1
|
Jajodia A, Mishra A, Doni Jayavelu N, Lambert K, Moss N, Yang Z, Cerosaletti K, Buckner JH, Hawkins RD. Functional dissection of noncoding variants associated with rheumatoid arthritis. Ann Rheum Dis 2025:S0003-4967(25)00890-8. [PMID: 40318978 DOI: 10.1016/j.ard.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVES Noncoding variants are critical to our understanding of the genetic basis of diseases and disorders such as rheumatoid arthritis (RA). While genome-wide association studies have identified regions of the genome associated with disease, functional studies are still lagging that can identify potentially causative variants. METHODS In order to functionally fine-map RA-associated variants, we identified variants at enhancers marked in primary activated T helper cells and conducted massively parallel reporter assay in these cells. RESULTS We found that combinations of functional variant genotypes are often exclusive to patients with RA. We leveraged 3-dimensional genome architecture and expression quantitative trait loci data to identify target genes of enhancers exhibiting allelic differences in activity. We confirmed enhancer activity and target gene interactions by Clustered Regularly Interpaced Short Palindromic Repeats Cas9 (CRISPR-Cas9) deletion in primary T cells. CONCLUSIONS The identification of functional enhancer variants suggests possible causal variants, and their target genes reveal known and novel genes as likely drivers of RA, as well as a means for therapeutic intervention.
Collapse
Affiliation(s)
- Ajay Jajodia
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Arpit Mishra
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Naresh Doni Jayavelu
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Nicholas Moss
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Zongchen Yang
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Jane H Buckner
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - R David Hawkins
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA; Benaroya Research Institute at Virginia Mason, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
2
|
Martin BT, Clarkson MW, Wu P, Di Lello P. 1H, 13C and 15N backbone resonance assignment of the catalytic domain of human PTPN22. BIOMOLECULAR NMR ASSIGNMENTS 2025:10.1007/s12104-025-10233-6. [PMID: 40281380 DOI: 10.1007/s12104-025-10233-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Protein Tyrosine Phosphatase Non-receptor type 22 (PTPN22) is a tyrosine-phosphatase that plays a major role in inhibiting T-cell activation in immune cells. Recent studies have revealed that downregulation of PTPN22 triggers T-cell activation and enhances antitumor immune response, thereby identifying PTPN22 as a potential pharmacological target in cancer-immunology.Here we report the 1H, 15N and 13C backbone resonance assignment for the 35.6 kDa catalytic domain of human PTPN22. This assignment will provide an essential experimental tool to identify and characterize potential PTPN22 inhibitors.
Collapse
Affiliation(s)
- Bryan T Martin
- Department of Structural Biology-Protein Sciences, Genentech, South San Francisco, CA, 94080, USA
| | - Michael W Clarkson
- Department of Structural Biology-Protein Sciences, Genentech, South San Francisco, CA, 94080, USA
| | - Ping Wu
- Department of Structural Biology-Protein Sciences, Genentech, South San Francisco, CA, 94080, USA
| | - Paola Di Lello
- Department of Structural Biology-Protein Sciences, Genentech, South San Francisco, CA, 94080, USA.
| |
Collapse
|
3
|
Wang WH, Xia MH, Liu XR, Lei SF, He P. A Bibliometric Analysis of GWAS on Rheumatoid Arthritis from 2002 to 2024. Hum Hered 2025; 90:18-32. [PMID: 40179854 DOI: 10.1159/000543947] [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: 07/29/2024] [Accepted: 01/24/2025] [Indexed: 04/05/2025] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) has become a serious threat to human health and quality of life worldwide. Previous studies have demonstrated that genetic factors play a crucial role in the onset and progression of RA. Due to the rapid development of genome-wide association study (GWAS) and large-scale genetic analysis, GWAS research on RA has received widespread attention in recent years. Therefore, we conducted a comprehensive visualization and bibliometric analysis of publications to identify hotspots and future trends in GWAS research on RA. METHODS Literature on RA and GWAS published between 2002 and 2024 was extracted from the Web of Science Core Collection database by strategic screening. Collected data were further analyzed by using VOSviewer, CiteSpace, and Excel. The collaborations networks of countries, authors, institutions, and the co-citation networks of publications were visualized. Finally, research hotspots and fronts were examined. RESULTS A total of 713 publications with 45,773 citations were identified. The number of publications and citations has had a significant surge since 2007. The United States contributed the most publications globally. Okada, Yukinori, was the most influential author. The most productive institution in this field was the University of Manchester. The analysis of keywords revealed that "mendelian randomization analysis", "association", "innate", "instruments", "bias", "pathogenesis", and "genome-wide association study" are likely to be the frontiers of research in this field. CONCLUSION This study can be used to predict future research advances in the fields of GWAS on RA and helps to promote academic collaboration among scholars.
Collapse
Affiliation(s)
- Wen-Hui Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China,
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China,
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China,
| | - Ming-Hui Xia
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Xin-Ru Liu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
- Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou, China
| | - Pei He
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
4
|
Wang S, Liu Y, Zou X, Pan M, Wan Q, Chu X. Exploring the pathogenesis of RA through the gut-articular axis-dysbiosis a potential factor. Clin Anat 2025; 38:134-145. [PMID: 39189295 DOI: 10.1002/ca.24215] [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: 05/29/2024] [Accepted: 08/04/2024] [Indexed: 08/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with a complex etiology. It has been suggested that the pathogenesis of RA begins in the mucosa and then transitions to the joints when many factors interact, including microbial dysbiosis, inflammatory responses, and immune abnormalities at the mucosal site. Data from RA animals and patients suggest there are changes in the mucosal microflora before the onset of RA, and that dysbiosis of the mucosal ecology continues to play a role in the development of arthritis. Microbial dysbiosis of the mucosa reduces the normal barrier function of the intestinal tract, promotes inflammatory reactions in the mucosal areas of the intestines, and then activates the intestinal immune cells abnormally to produce a large number of auto-reactive antibodies that exacerbate arthritis. Current findings do not clarify whether dysbiosis is only a potential trigger for the development of RA. If it is possible to intervene in such microbial changes before the onset of RA, could the clinical symptoms of arthritis be prevented or reduced? Finding new ways to regulate gut flora composition to maintain gut barrier function is an ongoing challenge for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Shuai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xingyu Zou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Mengjun Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qing Wan
- Tongling Institutes for Food and Drug Control, Tongling, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Anhui Province, China
| |
Collapse
|
5
|
Xie X, Wei G, Tang Z, Chen H, Lin X, Huang C, Yu H, He Y, Li M, Zhang X, He C, He Y, Chen J. Investigating the causal relationship between rheumatoid arthritis and cardiovascular disease: A Mendelian randomization study. Clin Rheumatol 2025; 44:1057-1067. [PMID: 39909965 DOI: 10.1007/s10067-025-07357-4] [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: 06/11/2024] [Revised: 01/13/2025] [Accepted: 01/18/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVE Previous research has revealed a positive correlation between rheumatoid arthritis (RA) and cardiovascular diseases, but the causal relationship is unclear. This study applies Mendelian randomization to examine whether RA causally contributes to the likelihood of various cardiovascular diseases, such as heart failure, coronary artery disease, and atrial fibrillation. METHODS Using genome-wide association data, we conducted a univariable MR (UVMR) analysis to evaluate the causal impact of RA on CVD, primarily utilizing the inverse variance weighted method. Additional MR methods were used to test the robustness of the results. Multivariable MR (MVMR) was applied to explore potential confounders. RESULTS In the European population, genetically predicted RA had a harmful causal effect on HF, with the IVW analysis indicating an OR of 1.06 (95% CI: 1.02-1.10, P < 0.01) based on 23 SNPs. No causal relationships were found between RA and other CVDs. The MVMR analysis did not identify significant causal impact of rheumatoid arthritis on HF after controlling for traditional risk factors. In the Asian population, RA was associated with an adverse effect on AF, with the IVW method reporting an OR of 1.20 (95% CI: 1.01-1.41, P = 0.03) for 5 SNPs. No other CVD relationships were found. CONCLUSIONS Our MR analysis indicates that genetic susceptibility to rheumatoid arthritis increases the likelihood of heart failure in European populations and atrial fibrillation in East Asian populations. However, established CVD risk factors, such as smoking, overweight, and physical inactivity, remain critically important in the management of RA. Key Points • Multiple studies have highlighted a marked increase in the cardiovascular event risk among individuals with RA. However, additional RCTs are needed for confirmation. • We applied Mendelian randomization to explore the potential causal relationship between rheumatoid arthritis and cardiovascular conditions. The findings demonstrated a causal link between RA and heart failure among European populations, as well as an association between RA and atrial fibrillation in East Asian groups. • Further adjustments using multivariable Mendelian randomization to account for the influence of traditional cardiovascular risk factors revealed that the causal association between RA and heart failure disappeared.
Collapse
Affiliation(s)
- Xintong Xie
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Guangliang Wei
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Zhenboyang Tang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Huidong Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Xiru Lin
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Chunyan Huang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Hao Yu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Youxian He
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Mengxiang Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Xue Zhang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Chengsong He
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China
| | - Yue He
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Jie Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, People's Republic of China.
- Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, People's Republic of China.
| |
Collapse
|
6
|
Wu T, Li Y, Liu Y, Chu CQ. Preclinical RA: How to halt its progression. Best Pract Res Clin Rheumatol 2025; 39:102030. [PMID: 39721896 DOI: 10.1016/j.berh.2024.102030] [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: 12/04/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder with a complex pathogenesis that evolves through various stages before clinical symptoms emerge. This review outlines the natural history of RA, starting from genetic predisposition and environmental triggers to preclinical autoimmunity and subsequent joint inflammation. Key genetic factors interact with environmental elements like smoking and infections, producing autoantibodies such as anti-citrullinated protein antibodies (ACPA) and rheumatoid factor, which precede clinical manifestations by several years. The preclinical phases offer critical opportunities for intervention aiming at halting disease progression. Preventive strategies including lifestyle modifications, dietary interventions, and targeted immune modulation may halt the progression to clinical RA in those at-risk individuals.
Collapse
Affiliation(s)
- Tong Wu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Lecheng Hospital, Sichuan University, Boao, Hainan, 571435, China.
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, 97239, USA; Rheumatology Section, VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
7
|
Wang X, Yue J, Guo S, Rahmatulla A, Li S, Liu Y, Chen Y. Dissolving microneedles: A transdermal drug delivery system for the treatment of rheumatoid arthritis. Int J Pharm 2025; 671:125206. [PMID: 39799999 DOI: 10.1016/j.ijpharm.2025.125206] [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: 11/16/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder that impacts around 1% of the global population. Up to 20% of people become disabled within a year, which has a severely negative impact on their health and quality of life. RA has a complicated pathogenic mechanism, which initially affects small joints and progresses to larger ones over time. It can damage the skin, eyes, heart, kidney, and lung. Oral medications, intra-articular injections, and other treatments are being used; nevertheless, they have drawbacks, including low bioavailability, numerous adverse effects, and poor patient compliance. Dissolving microneedles (DMNs) are a safe and painless method for transdermal drug delivery, achieved through their ability to physically penetrate the epidermal barrier. They enable targeted drug delivery, significantly enhancing the bioavailability of medications and improving patient compliance. DMNs are particularly effective in delivering both lipophilic and high molecular weight biomolecules. The superior bioavailability of DMNs is demonstrated by the fact that low-dose DMN administration can achieve up to 25.8 times higher bioavailability compared to oral administration. This paper provides a comprehensive review of recent advancements in the use of DMNs for RA treatment, encompassing various materials (such as hyaluronic acid, chitosan, etc.), fabrication techniques (such as the two-step casting method, photopolymerization), and performance evaluations (including morphology, mechanical properties, skin penetration capability, solubility, and pharmacodynamics). Additionally, a thorough safety assessment has been conducted, revealing that DMNs cause minimal skin irritation and exhibit low cytotoxicity, ensuring their safety for clinical application. DMNs provide a highly effective and promising alternative to oral and injectable drug delivery systems, offering a novel therapeutic approach for RA patients that significantly improves treatment outcomes and enhances their quality of life.
