. gene/chromatin accessibility/protein/transcription factor states within known cell types, cell-to-cell communication and co-expression networks) and immune cell responses to cytokines. Collectively, scImmOmics is a comprehensive and valuable platform for unraveling the heterogeneity and diversity of immune cells and elucidating the specific regulatory mechanisms at the single-cell level.
Collapse
Affiliation(s)
- Yan-Yu Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan 421001, China
| | - Li-Wei Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng-Cui Qian
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Qiao-Li Fang
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Zheng-Min Yu
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Ting Cui
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Fu-Juan Dong
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Fu-Hong Cai
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Ting-Ting Yu
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Li-Dong Li
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Qiu-Yu Wang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Yan-Bing Zhu
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Hui-Fang Tang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan 421001, China
| | - Bao-Yang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chun-Quan Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics and Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Myocardial Injury in Hunan Province, Hengyang, Hunan 421001, China
| |
Collapse
|
2
|
Zhou S, Ju S, Li X, Ruan C, Dong Z. Revealing the roles of IL-7R in abdominal aortic aneurysm through integrated analysis of single-cell RNA-seq and bulk RNA-seq. Biochem Biophys Res Commun 2024; 741:151042. [PMID: 39586133 DOI: 10.1016/j.bbrc.2024.151042] [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: 09/12/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a common cardiovascular disease in the elderly, but there are still no therapeutic targets for this disease. In this study, we collected and analyzed bulk RNA sequencing (bulk RNA-seq) and single-cell RNA sequencing (scRNA-seq) data of AAA from the Gene Expression Omnibus (GEO) database. The immune infiltration-related genes were identified and categorized into various cell types, revealing potential key genes and pathways. Examination of three bulk RNA-seq datasets revealed a total of 4087 differentially expressed genes. The expression levels of the immune-related genes IL-7R were significantly elevated in AAA tissues across all three datasets. Furthermore, scRNA-Seq analysis revealed increased expression of IL-7R in CD4+ memory cells within AAA tissues. Immunofluorescence staining corroborated these findings, demonstrating increased expression of IL-7R in CD4+ T cells in AAA tissues. In vitro, activation of IL-7R elevated the activation of JAK/STAT pathway and phenotypic switching in SMCs, while inhibition of IL-7R abolished these effects and suppressed the secretion of IFN-γ. In conclusion, the activation of IL-7R in CD4+ T cells is a key contributor to the pathogenesis of AAA, as it can promote secretion of IFN-γ via JAK/STAT pathway and induce phenotypic switching of SMCs.
Collapse
Affiliation(s)
- Siyuan Zhou
- Jinshan Hospital, Fudan University, Shanghai, China; Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Shuai Ju
- Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Li
- Jinshan Hospital, Fudan University, Shanghai, China
| | - Chengchao Ruan
- Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Zhihui Dong
- Jinshan Hospital, Fudan University, Shanghai, China; Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Wu T, Su D, Zhang L, Liu T, Wang Q, Yan C, Liu M, Ji H, Lei J, Zheng M, Wen Z. Mitochondrial Control of Proteasomal Psmb5 Drives the Differentiation of Tissue-Resident Memory T Cells in Patients with Rheumatoid Arthritis. Arthritis Rheumatol 2024; 76:1743-1757. [PMID: 39037181 DOI: 10.1002/art.42954] [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: 01/01/2024] [Revised: 06/13/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVE To explore T cell-intrinsic mechanisms underpinning the mal-differentiation of tissue-resident memory T (Trm) cells in patients with rheumatoid arthritis (RA). METHODS Circulating T cells from patient with RA and healthy individuals were used for Trm cell differentiation. The role of Hobit in Trm differentiation was investigated through targeted silencing experiments. Psmb5 expression regulation was explored by identifying BRD2 as a key transcription factor, with the interaction validated through chromatin immunoprecipitation-quantitative polymerase chain reaction. The impact of BRD2 succinylation on Trm differentiation was examined by manipulating succinyl-CoA levels in T cells. Humanized NSG chimeras representing synovitis provided insights into Trm infiltration in RA synovitis and were used for translational experiments. RESULTS In patients with RA, a notable predisposition of CD4+ T cells toward differentiation into Trm cells was observed, demonstrating a positive correlation with the disease activity score 28. Remarkably, Hobit was a pivotal facilitator in the formation of RA CD4+ Trm cells. Mechanistic studies unveiled the dysregulation of proteasomal Psmb5 in T cells of patients with RA as the key factor contributing to elevated Hobit protein levels. The deficiency of proteasomal Psmb5 was intricately linked to BRD2, with succinylation exerting a significant impact on Psmb5 transcription and Trm cell differentiation. This heightened BRD2 succinylation was attributed to elevated levels of mitochondrial succinyl-CoA in RA T cells. Consequently, targeting succinyl-CoA within CD4+ T cells controlled the inflammation of synovial tissues in humanized chimeras. CONCLUSION Mitochondrial succinyl-CoA fosters the succinylation of BRD2, resulting in compromised transcription of proteasomal Psmb5 and the differentiation of Trm cells in RA.
