51
|
Huang SUS, O’Sullivan KM. The Expanding Role of Extracellular Traps in Inflammation and Autoimmunity: The New Players in Casting Dark Webs. Int J Mol Sci 2022; 23:ijms23073793. [PMID: 35409152 PMCID: PMC8998317 DOI: 10.3390/ijms23073793] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
The first description of a new form of neutrophil cell death distinct from that of apoptosis or necrosis was discovered in 2004 and coined neutrophil extracellular traps "(NETs)" or "NETosis". Different stimuli for NET formation, and pathways that drive neutrophils to commit to NETosis have been elucidated in the years that followed. Critical enzymes required for NET formation have been discovered and targeted therapeutically. NET formation is no longer restricted to neutrophils but has been discovered in other innate cells: macrophages/monocytes, mast Cells, basophils, dendritic cells, and eosinophils. Furthermore, extracellular DNA can also be extruded from both B and T cells. It has become clear that although this mechanism is thought to enhance host defense by ensnaring bacteria within large webs of DNA to increase bactericidal killing capacity, it is also injurious to innocent bystander tissue. Proteases and enzymes released from extracellular traps (ETs), injure epithelial and endothelial cells perpetuating inflammation. In the context of autoimmunity, ETs release over 70 well-known autoantigens. ETs are associated with pathology in multiple diseases: lung diseases, vasculitis, autoimmune kidney diseases, atherosclerosis, rheumatoid arthritis, cancer, and psoriasis. Defining these pathways that drive ET release will provide insight into mechanisms of pathological insult and provide potential therapeutic targets.
Collapse
|
52
|
Zhang YP, Wang X, Jie LG, Qu Y, Zhu XT, Wu J, Yu QH. Osteoarticular Involvement-Associated Biomarkers and Pathways in Psoriasis: The Shared Pathway With Ankylosing Spondylitis. Front Immunol 2022; 13:836533. [PMID: 35371093 PMCID: PMC8969572 DOI: 10.3389/fimmu.2022.836533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Psoriatic arthritis (PsA) is a unique immune-mediated disease with cutaneous and osteoarticular involvement. However, only a few studies have explored the susceptibility of osteoarticular involvement in psoriasis (Ps) at the genetic level. This study investigated the biomarkers associated with osteoarticular participation and potential shared molecular mechanisms for PsA and ankylosing spondylitis (AS).
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Wu
- *Correspondence: Jing Wu, ; Qing-Hong Yu,
| | | |
Collapse
|
53
|
Liu J, Kumar S, Hong J, Huang ZM, Paez D, Castillo M, Calvo M, Chang HW, Cummins DD, Chung M, Yeroushalmi S, Bartholomew E, Hakimi M, Ye CJ, Bhutani T, Matloubian M, Gensler LS, Liao W. Combined Single Cell Transcriptome and Surface Epitope Profiling Identifies Potential Biomarkers of Psoriatic Arthritis and Facilitates Diagnosis via Machine Learning. Front Immunol 2022; 13:835760. [PMID: 35309349 PMCID: PMC8924042 DOI: 10.3389/fimmu.2022.835760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Early diagnosis of psoriatic arthritis (PSA) is important for successful therapeutic intervention but currently remains challenging due, in part, to the scarcity of non-invasive biomarkers. In this study, we performed single cell profiling of transcriptome and cell surface protein expression to compare the peripheral blood immunocyte populations of individuals with PSA, individuals with cutaneous psoriasis (PSO) alone, and healthy individuals. We identified genes and proteins differentially expressed between PSA, PSO, and healthy subjects across 30 immune cell types and observed that some cell types, as well as specific phenotypic subsets of cells, differed in abundance between these cohorts. Cell type-specific gene and protein expression differences between PSA, PSO, and healthy groups, along with 200 previously published genetic risk factors for PSA, were further used to perform machine learning classification, with the best models achieving AUROC ≥ 0.87 when either classifying subjects among the three groups or specifically distinguishing PSA from PSO. Our findings thus expand the repertoire of gene, protein, and cellular biomarkers relevant to PSA and demonstrate the utility of machine learning-based diagnostics for this disease.
Collapse
Affiliation(s)
- Jared Liu
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Sugandh Kumar
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Julie Hong
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Zhi-Ming Huang
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Diana Paez
- Division of Rheumatology, Department of Medicine, University of California at San Francisco, San Francisco, CA, United States
| | - Maria Castillo
- Division of Rheumatology, Department of Medicine, University of California at San Francisco, San Francisco, CA, United States
| | - Maria Calvo
- Division of Rheumatology, Department of Medicine, University of California at San Francisco, San Francisco, CA, United States
| | - Hsin-Wen Chang
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Daniel D. Cummins
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Mimi Chung
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Samuel Yeroushalmi
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Erin Bartholomew
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Marwa Hakimi
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Chun Jimmie Ye
- Division of Rheumatology, Department of Medicine, University of California at San Francisco, San Francisco, CA, United States
- Institute for Human Genetics, University of California at San Francisco, San Francisco, CA, United States
- Department of Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA, United States
- Institute of Computational Health Sciences, University of California at San Francisco, San Francisco, CA, United States
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Tina Bhutani
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
| | - Mehrdad Matloubian
- Division of Rheumatology, Department of Medicine, University of California at San Francisco, San Francisco, CA, United States
- Rosalind Russell/Ephraim P Engleman Rheumatology Research Center, University of California at San Francisco, San Francisco, CA, United States
| | - Lianne S. Gensler
- Division of Rheumatology, Department of Medicine, University of California at San Francisco, San Francisco, CA, United States
| | - Wilson Liao
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, United States
- Institute for Human Genetics, University of California at San Francisco, San Francisco, CA, United States
| |
Collapse
|
54
|
Liu P, Kuang Y, Ye L, Peng C, Chen W, Shen M, Zhang M, Zhu W, Lv C, Chen X. Predicting the Risk of Psoriatic Arthritis in Plaque Psoriasis Patients: Development and Assessment of a New Predictive Nomogram. Front Immunol 2022; 12:740968. [PMID: 35126345 PMCID: PMC8810526 DOI: 10.3389/fimmu.2021.740968] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/20/2021] [Indexed: 12/31/2022] Open
Abstract
ObjectiveThis study aimed to develop a risk of psoriatic arthritis (PsA) predictive model for plaque psoriasis patients based on the available features.MethodsPatients with plaque psoriasis or PsA were recruited. The characteristics, skin lesions, and nail clinical manifestations of the patients have been collected. The least absolute shrinkage was used to optimize feature selection, and logistic regression analysis was applied to further select features and build a PsA risk predictive model. Calibration, discrimination, and clinical utility of the prediction model were evaluated by using the calibration plot, C-index, the area under the curve (AUC), and decision curve analysis. Internal validation was performed using bootstrapping validation. The model was subjected to external validation with two separate cohorts.ResultsAge at onset, duration, nail involvement, erythematous lunula, onychorrhexis, oil drop, and subungual hyperkeratosis were presented as predictors to perform the prediction nomogram. The predictive model showed good calibration and discrimination (C-index: 0.759; 95% CI: 0.707–0.811). The AUC of this prediction model was 0.7578092. Excellent performances of the C-index were reached in the internal validation and external cohort validation (0.741, 0.844, and 0.845). The decision curve indicated good effect of the PsA nomogram in guiding clinical practice.ConclusionThis novel PsA nomogram could assess the risk of PsA in plaque psoriasis patients with good efficiency.
Collapse
Affiliation(s)
- Panpan Liu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Yehong Kuang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Li Ye
- Dalian Dermatosis Hospital, Dalian, China
| | - Cong Peng
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Wangqing Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Minxue Shen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Mi Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
| | - Wu Zhu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
- Gerontology Center of Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wu Zhu, ; Chengzhi Lv, ; Xiang Chen,
| | - Chengzhi Lv
- Dalian Dermatosis Hospital, Dalian, China
- *Correspondence: Wu Zhu, ; Chengzhi Lv, ; Xiang Chen,
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, China
- Gerontology Center of Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Wu Zhu, ; Chengzhi Lv, ; Xiang Chen,
| |
Collapse
|
55
|
Stuart PE, Tsoi LC, Nair RP, Ghosh M, Kabra M, Shaiq PA, Raja GK, Qamar R, Thelma B, Patrick MT, Parihar A, Singh S, Khandpur S, Kumar U, Wittig M, Degenhardt F, Tejasvi T, Voorhees JJ, Weidinger S, Franke A, Abecasis GR, Sharma VK, Elder JT. Transethnic analysis of psoriasis susceptibility in South Asians and Europeans enhances fine-mapping in the MHC and genomewide. HGG ADVANCES 2022; 3:100069. [PMID: 34927100 PMCID: PMC8682265 DOI: 10.1016/j.xhgg.2021.100069] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 10/24/2021] [Indexed: 02/06/2023] Open
Abstract
Because transethnic analysis may facilitate prioritization of causal genetic variants, we performed a genomewide association study (GWAS) of psoriasis in South Asians (SAS), consisting of 2,590 cases and 1,720 controls. Comparison with our existing European-origin (EUR) GWAS showed that effect sizes of known psoriasis signals were highly correlated in SAS and EUR (Spearman ρ = 0.78; p < 2 × 10-14). Transethnic meta-analysis identified two non-MHC psoriasis loci (1p36.22 and 1q24.2) not previously identified in EUR, which may have regulatory roles. For these two loci, the transethnic GWAS provided higher genetic resolution and reduced the number of potential causal variants compared to using the EUR sample alone. We then explored multiple strategies to develop reference panels for accurately imputing MHC genotypes in both SAS and EUR populations and conducted a fine-mapping of MHC psoriasis associations in SAS and the largest such effort for EUR. HLA-C*06 was the top-ranking MHC locus in both populations but was even more prominent in SAS based on odds ratio, disease liability, model fit and predictive power. Transethnic modeling also substantially boosted the probability that the HLA-C*06 protein variant is causal. Secondary MHC signals included coding variants of HLA-C and HLA-B, but also potential regulatory variants of these two genes as well as HLA-A and several HLA class II genes, with effects on both chromatin accessibility and gene expression. This study highlights the shared genetic basis of psoriasis in SAS and EUR populations and the value of transethnic meta-analysis for discovery and fine-mapping of susceptibility loci.
