1
|
Moysidou E, Christodoulou M, Lioulios G, Stai S, Karamitsos T, Dimitroulas T, Fylaktou A, Stangou M. Lymphocytes Change Their Phenotype and Function in Systemic Lupus Erythematosus and Lupus Nephritis. Int J Mol Sci 2024; 25:10905. [PMID: 39456692 PMCID: PMC11508046 DOI: 10.3390/ijms252010905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease, characterized by considerable changes in peripheral lymphocyte structure and function, that plays a critical role in commencing and reviving the inflammatory and immune signaling pathways. In healthy individuals, B lymphocytes have a major role in guiding and directing defense mechanisms against pathogens. Certain changes in B lymphocyte phenotype, including alterations in surface and endosomal receptors, occur in the presence of SLE and lead to dysregulation of peripheral B lymphocyte subpopulations. Functional changes are characterized by loss of self-tolerance, intra- and extrafollicular activation, and increased cytokine and autoantibody production. T lymphocytes seem to have a supporting, rather than a leading, role in the disease pathogenesis. Substantial aberrations in peripheral T lymphocyte subsets are evident, and include a reduction of cytotoxic, regulatory, and advanced differentiated subtypes, together with an increase of activated and autoreactive forms and abnormalities in follicular T cells. Up-regulated subpopulations, such as central and effector memory T cells, produce pre-inflammatory cytokines, activate B lymphocytes, and stimulate cell signaling pathways. This review explores the pivotal roles of B and T lymphocytes in the pathogenesis of SLE and Lupus Nephritis, emphasizing the multifaceted mechanisms and interactions and their phenotypic and functional dysregulations.
Collapse
Affiliation(s)
- Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Stamatia Stai
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Theodoros Karamitsos
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Cardiology, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theodoros Dimitroulas
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 4th Department of Medicine, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| |
Collapse
|
2
|
Chun KH, Park YC, Hwang N, Yoon BK, Kim JW, Fang S. Gene signature from cutaneous autoimmune diseases provides potential immunotherapy-relevant biomarkers in melanoma. Sci Rep 2023; 13:15023. [PMID: 37700026 PMCID: PMC10497583 DOI: 10.1038/s41598-023-42238-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/07/2023] [Indexed: 09/14/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are promising agents for treating melanoma. Given that autoimmune skin diseases exhibit hyper immune reaction, investigation of immune cells from autoimmune skin disease is crucial to validate the effectiveness of ICIs in melanoma treatment. We employed multipanel markers to predict the response to immune checkpoint inhibitors by characterizing the gene expression signatures of skin immune cells in systemic lupus erythematosus (SLE), atopic dermatitis (AD), and psoriasis (PS). By analyzing single-cell RNA sequencing data from each dataset, T cell gene signatures from autoimmune skin diseases exhibit a complex immune response in tumors that responded to immunotherapy. Based on that CD86 and CD80 provide essential costimulatory signals for T cell activation, we observed that interaction of CD86 signaling has been enhanced in the T cells of patients with SLE, AD, and PS. Our analysis revealed a common increase in CD86 signals from dendritic cells (DCs) to T cells in patients with SLE, AD, and PS, confirming that dendritic cells produce pro-inflammatory cytokines to activate T cells. Thus, we hypothesize that T cell gene signatures from autoimmune skin diseases exhibit a pro-inflammatory response and have the potential to predict cancer immunotherapy. Our study demonstrated that T cell gene signatures derived from inflammatory skin diseases, particularly SLE and PS, hold promise as potential biomarkers for predicting the response to immune checkpoint blockade therapy in patients with melanoma. Our data provide an understanding of the immune-related characteristics and differential gene expression patterns in autoimmune skin diseases, which may represent promising targets for melanoma immunotherapy.
Collapse
Affiliation(s)
- Kyu-Hye Chun
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Ye-Chan Park
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Nahee Hwang
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Bo Kyung Yoon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jae-Woo Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, 03722, Korea.
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, 03722, Korea.