Collapse
Affiliation(s)
- Tong Wu
- Soochow University, Suzhou, China
| | | | | | - Ting Liu
- Nanjing Medical University, Wuxi, China
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Yin H, Li L, Feng X, Wang Z, Zheng M, Zhao J, Fan X, Wu W, Gao L, Zhan Y, Zhao M, Lu Q. 2D4, a humanized monoclonal antibody targeting CD132, is a promising treatment for systemic lupus erythematosus. Signal Transduct Target Ther 2024; 9:323. [PMID: 39551768 PMCID: PMC11570697 DOI: 10.1038/s41392-024-02017-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Current therapies for systemic lupus erythematosus that target a particular factor or cell type exhibit limited effectiveness. To address this limitation, our focus was on CD132, a subunit common to six inflammatory factor receptors implicated in SLE. Our study revealed heightened CD132 expression in SLE patients' lymphocytes, contributing to the production of pro-inflammatory cytokines and immunoglobulins. We developed a novel humanized anti-CD132 monoclonal antibody, named as 2D4. 2D4 efficiently blocked IL-21 and IL-15, with limited effectiveness against IL-2, thereby suppressing T and B cells without disrupting immune tolerance. In the mouse immunization model, 2D4 virtually inhibited T cell-dependent, antigen-specific B-cell response. In lupus murine models, 2D4 mitigated inflammation by suppressing multiple pro-inflammatory cytokines and anti-dsDNA antibody titers, also diminishing proteinuria and glomerulonephritis. Compared to Belimumab, 2D4 exhibited superior efficacy in ameliorating the inflammatory state and preserving renal function. Moreover, 2D4 exhibited the ability to inhibit the production of pro-inflammatory factors and autoantibodies in PBMCs from individuals with SLE, highlighting its therapeutic potential for SLE individuals. Potent, 2D4 has the potential to significantly improve clinical outcomes in SLE and other complex autoimmune disorders.
Collapse
Affiliation(s)
- Huiqi Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiwei Feng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zijun Wang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Meiling Zheng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Junpeng Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinyu Fan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Wu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lingyu Gao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yijing Zhan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Ren Y, Tian J, Shi W, Feng J, Liu Y, Kang H, He Y. Evaluation of ocular surface inflammation and systemic conditions in patients with systemic lupus erythematosus: a cross-sectional study. BMC Ophthalmol 2024; 24:492. [PMID: 39533209 PMCID: PMC11556210 DOI: 10.1186/s12886-024-03760-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The cross-sectional study was designed to evaluate the association of ocular surface inflammation with systemic conditions in patients with systemic lupus erythematosus (SLE). METHODS The study enrolled 30 SLE patients and 30 controls. Ocular symptoms were evaluated using the Ocular Surface Disease Index (OSDI) questionnaire. Tear samples from all participants were collected for tear multi-cytokine and chemokine concentration analysis. All participants were assessed for dry eye disease (DED), including Schirmer I test, tear break-up time (TBUT), corneal fluorescein staining (CFS), meibomian gland secretion (MGS), lid-parallel conjunctival folds (LIPCOF), corneal clarity, and symblepharon. Besides, all participants were also examined for conjunctival impression cytology to measure the density of conjunctival goblet cells (CGCs). The peripheral blood indicators from SLE patients were also collected to measure the SLE-associated autoantibody specificities and systemic inflammatory indicators. Pearson and Spearman's analysis were uesd to examine the correlation between tear cytokines, CGCs, DED-related indicators, and systemic conditions. RESULTS The two groups were matched for age and gender in this study. 36.67% of eyes (11 in 30) of SLE patients and 13.33% of eyes (4 in 30) of controls were diagnosed with DED. OSDI scores, abnormal TBUT percentages, CFS percentages, and DED grading were all higher in SLE patients than in control group, while density of CGCs was lower. There were no significant differences in Schirmer I test, MGS, LIPCOF, corneal clarity, and symblepharon between SLE patients and controls. The levels of tear chemokine (C-X-C motif) ligand 11 (CXCL11) and cytokine interleukin-7 (IL-7) in patients with SLE were significantly higher than those in control group. Moreover, among SLE patients, the severity of DED and the level of tear chemokine CXCL11 were significantly positively correlated with SLE-associated autoantibody specificities. CONCLUSION Dry eye and tear cytokines and chemokines-mediated ocular surface inflammation persist in SLE patients and are associated with systemic conditions. Therefore, it is necessary for patients with SLE to combine systemic and ocular assessments.
Collapse
Affiliation(s)
- Yuerong Ren
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jing Tian
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Shi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jianing Feng
- Xi'an People's Hospital (Xi'an Fourth Hospital), Shaanxi Eye Hospital, Northwest University Affiliated People's Hospital, Xi'an, Shaanxi Province, China
| | - Yingyi Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Huanmin Kang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yan He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China.
| |
Collapse
|
6
|
He Y, Liu X, Wang R, Pang J, Tang Z, Zhong Q, Lin X. CD2 glycoprotein and CD44 structure and prevention of diabetes nephropathy: Central characteristics of related genes based on WGCNA and PPI. Int J Biol Macromol 2024; 279:135393. [PMID: 39245097 DOI: 10.1016/j.ijbiomac.2024.135393] [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: 07/14/2024] [Revised: 08/27/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Diabetic nephropathy (DN) is a prevalent complication of diabetes mellitus, characterized by complex pathogenesis that involves numerous molecules and signaling pathways. Among these, CD2 glycoprotein and CD44 play pivotal roles in cell adhesion, signal transduction, and inflammatory responses, potentially contributing significantly to the onset and progression of DN. This study aimed to investigate the central features of CD2 glycoprotein and CD44 in preventing diabetic nephropathy. To achieve this, kidney tissue sample data from DN patients were sourced from a public gene expression database. The roles of CD2 glycoprotein and CD44 within the PPI network were then analyzed, focusing on their interactions with other related genes. WGCNA analysis identified several significant gene modules associated with DN, including CD2 glycoprotein and CD44. PPI network analysis showed that these two proteins had a high degree of connectivity in the network, suggesting that they may be central regulatory molecules of DN. Further functional enrichment analysis revealed the potentially important role of CD2 glycoprotein and CD44 in diabetic nephropathy.