Collapse
Affiliation(s)
- Philip E. Stuart
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor MI, USA
| | - Rajan P. Nair
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Manju Ghosh
- Department of Pediatrics Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Madhulika Kabra
- Department of Pediatrics Genetics, All India Institute of Medical Sciences, New Delhi, India
| | - Pakeeza A. Shaiq
- Department of Biochemistry, PMASAA University, Rawalpindi, Pakistan
| | - Ghazala K. Raja
- Department of Biochemistry, PMASAA University, Rawalpindi, Pakistan
| | - Raheel Qamar
- COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - B.K. Thelma
- Department of Genetics, University of Delhi South Campus, 110021 New Delhi, India
| | - Matthew T. Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anita Parihar
- Department of Dermatology, All India Institute of Medical Sciences, New Delhi, India
| | - Sonam Singh
- Department of Dermatology, All India Institute of Medical Sciences, New Delhi, India
| | - Sujay Khandpur
- Department of Dermatology, All India Institute of Medical Sciences, New Delhi, India
| | - Uma Kumar
- Department of Rheumatology, All India Institute of Medical Sciences, New Delhi, India
| | - Michael Wittig
- Institute of Clinical Molecular Biology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel 24105, Germany
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel 24105, Germany
| | - Trilokraj Tejasvi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA
| | - John J. Voorhees
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stephan Weidinger
- Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel 24105, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel 24105, Germany
| | - Goncalo R. Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Vinod K. Sharma
- Department of Dermatology, All India Institute of Medical Sciences, New Delhi, India
| | - James T. Elder
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA
| |
Collapse
|
56
|
Laborde CM, Larzabal L, González-Cantero Á, Castro-Santos P, Díaz-Peña R. Advances of Genomic Medicine in Psoriatic Arthritis. J Pers Med 2022; 12:jpm12010035. [PMID: 35055350 PMCID: PMC8780979 DOI: 10.3390/jpm12010035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Psoriatic arthritis (PsA) is a common type of inflammatory arthritis found in up to 40% of patients with psoriasis. Although early diagnosis is important for reducing the risk of irreversible structural damage, there are no adequate screening tools for this purpose, and there are no clear markers of predisposition to the disease. Much evidence indicates that PsA disorder is complex and heterogeneous, where genetic and environmental factors converge to trigger inflammatory events and the development of the disease. Nevertheless, the etiologic events that underlie PsA are complex and not completely understood. In this review, we describe the existing data in PsA in order to highlight the need for further research in this disease to progress in the knowledge of its pathobiology and to obtain early diagnosis tools for these patients.
Collapse
Affiliation(s)
| | | | - Álvaro González-Cantero
- Department of Dermatology, Hospital Universitario Ramon y Cajal, 28034 Madrid, Spain;
- Faculty of Medicine, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda, 28223 Pozuelo de Alarcón, 28034 Madrid, Spain
| | - Patricia Castro-Santos
- Immunology, Centro de Investigaciones Biomédicas (CINBIO), Universidad de Vigo, 36310 Vigo, Spain;
| | - Roberto Díaz-Peña
- Faculty of Health Sciences, Universidad Autónoma de Chile, Talca 3460000, Chile
- Correspondence: or ; Tel.: +34-981-955-073
| |
Collapse
|
57
|
Batko B. Exploring the Diverse Immune and Genetic Landscape of Psoriatic Arthritis. J Clin Med 2021; 10:jcm10245926. [PMID: 34945224 PMCID: PMC8706996 DOI: 10.3390/jcm10245926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Psoriatic arthritis (PsA) is characterized by delays in diagnosis and modest effect of treatment in terms of joint response. An understanding of molecular pathomechanisms may aid in developing diagnostic and prognostic models. Genetic susceptibility (e.g., HLA class I genes, IL-23-related genes) can be responsible for the pattern of psoriatic manifestations and affinity for tissue involvement. Gene expression analysis indicates an inflammatory profile that is distinct for PsA, but disparate across tissues. This has clinical implications, as for example, dual blockade of IL-17A and IL-17F can lead to superior clinical effects if there is differential expression of IL-17 receptors in tissues. Structural and functional impairment of barrier tissue, including host-microbiome interactions, may be the source of immune activation. Interplay between different cell populations of innate and adaptive immunity is emerging, potentially providing a link between the transition of skin-to-joint disease. Th17 subsets, IL-17A, IL-17F and IL-23 are crucial in PsA pathogenesis, with both clinical and experimental evidence suggesting a differential molecular landscape in cutaneous and articular compartments.
Collapse
Affiliation(s)
- Bogdan Batko
- Department of Rheumatology and Immunology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, 30-705 Krakow, Poland
| |
Collapse
|
58
|
Qi F, Tan Y, Yao A, Yang X, He Y. Psoriasis to Psoriatic Arthritis: The Application of Proteomics Technologies. Front Med (Lausanne) 2021; 8:681172. [PMID: 34869404 PMCID: PMC8635007 DOI: 10.3389/fmed.2021.681172] [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: 03/16/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Psoriatic disease (PsD) is a spectrum of diseases that affect both skin [cutaneous psoriasis (PsC)] and musculoskeletal features [psoriatic arthritis (PsA)]. A considerable number of patients with PsC have asymptomatic synovio-entheseal inflammations, and approximately one-third of those eventually progress to PsA with an enigmatic mechanism. Published studies have shown that early interventions to the very early-stage PsA would effectively prevent substantial bone destructions or deformities, suggesting an unmet goal for exploring early PsA biomarkers. The emergence of proteomics technologies brings a complete view of all involved proteins in PsA transitions, offers a unique chance to map all potential peptides, and allows a direct head-to-head comparison of interaction pathways in PsC and PsA. This review summarized the latest development of proteomics technologies, highlighted its application in PsA biomarker discovery, and discussed the possible clinical detectable PsA risk factors in patients with PsC.
Collapse
Affiliation(s)
- Fei Qi
- Department of Dermatology, Capital Medical University Affiliated Beijing Chaoyang Hospital, Beijing, China
| | - Yaqi Tan
- Department of Dermatology, Capital Medical University Affiliated Beijing Chaoyang Hospital, Beijing, China
| | - Amin Yao
- Department of Dermatology, Capital Medical University Affiliated Beijing Chaoyang Hospital, Beijing, China
| | - Xutong Yang
- Department of Dermatology, Capital Medical University Affiliated Beijing Chaoyang Hospital, Beijing, China
| | - Yanling He
- Department of Dermatology, Capital Medical University Affiliated Beijing Chaoyang Hospital, Beijing, China
| |
Collapse
|
59
|
Manou-Stathopoulou S, Lewis MJ. Diversity of NF-κB signalling and inflammatory heterogeneity in Rheumatic Autoimmune Disease. Semin Immunol 2021; 58:101649. [PMID: 36064646 DOI: 10.1016/j.smim.2022.101649] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systemic Autoimmune Rheumatic Diseases, including Rheumatoid Arthritis, Systemic Lupus Erythematosus and Sjogren's syndrome, are characterised by a loss of immune tolerance and chronic inflammation. There is marked heterogeneity in clinical and molecular phenotypes in each condition, and the aetiology of these is unclear. NF-κB is an inducible transcription factor that is critical in the physiological inflammatory response, and which has been implicated in chronic inflammation. Genome-wide association studies have linked risk alleles related to the NF-κB pathway to the pathogenesis of multiple Systemic Autoimmune Rheumatic Diseases. This review describes how cell- and pathway-specific NF-κB activation contribute to the spectrum of clinical phenotypes and molecular pathotypes in rheumatic disease. Potential clinical applications are explored, including therapeutic interventions and utilisation of NF-κB as a biomarker of disease subtypes and treatment response.
Collapse
Affiliation(s)
- Sotiria Manou-Stathopoulou
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
60
|
Li YK, Zhang XX, Yang Y, Gao J, Shi Q, Liu SD, Fu WP, Sun C. Convergent Evidence Supports TH2LCRR as a Novel Asthma Susceptibility Gene. Am J Respir Cell Mol Biol 2021; 66:283-292. [PMID: 34851809 DOI: 10.1165/rcmb.2020-0481oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Asthma is a common complex disease with apparent genetic predispositions, and previous genome-wide association studies suggest that rs1295686 within the IL13 (interleukin 13) gene is significantly associated with asthma. Analysis of the data provided by the 1000 Genomes Project indicated that there are additional four SNPs in nearly complete linkage disequilibrium with rs1295686 in Caucasians. However, the causal SNPs and the associated mechanism remain unclear. To investigate this issue, functional genomics approaches were utilized to analyze the functions of these SNPs. Dual-luciferase assays indicated that the functional SNP is not rs1295686 but a haplotype consisting of other three SNPs, rs1295685, rs848 and rs847. Through chromosome conformation capture, it was found that the enhancer containing the three functional SNPs interacts with the promoter of TH2LCRR (T helper type 2 locus control region associated RNA), a recently identified long non-coding RNA. RNA-seq data analysis indicated that TH2LCRR expression is significantly increased in asthma patients and is dependent on the genotype at this locus, indicating that TH2LCRR is a novel susceptibility gene for asthma and that these SNPs confer asthma risk by regulating TH2LCRR expression. By chromatin immunoprecipitation, the related transcription factors that bind in the region surrounding these three SNPs were identified, and their interactions were investigated by functional genomics approaches. Our effort identified a novel mechanism through which genetic variations at this locus could influence asthma susceptibility.
Collapse
Affiliation(s)
- Yi-Kun Li
- Shaanxi Normal University, 12401, College of Life Sciences, Xi'an, China
| | - Xin-Xin Zhang
- Shaanxi Normal University, 12401, College of Life Sciences, Xi'an, China
| | - Yuan Yang
- Shaanxi Normal University, 12401, College of Life Sciences, Xi'an, China
| | - Jing Gao
- Shaanxi Normal University, 12401, College of Life Sciences, Xi'an, China
| | - Qiang Shi
- Shaanxi Normal University, 12401, College of Life Sciences, Xi'an, China
| | - Shao-Dong Liu
- Shaanxi Normal University, 12401, College of Life Sciences, Xi'an, China
| | - Wei-Ping Fu
- Kunming Medical University First Affilliated Hospital, 36657, Kunming, China
| | - Chang Sun
- Shaanxi Normal University, 12401, College of Life Sciences, Xi'an, China;
| |
Collapse
|
61
|
Advanced genomics and clinical phenotypes in psoriatic arthritis. Semin Immunol 2021; 58:101665. [PMID: 36307312 DOI: 10.1016/j.smim.2022.101665] [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] [Indexed: 12/13/2022]
Abstract
Psoriatic Arthritis (PsA) is a complex polygenic inflammatory disease showing a variable musculoskeletal involvement in patients with skin psoriasis. PsA coexist in 25-40 % of patients with the dermatological manifestations, but PsA may also predate the appearance of psoriasis. Nonetheless, the immunopathogenesis of psoriasis and PsA manifest significant similarities, with a major role of the individual susceptibility in both cases. Genome wide association studies (GWAS) identified several genes/loci associated with the risk to develop PsA, both dependent and independent of psoriasis. The major challenge is thus represented by the need to translate the identification of functional polymorphisms and other genetics findings into biological mechanisms along with the identification of novel putative drug targets. A functional genomics approach aims to increase GWAS power and recent evidence supports the use of a multilayer process, including eQTL, methylome, chromatin conformation analysis and genome editing to discover novel genes that can be affected by disease-associated variants, such as PsA. The available data have considered PsA as a unique homogeneous clinical entity while the clinical experience supports a wide variability of skin and joint manifestations coexisting in diverse patients with different mechanisms underlying the musculoskeletal and dermatological domains. A better discrimination of the patient features is encouraged by the limited data on functional genomics. We provide herein a review of the latest findings on PsA functional genomics highlighting the exciting developments in the field and how these might lead to a better understanding of gene regulation underpinning disease mechanisms and ultimately refine clinical phenotyping.