Collapse
Affiliation(s)
- Xueni Wang
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Jiang Yue
- Department of Endocrinology and Metabolism Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Shijie Guo
- Shengzhou Silk Protein Biotechnology Application Research Institute Zhejiang China
| | - Aysha Rahmatulla
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Shuangshuang Li
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Yang Liu
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China.
| | - Yuzhou Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China.
| |
Collapse
|
8
|
Gardette A, Marzaioli V, Bedouhene S, Hayem G, Hurtado-Nedelec M, He Y, Dang PMC, Dieudé P, Zhang ZY, Marie JC, El-Benna J. The protein tyrosine phosphatase Lyp/PTPN22 drives TNFα-induced priming of superoxide anions production by neutrophils and arthritis. Free Radic Biol Med 2025; 228:68-78. [PMID: 39724988 DOI: 10.1016/j.freeradbiomed.2024.12.046] [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: 10/08/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Neutrophils are essential for host defense against infections, but they also play a key role in acute and chronic inflammation. The protein tyrosine phosphatase non-receptor type 22 (PTPN22) gene encodes the lymphoid-specific tyrosine phosphatase (Lyp) and a genetic single-nucleotide polymorphism of PTPN22 rs2476601 (R620W) has been associated with several human autoimmune diseases, including rheumatoid arthritis (RA). Here, we investigated the role of Lyp in TNFα-induced priming of neutrophil ROS production and in the development of arthritis using new selective Lyp inhibitors. Results show that Lyp-selective inhibitors (IC-11 and compound 8b), inhibited TNFα-induced priming of neutrophil superoxide anion production. TNFα induced an increase of Lyp protein expression in human neutrophils isolated from healthy donors. Key pathways involved in neutrophil priming were investigated. Lyp-selective inhibitors, inhibited TNFα-induced p47phox phosphorylation on Ser345, ERK1/2 phosphorylation and Pin1 activation. Interestingly, Lyp expression was increased in neutrophils isolated from synovial fluid of RA patients and Lyp inhibitors, I-C11 and compound 8b prevented superoxide anion production by endogenously primed neutrophils isolated from synovial fluid of RA. Moreover, IC-11 significantly prevented collagen antibody-induced arthritis in mice. These results show that Lyp expression is increased in inflammatory neutrophils, Lyp is involved in TNFα-induced excessive ROS production by neutrophils and its inhibition protected mice against arthritis. Inhibition of Lyp could be a therapeutic strategy in inflammatory arthritis.
Collapse
Affiliation(s)
- Anaïs Gardette
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France; Service de Rhumatologie, Hôpital Bichat, APHP, Paris, France
| | - Viviana Marzaioli
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France; Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Samia Bedouhene
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Gilles Hayem
- Rheumatology Department, Paris-Saint Joseph Hospital Group, Paris, France
| | - Margarita Hurtado-Nedelec
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France; Service d'Hématologie et Immunologie, Hôpital Bichat, APHP, Paris, France
| | - Yantao He
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Pham My-Chan Dang
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Philippe Dieudé
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France; Service de Rhumatologie, Hôpital Bichat, APHP, Paris, France
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Jean-Claude Marie
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Jamel El-Benna
- INSERM-U1149, CNRS-ERL8252, Université de Paris-Cité, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Paris, France.
| |
Collapse
|
9
|
Qin Q, Jiang Y, Fan H, Yuan R, Zhong B, Zhang Y, Zhang Z, Lei X, Cai J, Cheng S. Investigating the shared genetic structure between rheumatoid arthritis and stroke. Hereditas 2025; 162:23. [PMID: 39953635 PMCID: PMC11827134 DOI: 10.1186/s41065-025-00386-8] [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: 10/15/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) increases the risk of stroke. However, the relationship between RA and stroke remains unclear. This study aimed to explore the shared genetics architecture (i.e., common genetic basis between different traits, diseases, or phenotypes) of RA and stroke, aiming to improve the intervention and management of patients with RA and stroke. METHODS Pooled statistics from publicly available genome-wide association studies for RA (8,255 cases and 409,001 controls) and stroke (43,132 cases and 43,132 controls) were used. A genome-wide positive association was conducted to (examine the comprehensive effects of genetic variants on a particular trait, disease, or phenotype at the genome-wide scale). Local genetic correlation studies used linkage disequilibrium score regression and super genetic covariance analyzer. Single nucleotide polymorphisms (SNPs) at risk were identified using genome-wide association study multiple trait analysis and PLINK software (Psnp <5e-08), followed by functional localization and annotation using Functional Mapping and Annotation of Genome-Wide Association Studies to identify specific genes and genetic variants that may contribute to the disease. Finally, a transcriptome-wide association study explored the relationship between genes and their association with RA risk. RESULTS A genome-wide significant positive correlation was evident between RA and stroke (genetic correlation = 0.3756). Among the localized genomic regions, the correlation between RA and stroke in the region of chr2:201572564-202,829,668 was the most significant (p = 0.0015). We identified 179 significant SNPs and five common risk genes for RA and stroke (IRF5, RNASET2, ZNF438, UBE2LS, and SYNGR1). These genes are involved in the immune-inflammatory pathway. CONCLUSIONS The findings suggest a shared genetic structure between RA and stroke. These findings may provide new insights into RA and stroke pathogenesis, and contribute to the development of new diagnostic markers and therapeutic targeted drugs to improve the clinical outcomes of patients with RA and stroke.
Collapse
Affiliation(s)
- Qian Qin
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
- Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
| | - Yong'An Jiang
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
- Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
| | - Hengyi Fan
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
| | - Raorao Yuan
- Department of Critical Care Medicine, Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bo Zhong
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
- Department of Neurosurgery, Xinyu People's Hospital, Xinyu, 338000, Jiangxi, P. R. China
| | - Yichen Zhang
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
- Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
| | - Zile Zhang
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
- Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
| | - Xin Lei
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
- Nanchang University, Nanchang, 330006, Jiangxi, P. R. China
| | - Jianhui Cai
- Department of Neurosurgery, Nanchang County People's Hospital, Nanchang, 330200, Jiangxi, P. R. China.
- Nanchang Cranio-Cerebral Trauma Laboratory, Nanchang, 330200, Jiangxi, P. R. China.
| | - Shiqi Cheng
- Department of Neurosurgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, P. R. China.
| |
Collapse
|
10
|
Jassim BA, Bai Y, Qu Z, Sander CJ, Lin J, Miao J, Zhang ZY. Structure-activity relationship studies and design of a PTPN22 inhibitor with enhanced isozyme selectivity and cellular efficacy. Eur J Med Chem 2025; 283:117129. [PMID: 39693863 PMCID: PMC11709133 DOI: 10.1016/j.ejmech.2024.117129] [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: 10/15/2024] [Revised: 11/28/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024]
Abstract
Protein tyrosine phosphatase non-receptor type 22 (PTPN22) lies downstream of the T cell receptor (TCR) and attenuates T cell signaling by dephosphorylating key effector proteins such as LCK, Zap70, and the intracellular region of the TCR. Recent evidence implicates PTPN22 as an exciting target for enabling immunotherapeutic efficacy against cancer. We carried out structural optimization of a benzofuran salicylic acid-based orthosteric PTPN22 inhibitor 8b, using a combination of crystal structure analysis, synthesis, matched molecular pairs analysis, and biochemical and cell-based assays. Herein, we report structure-activity relationship studies, lead optimization based on the 8b-PTPN22 co-crystal structure, and cellular evaluation of the top analog. Notably, our efforts yielded compound 8b-19, an essentially equipotent scaffold with superior isozyme selectivity, improved aqueous solubility, and significantly enhanced cellular efficacy compared to the parent 8b. This compound may serve as a lead for further optimization of PTPN22-targeting immunotherapies or as a chemical probe for interrogation for additional links between PTPN22 and immunomodulation in cells.
Collapse
Affiliation(s)
- Brenson A Jassim
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Yunpeng Bai
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Zihan Qu
- James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Conrad J Sander
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Jianping Lin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Jinmin Miao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; Institute for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
11
|
Shakeel L, Shaukat A, Khaliq N, Kashif A, Mujeeb A, Adnan Z, Taj J, Akilimali A. Rheumatoid arthritis: a comprehensive overview of genetic markers, emerging therapies, and personalized medicine. Ann Med Surg (Lond) 2025; 87:696-710. [PMID: 40110258 PMCID: PMC11918739 DOI: 10.1097/ms9.0000000000002890] [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: 09/13/2024] [Accepted: 12/09/2024] [Indexed: 03/22/2025] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent autoimmune disorder marked by chronic inflammatory arthritis and systemic effects. The etiology of RA is complex, involving genetic factors like HLA-DR4 and HLA-DR1, as well as environmental influences, particularly smoking, which heightens disease risk. Affecting approximately 1% of the global population, RA is associated with considerable morbidity and mortality, with its prevalence expected to increase due to demographic shifts, especially in certain regions. RA symptoms commonly manifest between ages 35 and 60 but can also affect children under 16 in cases of juvenile RA. Symptoms include prolonged joint stiffness, pain, fatigue, and, in advanced cases, joint deformities. Current treatment approaches involve disease-modifying antirheumatic drugs, biologics, and glucocorticoids to manage symptoms and slow disease progression, though these treatments often present limitations due to adverse effects and varied patient response. The identification of genetic markers, such as HLA-DRB1 and PTPN22, supports the growing emphasis on personalized treatment strategies that account for genetic and lifestyle factors. Non-pharmacological approaches - diet adjustments, physical activity, and stress management - are increasingly valued for their complementary role in RA management. Lifestyle interventions, including whole-food, plant-based diets and physical therapy, show promise in reducing inflammation and improving joint function. Technological advancements, like telemedicine, mobile health applications, and artificial intelligence, are enhancing RA diagnosis and treatment, making care more precise and accessible. Despite these advancements, RA remains incurable, necessitating continued research into novel therapeutic targets and approaches. A comprehensive, patient-centered approach that integrates lifestyle modifications, preventive strategies, and innovative treatments is essential for improving RA management and patient outcomes.
Collapse
Affiliation(s)
- Laiba Shakeel
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Ayesha Shaukat
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Nawal Khaliq
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Aayat Kashif
- Department of Internal Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Azka Mujeeb
- Department of Internal Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Zahabia Adnan
- Department of Internal Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Javeria Taj
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Aymar Akilimali
- Department of research, Medical Research circle, Goma, Democratic Republic of the Congo
| |
Collapse
|
12
|
Ates I, Terzi U, Suzen S, Irham LM. An overview on Sjögren's syndrome and systemic lupus erythematosus' genetics. Toxicol Res (Camb) 2025; 14:tfae194. [PMID: 39991010 PMCID: PMC11847510 DOI: 10.1093/toxres/tfae194] [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: 08/19/2024] [Revised: 10/13/2024] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
Major autoimmune rheumatic disorders, such as systemic lupus erythematosus and Sjögren's syndrome, are defined by the presence of autoantibodies. These diseases are brought on by immune system dysregulation, which can present clinically in a wide range of ways. The etiologies of these illnesses are complex and heavily impacted by a variety of genetic and environmental variables. The most powerful susceptibility element for each of these disorders is still the human leukocyte antigen (HLA) area, that was the initial locus found to be associated. This region is primarily responsible for the HLA class II genes, such as DQA1, DQB1, and DRB1, however class I genes have also been linked. Numerous genetic variants that do not pose a risk to HLA have been found as a result of intensive research into the genetic component of these diseases conducted over the last 20 years. Furthermore, it is generally acknowledged that autoimmune rheumatic illnesses have similar genetic backgrounds and share molecular pathways of disease, including the interferon (IFN) type I routes. Pleiotropic sites for autoimmune rheumatic illnesses comprise TNIP1, DNASEL13, IRF5, the HLA region, and others. It remains a challenge to determine the causative biological mechanisms beneath the genetic connections. Nonetheless, functional analyses of the loci and mouse models have produced recent advancements. With an emphasis on the HLA region, we present an updated summary of the structure of genes underpinning both of these autoimmune rheumatic illnesses here.