Collapse
Affiliation(s)
- Yi He
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi, Baise 533000, China
| | - Xin Liu
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi, Baise 533000, China
| | - Rong Wang
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi, Baise 533000, China
| | - Jun Pang
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi, Baise 533000, China
| | - Zhiming Tang
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi, Baise 533000, China
| | - Qiuhong Zhong
- Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi, Baise 533000, China; Department of ultrasound, The Affilated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China.
| | - Xu Lin
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China; Key Laboratory of Medical Research Basic Guarantee for Immune-Related Diseases Research of Guangxi, Baise 533000, China.
| |
Collapse
|
7
|
Wang B, Li J, Huang Y, Wu R. Synoviocyte detachment: an overlooked yet crucial histological aspect in rheumatoid arthritis. BMC Musculoskelet Disord 2024; 25:829. [PMID: 39434039 PMCID: PMC11492537 DOI: 10.1186/s12891-024-07935-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a prevalent autoimmune disorder that leads to chronic joint inflammation, deformity, disability, and systemic complications. This study aimed to analyze the clinical characteristics and synovial pathology of RA patients with synoviocyte detachment, and explore the factors associated with this phenomenon. METHODS This was a retrospective cohort study included RA patients who underwent synovial biopsy at our center from April to September 2023. Demographic, clinical, laboratory, and synovial histological data were retrospectively collected from medical records at the time of joint synovial biopsy in patients. Microscopic examination of hematoxylin and eosin (HE)-stained synovial tissue sections categorized the samples into synoviocyte detachment and no-synoviocyte detachment groups. Clinical characteristics and synovial pathological changes were compared between the two groups, and the factors associated with synoviocyte detachment were explored through logistic regression analysis. RESULTS Fifty-five RA patients were enrolled; 45 were females, and the mean age was 53.4 ± 11.8 years. Nine RA patients exhibited synoviocyte detachment. A total of 46 RA patients in the no-synoviocyte detachment group (15 with a normal lining layer and 31 with synovial cell proliferation) were included. Compared with the no-synoviocyte detachment group, the synoviocyte detachment group presented higher RF, ESR, CRP and DAS28-CRP levels (P < 0.05). The synoviocyte detachment group exhibited more prominent neovascularization (P < 0.05). ESR, DAS28-CRP and synovial neovascularization were risk factors associated with synoviocyte detachment in RA patients. CONCLUSION RA patients with synoviocyte detachment exhibit elevated clinical disease activity, marked by pronounced synovial pathology featuring increased neovascularization and less inflammatory cell infiltration. A significant reduction in lymphocyte count compared with patients with synovial cell proliferation was also observed.
Collapse
Affiliation(s)
- Bihua Wang
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, China
| | - Jianbin Li
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, China
| | - Yiping Huang
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, China
| | - Rui Wu
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
8
|
Meyer A. Illuminating the impact of γδ T cells in man and mice in spondylarthritides. Eur J Immunol 2024; 54:e2451071. [PMID: 39077953 DOI: 10.1002/eji.202451071] [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: 03/28/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/31/2024]
Abstract
Spondylarthritides (SpA) are a group of autoinflammatory diseases affecting the spine, peripheral joints, and entheses, including axial spondyloarthritis (axSpA) and psoriatic arthritis. AxSpA has a multifactorial etiology that involves genetic predispositions, such as HLA-B27 and IL-23R. Although HLA-B27 is strongly associated with axSpA, its role remains unclear. GWAS studies have demonstrated that genetic polymorphisms related to the IL-23 pathway occur throughout the spectrum of SpA, including but not limited to axSpA and PsA. IL-23 promotes the production of IL-17, which drives inflammation and tissue damage. This pathway contributes not only to peripheral enthesitis but also to spinal inflammation. γδ T cells in axSpA express IL-23R and RORγt, crucial for their activation, although specific pathogenic cells and factors remain elusive. Despite drug efficacy in PsA, IL-23R inhibition is ineffective in axSpA. Murine models provide valuable insights into the intricate cellular and molecular interactions that contribute to the development and progression of SpA. Those models are useful tools to elucidate the dynamics of γδ T cell involvement, offering insights into disease mechanisms and potential therapeutic targets. This review aims to illuminate the complex interplay between IL-23 and γδ T cells in SpA pathogenesis, emphasizing their roles in chronic inflammation, tissue damage, and disease heterogeneity.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Disease Models, Animal
- Interleukin-23/immunology
- Interleukin-23/metabolism
- Interleukin-23/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- HLA-B27 Antigen/genetics
- HLA-B27 Antigen/immunology
- Genetic Predisposition to Disease
- Spondylarthritis/immunology
- Receptors, Interleukin/genetics
- Receptors, Interleukin/metabolism
- Receptors, Interleukin/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
Collapse
Affiliation(s)
- Anja Meyer
- Center for Molecular Neurobiology Hamburg, Institute for Systems Immunology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
9
|
Yazdani R, Askari M, Ahmadi AM, Azizi G, Ciric B, Boehm A, Zhang GX, Rostami A. Mouse monocytes express CD127 by immune cells, not LPS. Front Immunol 2024; 15:1356004. [PMID: 39328413 PMCID: PMC11424446 DOI: 10.3389/fimmu.2024.1356004] [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: 12/14/2023] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The essential role of interleukin 7 (IL-7) signaling via its receptor (IL-7Rα; CD127) in T cell development and function has been well documented. However, CD127 expression and function in myeloid cells, including monocytes, are less clear, especially in mice. In the present study we report an inducible CD127 expression in mouse monocytes/macrophages. This induction is dependent on the presence of other immune cells, highlighting that regulation of CD127 expression on monocytes differs in mice and humans. We demonstrate that CD127 is functional, as IL-7 downregulated its expression. We also saw decreased CD127 expression during inflammation in vivo. Overall, upregulation of CD127 expression in vitro and its downregulation in vivo confirm that CD127 is an inducible marker on mouse monocyte/macrophage cells, in contrast to findings recently published by others. Characterizing the role of CD127 signaling in myeloid cells in inflammatory disorders would be worthwhile in future study.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University,