Collapse
|
62
|
Queiro R, Coto P, González-Lara L, Coto E. Genetic Variants of the NF-κB Pathway: Unraveling the Genetic Architecture of Psoriatic Disease. Int J Mol Sci 2021; 22:ijms222313004. [PMID: 34884808 PMCID: PMC8657577 DOI: 10.3390/ijms222313004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 11/27/2021] [Accepted: 11/29/2021] [Indexed: 01/02/2023] Open
Abstract
Psoriasis is a multifactorial genetic disease for which the genetic factors explain about 70% of disease susceptibility. Up to 30–40% of psoriasis patients develop psoriatic arthritis (PsA). However, PsA can be considered as a “disease within a disease”, since in most cases psoriasis is already present when joint complaints begin. This has made studies that attempt to unravel the genetic basis for both components of psoriatic disease enormously difficult. Psoriatic disease is also accompanied by a high burden of comorbid conditions, mainly of the cardiometabolic type. It is currently unclear whether these comorbidities and psoriatic disease have a shared genetic basis or not. The nuclear factor of kappa light chain enhancer of activated B cells (NF-κB) is a transcription factor that regulates a plethora of genes in response to infection, inflammation, and a wide variety of stimuli on several cell types. This mini-review is focused on recent findings that highlight the importance of this pathway both in the susceptibility and in the determinism of some features of psoriatic disease. We also briefly review the importance of genetic variants of this pathway as biomarkers of pharmacological response. All the above may help to better understand the etiopathogenesis of this complex entity.
Collapse
Affiliation(s)
- Rubén Queiro
- Rheumatology & ISPA Translational Immunology Division, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Department of Medicine, Oviedo University School of Medicine, 33011 Oviedo, Spain;
- Correspondence:
| | - Pablo Coto
- Dermatology Division, Hospital Vital Alvarez Buylla, 33611 Mieres, Spain;
| | - Leire González-Lara
- Dermatology Division, Hôpital Ambroise-Paré, 92100 Boulogne-Billancourt, France;
| | - Eliecer Coto
- Department of Medicine, Oviedo University School of Medicine, 33011 Oviedo, Spain;
- Molecular Genetics Unit, Hospital Universitario Central Asturias, 33011 Oviedo, Spain
| |
Collapse
|
63
|
Gao J, Shen X, Ko R, Huang C, Shen C. Cognitive Process of Psoriasis and Its Comorbidities: From Epidemiology to Genetics. Front Genet 2021; 12:735124. [PMID: 34899832 PMCID: PMC8662384 DOI: 10.3389/fgene.2021.735124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Psoriasis (PsO) is a chronic inflammatory skin disease that affects approximately 2% of the population all over the world. Comorbidities of PsO have increasingly garnered more interest in the past decades. Compared with the normal population, the incidences of comorbidities are higher among patients with PsO. In the last 20 years, researchers have focused on studying the genetic components of PsO, and genetic associations between PsO and its comorbidities were elucidated. This review provides an in-depth understanding and summarization of the connection between PsO and its comorbidities from the perspectives of epidemiology and genetics. Further understanding of PsO and its comorbidities will promote research on the pathogenesis, drug development, novel therapy methods, and personalized and precision treatment of PsO and its comorbidities.
Collapse
Affiliation(s)
- Jing Gao
- Department of Dermatology, the Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Xue Shen
- Department of Dermatology, Chengdu Second People’s Hospital, Chengdu, China
| | - Randy Ko
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Cong Huang
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University–the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Changbing Shen
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University–the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
64
|
Kubanov AA, Karamova AE, Chikin VV, Verbenko DA, Znamenskaya LF, Artamonova OG. Genetic markers for psoriatic arthritis in patients with psoriasis. Part I: non-HLA genes. VESTNIK DERMATOLOGII I VENEROLOGII 2021. [DOI: 10.25208/vdv1260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Psoriatic arthritis often develops in patients with psoriasis and can lead to joint deformity, stiffness, dysfunction, and disability. Psoriatic arthritis is a polygenic disease. and the issue of personalizing the prognosis of its development can only be resolved taking into account the variability of plenty genomic loci associated with the development of the disease. The personification of the prognosis of the disease can be solved taking into account the variability of the set of genomic loci with which its development is associated. The review examines genomic polymorphisms associated with the development of psoriatic arthritis not psoriasis, except of HLA polymorphisms. Genome regions containing polymorphisms, allelic variants of which are associated both with the development of psoriatic arthritis and reducing the likelihood of its occurrence, are described. It has been reported that the predisposition to the development of psoriatic arthritis in patients with psoriasis is determined by genes encoding proteins involved in inflammation and bone metabolism.
Collapse
|
65
|
McInnes IB, Szekanecz Z, McGonagle D, Maksymowych WP, Pfeil A, Lippe R, Song IH, Lertratanakul A, Sornasse T, Biljan A, Deodhar A. A review of JAK-STAT signalling in the pathogenesis of spondyloarthritis and the role of JAK inhibition. Rheumatology (Oxford) 2021; 61:1783-1794. [PMID: 34668515 PMCID: PMC9071532 DOI: 10.1093/rheumatology/keab740] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
Spondyloarthritis (SpA) comprises a group of chronic inflammatory diseases with overlapping clinical, genetic and pathophysiological features including back pain, peripheral arthritis, psoriasis, enthesitis and dactylitis. Several cytokines are involved in the pathogenesis of SpA, variously contributing to each clinical manifestation. Many SpA-associated cytokines, including IL-23, IL-17, IL-6, type I/II interferon and tumour necrosis factor signal directly or indirectly via the Janus kinase (JAK)–signal transducer and activator of transcription pathway. JAK signalling also regulates development and maturation of cells of the innate and adaptive immune systems. Accordingly, disruption of this signalling pathway by small molecule oral JAK inhibitors can inhibit signalling implicated in SpA pathogenesis. Herein we discuss the role of JAK signalling in the pathogenesis of SpA and summarize the safety and efficacy of JAK inhibition by reference to relevant SpA clinical trials.
Collapse
Affiliation(s)
- Iain B McInnes
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Zoltán Szekanecz
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,National Institute for Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, UK
| | - Walter P Maksymowych
- Division of Rheumatology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Pfeil
- Department of Internal Medicine III, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Ralph Lippe
- AbbVie Deutschland GmbH & Co. KG, Wiesbaden, Germany
| | | | | | | | | | - Atul Deodhar
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
66
|
Chia AYT, Ang GWX, Chan ASY, Chan W, Chong TKY, Leung YY. Managing Psoriatic Arthritis With Inflammatory Bowel Disease and/or Uveitis. Front Med (Lausanne) 2021; 8:737256. [PMID: 34604268 PMCID: PMC8481670 DOI: 10.3389/fmed.2021.737256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease that presents with psoriasis (PsO), peripheral and axial arthropathy. The heterogeneity of disease presentation leads to the term "psoriatic disease (PsD)" which is thought to better encompass the range of clinical manifestations. PsA is associated with several comorbidities such as cardiovascular diseases, metabolic syndrome and other extra-articular manifestations including uveitis, and inflammatory bowel disease (IBD). While novel therapeutics are being developed following advances in our understanding of the pathogenesis of the disease, the diverse combinations of PsA with its various comorbidities still pose a clinical challenge in managing patients with PsA. This article reviews our current understanding of the pathogenesis of PsA and how various pathways in the pathogenesis lead to the two comorbid extra-articular manifestations - uveitis and IBD. We also review current evidence of treatment strategies in managing patients with PsA with comorbidities of uveitis and/or IBD.
Collapse
Affiliation(s)
- Alfred Yu Ting Chia
- Duke-NUS Medical School, Singapore, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Duke-NUS Medical School, Singapore, Singapore
| | - Gladys Wei Xin Ang
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Duke-NUS Medical School, Singapore, Singapore
| | - Anita Sook Yee Chan
- Duke-NUS Medical School, Singapore, Singapore
- Singapore National Eye Center and Singapore Eye Research Center, Singapore, Singapore
| | - Webber Chan
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
| | | | - Ying Ying Leung
- Duke-NUS Medical School, Singapore, Singapore
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
67
|
Yadav K, Singh D, Singh MR. Novel archetype in psoriasis management bridging molecular dynamics in exploring novel therapies. Eur J Pharmacol 2021; 907:174254. [PMID: 34118225 DOI: 10.1016/j.ejphar.2021.174254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Psoriasis is an autoimmune chronic inflammatory condition of skin affecting 125 million populaces around the globe. It is implicated as a result of multifaceted phenomena involving various cell and subcell activities with the aid of numerous cellular and molecular components including signaling aisle and regulatory proteins owing to the development of such hyperproliferative dermatological conditions. This involves a deeply complex and conflicting pathology owing to genetic and immunological deviations resulting from the unusual presentation of different signaling pathways and regulatory proteins. Explorations of these biomarkers and intervention of molecular and cellular processes in psoriasis are yet to be investigated and could be an exceptional aspect for understanding pathology with successful targeting of disease. In the presented study, we have integrated molecular insights, including signaling molecules, pathways, and proteins implicated in pathogenesis, and we have attempted to link this knowledge to the targeting of these phenomena in order to manage the conditions precisely. Further, therapeutic delivery approaches for targeting distinct layers of skin have also been investigated based on the application of different nanocarriers for successful psoriasis treatment.
Collapse
Affiliation(s)
- Krishna Yadav
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492010, India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492010, India.
| |
Collapse
|
68
|
Grivas A, Fragoulis G, Garantziotis P, Banos A, Nikiphorou E, Boumpas D. Unraveling the complexities of psoriatic arthritis by the use of -Omics and their relevance for clinical care. Autoimmun Rev 2021; 20:102949. [PMID: 34509654 DOI: 10.1016/j.autrev.2021.102949] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022]
Abstract
-Omic technologies represent a novel approach to unravel ill-defined aspects of psoriatic arthritis (PsA). Large-scale information can be acquired from analysis of affected tissues in PsA via high-throughput studies in the domains of genomics, transcriptomics, epigenetics, proteomics and metabolomics. This is a critical overview of the current knowledge of -omics in PsA, with emphasis on the pathophysiological insights of diagnostic and therapeutic relevance, the advent of novel biomarkers and their potential use for precision medicine in PsA.
Collapse
Affiliation(s)
- Alexandros Grivas
- National and Kapodistrian University of Athens, Faculty of medicine, Athens, Greece; Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
| | - George Fragoulis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, Greece
| | - Panagiotis Garantziotis
- Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece; Division of Immunology and Rheumatology, Hannover Medical University, 30,625 Hannover, Germany
| | - Aggelos Banos
- Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, School of Immunology and Microbial Sciences, King's College London, King's Hospital, London, United Kingdom
| | - Dimitrios Boumpas
- National and Kapodistrian University of Athens, Faculty of medicine, Athens, Greece; Inflammation & Autoimmunity Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| |
Collapse
|
69
|
Pennington SR, FitzGerald O. Early Origins of Psoriatic Arthritis: Clinical, Genetic and Molecular Biomarkers of Progression From Psoriasis to Psoriatic Arthritis. Front Med (Lausanne) 2021; 8:723944. [PMID: 34485351 PMCID: PMC8416317 DOI: 10.3389/fmed.2021.723944] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/23/2021] [Indexed: 12/26/2022] Open
Abstract
Greater than 90% of patients with psoriatic arthritis (PsA) first develop their arthritis on a background of known psoriasis (Pso). Thus, having skin/nail Pso certainly is an important risk factor for PsA but as PsA develops in <30% of those affected with Pso, the presence of Pso alone is insufficient as a means of identifying which patients with Pso will develop PsA. It is hoped that with further molecular assessment of Pso patients who do not have any evidence of inflammatory musculoskeletal disease compared to those with early PsA features, that the “at risk” profile of Pso patients destined to develop PsA can be refined such that disease prevention studies can be designed and a new era of treatment for PsA can emerge. In this article, the early stages in the development of PsA are outlined and what is currently known about clinical features, genetic factors and soluble or tissue biomarkers associated with the development of PsA in patients with Pso is reviewed in detail. Finally, proposals are outlined regarding the approaches required in order to address this important research area.