Collapse
Affiliation(s)
- Ilker Ates
- Department of Toxicology, Ankara University, Faculty of Pharmacy, Emniyet Distr, Degol Str, No. 4, 06560 Yenimahalle, Ankara, Turkey
| | - Ulku Terzi
- Department of Toxicology, Ankara University, Faculty of Pharmacy, Emniyet Distr, Degol Str, No. 4, 06560 Yenimahalle, Ankara, Turkey
| | - Sinan Suzen
- Department of Toxicology, Ankara University, Faculty of Pharmacy, Emniyet Distr, Degol Str, No. 4, 06560 Yenimahalle, Ankara, Turkey
| | - Lalu Muhammad Irham
- Department of Toxicology, Ahmad Dahlan University, Faculty of Pharmacy, Prof. Dr. Soepomo, S.H., Street, Warungboto, 55164, Yogyakarta, Indonesia
| |
Collapse
|
13
|
Sahin D, Di Matteo A, Emery P. Biomarkers in the diagnosis, prognosis and management of rheumatoid arthritis: A comprehensive review. Ann Clin Biochem 2025; 62:3-21. [PMID: 39242085 PMCID: PMC11707974 DOI: 10.1177/00045632241285843] [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] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic, autoimmune condition that primarily affects the joints and periarticular soft tissues. In the past two decades, the discovery of new biomarkers has contributed to advances in the understanding of the pathogenesis and natural history of RA. These biomarkers, including genetic, clinical, serological and imaging biomarkers, play a key role in the different stages and aspects of RA, from the so called 'pre-clinical RA', which is characterized by subclinical pathological events, such as autoimmunity and inflammation, to diagnosis (including differential diagnosis), treatment decision making and disease monitoring.This review will provide an overview on the current role of traditional and newer biomarkers in the main aspects of RA management, from the identification of individuals 'at-risk' of RA who are likely to progress to clinically evident disease, to 'early' diagnosis of RA, prognosis, precision medicine, and prediction of response to treatment.
Collapse
Affiliation(s)
- Didem Sahin
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Andrea Di Matteo
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Paul Emery
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals, NHS Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
14
|
Makhija M, Manchanda D, Sharma M. Nano-based Therapeutics for Rheumatoid Arthritis: Recent Patents and Development. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:56-75. [PMID: 37691226 DOI: 10.2174/1872210518666230905155459] [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: 02/22/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease marked by inflammation of synovium and generation of autoantibodies. Bone and cartilage are frequently damaged along with weakening of tendons and ligaments resulting in disability. An effective RA treatment needs a multi-disciplinary approach which relies upon pathophysiology that is still partially understood. In RA patients, inflammation was induced by pro-inflammatory cytokines including IL-1, IL-6 & IL-10. The conventional dosage regimens for treating RA have drawbacks such as ineffectiveness, greater doses, frequent dosing, relatively expensive and serious adverse effects. To formulate an effective treatment plan for RA, research teams have recently focused on producing several nanoformulations containing anti-inflammatory APIs with an aim to target the inflamed area. Nanomedicines have recently gained popularity in the treatment of RA. Interestingly, unbelievable improvements have been observed in current years in diagnosis and management of RA utilizing nanotechnology. Various patents and clinical trial data have been reported in relevance to RA treatment.
Collapse
Affiliation(s)
- Manish Makhija
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Deeksha Manchanda
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Manu Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
| |
Collapse
|
15
|
Hasan M, Sarker MN, Jabin T, Sarker S, Ahmed S, Abdullah-Al-Shoeb M, Hossain T. Pathogenic single nucleotide polymorphisms in RhoA gene: Insights into structural and functional impacts on RhoA-PLD1 interaction through molecular dynamics simulation. Curr Res Struct Biol 2024; 8:100159. [PMID: 39698059 PMCID: PMC11653153 DOI: 10.1016/j.crstbi.2024.100159] [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: 08/21/2024] [Revised: 11/03/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
Molecular switches serve as key regulators of biological systems by acting as one of the crucial driving forces in the initiation of signal transduction pathway cascades. The Ras homolog gene family member A (RhoA) is one of the molecular switches that binds with GTP in order to cycle between an active GTP-bound state and an inactive GDP-bound state. Any aberrance in control over this circuit, particularly due to any perturbation in switching, leads to the development of different pathogenicity. Consequently, the single nucleotide polymorphisms (SNPs) within the RhoA gene, especially deleterious genetic variations, are crucial to study to forecast structural alteration and their functional impacts in light of disease onset. In this comprehensive study, we employed a range of computational tools to screen the deleterious SNPs of RhoA from 207 nonsynonymous SNPs (nsSNPs). By utilizing 7 distinct tools for further analysis, 8 common deleterious SNPs were sorted, among them 5 nsSNPs (V9G, G17E, E40K, A61T, F171L) were found to be in the highly conserved regions, with E40K and A61T at G2 and G3 motif of the GTP-binding domain respectively, indicating potential perturbation in GTP/GDP binding ability of the protein. RhoA-GDP complex interacts with the enzyme phospholipase, specifically PLD1, to regulate different cellular activities. PLD1 is also a crucial regulator of thrombosis and cancer. In that line of focus, our initial structural analysis of Y66H, A61T, G17E, I86N, and I151T mutations of RhoA revealed remarkable decreased hydrophobicity from which we further filtered out G17E and I86N which may have potential impact on the RhoA-GDP-PLD1 complex. Intriguingly, the comparative 250 ns (ns) molecular dynamics (MD) simulation of these two mutated complexes revealed overall structural instability and altered interaction patterns. Therefore, further investigation into these deleterious mutations with in vitro and in vivo studies could lead to the identification of potential biomarkers in terms of different pathogenesis and could also be utilized in personalized therapeutic targets in the long run.
Collapse
Affiliation(s)
| | | | | | - Saifuddin Sarker
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Shamim Ahmed
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mohammad Abdullah-Al-Shoeb
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Tanvir Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| |
Collapse
|
16
|
Srivastava S, Rasool M. Genetics, epigenetics and autoimmunity constitute a Bermuda triangle for the pathogenesis of rheumatoid arthritis. Life Sci 2024; 357:123075. [PMID: 39341491 DOI: 10.1016/j.lfs.2024.123075] [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: 07/23/2024] [Revised: 09/13/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
Rheumatoid arthritis (RA), a multigene disorder with a heritability rate of 60 %, is characterized by persistent pain, synovial hyperplasia, and cartilage and bone destruction, ultimately causing irreversible joint deformity. The etiology and pathogenesis of rheumatoid arthritis (RA) are primarily influenced by specific genetic variants, particularly HLA alleles such as HLA-DRB1*01 and DRB1*04. However, other HLA alleles such as HLA-DRB1*10 and DPB*1 have also been found to contribute to increased susceptibility to RA. However, non-HLA genes also confer a comparatively high risk of RA disease manifestation. The most relevant single nucleotide polymorphisms (SNPs) associated with non-HLA genes are PTPN22, TRAF1, CXCL-12, TBX-5, STAT4, FCGR, PADI4, and MTHFR. In conjunction with genetic susceptibility, epigenetic alterations orchestrate paramount involvement in regulating RA pathogenesis. Increasing evidence implicates DNA methylation and histone protein modifications, including acetylation and methylation, as the primary epigenetic mechanisms that drive the pathogenesis and clinical progression of the disease. In addition to genetic and epigenetic changes, autoimmune inflammation also determines the pathological progression of the synovial membrane in joints with RA. Glycosylation changes, such as sialylation and fucosylation, in immune cells have been shown to be relevant to disease progression. Genetic heterogeneity, epigenetic factors, and changes in glycosylation do not fully explain the features of RA. Therefore, investigating the interplay between genetics, epigenetics, and autoimmunity is crucial. This review highlights the significance and interaction of these elements in RA pathophysiology, suggesting their diagnostic potential and opening new avenues for novel therapeutic approaches.
Collapse
Affiliation(s)
- Susmita Srivastava
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, Tamil Nadu, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
17
|
Acero-Bedoya S, Higgs EF, Martinez AC, Tonea R, Gajewski TF. Dendritic cell-intrinsic PTPN22 negatively regulates antitumor immunity and impacts anti-PD-L1 efficacy. J Immunother Cancer 2024; 12:e009588. [PMID: 39461876 PMCID: PMC11529514 DOI: 10.1136/jitc-2024-009588] [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] [Accepted: 10/02/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Individuals with a loss-of-function single-nucleotide polymorphism in the gene encoding PTPN22 have an increased risk for autoimmune diseases, and patients with cancer with such alleles may respond better to checkpoint blockade immunotherapy. Studies in PTPN22 knockout (KO) mice have established it as a negative regulator of T cell responses in cancer models. However, the role of PTPN22 in distinct immune cell compartments, such as dendritic cells (DCs), remains undefined. METHODS We developed a novel PTPN22 conditional KO (cKO) mouse model that enables specific deletion in CD11c+ DCs by crossing to CD11c-Cre transgenic mice. Antitumor immunity was characterized using the B16.SIY and MC38.SIY cancer models and immune profiles of relevant tissues were evaluated by spectral flow cytometry. Antigen uptake, processing, and presentation, as well as DC proliferation to Flt3L, were characterized ex vivo. RESULTS Deletion of PTPN22 in DCs resulted in augmented antitumor immunity in multiple syngeneic tumor models. Tumor antigen-specific CD8+ T cells were increased in the tumor microenvironment (TME) of PTPN22 cKO mice and improved tumor control was CD8+ T cell-dependent. Augmented T cell priming was also detected at early time points in the draining lymph nodes, and these effects were correlated with an increased number of proliferating CD103+ DCs, also seen in the TME. In vitro studies revealed increased DC proliferation in response to Flt3L, as well as increased antigen processing and presentation. PTPN22 cKO mice bearing MC38 parental tumors showed combinatorial benefit with anti-PD-L1 therapy. CONCLUSIONS Deletion of PTPN22 in DCs is sufficient to drive an augmented tumor antigen-specific T cell response, resulting in enhanced tumor control. PTPN22 negatively regulates DC proliferation and antigen processing and presentation. Our work argues that PTPN22 is an attractive therapeutic target for cancer immunotherapy and highlights the potential to modulate antitumor immunity through the manipulation of DC signaling.
Collapse
Affiliation(s)
- Santiago Acero-Bedoya
- Pathology, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Emily F Higgs
- Pathology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Anna C Martinez
- Pathology, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Ruxandra Tonea
- Pathology, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - Thomas F Gajewski
- Pathology and Medicine, The University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| |
Collapse
|
18
|
Tanshee RR, Mahmud Z, Nabi AHMN, Sayem M. A comprehensive in silico investigation into the pathogenic SNPs in the RTEL1 gene and their biological consequences. PLoS One 2024; 19:e0309713. [PMID: 39240887 PMCID: PMC11379182 DOI: 10.1371/journal.pone.0309713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/16/2024] [Indexed: 09/08/2024] Open
Abstract
The Regulator of Telomere Helicase 1 (RTEL1) gene encodes a critical DNA helicase intricately involved in the maintenance of telomeric structures and the preservation of genomic stability. Germline mutations in the RTEL1 gene have been clinically associated with Hoyeraal-Hreidarsson syndrome, a more severe version of Dyskeratosis Congenita. Although various research has sought to link RTEL1 mutations to specific disorders, no comprehensive investigation has yet been conducted on missense mutations. In this study, we attempted to investigate the functionally and structurally deleterious coding and non-coding SNPs of the RTEL1 gene using an in silico approach. Initially, out of 1392 nsSNPs, 43 nsSNPs were filtered out through ten web-based bioinformatics tools. With subsequent analysis using nine in silico tools, these 43 nsSNPs were further shortened to 11 most deleterious nsSNPs. Furthermore, analyses of mutated protein structures, evolutionary conservancy, surface accessibility, domains & PTM sites, cancer susceptibility, and interatomic interaction revealed the detrimental effect of these 11 nsSNPs on RTEL1 protein. An in-depth investigation through molecular docking with the DNA binding sequence demonstrated a striking change in the interaction pattern for F15L, M25V, and G706R mutant proteins, suggesting the more severe consequences of these mutations on protein structure and functionality. Among the non-coding variants, two had the highest likelihood of being regulatory variants, whereas one variant was predicted to affect the target region of a miRNA. Thus, this study lays the groundwork for extensive analysis of RTEL1 gene variants in the future, along with the advancement of precision medicine and other treatment modalities.