Philadelphia, PA, United States
| |
Collapse
|
10
|
Liu J, Pan R. Causal effects of systemic inflammatory proteins on Guillain-Barre Syndrome: insights from genome-wide Mendelian randomization, single-cell RNA sequencing analysis, and network pharmacology. Front Immunol 2024; 15:1456663. [PMID: 39315093 PMCID: PMC11416972 DOI: 10.3389/fimmu.2024.1456663] [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: 07/02/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Background Evidence from observational studies indicates that inflammatory proteins play a vital role in Guillain-Barre Syndrome (GBS). Nevertheless, it is unclear how circulating inflammatory proteins are causally associated with GBS. Herein, we conducted a two-sample Mendelian randomization (MR) analysis to systematically explore the causal links of genetically determined systemic inflammatory proteins on GBS. Methods A total of 8,293 participants of European ancestry were included in a genome-wide association study of 41 inflammatory proteins as instrumental variables. Five MR approaches, encompassing inverse-variance weighted, weighted median, MR-Egger, simple model, and weighted model were employed to explore the causal links between inflammatory proteins and GBS. MR-Egger regression was utilized to explore the pleiotropy. Cochran's Q statistic was implemented to quantify the heterogeneity. Furthermore, we performed single-cell RNA sequencing analysis and predicted potential drug targets through molecular docking technology. Results By applying MR analysis, four inflammatory proteins causally associated with GBS were identified, encompassing IFN-γ (OR:1.96, 95%CI: 1.02-3.78, PIVW=0.045), IL-7 (OR:1.86, 95%CI: 1.07-3.23, PIVW=0.029), SCGF-β (OR:1.56, 95%CI: 1.11-2.19, PIVW=0.011), and Eotaxin (OR:1.99, 95%CI: 1.01-3.90, PIVW=0.046). The sensitivity analysis revealed no evidence of pleiotropy or heterogeneity. Additionally, significant genes were found through single-cell RNA sequencing analysis and several anti-inflammatory or neuroprotective small molecular compounds were identified by utilizing molecular docking technology. Conclusions Our MR analysis suggested that IFN-γ, IL-7, SCGF-β, and Eotaxin were causally linked to the occurrence and development of GBS. These findings elucidated potential causal associations and highlighted the significance of these inflammatory proteins in the pathogenesis and prospective therapeutic targets for GBS.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Neurology and Psychiatry, Longyou People’s Hospital Affiliated with Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Quzhou, Zhejiang, China
| | - Renbing Pan
- Department of Urology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| |
Collapse
|
11
|
Ziaka M, Exadaktylos A. Gut-derived immune cells and the gut-lung axis in ARDS. Crit Care 2024; 28:220. [PMID: 38965622 PMCID: PMC11225303 DOI: 10.1186/s13054-024-05006-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024] Open
Abstract
The gut serves as a vital immunological organ orchestrating immune responses and influencing distant mucosal sites, notably the respiratory mucosa. It is increasingly recognized as a central driver of critical illnesses, with intestinal hyperpermeability facilitating bacterial translocation, systemic inflammation, and organ damage. The "gut-lung" axis emerges as a pivotal pathway, where gut-derived injurious factors trigger acute lung injury (ALI) through the systemic circulation. Direct and indirect effects of gut microbiota significantly impact immune responses. Dysbiosis, particularly intestinal dysbiosis, termed as an imbalance of microbial species and a reduction in microbial diversity within certain bodily microbiomes, influences adaptive immune responses, including differentiating T regulatory cells (Tregs) and T helper 17 (Th17) cells, which are critical in various lung inflammatory conditions. Additionally, gut and bone marrow immune cells impact pulmonary immune activity, underscoring the complex gut-lung interplay. Moreover, lung microbiota alterations are implicated in diverse gut pathologies, affecting local and systemic immune landscapes. Notably, lung dysbiosis can reciprocally influence gut microbiota composition, indicating bidirectional gut-lung communication. In this review, we investigate the pathophysiology of ALI/acute respiratory distress syndrome (ARDS), elucidating the role of immune cells in the gut-lung axis based on recent experimental and clinical research. This exploration aims to enhance understanding of ALI/ARDS pathogenesis and to underscore the significance of gut-lung interactions in respiratory diseases.
Collapse
Affiliation(s)
- Mairi Ziaka
- Clinic of Geriatric Medicine, Center of Geriatric Medicine and Rehabilitation, Kantonsspital Baselland, Bruderholz, Switzerland.
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
12
|
Baxter J, Waters A. CVID With Unusual Peripheral Mononeuropathy and Associated IL-7 Receptor Mutation. Mil Med 2024; 189:e1819-e1822. [PMID: 38126802 DOI: 10.1093/milmed/usad480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 11/09/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Common variable immunodeficiency (CVID) is the most common symptomatic primary immunodeficiency. It is characterized by hypogammaglobulinemia and can present with a broad range of symptoms including recurrent bacterial infections, autoimmunity, and malignancy. Rarely, it has been implicated with peripheral neuropathy. We present a case of CVID with peripheral neuropathy and a pathogenic heterozygous variant of IL-7 receptor gene. The patient is a 38-year-old female with a history of recurrent infections since childhood including pneumonia and sinus infections status post tonsillectomy and sinus surgery. She subsequently developed severe left leg and lower back pain that progressed to left foot drop and decreased sensation over the left leg. She was found to have severe hypogammaglobulinemia and poor polysaccharide and protein response, thus meeting criteria for CVID. Mononeuropathy is a rare finding in CVID. Genetic panel was performed and was significant for a single pathogenic variant in IL-7 receptor. Disruptions in the IL-7 and IL-7 receptor signaling pathway have been associated with autoimmunity such as rheumatoid arthritis and multiple sclerosis. Further investigation is indicated to determine the clinical significance of this variant.