Collapse
Affiliation(s)
- Stephen R Pennington
- Conway Institute for Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| | - Oliver FitzGerald
- Conway Institute for Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
70
|
Yan D, Gudjonsson JE, Le S, Maverakis E, Plazyo O, Ritchlin C, Scher JU, Singh R, Ward NL, Bell S, Liao W. New Frontiers in Psoriatic Disease Research, Part I: Genetics, Environmental Triggers, Immunology, Pathophysiology, and Precision Medicine. J Invest Dermatol 2021; 141:2112-2122.e3. [PMID: 34303522 PMCID: PMC8384663 DOI: 10.1016/j.jid.2021.02.764] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
Psoriasis is a chronic inflammatory condition characterized by systemic immune dysregulation. Over the past several years, advances in genetics, microbiology, immunology, and mouse models have revealed the complex interplay between the heritable and microenvironmental factors that drive the development of psoriatic inflammation. In the first of this two-part review series, the authors will discuss the newest insights into the pathogenesis of psoriatic disease and highlight how the evolution of these scientific fields has paved the way for a more personalized approach to psoriatic disease treatment.
Collapse
Affiliation(s)
- Di Yan
- Ronald O. Perelman Department of Dermatology, NYU Langone Health, New York, New York, USA
| | | | - Stephanie Le
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Olesya Plazyo
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Ritchlin
- Center for Musculoskeletal Research, Division of Allergy, Immunology and Rheumatology, University of Rochester School of Medicine & Dentistry, Rochester, New York, USA
| | - Jose U Scher
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Roopesh Singh
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nicole L Ward
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Stacie Bell
- National Psoriasis Foundation, Portland, Oregon, USA
| | - Wilson Liao
- UCSF Department of Dermatology, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
71
|
EMMUNGİL H, İLGEN U, DİRESKENELİ RH. Autoimmunity in psoriatic arthritis: pathophysiological and clinical aspects. Turk J Med Sci 2021; 51:1601-1614. [PMID: 33581710 PMCID: PMC8569784 DOI: 10.3906/sag-2011-235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/13/2021] [Indexed: 11/03/2022] Open
Abstract
Psoriatic arthritis (PsA) is an underdiagnosed entity with a broad impact on the quality of life. Although the pathogenesis is largely unknown, autoimmune footprints of the inflammation in PsA have increasingly been recognized. Most of the genetic variation predisposing to PsA is mapped to the class I major histocompatibility complex (MHC) region and shared by a variety of autoimmune diseases. Polymorphisms in the genes IL12B, IL23R, IL13, TNIP1, TRAF3IP2, TYK2, and many others explain the non- HLA genetic risk with little known functional consequences. Entheseal and synovial cellular infiltrate with oligoclonal CD8+ T cells and occasional germinal centers, loss of regulatory T cell function, and specific autoantibodies such as anti-PsA peptide, anti-LL-37, and anti-ADAMTSL5 are the immunopathological findings suggestive of autoimmunity. These were supported by clinical observations of autoimmune multimorbidity and treatment response to calcineurin/mTOR and co-stimulation inhibition.
Collapse
Affiliation(s)
- Hakan EMMUNGİL
- Division of Rheumatology, Department of Rheumatology, Trakya University Medical Faculty, EdirneTurkey
| | - Ufuk İLGEN
- Division of Rheumatology, Department of Rheumatology, Trakya University Medical Faculty, EdirneTurkey
| | - Rafi Haner DİRESKENELİ
- Division of Rheumatology, Department of Rheumatology, Marmara University Medical Faculty, İstanbulTurkey
| |
Collapse
|
72
|
Volumetric Bone Mineral Density Measured by HR-pQCT in Patients with Psoriasis or Psoriatic Arthritis: A Systematic Review and Meta-Analysis with Trial Sequential Analysis. Healthcare (Basel) 2021; 9:healthcare9081056. [PMID: 34442193 PMCID: PMC8393585 DOI: 10.3390/healthcare9081056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/20/2022] Open
Abstract
Bone health in psoriasis and psoriatic arthritis has been emphasized in recent years. Novel imaging modalities allow investigations into volumetric bone mineral density (vBMD) and bone microstructure in psoriatic patients. However, literature regarding vBMD measured by high-resolution peripheral quantitative computed tomography (HR-pQCT) is inconclusive. We conducted a systematic review and meta-analysis to evaluate vBMD in patients with psoriatic disease. We searched PubMed, EMBASE, Web of Science, and Cochrane Library for relevant observational studies. A random-effects meta-analysis with trial sequential analysis (TSA) was performed. The pooled mean difference (MD) and 95% confidence interval (CI) were calculated. Five studies with 780 patients were included. Patients with psoriatic disease showed a lower average vBMD than controls (MD -14.90; 95% CI -22.90 to -6.89; TSA-adjusted CI -23.77 to -6.03; I2 = 41%). Trabecular vBMD and cortical vBMD results were inconclusive because of the small sample size. Patients recruited in Asia and those whose vBMD were measured at the distal radius exhibited a lower average vBMD than controls. Further research should clarify the association of psoriasis with bone microstructure and the underlying pathophysiology.
Collapse
|
73
|
Arenella M, Cadby G, De Witte W, Jones RM, Whitehouse AJ, Moses EK, Fornito A, Bellgrove MA, Hawi Z, Johnson B, Tiego J, Buitelaar JK, Kiemeney LA, Poelmans G, Bralten J. Potential role for immune-related genes in autism spectrum disorders: Evidence from genome-wide association meta-analysis of autistic traits. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2021; 26:361-372. [PMID: 34344231 PMCID: PMC8814945 DOI: 10.1177/13623613211019547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The clinical heterogeneity of autism spectrum disorders majorly challenges their genetic study. Autism spectrum disorders symptoms occur in milder forms in the general population, as autistic-like traits, and share genetic factors with autism spectrum disorders. Here, we investigate the genetics of individual autistic-like traits to improve our understanding of autism spectrum disorders. We meta-analysed four population-based genome-wide association studies investigating four autistic-like traits – ‘attention-to-detail’, ‘imagination’, ‘rigidity’ and ‘social-skills’ (n = 4600). Using autism spectrum disorder summary statistics from the Psychiatric Genomic Consortium (N = 46,350), we applied polygenic risk score analyses to understand the genetic relationship between autism spectrum disorders and autistic-like traits. Using MAGMA, we performed gene-based and gene co-expression network analyses to delineate involved genes and pathways. We identified two novel genome-wide significant loci – rs6125844 and rs3731197 – associated with ‘attention-to-detail’. We demonstrated shared genetic aetiology between autism spectrum disorders and ‘rigidity’. Analysing top variants and genes, we demonstrated a role of the immune-related genes RNF114, CDKN2A, KAZN, SPATA2 and ZNF816A in autistic-like traits. Brain-based genetic expression analyses further linked autistic-like traits to genes involved in immune functioning, and neuronal and synaptic signalling. Overall, our findings highlight the potential of the autistic-like trait–based approach to address the challenges of genetic research in autism spectrum disorders. We provide novel insights showing a potential role of the immune system in specific autism spectrum disorder dimensions.
Collapse
Affiliation(s)
- Martina Arenella
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.,Radboud University Medical Center, The Netherlands
| | - Gemma Cadby
- The University of Western Australia, Australia
| | | | | | | | - Eric K Moses
- The University of Western Australia, Australia.,University of Tasmania, Australia
| | - Alex Fornito
- Turner Institute of Brain and Mental Health, Australia.,Monash University, Australia
| | - Mark A Bellgrove
- Turner Institute of Brain and Mental Health, Australia.,Monash University, Australia
| | - Ziarih Hawi
- Turner Institute of Brain and Mental Health, Australia.,Monash University, Australia
| | - Beth Johnson
- Turner Institute of Brain and Mental Health, Australia.,Monash University, Australia
| | - Jeggan Tiego
- Turner Institute of Brain and Mental Health, Australia.,Monash University, Australia
| | - Jan K Buitelaar
- Radboud University Medical Center, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, The Netherlands.,Karakter Child and Adolescent Psychiatry University Centre, The Netherlands
| | | | | | - Janita Bralten
- Radboud University Medical Center, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, The Netherlands
| |
Collapse
|
74
|
Shi C, Ray-Jones H, Ding J, Duffus K, Fu Y, Gaddi VP, Gough O, Hankinson J, Martin P, McGovern A, Yarwood A, Gaffney P, Eyre S, Rattray M, Warren RB, Orozco G. Chromatin Looping Links Target Genes with Genetic Risk Loci for Dermatological Traits. J Invest Dermatol 2021; 141:1975-1984. [PMID: 33607115 PMCID: PMC8315765 DOI: 10.1016/j.jid.2021.01.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 01/12/2021] [Accepted: 01/21/2021] [Indexed: 02/08/2023]
Abstract
Chromatin looping between regulatory elements and gene promoters presents a potential mechanism whereby disease risk variants affect their target genes. In this study, we use H3K27ac HiChIP, a method for assaying the active chromatin interactome in two cell lines: keratinocytes and skin lymphoma-derived CD8+ T cells. We integrate public datasets for a lymphoblastoid cell line and primary CD4+ T cells and identify gene targets at risk loci for skin-related disorders. Interacting genes enrich for pathways of known importance in each trait, such as cytokine response (psoriatic arthritis and psoriasis) and replicative senescence (melanoma). We show examples of how our analysis can inform changes in the current understanding of multiple psoriasis-associated risk loci. For example, the variant rs10794648, which is generally assigned to IFNLR1, was linked to GRHL3, a gene essential in skin repair and development, in our dataset. Our findings, therefore, indicate a renewed importance of skin-related factors in the risk of disease.
Collapse
Affiliation(s)
- Chenfu Shi
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| | - Helen Ray-Jones
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - James Ding
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Kate Duffus
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Yao Fu
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Vasanthi Priyadarshini Gaddi
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Oliver Gough
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Jenny Hankinson
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Paul Martin
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| | - Amanda McGovern
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Annie Yarwood
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Patrick Gaffney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Steve Eyre
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Magnus Rattray
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Richard B Warren
- Dermatology Centre, Salford Royal NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Gisela Orozco
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
75
|
Deshayes S, Bazille C, El Khouri E, Kone-Paut I, Giurgea I, Georgin-Lavialle S, Martin Silva N, Dumont A, Ollivier I, Amselem S, de Boysson H, Aouba A. Chronic hepatic involvement in the clinical spectrum of A20 haploinsufficiency. Liver Int 2021; 41:1894-1900. [PMID: 33966343 DOI: 10.1111/liv.14935] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/09/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Secondary to tumour necrosis factor-alpha induced protein 3 (TNFAIP3) mutations, A20 haploinsufficiency (HA20) is a recently described autoinflammatory disease with clinical features similar to those of Behçet's and Crohn's diseases but with a constantly expanding clinical spectrum. Here, we describe HA20 liver involvement in three new patients from the same family. METHODS We retrospectively assessed clinical, biological and/or histological findings for eight patients over three generations of the same family with heterozygous mutations in the TNFAIP3 gene (c.259C > T, p.Arg87*). RESULTS The eight patients exhibited the following: aphthous ulcers (8/8, bipolar in 7), autoimmune features (6/8, including 5 with definitive autoimmune disease diagnoses, ie, type I diabetes, Hashimoto thyroiditis, pernicious anaemia, and/or 5 with antinuclear antibodies ≥320), pustulosis/folliculitis (5/8), abdominal pain (4/8), arthralgia (3/8), enlarged cervical lymph nodes (3/8) and pericarditis (1/8). In addition, three patients (twin sisters and their grandmother aged 23 and 70 years, respectively) exhibited persistent mild hepatic cytolysis associated with splenomegaly (n = 3), hepatomegaly (n = 1) and/or liver atrophy (n = 1) on echography. We could not detect any other causes of chronic liver diseases. Liver biopsies from three patients displayed hepatic fibrosis, hepatocyte injury and/or CD4+ /CD8+ T lymphocyte infiltration, and patterns of inflammatory cells and NLRP3 or NF-κB immunostaining differed from the predominant neutrophil infiltration observed in skin or some digestive tract biopsies. CONCLUSIONS This study reinforces the dual involvement of innate and adaptive immunity in HA20 according to both acute and chronic injury and the organ involved and widens its clinical spectrum to include chronic hepatic involvement.