Collapse
Affiliation(s)
- Rifah Rownak Tanshee
- Department of Mathematics and Natural Sciences, BRAC University, Badda, Dhaka, Bangladesh
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - A. H. M. Nurun Nabi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Mohammad Sayem
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
19
|
Lee J, Yurkovetskiy LA, Reiman D, Frommer L, Strong Z, Chang A, Kahaly GJ, Khan AA, Chervonsky AV. Androgens contribute to sex bias of autoimmunity in mice by T cell-intrinsic regulation of Ptpn22 phosphatase expression. Nat Commun 2024; 15:7688. [PMID: 39227386 PMCID: PMC11372096 DOI: 10.1038/s41467-024-51869-7] [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: 05/04/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024] Open
Abstract
Autoimmune diseases such as systemic lupus erythematosus (SLE) display a strong female bias. Although sex hormones have been associated with protecting males from autoimmunity, the molecular mechanisms are incompletely understood. Here we report that androgen receptor (AR) expressed in T cells regulates genes involved in T cell activation directly, or indirectly via controlling other transcription factors. T cell-specific deletion of AR in mice leads to T cell activation and enhanced autoimmunity in male mice. Mechanistically, Ptpn22, a phosphatase and negative regulator of T cell receptor signaling, is downregulated in AR-deficient T cells. Moreover, a conserved androgen-response element is found in the regulatory region of Ptpn22 gene, and the mutation of this transcription element in non-obese diabetic mice increases the incidence of spontaneous and inducible diabetes in male mice. Lastly, Ptpn22 deficiency increases the disease severity of male mice in a mouse model of SLE. Our results thus implicate AR-regulated genes such as PTPN22 as potential therapeutic targets for autoimmune diseases.
Collapse
MESH Headings
- Animals
- Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism
- Male
- Female
- Autoimmunity
- Receptors, Androgen/metabolism
- Receptors, Androgen/genetics
- Mice
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/genetics
- Androgens/metabolism
- Mice, Knockout
- Lymphocyte Activation
- Mice, Inbred NOD
- Mice, Inbred C57BL
- Disease Models, Animal
- Signal Transduction
Collapse
Affiliation(s)
- Jean Lee
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, 60637, USA
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - Leonid A Yurkovetskiy
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
- Committee on Microbiology, The University of Chicago, Chicago, IL, 60637, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Derek Reiman
- Toyota Technological Institute at Chicago, Chicago, IL, 60637, USA
| | - Lara Frommer
- Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, 55101, Germany
| | - Zoe Strong
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - Anthony Chang
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - George J Kahaly
- Department of Medicine I, Johannes Gutenberg University (JGU) Medical Center, Mainz, 55101, Germany
| | - Aly A Khan
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA.
- Toyota Technological Institute at Chicago, Chicago, IL, 60637, USA.
- Department of Family Medicine, The University of Chicago, Chicago, IL, 60637, USA.
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA.
| | - Alexander V Chervonsky
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA.
- Committee on Microbiology, The University of Chicago, Chicago, IL, 60637, USA.
- Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
20
|
Shen J, Pan J, Yu G, Cai H, Xu H, Yan H, Feng Y. Genetic interactions and pleiotropy in metabolic diseases: Insights from a comprehensive GWAS analysis. J Cell Mol Med 2024; 28:e70045. [PMID: 39238070 PMCID: PMC11377178 DOI: 10.1111/jcmm.70045] [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: 01/29/2024] [Revised: 07/27/2024] [Accepted: 08/16/2024] [Indexed: 09/07/2024] Open
Abstract
This study offers insights into the genetic and biological connections between nine common metabolic diseases using data from genome-wide association studies. Our goal is to unravel the genetic interactions and biological pathways of these complex diseases, enhancing our understanding of their genetic architecture. We employed a range of advanced analytical techniques to explore the genetic correlations and shared genetic variants of these diseases. These methods include Linked Disequilibrium Score Regression, High-Definition Likelihood (HDL), genetic analysis combining multiplicity and annotation (GPA), two-sample Mendelian randomization analyses, analysis under the multiplicity-complex null hypothesis (PLACO), and Functional mapping and annotation of genetic associations (FUMA). Additionally, Bayesian co-localization analyses were used to examine associations of specific loci across traits. Our study discovered significant genomic correlations and shared loci, indicating complex genetic interactions among these metabolic diseases. We found several shared single nucleotide variants and risk loci, notably highlighting the role of the immune system and endocrine pathways in these diseases. Particularly, rs2476601 and its associated gene PTPN22 appear to play a crucial role in the connection between type 2 diabetes mellitus, hypothyroidism/mucous oedema and hypoglycaemia. These findings enhance our understanding of the genetic underpinnings of these diseases and open new potential avenues for targeted therapeutic and preventive strategies. The results underscore the importance of considering pleiotropic effects in deciphering the genetic architecture of complex diseases, especially metabolic ones.
Collapse
Affiliation(s)
- Jing Shen
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Julong Pan
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Gang Yu
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Hui Cai
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Hua Xu
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Hanfei Yan
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Yu Feng
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
- The University of New South Wales, Sydney, New South Wales, Australia
- The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Binti S, Linder AG, Edeen PT, Fay DS. A conserved protein tyrosine phosphatase, PTPN-22, functions in diverse developmental processes in C. elegans. PLoS Genet 2024; 20:e1011219. [PMID: 39173071 PMCID: PMC11373843 DOI: 10.1371/journal.pgen.1011219] [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: 03/11/2024] [Revised: 09/04/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Abstract
Protein tyrosine phosphatases non-receptor type (PTPNs) have been studied extensively in the context of the adaptive immune system; however, their roles beyond immunoregulation are less well explored. Here we identify novel functions for the conserved C. elegans phosphatase PTPN-22, establishing its role in nematode molting, cell adhesion, and cytoskeletal regulation. Through a non-biased genetic screen, we found that loss of PTPN-22 phosphatase activity suppressed molting defects caused by loss-of-function mutations in the conserved NIMA-related kinases NEKL-2 (human NEK8/NEK9) and NEKL-3 (human NEK6/NEK7), which act at the interface of membrane trafficking and actin regulation. To better understand the functions of PTPN-22, we carried out proximity labeling studies to identify candidate interactors of PTPN-22 during development. Through this approach we identified the CDC42 guanine-nucleotide exchange factor DNBP-1 (human DNMBP) as an in vivo partner of PTPN-22. Consistent with this interaction, loss of DNBP-1 also suppressed nekl-associated molting defects. Genetic analysis, co-localization studies, and proximity labeling revealed roles for PTPN-22 in several epidermal adhesion complexes, including C. elegans hemidesmosomes, suggesting that PTPN-22 plays a broad role in maintaining the structural integrity of tissues. Localization and proximity labeling also implicated PTPN-22 in functions connected to nucleocytoplasmic transport and mRNA regulation, particularly within the germline, as nearly one-third of proteins identified by PTPN-22 proximity labeling are known P granule components. Collectively, these studies highlight the utility of combined genetic and proteomic approaches for identifying novel gene functions.
Collapse
Affiliation(s)
- Shaonil Binti
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Adison G. Linder
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Philip T. Edeen
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - David S. Fay
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| |
Collapse
|
22
|
Mastana S, Knight E, Hampson A, Akam L, Hunter DJ, Ghelani A, Samanta A, Singh P. Role of Selected Genetic Polymorphisms in the Development of Rheumatoid Arthritis in a British White Population. Genes (Basel) 2024; 15:1009. [PMID: 39202369 PMCID: PMC11354150 DOI: 10.3390/genes15081009] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a complex autoimmune disease that negatively affects synovial joints, leading to the deterioration of movement and mobility of patients. This chronic disease is considered to have a strong genetic inheritance, with genome-wide association studies (GWAS) highlighting many genetic loci associated with the disease. Moreover, numerous confounding and non-genetic factors also contribute to the risk of the disease. AIMS This study investigates the association of selected genetic polymorphisms with rheumatoid arthritis risk and develops a polygenic risk score (PRS) based on selected genes. METHODS A case-control study recruited fully consenting participants from the East Midlands region of the UK. DNA samples were genotyped for a range of polymorphisms and genetic associations were calculated under several inheritance models. PRS was calculated at crude (unweighted) and weighted levels, and its associations with clinical parameters were determined. RESULTS There were significant associations with the risk of RA at six genetic markers and their associated risk alleles (TNRF2*G, TRAF1*A, PTPN22*T, HLA-DRB1*G, TNFα*A, and IL4-590*T). The TTG haplotype at the VDR locus increased the risk of RA with an OR of 3.05 (CI 1.33-6.98, p = 0.009). The GA haplotype of HLADRB1-TNFα-308 was a significant contributor to the risk of RA in this population (OR = 2.77, CI 1.23-6.28, p = 0.01), although linkage disequilibrium was low. The polygenic risk score was significantly higher in cases over controls in both unweighted (mean difference = 1.48, t285 = 5.387, p < 0.001) and weighted (mean difference = 2.75, t285 = 6.437, p < 0.001) results. CONCLUSION Several genetic loci contribute to the increased risk of RA in the British White sample. The PRS is significantly higher in those with RA and can be used for clinical applications and personalised prevention of disease.
Collapse
Affiliation(s)
- Sarabjit Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Ella Knight
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Abigail Hampson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Liz Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - David John Hunter
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Anant Ghelani
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK; (E.K.); (A.H.); (L.A.); (D.J.H.); (A.G.)
| | - Ash Samanta
- Rheumatology, University Hospitals of Leicester NHS Trust, Leicester LE1 5WW, UK;
| | - Puneetpal Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India;
| |
Collapse
|
23
|
Chasov V, Ganeeva I, Zmievskaya E, Davletshin D, Gilyazova E, Valiullina A, Bulatov E. Cell-Based Therapy and Genome Editing as Emerging Therapeutic Approaches to Treat Rheumatoid Arthritis. Cells 2024; 13:1282. [PMID: 39120313 PMCID: PMC11312096 DOI: 10.3390/cells13151282] [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: 06/17/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of the joints. Although much remains unknown about the pathogenesis of RA, there is evidence that impaired immune tolerance and the development of RA are related. And it is precisely the restoration of immune tolerance at the site of the inflammation that is the ultimate goal of the treatment of RA. Over the past few decades, significant progress has been made in the treatment of RA, with higher rates of disease remission and improved long-term outcomes. Unfortunately, despite these successes, the proportion of patients with persistent, difficult-to-treat disease remains high, and the task of improving our understanding of the basic mechanisms of disease development and developing new ways to treat RA remains relevant. This review focuses on describing new treatments for RA, including cell therapies and gene editing technologies that have shown potential in preclinical and early clinical trials. In addition, we discuss the opportunities and limitations associated with the use of these new approaches in the treatment of RA.
Collapse
Affiliation(s)
- Vitaly Chasov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Irina Ganeeva
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Ekaterina Zmievskaya
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Damir Davletshin
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Elvina Gilyazova
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Aygul Valiullina
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Emil Bulatov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow 119048, Russia
| |
Collapse
|
24
|
Brownlie RJ, Salmond RJ. Regulation of T Cell Signaling and Immune Responses by PTPN22. Mol Cell Biol 2024; 44:443-452. [PMID: 39039893 PMCID: PMC11486154 DOI: 10.1080/10985549.2024.2378810] [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] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 07/24/2024] Open
Abstract
Protein tyrosine phosphatases (PTPs) play central roles in the regulation of cell signaling, organismal development, cellular differentiation and proliferation, and cancer. In the immune system, PTPs regulate the activation, differentiation and effector function of lymphocytes and myeloid cells whilst single-nucleotide polymorphisms (SNPs) in PTP-encoding genes have been identified as risk factors for the development of autoimmunity. In this review we describe the roles for PTP nonreceptor type 22 (PTPN22) in the regulation of T lymphocyte signaling and activation in autoimmunity, infection and cancer. We summarize recent progress in our understanding of the regulation of PTPN22 activity, the impact of autoimmune disease-associated PTPN22 SNPs on T cell responses and describe approaches to harness PTPN22 as a target to improve T cell-based immunotherapies in cancer.
Collapse
|
25
|
Terashima A, Ono K, Omata Y, Tanaka S, Saito T. Inflammatory diseases causing joint and bone destruction: rheumatoid arthritis and hemophilic arthropathy. J Bone Miner Metab 2024; 42:455-462. [PMID: 38856919 DOI: 10.1007/s00774-024-01520-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
Various diseases and conditions cause joint disorders. Osteoarthritis (OA) is characterized by the degeneration of articular cartilage, synovitis, and anabolic changes in surrounding bone tissues. In contrast, rheumatoid arthritis (RA) and hemophilic arthropathy (HA) display marked destruction of bone tissues caused by synovitis. RA is a representative autoimmune disease. The primary tissue of RA pathogenesis is the synovial membrane and involves various immune cells that produce catabolic cytokines and enzymes. Hemophilia is a genetic disorder caused by a deficiency in blood clotting factors. Recurrent intra-articular bleeding leads to chronic synovitis through excessive iron deposition and results in the destruction of affected joints. Although the triggers for these two joint diseases are completely different, many cytokines and enzymes are common in the pathogenesis of both RA and HA. This review focuses on the similarities between joint and bone destruction in RA and HA. The insights may be useful in developing better treatments for hemophilia patients with arthropathy and osteoporosis by leveraging advanced therapeutics for RA.