Collapse
Affiliation(s)
- Joseph Baxter
- Department of Allergy and Immunology, Wilford Hall Ambulatory Surgical Center, JBSA-Lackland AFB, TX 78236, USA
| | - Aubri Waters
- Department of Allergy and Immunology, Brooke Army Medical Center, Fort Sam Houston, TX 78234, USA
| |
Collapse
|
13
|
Huang Z, Braunstein Z, Chen J, Wei Y, Rao X, Dong L, Zhong J. Precision Medicine in Rheumatic Diseases: Unlocking the Potential of Antibody-Drug Conjugates. Pharmacol Rev 2024; 76:579-598. [PMID: 38622001 DOI: 10.1124/pharmrev.123.001084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/25/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
In the era of precision medicine, antibody-drug conjugates (ADCs) have emerged as a cutting-edge therapeutic strategy. These innovative compounds combine the precision of monoclonal antibodies with the potent cell-killing or immune-modulating abilities of attached drug payloads. This unique strategy not only reduces off-target toxicity but also enhances the therapeutic effectiveness of drugs. Beyond their well established role in oncology, ADCs are now showing promising potential in addressing the unmet needs in the therapeutics of rheumatic diseases. Rheumatic diseases, a diverse group of chronic autoimmune diseases with varying etiologies, clinical presentations, and prognoses, often demand prolonged pharmacological interventions, creating a pressing need for novel, efficient, and low-risk treatment options. ADCs, with their ability to precisely target the immune components, have emerged as a novel therapeutic strategy in this context. This review will provide an overview of the core components and mechanisms behind ADCs, a summary of the latest clinical trials of ADCs for the treatment of rheumatic diseases, and a discussion of the challenges and future prospects faced by the development of next-generation ADCs. SIGNIFICANCE STATEMENT: There is a lack of efficient and low-risk targeted therapeutics for rheumatic diseases. Antibody-drug conjugates, a class of cutting-edge therapeutic drugs, have emerged as a promising targeted therapeutic strategy for rheumatic disease. Although there is limited literature summarizing the progress of antibody-drug conjugates in the field of rheumatic disease, updating the advancements in this area provides novel insights into the development of novel antirheumatic drugs.
Collapse
Affiliation(s)
- Zhiwen Huang
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Zachary Braunstein
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Jun Chen
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Yingying Wei
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Xiaoquan Rao
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Lingli Dong
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Jixin Zhong
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| |
Collapse
|
14
|
Liu Z, Peng Z, Lin H, Zhou K, Liang L, Cao J, Huang Z, Mei J. Identifying potential drug targets for idiopathic pulmonary fibrosis: a mendelian randomization study based on the druggable genes. Respir Res 2024; 25:217. [PMID: 38783236 PMCID: PMC11118997 DOI: 10.1186/s12931-024-02848-5] [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: 09/26/2023] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic fibrotic interstitial lung disease characterized by progressive dyspnea and decreased lung function, yet its exact etiology remains unclear. It is of great significance to discover new drug targets for IPF. METHODS We obtained the cis-expression quantitative trait locus (cis-eQTL) of druggable genes from eQTLGen Consortium as exposure and the genome wide association study (GWAS) of IPF from the International IPF Genetics Consortium as outcomes to simulate the effects of drugs on IPF by employing mendelian randomization analysis. Then colocalization analysis was performed to calculate the probability of both cis-eQTL of druggable genes and IPF sharing a causal variant. For further validation, we conducted protein quantitative trait locus (pQTL) analysis to reaffirm our findings. RESULTS The expression of 45 druggable genes was significantly associated with IPF susceptibility at FDR < 0.05. The expression of 23 and 15 druggable genes was significantly associated with decreased forced vital capacity (FVC) and diffusing capacity of the lungs for carbon monoxide (DLco) in IPF patients, respectively. IPF susceptibility and two significant genes (IL-7 and ABCB2) were likely to share a causal variant. The results of the pQTL analysis demonstrated that high levels of IL-7 in plasma are associated with a reduced risk of IPF (OR = 0.67, 95%CI: 0.47-0.97). CONCLUSION IL-7 stands out as the most promising potential drug target to mitigate the risk of IPF. Our study not only sheds light on potential drug targets but also provides a direction for future drug development in IPF.
Collapse
Affiliation(s)
- Zetao Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Zhiyu Peng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Huahang Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Ke Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Linchuan Liang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Jie Cao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Zhaokang Huang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Jiandong Mei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Dai Y, Chen L, Zhang Z, Liu X. Identification and validation of immune-related genes in osteoarthritic synovial fibroblasts. Heliyon 2024; 10:e28330. [PMID: 38571590 PMCID: PMC10988018 DOI: 10.1016/j.heliyon.2024.e28330] [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: 07/10/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
Objective OA was generally considered as a non-inflammatory disease dominated by articular cartilage degeneration. However, the role of synovitis in OA pathogenesis has received increasing attention. Recent studies support that OA patients have a pro-inflammatory/catabolic synovial environment similar to RA patients, promoting the occurrence and development of OA. Therefore, we investigated the co-immune-related genes and pathways of OA and RA to explore whether part of the pathogenesis of RA synovitis can be used to explain OA synovitis. Methods Data of GSE29746 and GSE12021 were downloaded from the Gene Expression Omnibus (GEO) database. Compared with control group, differentially expressed genes (DEGs) of OA and RA groups were screened separately by R software, Venny website was used to screen co-DEGs. Metascape was used to screen the common enriched terms and pathways between OA and RA. STRING website and Cytoscape software were used to map protein-protein interaction (PPI) networks and screen co-hub genes. GSE29746 was selected as the test dataset, and GSE12021 as the validation dataset for validate the co-hub genes. The results were validated by western blotting (WB) and real-time quantitative polymerase chain reaction (qPCR) of clinical synovial samples. Results We identified 573 OA-related DEGs, 148 RA-related DEGs, and 52 co-DEGs, revealing 14 common enriched terms, most of which were related to immune inflammation. IL7R was the only upregulated co-hub gene between OA and RA in the PPI network, consistent with the validation dataset. IL7R was highly expressed in clinical osteoarthritic synovial samples (P < 0.001). Conclusion Our findings suggested that IL7R is a critical co-DEG in OA and RA and confirmed the involvement of immune inflammation in disease pathogenesis. Furthermore, it confirms the role of IL7R in synovial inflammation in RA and OA synovitis and provides evidence for further investigation of OA immune inflammation.