Collapse
Affiliation(s)
- Samuel Deshayes
- Department of Internal Medicine, CHU de Caen Normandie, Caen, France.,Normandie Univ, UNICAEN, Caen, France
| | - Céline Bazille
- Department of Pathology, CHU de Caen Normandie, Caen, France
| | - Elma El Khouri
- Department of Genetics, Sorbonne Université, UPMC University, Paris, France
| | - Isabelle Kone-Paut
- Department of Pediatric Rheumatology, Bicêtre Hospital, AP-HP, University of Paris Sud Saclay, CEREMAIA, Le Kremlin-Bicêtre, France
| | - Irina Giurgea
- Department of Genetics, Sorbonne Université, UPMC University, Paris, France
| | - Sophie Georgin-Lavialle
- Department of Internal Medicine, Sorbonne Université, UPMC University, CEREMAIA, Paris, France
| | | | - Anaël Dumont
- Department of Internal Medicine, CHU de Caen Normandie, Caen, France.,Normandie Univ, UNICAEN, Caen, France
| | - Isabelle Ollivier
- Department of Hepatogastroenterology, CHU de Caen Normandie, Caen, France
| | - Serge Amselem
- Department of Genetics, Sorbonne Université, UPMC University, Paris, France
| | - Hubert de Boysson
- Department of Internal Medicine, CHU de Caen Normandie, Caen, France.,Normandie Univ, UNICAEN, Caen, France
| | - Achille Aouba
- Department of Internal Medicine, CHU de Caen Normandie, Caen, France.,Normandie Univ, UNICAEN, Caen, France
| |
Collapse
|
76
|
ALrefai A, Dawood A, Shehata W, Elhelbawy M, Elhelbawy N. Evaluation of NLRP3 (rs10754558) and PTPN22 (1858C/T) (rs2476601) Functional Polymorphisms in Psoriasis Susceptibility in Egypt. Appl Clin Genet 2021; 14:331-339. [PMID: 34345180 PMCID: PMC8324977 DOI: 10.2147/tacg.s319065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Psoriasis is a complex autoimmune multifactorial disease induced by interaction of environmental and genetic factors. This research aimed to clarify the association of NLRP3 (rs10754558) and PTPN22 (1858C/T) (rs2476601) polymorphisms with susceptibility to psoriasis. METHODS This case-control study involved 150 patients diagnosed with psoriasis and 100 age- and gender-matched apparently healthy individuals. NLRP3 (rs10754558) polymorphism was done by real time PCR and PTPN22 1858C/T (rs2476601) genotype was identified by tetra-primer amplification refractory mutation system-polymerase chain reaction (PCR) method. RESULTS The genotypes distribution of NLRP3 (rs10754558) were significantly associated with psoriasis (p<0.0001). Whereas for PTPN22 (1858C/T) (rs2476601), no significance was found (p=0.09). NLRP3 (rs10754558) GC genotype revealed a significant association with psoriasis (p<0.0001), mainly among male (p=0.004) patients with mild psoriasis (p=0.001) and affected extremities (p=0.0001). CONCLUSION We can conclude that the NLRP3 (rs10754558) GC genotype may play a role in psoriasis susceptibility among male Egyptian populations with affected extremities. Future studies must evaluate its role in the prevention or the treatment of psoriasis.
Collapse
Affiliation(s)
- Abeer ALrefai
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin Elkom City, Egypt
- Biochemistry Department, Faculty of Medicine, Umm Al-Qura University, Makah City, Saudi Arabia
| | - Ashraf Dawood
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin Elkom City, Egypt
| | - Wafaa Shehata
- Dermatology, Andrology & Sexually Transmitted Diseases (STDs) Department, Faculty of Medicine, Menoufia University, Shebin Elkom City, Egypt
| | - Mohammed Elhelbawy
- Clinical Pathology Department, Faculty of Medicine, Menoufia University, Shebin Elkom City, Egypt
| | - Nesreen Elhelbawy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Shebin Elkom City, Egypt
| |
Collapse
|
77
|
Prinz JC. Antigen Processing, Presentation, and Tolerance: Role in Autoimmune Skin Diseases. J Invest Dermatol 2021; 142:750-759. [PMID: 34294386 DOI: 10.1016/j.jid.2021.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/12/2021] [Accepted: 05/16/2021] [Indexed: 10/20/2022]
Abstract
Autoreactive T cells pose a constant risk for the emergence of autoimmune skin diseases in genetically predisposed individuals carrying certain HLA risk alleles. Immune tolerance mechanisms are opposed by broad HLA-presented self-immunopeptidomes, a predefined repertoire of polyspecific TCRs, the continuous generation of new antibody specificities by somatic recombination of Ig genes in B cells, and heightened proinflammatory reactivity. Increased autoantigen presentation by HLA molecules, cross-activation of pathogen-induced T cells against autologous structures, altered metabolism of self-proteins, and excessive production of proinflammatory signals may all contribute to the breakdown of immune tolerance and the development of autoimmune skin diseases.
Collapse
Affiliation(s)
- Jörg Christoph Prinz
- Department of Dermatology and Allergy, University Hospital, Ludwig-Maximilian-University of Munich, Munich, Germany.
| |
Collapse
|
78
|
Gonciarz M, Pawlak-Buś K, Leszczyński P, Owczarek W. TYK2 as a therapeutic target in the treatment of autoimmune and inflammatory diseases. Immunotherapy 2021; 13:1135-1150. [PMID: 34235974 DOI: 10.2217/imt-2021-0096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
JAKs are intracellular protein tyrosine kinases that, through activation of STATs, are responsible for signal transduction pathways that regulate cellular responses to numerous cytokines, growth factors and hormones in many different cells. JAK-STAT signaling plays a key role in regulating immune function, and cytokines - such as IL-23, IL-12 and type I interferons - are central to the pathogenesis of autoimmune diseases, including psoriasis, inflammatory bowel disease and systemic lupus erythematosus. Here the authors review the evidence for targeting TYK2 as a more specific approach to treating these conditions. TYK2 inhibitors are clinically effective in autoimmune and inflammatory diseases and may avoid some of the complications reported with nonselective JAK inhibitors.
Collapse
Affiliation(s)
- Maciej Gonciarz
- Department of Gastroenterology & Internal Medicine, Military Institute of Medicine, Warsaw, Poland
| | - Katarzyna Pawlak-Buś
- Department of Rheumatology, Rehabilitation & Internal Medicine, University of Medical Sciences, Poznan, Poland
| | - Piotr Leszczyński
- Department of Rheumatology, Rehabilitation & Internal Medicine, University of Medical Sciences, Poznan, Poland
| | - Witold Owczarek
- Department of Dermatology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
79
|
Hromadová D, Elewaut D, Inman RD, Strobl B, Gracey E. From Science to Success? Targeting Tyrosine Kinase 2 in Spondyloarthritis and Related Chronic Inflammatory Diseases. Front Genet 2021; 12:685280. [PMID: 34290741 PMCID: PMC8287328 DOI: 10.3389/fgene.2021.685280] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022] Open
Abstract
Spondyloarthritis (SpA) is a family of inflammatory arthritic diseases, which includes the prototypes of psoriatic arthritis and ankylosing spondylitis. SpA is commonly associated with systemic inflammatory diseases, such as psoriasis and inflammatory bowel disease. Immunological studies, murine models and the genetics of SpA all indicate a pathogenic role for the IL-23/IL-17 axis. Therapeutics targeting the IL-23/IL-17 pathway are successful at providing symptomatic relief, but may not provide complete protection against progression of arthritis. Thus there is still tremendous interest in the discovery of novel therapeutic targets for SpA. Tyrosine kinase 2 (TYK2) is a member of the Janus kinases, which mediate intracellular signaling of cytokines via signal transducer and activator of transcription (STAT) activation. TYK2 plays a crucial role in mediating IL-23 receptor signaling and STAT3 activation. A plethora of natural mutations in and around TYK2 have provided a wealth of data to associate this kinase with autoimmune/autoinflammatory diseases in humans. Induced and natural mutations in murine Tyk2 largely support human data; however, key inter-species differences exist, which means extrapolation of data from murine models to humans needs to be done with caution. Despite these reservations, novel selective TYK2 inhibitors are now proving successful in advanced clinical trials of inflammatory diseases. In this review, we will discuss TYK2 from basic biology to therapeutic targeting, with an emphasis on studies in SpA. Seminal studies uncovering the basic science of TYK2 have provided sound foundations for targeting it in SpA and related inflammatory diseases. TYK2 inhibitors may well be the next blockbuster therapeutic for SpA.
Collapse
Affiliation(s)
- Dominika Hromadová
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Robert D. Inman
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Departments of Medicine and Immunology, University of Toronto, Toronto, ON, Canada
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Eric Gracey
- Molecular Immunology and Inflammation Unit, VIB Centre for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
80
|
Motta F, Pederzani A, Carena MC, Ceribelli A, Wordsworth PB, De Santis M, Selmi C, Vecellio M. MicroRNAs in Axial Spondylarthritis: an Overview of the Recent Progresses in the Field with a Focus on Ankylosing Spondylitis and Psoriatic Arthritis. Curr Rheumatol Rep 2021; 23:59. [PMID: 34216293 PMCID: PMC8254706 DOI: 10.1007/s11926-021-01027-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/18/2022]
Abstract
Purpose of Review To highlight the recent discoveries and lines of evidence on the role of microRNAs in ankylosing spondylitis (AS) and psoriatic arthritis (PsA), focusing on their expression profiling and mechanisms of action. Recent Findings AS and PsA are chronic inflammatory musculoskeletal diseases with axial manifestations and represent an excellent model for studying microRNAs contribution to the disease pathogenesis, particularly through immunomodulation, inflammation, and bone remodelling, or their value as candidate diagnostic and prognostic biomarkers. Summary MicroRNAs are single-stranded nucleotides able to regulate gene expression. They are a key component of the epigenetic machinery, involved in physiological and pathological processes. The contribution of microRNAs in AS and PsA (such as miR-29a in regulating bone metabolism) is highlighted by several works in the field but their utility as possible markers must be still confirmed, particularly in larger patients’ cohorts.