Collapse
Affiliation(s)
- Asuka Terashima
- Bone and Cartilage Regenerative Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kumiko Ono
- Department of Joint Surgery, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Yasunori Omata
- Bone and Cartilage Regenerative Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Sakae Tanaka
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Taku Saito
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
26
|
Fiske BE, Wemlinger SM, Crute BW, Getahun A. The Src-family kinase Lyn plays a critical role in establishing and maintaining B cell anergy by suppressing PI3K-dependent signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595208. [PMID: 38826354 PMCID: PMC11142063 DOI: 10.1101/2024.05.21.595208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Although the Src family kinase (SFK) Lyn is known to be involved in induction and maintenance of peripheral B cell tolerance, the molecular basis of its action in this context remains unclear. This question has been approached using conventional as well as B cell-targeted knockouts of Lyn, with varied conclusions likely confused by collateral loss of Lyn functions in B cell and myeloid cell development and activation. Here we utilized a system in which Lyn gene deletion is tamoxifen inducible and B cell restricted. This system allows acute elimination of Lyn in B cells without off-target effects. This genetic tool was employed in conjunction with immunoglobulin transgenic mice in which peripheral B cells are autoreactive. DNA reactive Ars/A1 B cells require continuous inhibitory signaling, mediated by the inositol phosphatase SHIP-1 and the tyrosine phosphatase SHP-1, to maintain an unresponsive (anergic) state. Here we show that Ars/A1 B cells require Lyn to establish and maintain B cell unresponsiveness. Lyn primarily functions by restricting PI3K-dependent signaling pathways. This Lyn-dependent mechanism complements the impact of reduced mIgM BCR expression to restrict BCR signaling in Ars/A1 B cells. Our findings suggest that a subset of autoreactive B cells requires Lyn to become anergic and that the autoimmunity associated with dysregulated Lyn function may, in part, be due to an inability of these autoreactive B cells to become tolerized.
Collapse
|
27
|
Kronzer VL, Sparks JA, Raychaudhuri S, Cerhan JR. Low-frequency and rare genetic variants associated with rheumatoid arthritis risk. Nat Rev Rheumatol 2024; 20:290-300. [PMID: 38538758 DOI: 10.1038/s41584-024-01096-7] [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: 02/20/2024] [Indexed: 04/28/2024]
Abstract
Rheumatoid arthritis (RA) has an estimated heritability of nearly 50%, which is particularly high in seropositive RA. HLA alleles account for a large proportion of this heritability, in addition to many common single-nucleotide polymorphisms with smaller individual effects. Low-frequency and rare variants, such as those captured by next-generation sequencing, can also have a large role in heritability in some individuals. Rare variant discovery has informed the development of drugs such as inhibitors of PCSK9 and Janus kinases. Some 34 low-frequency and rare variants are currently associated with RA risk. One variant (19:10352442G>C in TYK2) was identified in five separate studies, and might therefore represent a promising therapeutic target. Following a set of best practices in future studies, including studying diverse populations, using large sample sizes, validating RA and serostatus, replicating findings, adjusting for other variants and performing functional assessment, could help to ensure the relevance of identified variants. Exciting opportunities are now on the horizon for genetics in RA, including larger datasets and consortia, whole-genome sequencing and direct applications of findings in the management, and especially treatment, of RA.
Collapse
Affiliation(s)
| | - Jeffrey A Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, 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 and Harvard Medical School, Boston, MA, USA
| | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
28
|
Liu S, Lagos J, Shumlak NM, Largent AD, Lewis ST, Holder U, Du SW, Liu Y, Hou B, Acharya M, Jackson SW. NADPH oxidase exerts a B cell-intrinsic contribution to lupus risk by modulating endosomal TLR signals. J Exp Med 2024; 221:e20230774. [PMID: 38442270 PMCID: PMC10913815 DOI: 10.1084/jem.20230774] [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: 05/08/2023] [Revised: 11/11/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024] Open
Abstract
Genome-wide association studies in systemic lupus erythematosus (SLE) have linked loss-of-function mutations in phagocytic NADPH oxidase complex (NOX2) genes, including NCF1 and NCF2, to disease pathogenesis. The prevailing model holds that reduced NOX2 activity promotes SLE via defective efferocytosis, the immunologically silent clearance of apoptotic cells. Here, we describe a parallel B cell-intrinsic mechanism contributing to breaks in tolerance. In keeping with an important role for B cell Toll-like receptor (TLR) pathways in lupus pathogenesis, NOX2-deficient B cells exhibit enhanced signaling downstream of endosomal TLRs, increased humoral responses to nucleic acid-containing antigens, and the propensity toward humoral autoimmunity. Mechanistically, TLR-dependent NOX2 activation promotes LC3-mediated maturation of TLR-containing endosomes, resulting in signal termination. CRISPR-mediated disruption of NCF1 confirmed a direct role for NOX2 in regulating endosomal TLR signaling in primary human B cells. Together, these data highlight a new B cell-specific mechanism contributing to autoimmune risk in NCF1 and NCF2 variant carriers.
Collapse
Affiliation(s)
- Shuozhi Liu
- Seattle Children’s Research Institute, Seattle, WA, USA
| | | | | | | | | | - Ursula Holder
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Samuel W. Du
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yifan Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Baidong Hou
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Mridu Acharya
- Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Shaun W. Jackson
- Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
29
|
Simula ER, Jasemi S, Cossu D, Manca PC, Sanna D, Scarpa F, Meloni G, Cusano R, Sechi LA. The Genetic Landscape of Systemic Rheumatic Diseases: A Comprehensive Multigene-Panel Study Identifying Key Gene Polymorphisms. Pharmaceuticals (Basel) 2024; 17:438. [PMID: 38675400 PMCID: PMC11054024 DOI: 10.3390/ph17040438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Systemic rheumatic diseases, including conditions such as rheumatoid arthritis, Sjögren's syndrome, systemic sclerosis, and systemic lupus erythematosus, represent a complex array of autoimmune disorders characterized by chronic inflammation and diverse clinical manifestations. This study focuses on unraveling the genetic underpinnings of these diseases by examining polymorphisms in key genes related to their pathology. Utilizing a comprehensive genetic analysis, we have documented the involvement of these genetic variations in the pathogenesis of rheumatic diseases. Our study has identified several key polymorphisms with notable implications in rheumatic diseases. Polymorphism at chr11_112020916 within the IL-18 gene was prevalent across various conditions with a potential protective effect. Concurrently, the same IL18R1 gene polymorphism located at chr2_103010912, coding for the IL-18 receptor, was observed in most rheumatic conditions, reinforcing its potential protective role. Additionally, a further polymorphism in IL18R1 at chr2_103013408 seems to have a protective influence against the rheumatic diseases under investigation. In the context of emerging genes involved in rheumatic diseases, like PARK2, a significant polymorphism at chr6_161990516 was consistently identified across different conditions, exhibiting protective characteristics in these pathological contexts. The findings underscore the complexity of the genetic landscape in rheumatic autoimmune disorders and pave the way for a deeper understanding of their etiology and the possible development of more targeted and effective therapeutic strategies.
Collapse
Affiliation(s)
- Elena Rita Simula
- Dipartimento di Scienze Biomediche, Università di Sassari, 07100 Sassari, Italy; (E.R.S.); (S.J.); (D.C.); (D.S.); (F.S.)
| | - Seyedesomaye Jasemi
- Dipartimento di Scienze Biomediche, Università di Sassari, 07100 Sassari, Italy; (E.R.S.); (S.J.); (D.C.); (D.S.); (F.S.)
| | - Davide Cossu
- Dipartimento di Scienze Biomediche, Università di Sassari, 07100 Sassari, Italy; (E.R.S.); (S.J.); (D.C.); (D.S.); (F.S.)
| | - Pietro Carmelo Manca
- S.C. Servizio Immunotrasfusionale, Azienda Ospedaliero-Universitaria di Sassari, 07100 Sassari, Italy;
| | - Daria Sanna
- Dipartimento di Scienze Biomediche, Università di Sassari, 07100 Sassari, Italy; (E.R.S.); (S.J.); (D.C.); (D.S.); (F.S.)
| | - Fabio Scarpa
- Dipartimento di Scienze Biomediche, Università di Sassari, 07100 Sassari, Italy; (E.R.S.); (S.J.); (D.C.); (D.S.); (F.S.)
| | - Gianfranco Meloni
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, 07100 Sassari, Italy;
| | - Roberto Cusano
- Centro di Ricerca, Sviluppo, Studi Superiori in Sardegna (CRS4), Pula, 09100 Cagliari, Italy;
| | - Leonardo Antonio Sechi
- Dipartimento di Scienze Biomediche, Università di Sassari, 07100 Sassari, Italy; (E.R.S.); (S.J.); (D.C.); (D.S.); (F.S.)
- Struttura Complessa di Microbiologia e Virologia, Azienda Ospedaliera Universitaria, 07100 Sassari, Italy
| |
Collapse
|
30
|
Binti S, Linder AG, Edeen PT, Fay DS. A conserved protein tyrosine phosphatase, PTPN-22, functions in diverse developmental processes in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584557. [PMID: 38559252 PMCID: PMC10980042 DOI: 10.1101/2024.03.12.584557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Protein tyrosine phosphatases non-receptor type (PTPNs) have been studied extensively in the context of the adaptive immune system; however, their roles beyond immunoregulation are less well explored. Here we identify novel functions for the conserved C. elegans phosphatase PTPN-22, establishing its role in nematode molting, cell adhesion, and cytoskeletal regulation. Through a non-biased genetic screen, we found that loss of PTPN-22 phosphatase activity suppressed molting defects caused by loss-of-function mutations in the conserved NIMA-related kinases NEKL-2 (human NEK8/NEK9) and NEKL-3 (human NEK6/NEK7), which act at the interface of membrane trafficking and actin regulation. To better understand the functions of PTPN-22, we carried out proximity labeling studies to identify candidate interactors of PTPN-22 during development. Through this approach we identified the CDC42 guanine-nucleotide exchange factor DNBP-1 (human DNMBP) as an in vivo partner of PTPN-22. Consistent with this interaction, loss of DNBP-1 also suppressed nekl-associated molting defects. Genetic analysis, co-localization studies, and proximity labeling revealed roles for PTPN-22 in several epidermal adhesion complexes, including C. elegans hemidesmosomes, suggesting that PTPN-22 plays a broad role in maintaining the structural integrity of tissues. Localization and proximity labeling also implicated PTPN-22 in functions connected to nucleocytoplasmic transport and mRNA regulation, particularly within the germline, as nearly one-third of proteins identified by PTPN-22 proximity labeling are known P granule components. Collectively, these studies highlight the utility of combined genetic and proteomic approaches for identifying novel gene functions.
Collapse
Affiliation(s)
- Shaonil Binti
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, 1000 E. University Ave., Laramie, Wyoming
| | - Adison G Linder
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, 1000 E. University Ave., Laramie, Wyoming
| | - Philip T Edeen
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, 1000 E. University Ave., Laramie, Wyoming
| | - David S Fay
- Department of Molecular Biology, College of Agriculture, Life Sciences and Natural Resources, University of Wyoming, 1000 E. University Ave., Laramie, Wyoming
| |
Collapse
|
31
|
Gao Y, Zhang Y, Liu X. Rheumatoid arthritis: pathogenesis and therapeutic advances. MedComm (Beijing) 2024; 5:e509. [PMID: 38469546 PMCID: PMC10925489 DOI: 10.1002/mco2.509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by the unresolved synovial inflammation for tissues-destructive consequence, which remains one of significant causes of disability and labor loss, affecting about 0.2-1% global population. Although treatments with disease-modifying antirheumatic drugs (DMARDs) are effective to control inflammation and decrease bone destruction, the overall remission rates of RA still stay at a low level. Therefore, uncovering the pathogenesis of RA and expediting clinical transformation are imminently in need. Here, we summarize the immunological basis, inflammatory pathways, genetic and epigenetic alterations, and metabolic disorders in RA, with highlights on the abnormality of immune cells atlas, epigenetics, and immunometabolism. Besides an overview of first-line medications including conventional DMARDs, biologics, and small molecule agents, we discuss in depth promising targeted therapies under clinical or preclinical trials, especially epigenetic and metabolic regulators. Additionally, prospects on precision medicine based on synovial biopsy or RNA-sequencing and cell therapies of mesenchymal stem cells or chimeric antigen receptor T-cell are also looked forward. The advancements of pathogenesis and innovations of therapies in RA accelerates the progress of RA treatments.