Collapse
Affiliation(s)
- Yaduan Dai
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Chen
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhan Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xueyong Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Yuan R, Wan X, Bao L, Long T, Li H, Zhou Y, Liu L, Shi P, Gong R, Jiang H. Tolerogenic dendritic cells alleviate collagen-induced arthritis by regulating T-cell differentiation and inhibiting NLRP3-mediated apoptosis. Int Immunopharmacol 2024; 130:111764. [PMID: 38452413 DOI: 10.1016/j.intimp.2024.111764] [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: 11/30/2023] [Revised: 02/12/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVE Tolerogenic dendritic cells (tolDCs) have emerged as a potential treatment for rheumatoid arthritis (RA). However, the detailed mechanism requires further investigation. In this study, we aimed to explore the effects of tolDCs on T-cell differentiation and NLRP3-mediated pyroptosis in a collagen-induced arthritis (CIA) rat model. METHODS TolDCs were induced using NF-κB ODN decoy. The efficacy of tolDCs intervention in alleviating arthritis symptoms was evaluated in CIA rats. Flow cytometry was employed to analyze CD4+ T-cell subpopulations, while scanning electron microscopy was utilized to observe pyroptosis morphology. Immunohistochemistry was used to assess the expression of pyroptosis-associated proteins. RESULTS TolDCs intervention significantly reduced joint inflammation and damage in CIA rats. Moreover, it successfully restored the balance of Th1/Th2 cells as well as the balance of Treg/Th17 cells. Furthermore, tolDCs intervention effectively suppressed NLRP3-mediated pyroptosis in the synovium, decreasing the release of IL-1β and IL-18. CONCLUSION Our findings underscore the efficacy of tolDCs in attenuating CIA progression through modulation of CD4+ T-cell subpopulations and inhibition of NLRP3-mediated pyroptosis.
Collapse
Affiliation(s)
- Rui Yuan
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, China; School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China; Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiufang Wan
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, China; School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China; Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Lunmin Bao
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, China; Department of Laboratory Medicine, People' Hospital of Anshun City, Anshun, China
| | - Tiaoyu Long
- Department of Laboratory Medicine, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Honghong Li
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Yan Zhou
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Lian Liu
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Ping Shi
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Rui Gong
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Hongmei Jiang
- Department of Immunology, School of Basic Medical Science, Guizhou Medical University, Guiyang, China; School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
17
|
Padovano C, Bianco SD, Sansico F, De Santis E, Tamiro F, Colucci M, Totti B, Di Iasio S, Bruno G, Panelli P, Miscio G, Mazza T, Giambra V. The Notch1 signaling pathway directly modulates the human RANKL-induced osteoclastogenesis. Sci Rep 2023; 13:21199. [PMID: 38040752 PMCID: PMC10692129 DOI: 10.1038/s41598-023-48615-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023] Open
Abstract
Notch signaling is an evolutionary conserved pathway with a key role in tissue homeostasis, differentiation and proliferation. It was reported that Notch1 receptor negatively regulates mouse osteoclast development and formation by inhibiting the expression of macrophage colony-stimulating factor in mesenchymal cells. Nonetheless, the involvement of Notch1 pathway in the generation of human osteoclasts is still controversial. Here, we report that the constitutive activation of Notch1 signaling induced a differentiation block in human mononuclear CD14+ cells directly isolated from peripheral blood mononuclear cells (PBMCs) upon in vitro stimulation to osteoclasts. Additionally, using a combined approach of single-cell RNA sequencing (scRNA-Seq) simultaneously with a panel of 31 oligo-conjugated antibodies against cell surface markers (AbSeq assay) as well as unsupervised learning methods, we detected four different cell stages of human RANKL-induced osteoclastogenesis after 5 days in which Notch1 signaling enforces the cell expansion of specific subsets. These cell populations were characterized by distinct gene expression and immunophenotypic profiles and active Notch1, JAK/STAT and WNT signaling pathways. Furthermore, cell-cell communication analyses revealed extrinsic modulators of osteoclast progenitors including the IL7/IL7R and WNT5a/RYK axes. Interestingly, we also report that Interleukin-7 receptor (IL7R) was a downstream effector of Notch1 pathway and that Notch1 and IL7R interplay promoted cell expansion of human RANKL-induced osteoclast progenitors. Taken together, these findings underline a novel cell pattern of human osteoclastogenesis, outlining the key role of Notch1 and IL-7R signaling pathways.
Collapse
Affiliation(s)
- Costanzo Padovano
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Salvatore Daniele Bianco
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy
| | - Francesca Sansico
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Elisabetta De Santis
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Francesco Tamiro
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Mattia Colucci
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Beatrice Totti
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Serena Di Iasio
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Gaja Bruno
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Patrizio Panelli
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Giuseppe Miscio
- Clinical Laboratory Analysis and Transfusional Medicine, Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy
| | - Vincenzo Giambra
- Hematopathology Laboratory, Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS "Casa Sollievo della Sofferenza", 71013, San Giovanni Rotondo (FG), Italy.