Collapse
Affiliation(s)
- Francesca Motta
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andrea Pederzani
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Angela Ceribelli
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Paul B Wordsworth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Matteo Vecellio
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy. .,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
81
|
Williams AL, Walton CB, Pinell B, Khadka VS, Dunn B, Lee K, Anagaran MCT, Avelar A, Shohet RV. Ischemic heart injury leads to HIF1-dependent differential splicing of CaMK2γ. Sci Rep 2021; 11:13116. [PMID: 34162925 PMCID: PMC8222303 DOI: 10.1038/s41598-021-92426-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 05/19/2021] [Indexed: 12/04/2022] Open
Abstract
Ischemic heart disease is a leading cause of heart failure and hypoxia inducible factor 1 (HIF1) is a key transcription factor in the response to hypoxic injury. Our lab has developed a mouse model in which a mutated, oxygen-stable form of HIF1α (HIF-PPN) can be inducibly expressed in cardiomyocytes. We observed rapid cardiac dilation and loss of contractility in these mice due to lower expression of excitation-contraction coupling genes and reduced calcium flux. As alternative splicing plays an underappreciated role in transcriptional regulation, we used RNA sequencing to search for splicing changes in calcium-handling genes of HIF-PPN hearts and compared them to previous sequencing data from a model of myocardial infarction (MI) to select for transcripts that are modified in a pathological setting. We found overlap between genes differentially expressed in HIF-PPN and post-MI mice (54/131 genes upregulated in HIF-PPN hearts at 1 day and/or 3 days post-MI, and 45/78 downregulated), as well as changes in alternative splicing. Interestingly, calcium/calmodulin dependent protein kinase II, gamma (CAMK2G) was alternatively spliced in both settings, with variant 1 (v1) substantially decreased compared to variants 2 (v2) and 3 (v3). These findings were also replicated in vitro when cells were transfected with HIF-PPN or exposed to hypoxia. Further analysis of CAMK2γ protein abundance revealed only v1 was detectable and substantially decreased up to 7 days post-MI. Rbfox1, a splicing factor of CAMK2G, was also decreased in HIF-PPN and post-MI hearts. Subcellular fractionation showed CAMK2γ v1 was found in the nuclear and cytoplasmic fractions, and abundance decreased in both fractions post-MI. Chromatin immunoprecipitation analysis of HIF1 in post-MI hearts also demonstrated direct HIF1 binding to CAMK2G. CaMK2 is a key transducer of calcium signals in both physiological and pathological settings. The predominantly expressed isoform in the heart, CaMK2δ, has been extensively studied in cardiac injury, but the specific role of CaMK2γ is not well defined. Our data suggest that loss of CaMK2γ after MI is HIF1-dependent and may play an important role in the heart's calcium signaling and transcriptional response to hypoxia.
Collapse
Affiliation(s)
- Allison Lesher Williams
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA
| | - Chad B Walton
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA
| | - Blake Pinell
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA
| | - Vedbar S Khadka
- Bioinformatics Core, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Brandyn Dunn
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA
| | - Katie Lee
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA
| | - M C Therese Anagaran
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA
| | - Abigail Avelar
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA
| | - Ralph V Shohet
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St. BSB 311, Honolulu, HI, 96813, USA.
| |
Collapse
|
82
|
Mulder MLM, van Hal TW, Wenink MH, Koenen HJPM, van den Hoogen FHJ, de Jong EMGJ, van den Reek JMPA, Vriezekolk JE. Clinical, laboratory, and genetic markers for the development or presence of psoriatic arthritis in psoriasis patients: a systematic review. Arthritis Res Ther 2021; 23:168. [PMID: 34127053 PMCID: PMC8201808 DOI: 10.1186/s13075-021-02545-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
Twenty to thirty percent of psoriasis (Pso) patients will develop psoriatic arthritis (PsA). Detection of Pso patients that are (at risk for) developing PsA is essential to prevent structural damage. We conducted a systematic search of five bibliographic databases, up to May 2020. We searched for studies assessing markers (clinical, laboratory, genetic) associated with the development or presence of PsA in Pso patients. Study selection and quality assessment of the included studies was performed, followed by a qualitative best evidence synthesis to determine the level of evidence for a marker and its association with concomitant/developing PsA in Pso. Overall, 259 possible markers were identified in 119 studies that met the inclusion criteria. Laboratory markers related to inflammation and bone metabolism reached a strong level of evidence for the association (not prediction) of PsA in Pso. Only CXCL10 showed strong evidence for a positive predictive value for PsA in Pso. The importance of timely detecting PsA in a Pso population, and finding more (bio)markers contributing to early detection, remains high.
Collapse
Affiliation(s)
- Michelle L M Mulder
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands. .,Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Tamara W van Hal
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands.,Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark H Wenink
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands
| | - Hans J P M Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Elke M G J de Jong
- Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud University, Nijmegen, The Netherlands.,Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Juul M P A van den Reek
- Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johanna E Vriezekolk
- Department of Rheumatology, Sint Maartenskliniek, PO box 9011, 6500 GM, Nijmegen, The Netherlands
| |
Collapse
|
83
|
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory arthritis with a highly variable clinical presentation that does not have a validated molecular or imaging test, making accurate diagnosis a challenge. Consequences of diagnostic delay include irreversible joint damage and significant morbidity. Over the past few decades, there have been major advances in the understanding and treatment of PsA, leading to more targeted therapies. However, there is no current method to predict optimal treatment strategy to achieve minimal disease activity and prevent medication-related adverse events in the management of early disease. PsA is also associated with other comorbidities that include metabolic syndrome and psychosocial burden; two areas that are often unaddressed in the clinical setting and have associated sequelae. This chapter focuses on key domains of unmet needs, which include diagnostic challenges, delay in diagnosis, prognostication systems and stratified medicine approaches and precision medicine strategies for established and emerging therapies.
Collapse
Affiliation(s)
- Beverly Cheok Kuan Ng
- Clinical Research Fellow, Department of Rheumatology, Addenbrooke's Hospital, Cambridge, UK.
| | - Deepak Rohit Jadon
- Consultant Rheumatologist, Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
84
|
|
85
|
O'Rielly DD, Rahman P. Clinical and molecular significance of genetic loci associated with psoriatic arthritis. Best Pract Res Clin Rheumatol 2021; 35:101691. [PMID: 34020887 DOI: 10.1016/j.berh.2021.101691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Psoriatic arthritis (PsA) is caused by a combination of environmental and multiple genetic factors, with clear evidence for a strong genetic basis. The remarkable accumulation of knowledge gained from genetic, pharmacogenetic, and therapeutic response of biologic agents in PsA has fundamentally changed and advanced our understanding of disease pathogenesis and has identified key signalling pathways. However, only one-quarter of the genetic contribution of PsA has been accounted for; and dissecting the genetic contributors of the cutaneous disease from those that would identify joint disease has been challenging. More importantly, the clinical utility of multiple proposed loci is unclear. In this review, we summarize the potential clinical relevance from established genetic associations and provide insight on the proposed molecular pathways that arise from these associations.
Collapse
Affiliation(s)
- Darren D O'Rielly
- Faculty of Medicine, Memorial University, Craig L Dobbin Genetics Research Centre, Suite 3M500, 300 Prince Philip Drive, St. John's, NL, A1B3V6, Canada
| | - Proton Rahman
- St. Clare's Mercy Hospital, 154 LeMarchant Rd, St. John's, Newfoundland, A1C5B8, Canada.
| |
Collapse
|
86
|
Haley EK, Matmusaev M, Hossain IN, Davin S, Martin TM, Ermann J. The impact of genetic background and sex on the phenotype of IL-23 induced murine spondyloarthritis. PLoS One 2021; 16:e0247149. [PMID: 33983951 PMCID: PMC8118278 DOI: 10.1371/journal.pone.0247149] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/27/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Overexpression of IL-23 in adult mice by means of hydrodynamic tail vein injection of IL-23 minicircles has been reported to result in spondyloarthritis-like disease. The impact of genetic background and sex on the disease phenotype in this model has not been investigated. METHODS We compared male B10.RIII mice with male C57BL/6 mice, and male with female B10.RIII mice after hydrodynamic injection of IL-23 enhanced episomal vector (EEV) at 8-12 weeks of age. We monitored clinical arthritis scores, paw swelling, and body weight. Animals were euthanized after two weeks and tissues were harvested for histology, flow cytometry and gene expression analysis. Serum cytokine levels were determined by ELISA. FINDINGS Male B10.RIII mice developed arthritis in the forepaws and feet within 6 days after IL-23 EEV injection; they also exhibited psoriasis-like skin disease, colitis, weight loss, and osteopenia. In contrast to previous reports, we did not observe spondylitis or uveitis. Male C57BL/6 mice injected with IL-23 EEV had serum IL-23 levels comparable with B10.RIII mice and developed skin inflammation, colitis, weight loss, and osteopenia but failed to develop arthritis. Female B10.RIII mice had more severe arthritis than male B10.RIII mice but did not lose weight. CONCLUSIONS The phenotype of IL-23 induced disease in mice is controlled by genetic background and sex of the animals. The development of extra-articular manifestations but absence of arthritis in C57BL/6 mice suggests that organ-specificity of IL-23 driven inflammation is genetically determined. The mechanisms behind the strain-specific differences and the sexual dimorphism observed in this study may be relevant for human spondyloarthritis and warrant further exploration.
Collapse
Affiliation(s)
- Emma K. Haley
- Brigham and Women’s Hospital, Boston, MA, United States of America
| | | | | | - Sean Davin
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States of America
| | - Tammy M. Martin
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States of America
| | - Joerg Ermann
- Brigham and Women’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
87
|
Wu D, Wong P, Lam SHM, Li EK, Qin L, Tam LS, Gu J. The causal effect of interleukin-17 on the risk of psoriatic arthritis: a Mendelian randomization study. Rheumatology (Oxford) 2021; 60:1963-1973. [PMID: 33188428 DOI: 10.1093/rheumatology/keaa629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/24/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To determine causal associations between genetically predicted TNF-α, IL-12p70 and IL-17 levels and risk of PsA. METHODS The publicly available summary-level findings from genome-wide association studies (GWAS) was used to identify loci influencing normal physiological concentrations of TNF-α, IL-12p70 and IL-17 (n = 8293) among healthy individuals as exposure and a GWAS for PsA from the UK Biobank (PsA = 900, control = 462 033) as the outcome. A two-sample Mendelian randomization (MR) analysis was performed using the inverse-variance weighted (IVW), weighted median and MR-Egger regression methods. Sensitivity analysis and MR-Egger regression analysis were performed to evaluate the heterogeneity and pleiotropic effects of each variant. RESULTS Single-nucleotide polymorphisms (SNPs) at genome-wide significance from GWASs on TNF-α, IL-12p70 and IL-17 were identified as the instrumental variables. The IVW method indicated a causal association between increased IL-17 level and risk of PsA (β = -0.00186 per allele, s.e. = 0.00043, P = 0.002). Results were consistent in the weighted median method (β = -0.00145 per allele, s.e. = 0.00059, P = 0.014) although the MR-Egger method suggested a non-significant association (β = -0.00133 per allele, s.e. = 0.00087; P = 0.087). Single SNP MR results revealed that the C allele of rs117556572 was robustly associated with risk of PsA (β = 0.00210, s.e. = 0.00069, P = 0.002). However, no evidence for a causal effect was observed between TNF-α, IL-12p70, decreased IL-17 levels and risk of PsA. CONCLUSION Our findings provide preliminary evidence that genetic variants predisposing to higher physiological IL-17 level are associated with decreased risk of PsA.