Collapse
Affiliation(s)
- Ying Gao
- Department of RheumatologyChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Yunkai Zhang
- Naval Medical CenterNaval Medical UniversityShanghaiChina
| | - Xingguang Liu
- National Key Laboratory of Immunity & InflammationNaval Medical UniversityShanghaiChina
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
| |
Collapse
|
32
|
Madahar SS, Gideon A, Abdul-Sater AA. Nod-like receptors in inflammatory arthritis. Biomed J 2024; 47:100655. [PMID: 37598797 PMCID: PMC10825342 DOI: 10.1016/j.bj.2023.100655] [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: 06/22/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023] Open
Abstract
Nod-like receptors (NLRs) are innate immune receptors that play a key role in sensing components from pathogens and from damaged cells or organelles. NLRs form signaling complexes that can lead to activation of transcription factors or effector caspases - by means of inflammasome activation -Inflammatory arthritis (IA) culminating in promoting inflammation. An increasing body of research supports the role of NLRs in driving pathogenesis of IA, a collection of diseases that include rheumatoid arthritis (RA), psoriatic arthritis (PsA), ankylosing spondylitis, and pediatric arthritis. In this review, we briefly discuss the main drivers of IA diseases and dive into the evidence for - and against - various NLRs in driving these diseases. We also review the studies examining the use of NLR and inflammasome inhibitors as potential therapies for IA.
Collapse
Affiliation(s)
- Sahib Singh Madahar
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada; Department of Biology, York University, Toronto, Ontario, Canada
| | - Alita Gideon
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Ali A Abdul-Sater
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada.
| |
Collapse
|
33
|
Salmond RJ. Targeting Protein Tyrosine Phosphatases to Improve Cancer Immunotherapies. Cells 2024; 13:231. [PMID: 38334623 PMCID: PMC10854786 DOI: 10.3390/cells13030231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/17/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Advances in immunotherapy have brought significant therapeutic benefits to many cancer patients. Nonetheless, many cancer types are refractory to current immunotherapeutic approaches, meaning that further targets are required to increase the number of patients who benefit from these technologies. Protein tyrosine phosphatases (PTPs) have long been recognised to play a vital role in the regulation of cancer cell biology and the immune response. In this review, we summarize the evidence for both the pro-tumorigenic and tumour-suppressor function of non-receptor PTPs in cancer cells and discuss recent data showing that several of these enzymes act as intracellular immune checkpoints that suppress effective tumour immunity. We highlight new data showing that the deletion of inhibitory PTPs is a rational approach to improve the outcomes of adoptive T cell-based cancer immunotherapies and describe recent progress in the development of PTP inhibitors as anti-cancer drugs.
Collapse
Affiliation(s)
- Robert J Salmond
- Leeds Institute of Medical Research at St. James's, School of Medicine, University of Leeds, Leeds LS9 7TF, UK
| |
Collapse
|
34
|
Shekh MR, Ahmed N, Kumar V. A Review of the Occurrence of Rheumatoid Arthritis and Potential Treatments through Medicinal Plants from an Indian Perspective. Curr Rheumatol Rev 2024; 20:241-269. [PMID: 38018201 DOI: 10.2174/0115733971268416231116184056] [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: 07/13/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 11/30/2023]
Abstract
Arthritis is a medical condition that affects the joints and causes inflammation, pain, and stiffness. There are different types of arthritis, and it can affect people of all ages, even infants and the elderly. Recent studies have found that individuals with diabetes, heart disease, and obesity are more likely to experience arthritis symptoms. According to the World Health Organization, over 21% of people worldwide suffer from musculoskeletal problems. Roughly 42.19 million individuals in India, constituting around 0.31% of the populace, have been documented as having Rheumatic Arthritis (RA). Compared to other common diseases like diabetes, cancer, and AIDS, arthritis is more prevalent in the general population. Unfortunately, there is no specific cure for arthritis, and treatment plans usually involve non-pharmacological methods, surgeries, and medications that target specific symptoms. Plant-based remedies have also been shown to be effective in managing inflammation and related complications. In addition to therapies, maintaining a healthy diet, exercise, and weight management are essential for managing arthritis. This review discusses the causes, prevalence, diagnostic methods, current and prospective future treatments, and potential medicinal plants that may act as anti-inflammatory or anti-rheumatic agents. However, more research is necessary to identify the underlying mechanisms and active molecules that could improve arthritis treatment.
Collapse
Affiliation(s)
- Mohammad Raeesh Shekh
- National Innovation Foundation (NIF), India, Grambharti, Amrapur, Gandhinagar, Mahudi Road, Gandhinagar, Gujarat, India
| | - Nasir Ahmed
- Forensic Anthropology-1, Department of Forensic Medicine, YMC, Yenepoya Deemed to be University, University Road, Deralakatte, Mangaluru, Karnataka, 575018, India
| | - Vivek Kumar
- National Innovation Foundation (NIF), India, Grambharti, Amrapur, Gandhinagar, Mahudi Road, Gandhinagar, Gujarat, India
| |
Collapse
|
35
|
Malik JA, Kaur G, Agrewala JN. Revolutionizing medicine with toll-like receptors: A path to strengthening cellular immunity. Int J Biol Macromol 2023; 253:127252. [PMID: 37802429 DOI: 10.1016/j.ijbiomac.2023.127252] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Toll-like receptors play a vital role in cell-mediated immunity, which is crucial for the immune system's defense against pathogens and maintenance of homeostasis. The interaction between toll-like-receptor response and cell-mediated immunity is complex and essential for effectively eliminating pathogens and maintaining immune surveillance. In addition to pathogen recognition, toll-like receptors serve as adjuvants in vaccines, as molecular sensors, and recognize specific patterns associated with pathogens and danger signals. Incorporating toll-like receptor ligands into vaccines can enhance the immune response to antigens, making them potent adjuvants. Furthermore, they bridge the innate and adaptive immune systems and improve antigen-presenting cells' capacity to process and present antigens to T cells. The intricate signaling pathways and cross-talk between toll-like-receptor and T cell receptor (TCR) signaling emphasize their pivotal role in orchestrating effective immune responses against pathogens, thus facilitating the development of innovative vaccine strategies. This article provides an overview of the current understanding of toll-like receptor response and explores their potential clinical applications. By unraveling the complex mechanisms of toll-like-receptor signaling, we can gain novel insights into immune responses and potentially develop innovative therapeutic approaches. Ongoing investigations into the toll-like-receptor response hold promise in the future in enhancing our ability to combat infections, design effective vaccines, and improve clinical outcomes.
Collapse
Affiliation(s)
- Jonaid Ahmad Malik
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology, Ropar, Punjab 140001, India
| | - Gurpreet Kaur
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology, Ropar, Punjab 140001, India; Department of Biotechnology, Chandigarh Group of Colleges, Landran, Mohali, Punjab 140055, India
| | - Javed N Agrewala
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology, Ropar, Punjab 140001, India.
| |
Collapse
|
36
|
Ansari MA, Chauhan W, Shoaib S, Alyahya SA, Ali M, Ashraf H, Alomary MN, Al-Suhaimi EA. Emerging therapeutic options in the management of diabetes: recent trends, challenges and future directions. Int J Obes (Lond) 2023; 47:1179-1199. [PMID: 37696926 DOI: 10.1038/s41366-023-01369-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/04/2023] [Accepted: 08/17/2023] [Indexed: 09/13/2023]
Abstract
Diabetes is a serious health issue that causes a progressive dysregulation of carbohydrate metabolism due to insufficient insulin hormone, leading to consistently high blood glucose levels. According to the epidemiological data, the prevalence of diabetes has been increasing globally, affecting millions of individuals. It is a long-term condition that increases the risk of various diseases caused by damage to small and large blood vessels. There are two main subtypes of diabetes: type 1 and type 2, with type 2 being the most prevalent. Genetic and molecular studies have identified several genetic variants and metabolic pathways that contribute to the development and progression of diabetes. Current treatments include gene therapy, stem cell therapy, statin therapy, and other drugs. Moreover, recent advancements in therapeutics have also focused on developing novel drugs targeting these pathways, including incretin mimetics, SGLT2 inhibitors, and GLP-1 receptor agonists, which have shown promising results in improving glycemic control and reducing the risk of complications. However, these treatments are often expensive, inaccessible to patients in underdeveloped countries, and can have severe side effects. Peptides, such as glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), are being explored as a potential therapy for diabetes. These peptides are postprandial glucose-dependent pancreatic beta-cell insulin secretagogues and have received much attention as a possible treatment option. Despite these advances, diabetes remains a major health challenge, and further research is needed to develop effective treatments and prevent its complications. This review covers various aspects of diabetes, including epidemiology, genetic and molecular basis, and recent advancements in therapeutics including herbal and synthetic peptides.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.
| | - Waseem Chauhan
- Department of Hematology, Duke University, Durham, NC, 27710, USA
| | - Shoaib Shoaib
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Sami A Alyahya
- Wellness and Preventive Medicine Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, 11442, Saudi Arabia
| | - Mubashshir Ali
- USF Health Byrd Alzheimer's Center and Neuroscience Institute, Department of Molecular Medicine, Tampa, FL, USA
| | - Hamid Ashraf
- Rajiv Gandhi Center for Diabetes and Endocrinology, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, 11442, Saudi Arabia.
| | - Ebtesam A Al-Suhaimi
- King Abdulaziz & his Companions Foundation for Giftedness & Creativity, Riyadh, Saudi Arabia.
| |
Collapse
|
37
|
Arshad M, Noor N, Iqbal Z, Jaleel H. In silico analysis of missense SNPs in TNFR1a and their possible therapeutic or pathogenic role in immune diseases. Hum Immunol 2023; 84:609-617. [PMID: 37748952 DOI: 10.1016/j.humimm.2023.09.003] [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: 05/19/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 09/27/2023]
Abstract
Tumor necrosis factor alpha (TNFa) is an inflammatory cytokine that is involved in the pathogenesis of various inflammatory disorders including rheumatoid arthritis. TNF-alpha receptor I (TNFR1a) is one of the receptors TNFa binds with for its activation. Any variation in this receptor might affect the role of TNFa in successive events. Amino acid residue substitutions might happen in TNFR1a through non-synonymous single nucleotide polymorphisms (nsSNPs) which may alter the functioning of TNFa, hence, identifying any such substitutions is of paramount significance. In this study, six nsSNPs at five different evolutionary conserved regions are predicted to be detrimental to the structure and/or function of TNFR1a by using numerous computational tools. Their 3D models are also proposed in this study. Besides, they were found to reduce the stability and affect the molecular mechanisms of this protein. Two contrasting possibilities might happen because of these substitutions. One, they might reduce the production of TNFa which is overexpressed in inflammatory diseases, hence can play therapeutic role in such diseases. Second, they might possibly hinder the apoptosis to occur which can effectuate the uncontrolled division of cells, hence can be pathogenic in diseases like cancer. Further investigations on these nsSNPs using animal models and at cellular level will open doors to understand the underlying mechanisms behind various diseases.
Collapse
Affiliation(s)
- Maria Arshad
- Department of Biochemistry and Molecular Biology, University of Iceland, Reykjavik, Iceland.
| | - Nabeel Noor
- Shalamar Medical & Dental College, Lahore, Pakistan
| | - Zunair Iqbal
- Shalamar Medical & Dental College, Lahore, Pakistan
| | - Hadiqa Jaleel
- Department of Research & Innovation, Shalamar Institute of Health Sciences, Lahore, Pakistan
| |
Collapse
|
38
|
Trier NH, Houen G. Antibody Cross-Reactivity in Auto-Immune Diseases. Int J Mol Sci 2023; 24:13609. [PMID: 37686415 PMCID: PMC10487534 DOI: 10.3390/ijms241713609] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Autoimmunity is defined by the presence of antibodies and/or T cells directed against self-components. Although of unknown etiology, autoimmunity commonly is associated with environmental factors such as infections, which have been reported to increase the risk of developing autoimmune diseases. Occasionally, similarities between infectious non-self and self-tissue antigens may contribute to immunological cross-reactivity in autoimmune diseases. These reactions may be interpreted as molecular mimicry, which describes cross-reactivity between foreign pathogens and self-antigens that have been reported to cause tissue damage and to contribute to the development of autoimmunity. By focusing on the nature of antibodies, cross-reactivity in general, and antibody-antigen interactions, this review aims to characterize the nature of potential cross-reactive immune reactions between infectious non-self and self-tissue antigens which may be associated with autoimmunity but may not actually be the cause of disease onset.