| |
Collapse
|
18
|
Benucci M, Bernardini P, Coccia C, De Luca R, Levani J, Economou A, Damiani A, Russo E, Amedei A, Guiducci S, Bartoloni E, Manfredi M, Grossi V, Infantino M, Perricone C. JAK inhibitors and autoimmune rheumatic diseases. Autoimmun Rev 2023; 22:103276. [PMID: 36649877 DOI: 10.1016/j.autrev.2023.103276] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
The four Janus kinase (JAK) proteins and the seven Signal Transducers of Activated Transcription (STAT) mediate intracellular signal transduction downstream of cytokine receptors, which are involved in the pathology of allergic, autoimmune, and inflammatory diseases. The development of targeted small-molecule treatments with diverse selective inhibitory profiles, such as JAK inhibitors (JAKi), has supported an important change in the treatment of multiple disorders. Indeed, JAKi inhibit intracellular signalling controlled by numerous cytokines implicated in the disease process of rheumatoid arthritis and several other inflammatory and immune diseases. Therefore, JAKi have the capacity to target multiple pathways of those diseases. Other autoimmune diseases treated with JAKi include systemic sclerosis, systemic lupus erythematosus, dermatomyositis, primary Sjogren's syndrome, and vasculitis. In all of these cases, innate immunity stimulation activates adaptive immunity, resulting in the production of autoreactive T cells as well as the stimulation and differentiation of B cells. Mechanism-based treatments that target JAK-STAT pathways have the possibility of improving outcomes by reducing the consumption of glucocorticoids and/or non-specific immunosuppressive drugs in the management of systemic immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Maurizio Benucci
- Rheumatology Unit, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, Florence, Italy
| | - Pamela Bernardini
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Carmela Coccia
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Riccardo De Luca
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Juela Levani
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Alessio Economou
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Arianna Damiani
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Serena Guiducci
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Mariangela Manfredi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, Azienda USL-Toscana Centro, Florence, Italy
| | - Valentina Grossi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, Azienda USL-Toscana Centro, Florence, Italy
| | - Maria Infantino
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, Azienda USL-Toscana Centro, Florence, Italy
| | - Carlo Perricone
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
19
|
Huang H, Dong X, Mao K, Pan W, Nie B, Jiang L. Identification of key candidate genes and pathways in rheumatoid arthritis and osteoarthritis by integrated bioinformatical analysis. Front Genet 2023; 14:1083615. [PMID: 36861127 PMCID: PMC9968929 DOI: 10.3389/fgene.2023.1083615] [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: 10/29/2022] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are the most common joint disorders. Although they have shown analogous clinical manifestations, the pathogenesis of RA and OA are different. In this study, we used the online Gene Expression Omnibus (GEO) microarray expression profiling dataset GSE153015 to identify gene signatures between RA and OA joints. The relevant data on 8 subjects obtained from large joints of RA patients (RA-LJ), 8 subjects obtained from small joints of RA patients (RA-SJ), and 4 subjects with OA were investigated. Differentially expressed genes (DEGs) were screened. Functional enrichment analysis of DEGs including the Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were identified, which were mainly associated with T cell activation or chemokine activity. Besides, protein-protein interaction (PPI) network analysis was performed, and key modules were identified. Hub genes of RA-LJ and OA groups were screened, they were CD8A, GZMB, CCL5, CD2, and CXCL9, whereas CD8A, CD2, IL7R, CD27, and GZMB were hub genes of RA-SJ and OA group. The novel DEGs and functional pathways between RA and OA identified in this study may provide new insight into the underlying molecular mechanisms and therapeutic strategies of RA and OA.
Collapse
Affiliation(s)
- Huijing Huang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyi Dong
- Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kaimin Mao
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wanwan Pan
- Yankuang New Journey General Hospital, Jingning, Shandong, China
| | - Bin’en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Lindi Jiang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China,*Correspondence: Lindi Jiang,
| |
Collapse
|
20
|
Hu H, Feng Z, Lin H, Zhao J, Zhang Y, Xu F, Chen L, Chen F, Ma Y, Su J, Zhao Q, Shuai J. Modeling and analyzing single-cell multimodal data with deep parametric inference. Brief Bioinform 2023; 24:6987655. [PMID: 36642414 DOI: 10.1093/bib/bbad005] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/11/2022] [Accepted: 01/02/2023] [Indexed: 01/17/2023] Open
Abstract
The proliferation of single-cell multimodal sequencing technologies has enabled us to understand cellular heterogeneity with multiple views, providing novel and actionable biological insights into the disease-driving mechanisms. Here, we propose a comprehensive end-to-end single-cell multimodal analysis framework named Deep Parametric Inference (DPI). DPI transforms single-cell multimodal data into a multimodal parameter space by inferring individual modal parameters. Analysis of cord blood mononuclear cells (CBMC) reveals that the multimodal parameter space can characterize the heterogeneity of cells more comprehensively than individual modalities. Furthermore, comparisons with the state-of-the-art methods on multiple datasets show that DPI has superior performance. Additionally, DPI can reference and query cell types without batch effects. As a result, DPI can successfully analyze the progression of COVID-19 disease in peripheral blood mononuclear cells (PBMC). Notably, we further propose a cell state vector field and analyze the transformation pattern of bone marrow cells (BMC) states. In conclusion, DPI is a powerful single-cell multimodal analysis framework that can provide new biological insights into biomedical researchers. The python packages, datasets and user-friendly manuals of DPI are freely available at https://github.com/studentiz/dpi.