Collapse
Affiliation(s)
- Dongze Wu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Priscilla Wong
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Steven H M Lam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Edmund K Li
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling Qin
- Bone Quality and Health Centre of the Department of Orthopedics & Traumatology, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
88
|
Carvalho AL, Hedrich CM. The Molecular Pathophysiology of Psoriatic Arthritis-The Complex Interplay Between Genetic Predisposition, Epigenetics Factors, and the Microbiome. Front Mol Biosci 2021; 8:662047. [PMID: 33869291 PMCID: PMC8047476 DOI: 10.3389/fmolb.2021.662047] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Psoriasis is a symmetric autoimmune/inflammatory disease that primarily affects the skin. In a significant proportion of cases, it is accompanied by arthritis that can affect any joint, the spine, and/or include enthesitis. Psoriasis and psoriatic arthritis are multifactor disorders characterized by aberrant immune responses in genetically susceptible individuals in the presence of additional (environmental) factors, including changes in microbiota and/or epigenetic marks. Epigenetic changes can be heritable or acquired (e.g., through changes in diet/microbiota or as a response to therapeutics) and, together with genetic factors, contribute to disease expression. In psoriasis, epigenetic alterations are mainly related to cell proliferation, cytokine signaling and microbial tolerance. Understanding the complex interplay between heritable and acquired pathomechanistic factors contributing to the development and maintenance of psoriasis is crucial for the identification and validation of diagnostic and predictive biomarkers, and the introduction of individualized effective and tolerable new treatments. This review summarizes the current understanding of immune activation, genetic, and environmental factors that contribute to the pathogenesis of psoriatic arthritis. Particular focus is on the interactions between these factors to propose a multifactorial disease model.
Collapse
Affiliation(s)
- Ana L Carvalho
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, United Kingdom
| |
Collapse
|
89
|
Insights into the pathogenesis of psoriatic arthritis from genetic studies. Semin Immunopathol 2021; 43:221-234. [PMID: 33712923 DOI: 10.1007/s00281-021-00843-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022]
Abstract
Psoriatic arthritis (PsA) is a relatively common inflammatory arthritis, a spondyloarthritis (SpA), that occurs most often in patients with psoriasis, a common immune-mediated inflammatory skin disease. Both psoriasis and PsA are highly heritable. Genetic and recent genomic studies have identified variants associated with psoriasis and PsA, but variants differentiating psoriasis from PsA are few. In this review, we describe recent developments in understanding the genetic burden of PsA, linkage, association and epigenetic studies. Using pathway analysis, we provide further insights into the similarities and differences between PsA and psoriasis, as well as between PsA and other immune-mediated inflammatory diseases, particularly ankylosing spondylitis, another SpA. Environmental factors that may trigger PsA in patients with psoriasis are also reviewed. To further understand the pathogenetic differences between PsA and psoriasis as well as other SpA, larger cohort studies of well-phenotyped subjects with integrated analysis of genomic, epigenomic, transcriptomic, proteomic and metabolomic data using interomic system biology approaches are required.
Collapse
|
90
|
Feng GJ, Wei XT, Zhang H, Yang XL, Shen H, Tian Q, Deng HW, Zhang L, Pei YF. Identification of pleiotropic loci underlying hip bone mineral density and trunk lean mass. J Hum Genet 2021; 66:251-260. [PMID: 32929176 PMCID: PMC7880826 DOI: 10.1038/s10038-020-00835-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 08/15/2020] [Accepted: 08/24/2020] [Indexed: 11/09/2022]
Abstract
Bone mineral density (BMD) and lean body mass (LBM) not only have a considerable heritability each, but also are genetically correlated. However, common genetic determinants shared by both traits are largely unknown. In the present study, we performed a bivariate genome-wide association study (GWAS) meta-analysis of hip BMD and trunk lean mass (TLM) in 11,335 subjects from 6 samples, and performed replication in estimated heel BMD and TLM in 215,234 UK Biobank (UKB) participants. We identified 2 loci that nearly attained the genome-wide significance (GWS, p < 5.0 × 10-8) level in the discovery GWAS meta-analysis and that were successfully replicated in the UKB sample: 11p15.2 (lead SNP rs12800228, discovery p = 2.88 × 10-7, replication p = 1.95 × 10-4) and 18q21.32 (rs489693, discovery p = 1.67 × 10-7, replication p = 1.17 × 10-3). The above 2 pleiotropic loci may play a pleiotropic role for hip BMD and TLM development. So our findings provide useful insights that further enhance our understanding of genetic interplay between BMD and LBM.
Collapse
Affiliation(s)
- Gui-Juan Feng
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Xin-Tong Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Hong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Xiao-Lin Yang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China
| | - Hui Shen
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Qing Tian
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Hong-Wen Deng
- Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA.
| | - Lei Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China.
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China.
| | - Yu-Fang Pei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, People's Republic of China.
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Jiangsu, People's Republic of China.
| |
Collapse
|
91
|
Dybska E, Adams AT, Duclaux-Loras R, Walkowiak J, Nowak JK. Waiting in the wings: RUNX3 reveals hidden depths of immune regulation with potential implications for inflammatory bowel disease. Scand J Immunol 2021; 93:e13025. [PMID: 33528856 DOI: 10.1111/sji.13025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/11/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Complex interactions between the environment and the mucosal immune system underlie inflammatory bowel disease (IBD). The involved cytokine signalling pathways are modulated by a number of transcription factors, one of which is runt-related transcription factor 3 (RUNX3). OBJECTIVE To systematically review the immune roles of RUNX3 in immune regulation, with a focus on the context of IBD. METHODS Relevant articles and reviews were identified through a Scopus search in April 2020. Information was categorized by immune cell types, analysed and synthesized. IBD transcriptome data sets and FANTOM5 regulatory networks were processed in order to complement the literature review. RESULTS The available evidence on the immune roles of RUNX3 allowed for its description in twelve cell types: intraepithelial lymphocyte, Th1, Th2, Th17, Treg, double-positive T, cytotoxic T, B, dendritic, innate lymphoid, natural killer and macrophages. In the gut, the activity of RUNX3 is multifaceted and context-dependent: it may promote homeostasis or exacerbated reactions via cytokine signalling and regulation of receptor expression. RUNX3 is mostly engaged in pathways involving ThPOK, T-bet, IFN-γ, TGF-β/IL-2Rβ, GATA/CBF-β, SMAD/p300 and a number of miRNAs. RUNX3 targets relevant to IBD may include RAG1, OSM and IL-17B. Moreover, in IBD RUNX3 expression correlates positively with GZMM, and negatively with IFNAR1, whereas in controls, it strongly associates with TGFBR3. CONCLUSIONS Dysregulation of RUNX3, mostly in the form of deficiency, likely contributes to IBD pathogenesis. More clinical research is needed to examine RUNX3 in IBD.
Collapse
Affiliation(s)
- Emilia Dybska
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Alex T Adams
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Experimental Medicine Division, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Rémi Duclaux-Loras
- INSERM U1111, Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1, Lyon, France
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Jan K Nowak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
92
|
Shi C, Rattray M, Barton A, Bowes J, Orozco G. Using functional genomics to advance the understanding of psoriatic arthritis. Rheumatology (Oxford) 2021; 59:3137-3146. [PMID: 32778885 PMCID: PMC7590405 DOI: 10.1093/rheumatology/keaa283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/17/2020] [Accepted: 04/28/2020] [Indexed: 01/03/2023] Open
Abstract
Psoriatic arthritis (PsA) is a complex disease where susceptibility is determined by genetic and environmental risk factors. Clinically, PsA involves inflammation of the joints and the skin, and, if left untreated, results in irreversible joint damage. There is currently no cure and the few treatments available to alleviate symptoms do not work in all patients. Over the past decade, genome-wide association studies (GWAS) have uncovered a large number of disease-associated loci but translating these findings into functional mechanisms and novel targets for therapeutic use is not straightforward. Most variants have been predicted to affect primarily long-range regulatory regions such as enhancers. There is now compelling evidence to support the use of chromatin conformation analysis methods to discover novel genes that can be affected by disease-associated variants. Here, we will review the studies published in the field that have given us a novel understanding of gene regulation in the context of functional genomics and how this relates to the study of PsA and its underlying disease mechanism.
Collapse
Affiliation(s)
- Chenfu Shi
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, Centre for Genetics and Genomics Versus Arthritis
| | - Magnus Rattray
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre
| | - Anne Barton
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, Centre for Genetics and Genomics Versus Arthritis.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre.,Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - John Bowes
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, Centre for Genetics and Genomics Versus Arthritis.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre
| | - Gisela Orozco
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, Centre for Genetics and Genomics Versus Arthritis.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre.,Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
93
|
Leijten E, Tao W, Pouw J, van Kempen T, Olde Nordkamp M, Balak D, Tekstra J, Muñoz-Elías E, DePrimo S, Drylewicz J, Pandit A, Boes M, Radstake T. Broad proteomic screen reveals shared serum proteomic signature in patients with psoriatic arthritis and psoriasis without arthritis. Rheumatology (Oxford) 2021; 60:751-761. [PMID: 32793974 PMCID: PMC7850582 DOI: 10.1093/rheumatology/keaa405] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To identify novel serum proteins involved in the pathogenesis of PsA as compared with healthy controls, psoriasis (Pso) and AS, and to explore which proteins best correlated to major clinical features of the disease. METHODS A high-throughput serum biomarker platform (Olink) was used to assess the level of 951 unique proteins in serum of patients with PsA (n = 20), Pso (n = 18) and AS (n = 19), as well as healthy controls (HC, n = 20). Pso and PsA were matched for Psoriasis Area and Severity Index (PASI) and other clinical parameters. RESULTS We found 68 differentially expressed proteins (DEPs) in PsA as compared with HC. Of those DEPs, 48 proteins (71%) were also dysregulated in Pso and/or AS. Strikingly, there were no DEPs when comparing PsA with Pso directly. On the contrary, hierarchical cluster analysis and multidimensional scaling revealed that HC clustered distinctly from all patients, and that PsA and Pso grouped together. The number of swollen joints had the strongest positive correlation to ICAM-1 (r = 0.81, P < 0.001) and CCL18 (0.76, P < 0.001). PASI score was best correlated to PI3 (r = 0.54, P < 0.001) and IL-17 receptor A (r = -0.51, P < 0.01). There were more proteins correlated to PASI score when analysing Pso and PsA patients separately, as compared with analysing Pso and PsA patients pooled together. CONCLUSION PsA and Pso patients share a serum proteomic signature, which supports the concept of a single psoriatic spectrum of disease. Future studies should target skin and synovial tissues to uncover differences in local factors driving arthritis development in Pso.