Collapse
Affiliation(s)
- Nicole Hartwig Trier
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens Vej 1-23, 2600 Glostrup, Denmark
| | - Gunnar Houen
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens Vej 1-23, 2600 Glostrup, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
39
|
Trier NH, Houen G. Anti-citrullinated protein antibodies as biomarkers in rheumatoid arthritis. Expert Rev Mol Diagn 2023; 23:895-911. [PMID: 37578277 DOI: 10.1080/14737159.2023.2247986] [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: 03/29/2023] [Revised: 07/15/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION The serological biomarker anti-citrullinated protein antibodies (ACPAs) may have several functions but is especially important for the diagnosis of rheumatoid arthritis (RA) along with clinical symptoms. AREAS COVERED This review provides an overview of ACPAs, which are useful in RA diagnostics and may improve our understanding of disease etiology. PubMed was searched with combinations of words related to antibodies recognizing epitopes containing the post-translationally modified amino acid citrulline in combination with rheumatoid arthritis; cyclic citrullinated peptide, CCP, anti-CCP, anti-citrullinated protein antibodies, ACPA, citrullination, peptide/protein arginine deiminase, PAD, filaggrin, vimentin, keratin, collagen, perinuclear factor, EBNA1, EBNA2, and others. From this search, we made a qualitative extract of publications relevant to the discovery, characterization, and clinical use of these antibodies in relation to RA. We highlight significant findings and identify areas for improvement. EXPERT OPINION ACPAs have high diagnostic sensitivity and specificity for RA and recognize citrullinated epitopes from several proteins. The best-performing single epitope originates from Epstein-Barr Virus nuclear antigen 2 and contains a central Cit-Gly motif, which is recognized by ACPAS when located in a flexible peptide structure. In addition, ACPAs may also have prognostic value, especially in relation to early treatment, although ACPAs' main function is to aid in the diagnosis of RA.
Collapse
Affiliation(s)
| | - Gunnar Houen
- Department of Neurology, Rigshospitalet Glostrup, Glostrup, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| |
Collapse
|
40
|
Fichna M, Małecki PP, Żurawek M, Furman K, Gębarski B, Fichna P, Ruchała M. Genetic variants and risk of endocrine autoimmunity in relatives of patients with Addison's disease. Endocr Connect 2023; 12:e230008. [PMID: 37010089 PMCID: PMC10235924 DOI: 10.1530/ec-23-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/03/2023] [Indexed: 04/04/2023]
Abstract
Since individuals with Addison's disease (AD) present considerable co-occurrence of additional autoimmune conditions, clustering of autoimmunity was also predicted among their relatives. The study was aimed to assess circulating autoantibodies in first-degree relatives of patients with AD and to correlate them with the established genetic risk factors (PTPN22 rs2476601, CTLA4 rs231775, and BACH2 rs3757247). Antibodies were evaluated using validated commercial assays, and genotyping was performed using TaqMan chemistry. The studied cohort comprised 112 female and 75 male relatives. Circulating autoantibodies were found in 69 relatives (36.9%). Thyroid autoantibodies, that is antibodies to thyroid peroxidase (aTPO) and thyroglobulin (aTg), were detectable in 25.1 and 17.1% relatives, respectively. Antibodies to 21-hydroxylase (a21OH) were found in 5.8% individuals, and beta cell-specific antibodies to ZnT8, GAD, and IA2 were found in 7.5, 8.0, and 2.7%, respectively. The prevalence of a21OH (P = 0.0075; odds ratio (OR) 7.68; 95% CI 1.903-36.0), aTPO (P < 0.0001; OR 3.85; 95% CI 1.873-7.495), and aTg (P < 0.0001; OR 7.73; 95% CI 3.112-19.65), as well as aGAD (P = 0.0303; OR 3.38; 95% CI 1.180-9.123) and aZnT8 (P = 0.032; OR 6.40; 95% CI 1.846-21.91), was significantly increased in carriers of rs2476601 T allele. Moreover, T allele appeared to be a risk factor for multiple circulating autoantibody specificities (P = 0.0009; OR 5.79; 95% CI 1.962-15.81). None of the studied autoantibodies demonstrated significant association with rs231775 in CTLA4 (P > 0.05), and only weak association was detected between BACH2 rs3757247 and circulating aTPO (P = 0.0336; OR 2.12; 95%CI 1.019-4.228). In conclusion, first-degree relatives of patients with AD, carriers of the PTPN22 rs2476601 T allele, are at particular risk of developing autoantibodies to endocrine antigens.
Collapse
Affiliation(s)
- Marta Fichna
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Piotr P Małecki
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Magdalena Żurawek
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | | | - Piotr Fichna
- Department of Paediatric Diabetes and Obesity, Poznan University of Medical Sciences, Poznan, Poland
| | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
41
|
Sardana Y, Bhatti GK, Singh C, Sharma PK, Reddy PH, Bhatti JS. Progression of pre-rheumatoid arthritis to clinical disease of joints: Potential role of mesenchymal stem cells. Life Sci 2023; 321:121641. [PMID: 36997059 DOI: 10.1016/j.lfs.2023.121641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023]
Abstract
Rheumatoid arthritis (RA) related autoimmunity is developed at mucosal sites due to the interplay between genetic risk factors and environmental triggers. The pre-RA phase that leads to anti-citrullinated protein antibodies, rheumatoid factor, and other autoantibodies spread in the systemic circulation may not affect articular tissue for years until a mysterious second hit triggers the localization of RA-related autoimmunity in joints. Several players in the joint microenvironment mediate the synovial innate and adaptive immunological processes, eventually leading to clinical synovitis. There still exists a gap in the early phase of RA pathogenesis, i.e., the progression of diseases from the systemic circulation to joints. The lack of better understanding of these events results in the inability to answer questions about why only after a certain point of time the disease appears in joints and why in some cases, it simply remains latent and doesn't affect joints at all. In the current review, we focused on the immunomodulatory and regenerative role of mesenchymal stem cells and associated exosomes in RA pathology. We also highlighted the age-related dysregulations in activities of mesenchymal stem cells and how that might trigger homing of systemic autoimmunity to joints.
Collapse
Affiliation(s)
- Yogesh Sardana
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, Hemvati Nandan Bahuguna Garhwal University, Uttarakhand, India
| | | | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
42
|
Budlewski T, Sarnik J, Galita G, Dragan G, Brzezińska O, Popławska M, Popławski T, Makowska J. SNP in PTPN22, PADI4, and STAT4 but Not TRAF1 and CD40 Increase the Risk of Rheumatoid Arthritis in Polish Population. Int J Mol Sci 2023; 24:ijms24087586. [PMID: 37108746 PMCID: PMC10145319 DOI: 10.3390/ijms24087586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Single nucleotide polymorphisms in non-HLA genes are involved in the development of rheumatoid arthritis (RA). SNPS in genes: PADI4 (rs2240340), STAT4 (rs7574865), CD40 (rs4810485), PTPN22 (rs2476601), and TRAF1 (rs3761847) have been described as risk factors for the development of autoimmune diseases, including RA. This study aimed to assess the prevalence of polymorphisms of these genes in the Polish population of patients with rheumatoid arthritis as compared to healthy controls. 324 subjects were included in the study: 153 healthy subjects and 181 patients from the Department of Rheumatology, Medical University of Lodz who fulfilled the criteria of rheumatoid arthritis diagnosis. Genotypes were determined by Taqman SNP Genotyping Assay. rs2476601 (G/A, OR = 2.16, CI = 1.27-3.66; A/A, OR = 10.35, CI = 1.27-84.21), rs2240340 (C/T, OR = 4.35, CI = 2.55-7.42; T/T, OR = 2.80, CI = 1.43-4.10) and rs7574865 (G/T, OR = 1.97, CI = 1.21-3.21; T/T, OR = 3.33, CI = 1.01-11.02) were associated with RA in the Polish population. Rs4810485 was also associated with RA, however after Bonferroni's correction was statistically insignificant. We also found an association between minor alleles of rs2476601, rs2240340, and rs7574865 and RA (OR = 2.32, CI = 1.47-3.66; OR = 2.335, CI = 1.64-3.31; OR = 1.88, CI = 1.27-2.79, respectively). Multilocus analysis revealed an association between CGGGT and rare (below 0.02 frequency) haplotypes (OR = 12.28, CI = 2.65-56.91; OR = 3.23, CI = 1.63-6.39). In the Polish population, polymorphisms of the PADI4, PTPN22, and STAT4 genes have been detected, which are also known risk factors for RA in various other populations.
Collapse
Affiliation(s)
- Tomasz Budlewski
- Department of Rheumatology, Medical University of Lodz, 92-115 Lodz, Poland
| | - Joanna Sarnik
- Department of Rheumatology, Medical University of Lodz, 92-115 Lodz, Poland
| | - Grzegorz Galita
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
- Doctoral Study in Molecular Genetics, Cytogenetics and Medical Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Grzegorz Dragan
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Olga Brzezińska
- Department of Rheumatology, Medical University of Lodz, 92-115 Lodz, Poland
| | - Marta Popławska
- Biobank, Department of Immunology and Allergy, Medical University of Lodz, 92-213 Lodz, Poland
| | - Tomasz Popławski
- Department of Pharmaceutical Microbiology and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Joanna Makowska
- Department of Rheumatology, Medical University of Lodz, 92-115 Lodz, Poland
| |
Collapse
|
43
|
Műzes G, Sipos F. Autoimmunity and Carcinogenesis: Their Relationship under the Umbrella of Autophagy. Biomedicines 2023; 11:1130. [PMID: 37189748 PMCID: PMC10135912 DOI: 10.3390/biomedicines11041130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system and autophagy share a functional relationship. Both innate and adaptive immune responses involve autophagy and, depending on the disease's origin and pathophysiology, it may have a detrimental or positive role on autoimmune disorders. As a "double-edged sword" in tumors, autophagy can either facilitate or impede tumor growth. The autophagy regulatory network that influences tumor progression and treatment resistance is dependent on cell and tissue types and tumor stages. The connection between autoimmunity and carcinogenesis has not been sufficiently explored in past studies. As a crucial mechanism between the two phenomena, autophagy may play a substantial role, though the specifics remain unclear. Several autophagy modifiers have demonstrated beneficial effects in models of autoimmune disease, emphasizing their therapeutic potential as treatments for autoimmune disorders. The function of autophagy in the tumor microenvironment and immune cells is the subject of intensive study. The objective of this review is to investigate the role of autophagy in the simultaneous genesis of autoimmunity and malignancy, shedding light on both sides of the issue. We believe our work will assist in the organization of current understanding in the field and promote additional research on this urgent and crucial topic.
Collapse
Affiliation(s)
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary;
| |
Collapse
|
44
|
Anderson W, Barahmand-pour-Whitman F, Linsley PS, Cerosaletti K, Buckner JH, Rawlings DJ. PTPN22 R620W gene editing in T cells enhances low-avidity TCR responses. eLife 2023; 12:e81577. [PMID: 36961507 PMCID: PMC10065793 DOI: 10.7554/elife.81577] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 03/21/2023] [Indexed: 03/25/2023] Open
Abstract
A genetic variant in the gene PTPN22 (R620W, rs2476601) is strongly associated with increased risk for multiple autoimmune diseases and linked to altered TCR regulation and T cell activation. Here, we utilize Crispr/Cas9 gene editing with donor DNA repair templates in human cord blood-derived, naive T cells to generate PTPN22 risk edited (620W), non-risk edited (620R), or knockout T cells from the same donor. PTPN22 risk edited cells exhibited increased activation marker expression following non-specific TCR engagement, findings that mimicked PTPN22 KO cells. Next, using lentiviral delivery of T1D patient-derived TCRs against the pancreatic autoantigen, islet-specific glucose-6 phosphatase catalytic subunit-related protein (IGRP), we demonstrate that loss of PTPN22 function led to enhanced signaling in T cells expressing a lower avidity self-reactive TCR, but not a high-avidity TCR. In this setting, loss of PTPN22 mediated enhanced proliferation and Th1 skewing. Importantly, expression of the risk variant in association with a lower avidity TCR also increased proliferation relative to PTPN22 non-risk T cells. Together, these findings suggest that, in primary human T cells, PTPN22 rs2476601 contributes to autoimmunity risk by permitting increased TCR signaling and activation in mildly self-reactive T cells, thereby potentially expanding the self-reactive T cell pool and skewing this population toward an inflammatory phenotype.