Collapse
Affiliation(s)
- Huan Hu
- Department of Physics, and Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen 361005, China.,National Institute for Data Science in Health and Medicine, and State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen 361005 China.,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), and Wenzhou Institute and Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Zhen Feng
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou 325000, China
| | - Hai Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), and Wenzhou Institute and Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Junjie Zhao
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou 510000, China
| | - Yaru Zhang
- Institute of Biomedical Big Data, School of Ophthalmology & Optometry and Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Fei Xu
- Department of Physics, and Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen 361005, China
| | - Lingling Chen
- Department of Physics, and Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen 361005, China
| | - Feng Chen
- Department of Physics, and Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen 361005, China
| | - Yunlong Ma
- Institute of Biomedical Big Data, School of Ophthalmology & Optometry and Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Jianzhong Su
- Institute of Biomedical Big Data, School of Ophthalmology & Optometry and Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou 325027, China
| | - Qi Zhao
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan 114051, China
| | - Jianwei Shuai
- Department of Physics, and Fujian Provincial Key Laboratory for Soft Functional Materials Research, Xiamen University, Xiamen 361005, China.,National Institute for Data Science in Health and Medicine, and State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen 361005 China.,Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), and Wenzhou Institute and Wenzhou Key Laboratory of Biophysics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| |
Collapse
|
21
|
Zhou TT, Sun JJ, Tang LD, Yuan Y, Wang JY, Zhang L. Potential diagnostic markers and therapeutic targets for rheumatoid arthritis with comorbid depression based on bioinformatics analysis. Front Immunol 2023; 14:1007624. [PMID: 36911710 PMCID: PMC9995708 DOI: 10.3389/fimmu.2023.1007624] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
Background Rheumatoid arthritis (RA) and depression are prevalent diseases that have a negative impact on the quality of life and place a significant economic burden on society. There is increasing evidence that the two diseases are closely related, which could make the disease outcomes worse. In this study, we aimed to identify diagnostic markers and analyzed the therapeutic potential of key genes. Methods We assessed the differentially expressed genes (DEGs) specific for RA and Major depressive disorder (MDD) and used weighted gene co-expression network analysis (WGCNA) to identify co-expressed gene modules by obtaining the Gene expression profile data from Gene Expression Omnibus (GEO) database. By using the STRING database, a protein-protein interaction (PPI) network constructed and identified key genes. We also employed two types of machine learning techniques to derive diagnostic markers, which were assessed for their association with immune cells and potential therapeutic effects. Molecular docking and in vitro experiments were used to validate these analytical results. Results In total, 48 DEGs were identified in RA with comorbid MDD. The PPI network was combined with WGCNA to identify 26 key genes of RA with comorbid MDD. Machine learning-based methods indicated that RA combined with MDD is likely related to six diagnostic markers: AURKA, BTN3A2, CXCL10, ERAP2, MARCO, and PLA2G7. CXCL10 and MARCO are closely associated with diverse immune cells in RA. However, apart from PLA2G7, the expression levels of the other five genes were associated with the composition of the majority of immune cells in MDD. Molecular docking and in vitro studies have revealed that Aucubin (AU) exerts the therapeutic effect through the downregulation of CXCL10 and BTN3A2 gene expression in PC12 cells. Conclusion Our study indicates that six diagnostic markers were the basis of the comorbidity mechanism of RA and MDD and may also be potential therapeutic targets. Further mechanistic studies of the pathogenesis and treatment of RA and MDD may be able to identify new targets using these shared pathways.
Collapse
Affiliation(s)
- Tao-Tao Zhou
- Department of Mathematics and Physics, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ji-Jia Sun
- Department of Mathematics and Physics, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Dong Tang
- Teaching and Research Section of Chinese Materia Medica, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Yuan
- Teaching and Research Section of Chinese Materia Medica, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian-Ying Wang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
IL-33 Deficiency Attenuates Lung Inflammation by Inducing Th17 Response and Impacting the Th17/Treg Balance in LPS-Induced ARDS Mice via Dendritic Cells. J Immunol Res 2022; 2022:9543083. [PMID: 36570798 PMCID: PMC9788894 DOI: 10.1155/2022/9543083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/23/2022] Open
Abstract
Objectives The characteristic pathophysiological feature of acute respiratory distress syndrome (ARDS) is a dysregulated inflammatory response. T helper 17 (Th17) cells in the lung are inflammatory cells that contribute to pulmonary inflammatory cascades. In addition, Th17/regulatory T cells (Treg cells) also play an important role in the inflammatory process. Dendritic cells (DCs) can regulate the differentiation of CD4+ T cells, including Th17 and Treg cells. Recent evidence revealed that interleukin-33 (IL-33) signaling could activate and mature DCs. Therefore, the aim of this study was to investigate the effects of IL-33 on inflammation and immunoregulation by inducing the Th17 response and influencing the Th17/Treg balance in LPS-induced ARDS. Methods IL-33 gene knockout mice and the administration of recombinant mouse IL-33 (rmIL-33) were used to investigate the role of IL-33 and the underlying mechanisms in an LPS-induced ARDS model. Hematoxylin and eosin (H&E) staining, wet/dry (W/D) weight ratios, cell counts, and the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-17 (IL-17), and interleukin-10 (IL-10) in bronchoalveolar lavage fluid (BALF) were investigated. The levels of IL-33, orphan nuclear receptor gamma t (RORγt), and forkhead transcription factor protein 3 (FOXP3) protein in lung tissue were evaluated by Western blotting. The mRNA expression levels of IL-33 and RORγt were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Th17 and Treg cell frequencies were determined by flow cytometry. The levels of IL-6 in the supernatant in a dendritic cell culture system were examined by ELISA. Results Increased expression of IL-33 was observed in mice with LPS-induced ARDS. IL-33 deficiency significantly inhibited inflammation and attenuated LPS-induced ARDS, whereas pretreatment with rmIL-33 aggravated pulmonary inflammatory response. Furthermore, depletion of IL-33 inhibited Th17 cells, significantly decreased RORγt mRNA and protein expression and IL-17 levels in BALF, and led to less differentiation of T cells into Th17 cells than Treg cells. Moreover, IL-33-/- DCs secreted less IL-6 and IL-23 than normal control DCs. Conclusion IL-33 deficiency alleviated lung injury in the LPS-induced ARDS model, which was closely related to suppressing Th17 responses and regulating the Th17/Treg balance. The expansion of Th17 cells and imbalance in Th17/Treg cells may be associated with IL-6 and IL-23 secreted from IL-33-activated DCs.
Collapse
|