Collapse
Affiliation(s)
- Emmerik Leijten
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands.,Center for Translational Immunology, Utrecht, The Netherlands
| | - Weiyang Tao
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands.,Center for Translational Immunology, Utrecht, The Netherlands
| | - Juliette Pouw
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands.,Center for Translational Immunology, Utrecht, The Netherlands
| | - Tessa van Kempen
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands.,Center for Translational Immunology, Utrecht, The Netherlands
| | - Michel Olde Nordkamp
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands.,Center for Translational Immunology, Utrecht, The Netherlands
| | - Deepak Balak
- Department of Dermatology, UMC Utrecht, Utrecht, The Netherlands
| | - J Tekstra
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands
| | - Ernesto Muñoz-Elías
- Immunology Biomarkers, Janssen Research & Development LLC, San Diego, CA, USA
| | - Samuel DePrimo
- Immunology Biomarkers, Janssen Research & Development LLC, San Diego, CA, USA
| | - Julia Drylewicz
- Center for Translational Immunology, Utrecht, The Netherlands
| | - Aridaman Pandit
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands.,Center for Translational Immunology, Utrecht, The Netherlands
| | - Marianne Boes
- Center for Translational Immunology, Utrecht, The Netherlands.,Department of Pediatrics, UMC Utrecht, Utrecht, The Netherlands
| | - Timothy Radstake
- Department of Rheumatology and Clinical Immunology, Utrecht, The Netherlands.,Center for Translational Immunology, Utrecht, The Netherlands
| |
Collapse
|
94
|
Brown MA, Xu H, Li Z. Genetics and the axial spondyloarthritis spectrum. Rheumatology (Oxford) 2021; 59:iv58-iv66. [PMID: 33053195 PMCID: PMC7566537 DOI: 10.1093/rheumatology/keaa464] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
The axial SpAs (axSpAs) are clearly clinically a heterogeneous set of diseases with markedly varying extra-articular features. These diseases are all highly heritable and have overlapping but differing genetic origins. Shared features include association with HLA class I alleles and genes of the IL-23 pathway, among other things. Significant differences do exist however, both in the genetic loci involved and at specific loci in the individual genetic variants associated with each disease. These similarities and differences are of great interest in regards to disease pathogenesis and treatment development, although individually they are too small in effect to be of prognostic or diagnostic value. Polygenic risk scores, which capture a high proportion of the genetic variation between disorders, have been shown to have clinically useful discriminatory capacity in axSpA. This suggests they have the potential to enable improved disease classification, incorporating basic pathogenic features such as genomics, and ultimately benefitting clinical care. The aim of this article is to review the genetic characteristics of the spectrum of axSpAs and to discuss how this influences our understanding of the disease pathogenesis and the clinical implications of this understanding.
Collapse
Affiliation(s)
- Matthew A Brown
- Guy's & St Thomas' NHS Foundation Trust and King's College London NIHR Biomedical Research Centre, London, UK
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Second Military Medical University, Shanghai, China.,Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China.,Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Zhixiu Li
- Translational Genomics Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
95
|
Brown MA, Aletaha D. Genetic risk scores in inflammatory arthritis: a new era? Nat Rev Rheumatol 2021; 16:545-546. [PMID: 32690928 DOI: 10.1038/s41584-020-0473-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matthew A Brown
- Guy's & St Thomas' NHS Foundation Trust and King's College London NIHR Biomedical Research Centre, London, England.
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
96
|
Vecellio M, Hake VX, Davidson C, Carena MC, Wordsworth BP, Selmi C. The IL-17/IL-23 Axis and Its Genetic Contribution to Psoriatic Arthritis. Front Immunol 2021; 11:596086. [PMID: 33574815 PMCID: PMC7871349 DOI: 10.3389/fimmu.2020.596086] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease belonging to the family of spondyloarthropathies (SpA). PsA commonly aggravates psoriasis of the skin and frequently manifests as an oligoarthritis with axial skeletal involvement and extraarticular manifestations including dactylitis, enthesitis, and uveitis. The weight of genetic predisposition to psoriasis and PsA is illustrated by the concordance rates in monozygotic twins which clearly demonstrate that genomics is insufficient to induce the clinical phenotype. The association of PsA with several single nucleotide polymorphisms (SNPs) at the IL23R locus and the involvement of Th17 cells in the immunopathogenesis of PsA clearly put the IL-23/IL-17 axis in the spotlight. The IL-23 and IL-17 cytokines have a pivotal role in the chronic inflammation of the synovium in PsA and are also prominent in the skin lesions of those with PsA. In this review, we focus on the genetic association of the IL-23/IL-17 axis with PsA and the contribution of these master cytokines in the pathophysiology of the disease, highlighting the main cell types incriminated in PsA and their specific role in the peripheral blood, lesional skin and joints of patients. We then provide an overview of the approved biologic drugs targeting the IL-23/IL-17 axis and discuss the advantages of genetic stratification to enhance personalized therapies in PsA.
Collapse
Affiliation(s)
- Matteo Vecellio
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Vivien Xanath Hake
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Connor Davidson
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | | | - B Paul Wordsworth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| |
Collapse
|
97
|
Pisani C, Onori A, Gabanella F, Di Certo MG, Passananti C, Corbi N. Identification of protein/mRNA network involving the PSORS1 locus gene CCHCR1 and the PSORS4 locus gene HAX1. Exp Cell Res 2021; 399:112471. [PMID: 33417922 DOI: 10.1016/j.yexcr.2021.112471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 01/22/2023]
Abstract
CCHCR1 (Coiled-Coil alpha-Helical Rod 1), maps to chromosomal region 6p21.3, within the major psoriasis susceptibility locus PSORS1. CCHCR1 itself is a plausible psoriasis candidate gene, however its role in psoriasis pathogenesis remains unclear. We previously demonstrated that CCHCR1 protein acts as a cytoplasmic docking site for RNA polymerase II core subunit 3 (RPB3) in cycling cells, suggesting a role for CCHCR1 in vesicular trafficking between cellular compartments. Here, we report a novel interaction between CCHCR1 and the RNA binding protein HAX1. HAX1 maps to chromosomal region 1q21.3 within the PSORS4 locus and is over-expressed in psoriasis. Both CCHCR1 and HAX1 share subcellular co-localization with mitochondria, nuclei and cytoplasmic vesicles as P-bodies. By a series of ribonucleoprotein immunoprecipitation (RIP) assays, we isolated a pool of mRNAs complexed with HAX1 and/or CCHCR1 proteins. Among the mRNAs complexed with both CCHCR1 and HAX1 proteins, there are Vimentin mRNA, previously described to be bound by HAX1, and CAMP/LL37 mRNA, whose gene product is over-expressed in psoriasis.
Collapse
Affiliation(s)
- Cinzia Pisani
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Italy.
| | - Annalisa Onori
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Italy.
| | - Francesca Gabanella
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Italy; CNR-Institute of Biochemistry and Cell Biology, Department of Sense Organs, Sapienza University of Rome, Italy.
| | - Maria Grazia Di Certo
- CNR-Institute of Biochemistry and Cell Biology, Department of Sense Organs, Sapienza University of Rome, Italy.
| | - Claudio Passananti
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Italy.
| | - Nicoletta Corbi
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University of Rome, Italy.
| |
Collapse
|
98
|
Verbenko DA, Karamova AE, Artamonova OG, Deryabin DG, Rakitko A, Chernitsov A, Krasnenko A, Elmuratov A, Solomka VS, Kubanov AA. Apremilast Pharmacogenomics in Russian Patients with Moderate-to-Severe and Severe Psoriasis. J Pers Med 2020; 11:jpm11010020. [PMID: 33383665 PMCID: PMC7823747 DOI: 10.3390/jpm11010020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/25/2020] [Indexed: 12/14/2022] Open
Abstract
One of the target drugs for plaque psoriasis treatment is apremilast, which is a selective phosphodiesterase 4 (PDE4) inhibitor. In this study, 34 moderate-to-severe and severe plaque psoriasis patients from Russia were treated with apremilast for 26 weeks. This allowed us to observe the effectiveness of splitting patient cohorts based on clinical outcomes, which were assessed using the Psoriasis Area Severity Index (PASI). In total, 14 patients (41%) indicated having an advanced outcome with delta PASI 75 after treatment; 20 patients indicated having moderate or no effects. Genome variability was investigated using the Illumina Infinium Global Screening Array. Genome-wide analysis revealed apremilast therapy clinical outcome associations at three compact genome regions with undefined functions situated on chromosomes 2, 4, and 5, as well as on a single single-nucleotide polymorphism (SNP) on chromosome 23. Pre-selected SNP sets were associated with psoriasis vulgaris analysis, which was used to identify four SNP-associated targeted therapy efficiencies: IL1β (rs1143633), IL4 (IL13) (rs20541), IL23R (rs2201841), and TNFα (rs1800629) genes. Moreover, we showed that the use of the global polygenic risk score allowed for the prediction of onset psoriasis in Russians. Therefore, these results can serve as a starting point for creating a predictive model of apremilast therapy response in the targeted therapy of patients with psoriasis vulgaris.
Collapse
Affiliation(s)
- Dmitry A. Verbenko
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
- Correspondence:
| | - Arfenya E. Karamova
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Olga G. Artamonova
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Dmitry G. Deryabin
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Alexander Rakitko
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Alexandr Chernitsov
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Anna Krasnenko
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Artem Elmuratov
- Genotek Ltd., Nastavnicheskiipereulok 17/1, 105120 Moscow, Russia; (A.R.); (A.C.); (A.K.); (A.E.)
| | - Victoria S. Solomka
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| | - Alexey A. Kubanov
- State Research Center of Dermatovenereology and Cosmetology, Korolenko St., 3, bldg 6, 107076 Moscow, Russia; (A.E.K.); (O.G.A.); (D.G.D.); (V.S.S.); (A.A.K.)
| |
Collapse
|
99
|
Bone Mineral Density, Osteoporosis, and Fracture Risk in Adult Patients with Psoriasis or Psoriatic Arthritis: A Systematic Review and Meta-Analysis of Observational Studies. J Clin Med 2020; 9:jcm9113712. [PMID: 33227975 PMCID: PMC7699147 DOI: 10.3390/jcm9113712] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Awareness of psoriasis-related comorbidities has been established in the current guidelines; however, evidence regarding the association of bone density or bone fragility with psoriatic disease remains inconclusive. METHODS We conducted a systematic review and meta-analysis to assess bone mineral density and the risk of osteoporosis and fractures in patients with psoriatic disease, including those with cutaneous psoriasis and psoriatic arthritis. We searched electronic databases for published observational studies. A meta-analysis was performed using the random-effect model. Pooled estimates and their confidence intervals (CIs) were calculated. Small-study effects were examined using the Doi plot and Luis Furuya-Kanamori index. RESULTS The analysis of the standardized mean difference in the absolute value of bone mineral density at different measuring sites (lumbar spine, femoral neck, and total hip) revealed no significant difference between patients with psoriatic disease and non-psoriatic controls. The pooled results of the adjusted odds ratios (ORs) demonstrated no increased risk of osteoporosis in patients with psoriatic disease. Notably, patients with psoriatic disease had a higher OR of developing bone fractures (adjusted OR: 1.09; 95% CI: 1.06 to 1.12; I2: 0%). CONCLUSION Patients with psoriatic disease may be more likely to develop fractures compared with non-psoriatic controls. This higher risk for fracture may not necessarily be associated with lower bone mineral density nor a higher risk for osteoporosis.
Collapse
|
100
|
Razani B, Malynn BA, Ma A. Preserving immune homeostasis with A20. Adv Immunol 2020; 148:1-48. [PMID: 33190732 DOI: 10.1016/bs.ai.2020.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
A20/TNFAIP3 is a TNF induced gene that plays a profound role in preserving cellular and organismal homeostasis (Lee, et al., 2000; Opipari etal., 1990). This protein has been linked to multiple human diseases via genetic, epigenetic, and an emerging series of patients with mono-allelic coding mutations. Diverse cellular functions of this pleiotropically expressed protein include immune-suppressive, anti-inflammatory, and cell protective functions. The A20 protein regulates ubiquitin dependent cell signals; however, the biochemical mechanisms by which it performs these functions is surprisingly complex. Deciphering these cellular and biochemical facets of A20 dependent biology should greatly improve our understanding of murine and human disease pathophysiology as well as unveil new mechanisms of cell and tissue biology.
Collapse
Affiliation(s)
- Bahram Razani
- Department of Dermatology, University of California, San Francisco, CA, United States
| | - Barbara A Malynn
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|