Collapse
Affiliation(s)
- Warren Anderson
- Center for Immunity and Immunotherapies, Seattle Children's Research InstituteSeattleUnited States
| | | | - Peter S Linsley
- Benaroya Research Institute at Virginia MasonSeattleUnited States
| | | | - Jane H Buckner
- Benaroya Research Institute at Virginia MasonSeattleUnited States
| | - David J Rawlings
- Department of Pediatrics and Immunology, University of WashingtonSeattleUnited States
| |
Collapse
|
45
|
Sundaresan B, Shirafkan F, Ripperger K, Rattay K. The Role of Viral Infections in the Onset of Autoimmune Diseases. Viruses 2023; 15:v15030782. [PMID: 36992490 PMCID: PMC10051805 DOI: 10.3390/v15030782] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Autoimmune diseases (AIDs) are the consequence of a breach in immune tolerance, leading to the inability to sufficiently differentiate between self and non-self. Immune reactions that are targeted towards self-antigens can ultimately lead to the destruction of the host's cells and the development of autoimmune diseases. Although autoimmune disorders are comparatively rare, the worldwide incidence and prevalence is increasing, and they have major adverse implications for mortality and morbidity. Genetic and environmental factors are thought to be the major factors contributing to the development of autoimmunity. Viral infections are one of the environmental triggers that can lead to autoimmunity. Current research suggests that several mechanisms, such as molecular mimicry, epitope spreading, and bystander activation, can cause viral-induced autoimmunity. Here we describe the latest insights into the pathomechanisms of viral-induced autoimmune diseases and discuss recent findings on COVID-19 infections and the development of AIDs.
Collapse
Affiliation(s)
- Bhargavi Sundaresan
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Fatemeh Shirafkan
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Kevin Ripperger
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| | - Kristin Rattay
- Institute of Pharmacology, Biochemical Pharmacological Center, University of Marburg, 35043 Marburg, Germany
| |
Collapse
|
46
|
Pasha U, Nisar H, Nisar H, Abid R, Ashraf NM, Sadaf S. Molecular Dynamic Simulations Unravel the Underlying Impact of Missense Mutation in Autoimmunity Gene PTPN22 on Predisposition to Rheumatoid Arthritis. J Interferon Cytokine Res 2023; 43:121-132. [PMID: 36811459 DOI: 10.1089/jir.2022.0216] [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] [Indexed: 02/24/2023] Open
Abstract
Genetic mutations in various proteins have been implicated with increased risk or severity of rheumatoid arthritis (RA) in different population groups. In the present case-control study, we have investigated the risk association of single nucleotide mutations present in some of the highly reported anti-inflammatory proteins and/or cytokines, with RA susceptibility in the Pakistani subjects. The study involves 310 ethnically and demographically similar participants from whom blood samples were taken and processed for DNA extraction. Through extensive data mining, 5 hotspot mutations reported in 4 genes, that is, interleukin (IL)-4 (-590; rs2243250), IL-10 (-592; rs1800872), IL-10 (-1082; rs1800896), PTPN22 (C1858T; rs2476601), and TNFAIP3 (T380G; rs2230926), were selected for RA susceptibility analyses using genotyping assays. The results demonstrated the association of only 2 DNA variants [rs2243250 (odds ratio, OR = 2.025, 95% confidence interval, CI = 1.357-3.002, P = 0.0005 Allelic) and rs2476601 (OR = 4.25, 95% CI = 1.569-11.55, P = 0.004 Allelic)] with RA susceptibility in the local population. The former single nucleotide mutation was nonfunctional, whereas the latter, residing in the exonic region of a linkage-proven autoimmunity gene PTPN22, was involved in R620→W620 substitution. Comparative molecular dynamic simulations and free-energy calculations revealed a radical impact on the geometry/confirmation of key functional moieties in the mutant protein leading to a rather weak binding of W620 variant with the interacting receptor (SRC kinase). The interaction imbalance and binding instabilities provide convincing clues about the insufficient inhibition of T cell activation and/or ineffective clearance of autoimmune clones-a hallmark of several autoimmune disorders. In conclusion, the present research describes the association of 2 hotspot mutations in IL-4 promoter and PTPN22 gene with RA susceptibility in the Pakistani study cohort. It also details how a functional mutation in PTPN22 impacts the overall protein geometry, charge, and/or receptor interactions to contribute to RA susceptibility.
Collapse
Affiliation(s)
- Usman Pasha
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Haseeb Nisar
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
- Department of Life Sciences, University of Management and Technology, Lahore, Pakistan
| | - Hajira Nisar
- Emergency and Out Patient Department, Ali Fatima Hospital, Lahore, Pakistan
| | - Rizwan Abid
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Naeem Mahmood Ashraf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Saima Sadaf
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
47
|
Expression of Non-T Cell Activation Linker (NTAL) in Jurkat Cells Negatively Regulates TCR Signaling: Potential Role in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24054574. [PMID: 36902005 PMCID: PMC10003381 DOI: 10.3390/ijms24054574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
T lymphocytes are key players in adaptive immune responses through the recognition of peptide antigens through the T Cell Receptor (TCR). After TCR engagement, a signaling cascade is activated, leading to T cell activation, proliferation, and differentiation into effector cells. Delicate control of activation signals coupled to the TCR is needed to avoid uncontrolled immune responses involving T cells. It has been previously shown that mice deficient in the expression of the adaptor NTAL (Non-T cell activation linker), a molecule structurally and evolutionarily related to the transmembrane adaptor LAT (Linker for the Activation of T cells), develop an autoimmune syndrome characterized by the presence of autoantibodies and enlarged spleens. In the present work we intended to deepen investigation into the negative regulatory functions of the NTAL adaptor in T cells and its potential relationship with autoimmune disorders. For this purpose, in this work we used Jurkat cells as a T cell model, and we lentivirally transfected them to express the NTAL adaptor in order to analyze the effect on intracellular signals associated with the TCR. In addition, we analyzed the expression of NTAL in primary CD4+ T cells from healthy donors and Rheumatoid Arthritis (RA) patients. Our results showed that NTAL expression in Jurkat cells decreased calcium fluxes and PLC-γ1 activation upon stimulation through the TCR complex. Moreover, we showed that NTAL was also expressed in activated human CD4+ T cells, and that the increase of its expression was reduced in CD4+ T cells from RA patients. Our results, together with previous reports, suggest a relevant role for the NTAL adaptor as a negative regulator of early intracellular TCR signaling, with a potential implication in RA.
Collapse
|
48
|
Rheumatoid arthritis and non-coding RNAs; how to trigger inflammation. Life Sci 2023; 315:121367. [PMID: 36639050 DOI: 10.1016/j.lfs.2023.121367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/31/2022] [Accepted: 12/31/2022] [Indexed: 01/12/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic and chronic inflammatory disease categorized by continuous synovitis in the joints and systemic inflammatory responses that can cause lifelong disability. The major cause of RA is the dysregulation of the immune response. The development of RA disease includes multiplex association of several interleukins and cells, which leads to synovial cell growth, cartilage and bone damage. The primary stage of RA disease is related to the modification of both the innate and adaptive immune systems, which leads to the formation of autoantibodies. This process results in many damaged molecules and epitope spreading. Both the innate (e.g., dendritic cells, macrophages, and neutrophils) and acquired immune cells (e.g., T and B lymphocytes) will increase and continue the chronic inflammatory condition in the next stages of the RA disease. In recent years, non-coding RNAs have been proved as significant controllers of biological functions, especially immune cell expansion and reactions. Non-coding RNAs were primarily containing microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA). Various studies confirmed non-coding RNAs as hopeful markers for diagnosing and curing RA. This review will describe and cover existing knowledge about RA pathogenesis, which might be favorable for discovering possible ncRNA markers for RA.
Collapse
|
49
|
Meitei HT, Lal G. T cell receptor signaling in the differentiation and plasticity of CD4 + T cells. Cytokine Growth Factor Rev 2023; 69:14-27. [PMID: 36028461 DOI: 10.1016/j.cytogfr.2022.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/17/2022] [Indexed: 02/07/2023]
Abstract
CD4+ T cells are critical components of the adaptive immune system. The T cell receptor (TCR) and co-receptor signaling cascades shape the phenotype and functions of CD4+ T cells. TCR signaling plays a crucial role in T cell development, antigen recognition, activation, and differentiation upon recognition of foreign- or auto-antigens. In specific autoimmune conditions, altered TCR repertoire is reported and can predispose autoimmunity with organ-specific inflammation and tissue damage. TCR signaling modulates various signaling cascades and regulates epigenetic and transcriptional regulation during homeostasis and disease conditions. Understanding the mechanism by which coreceptors and cytokine signals control the magnitude of TCR signal amplification will aid in developing therapeutic strategies to treat inflammation and autoimmune diseases. This review focuses on the role of the TCR signaling cascade and its components in the activation, differentiation, and plasticity of various CD4+ T cell subsets.
Collapse
Affiliation(s)
| | - Girdhari Lal
- National Centre for Cell Science, SPPU campus, Ganeshkhind, Pune, MH 411007, India.
| |
Collapse
|
50
|
La Bella S, Rinaldi M, Di Ludovico A, Di Donato G, Di Donato G, Salpietro V, Chiarelli F, Breda L. Genetic Background and Molecular Mechanisms of Juvenile Idiopathic Arthritis. Int J Mol Sci 2023; 24:ijms24031846. [PMID: 36768167 PMCID: PMC9916312 DOI: 10.3390/ijms24031846] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/07/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic disease in the paediatric population. JIA comprises a heterogeneous group of disorders with different onset patterns and clinical presentations with the only element in common being chronic joint inflammation. This review sought to evaluate the most relevant and up-to-date evidence on current knowledge regarding the pathogenesis of JIA subtypes to provide a better understanding of these disorders. Despite significant improvements over the past decade, the aetiology and molecular mechanisms of JIA remain unclear. It has been suggested that the immunopathogenesis is characterised by complex interactions between genetic background and environmental factors that may differ between JIA subtypes. Human leukocyte antigen (HLA) haplotypes and non-HLA genes play a crucial role in the abnormal activation of both innate and adaptive immune cells that cooperate in causing the inflammatory process. This results in the involvement of proinflammatory cytokines, including tumour necrosis factor (TNF)α, interleukin (IL)-1, IL-6, IL-10, IL-17, IL-21, IL-23, and others. These mediators, interacting with the surrounding tissue, cause cartilage stress and bone damage, including irreversible erosions. The purpose of this review is to provide a comprehensive overview of the genetic background and molecular mechanisms of JIA.
Collapse
Affiliation(s)
- Saverio La Bella
- Paediatric Department, University of Chieti “G. D’Annunzio”, 66100 Chieti, Italy
| | - Marta Rinaldi
- Paediatric Department, Buckinghamshire Healthcare NHS Trust, Aylesbury-Thames Valley Deanery, Aylesbury HP21 8AL, UK
| | - Armando Di Ludovico
- Paediatric Department, University of Chieti “G. D’Annunzio”, 66100 Chieti, Italy
| | - Giulia Di Donato
- Paediatric Department, University of Chieti “G. D’Annunzio”, 66100 Chieti, Italy
| | - Giulio Di Donato
- Paediatric Department, University of L’Aquila, 67100 L’Aquila, Italy
| | | | - Francesco Chiarelli
- Paediatric Department, University of Chieti “G. D’Annunzio”, 66100 Chieti, Italy
| | - Luciana Breda
- Paediatric Department, University of Chieti “G. D’Annunzio”, 66100 Chieti, Italy
- Correspondence: ; Tel.: +39-0871-357377
| |
Collapse
|