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
3
|
Sestan M, Kifer N, Arsov T, Cook M, Ellyard J, Vinuesa CG, Jelusic M. The Role of Genetic Risk Factors in Pathogenesis of Childhood-Onset Systemic Lupus Erythematosus. Curr Issues Mol Biol 2023; 45:5981-6002. [PMID: 37504294 PMCID: PMC10378459 DOI: 10.3390/cimb45070378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023] Open
Abstract
The pathogenesis of childhood-onset systemic lupus erythematosus (cSLE) is complex and not fully understood. It involves three key factors: genetic risk factors, epigenetic mechanisms, and environmental triggers. Genetic factors play a significant role in the development of the disease, particularly in younger individuals. While cSLE has traditionally been considered a polygenic disease, it is now recognized that in rare cases, a single gene mutation can lead to the disease. Although these cases are uncommon, they provide valuable insights into the disease mechanism, enhance our understanding of pathogenesis and immune tolerance, and facilitate the development of targeted treatment strategies. This review aims to provide a comprehensive overview of both monogenic and polygenic SLE, emphasizing the implications of specific genes in disease pathogenesis. By conducting a thorough analysis of the genetic factors involved in SLE, we can improve our understanding of the underlying mechanisms of the disease. Furthermore, this knowledge may contribute to the identification of effective biomarkers and the selection of appropriate therapies for individuals with SLE.
Collapse
Affiliation(s)
- Mario Sestan
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Nastasia Kifer
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Todor Arsov
- Faculty of Medical Sciences, University Goce Delchev, 2000 Shtip, North Macedonia
- The Francis Crick Institute, London NW1 1AT, UK
| | - Matthew Cook
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
- Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
| | - Julia Ellyard
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
| | | | - Marija Jelusic
- Department of Paediatrics, University of Zagreb School of Medicine, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
4
|
A p38α-BLIMP1 signalling pathway is essential for plasma cell differentiation. Nat Commun 2022; 13:7321. [PMID: 36443297 PMCID: PMC9703440 DOI: 10.1038/s41467-022-34969-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Plasma cells (PC) are antibody-secreting cells and terminal effectors in humoral responses. PCs differentiate directly from activated B cells in response to T cell-independent (TI) antigens or from germinal center B (GCB) cells in T cell-dependent (TD) antigen-induced humoral responses, both of which pathways are essentially regulated by the transcription factor BLIMP1. The p38 mitogen-activated protein kinase isoforms have already been implicated in B cell development, but the precise role of p38α in B cell differentiation is still largely unknown. Here we show that PC differentiation and antibody responses are severely impaired in mice with B cell-specific deletion of p38α, while B cell development and the GCB cell response are spared. By utilizing a Blimp1 reporter mouse model, we show that p38α-deficiency results in decreased BLIMP1 expression. p38α-driven BLIMP1 up-regulation is required for both TI and TD PCs differentiation. By combining CRISPR/Cas9 screening and other approaches, we identify TCF3, TCF4 and IRF4 as downstream effectors of p38α to control PC differentiation via Blimp1 transcription. This study thus identifies an important signalling pathway underpinning PC differentiation upstream of BLIMP1, and points to a highly specialized and non-redundant role for p38α among p38 isoforms.
Collapse
|
5
|
Xu N, Liu J, Li X. Lupus nephritis: The regulatory interplay between epigenetic and MicroRNAs. Front Physiol 2022; 13:925416. [PMID: 36187762 PMCID: PMC9523357 DOI: 10.3389/fphys.2022.925416] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous, small, non-coding RNA molecules that act as epigenetic modifiers to regulate the protein levels of target messenger RNAs without altering their genetic sequences. The highly complex role of miRNAs in the epigenetics of lupus nephritis (LN) is increasingly being recognized. DNA methylation and histone modifications are focal points of epigenetic research. miRNAs play a critical role in renal development and physiology, and dysregulation may result in abnormal renal cell proliferation, inflammation, and fibrosis of the kidneys in LN. However, epigenetic and miRNA-mediated regulation are not mutually exclusive. Further research has established a link between miRNA expression and epigenetic regulation in various disorders, including LN. This review summarizes the most recent evidence regarding the interaction between miRNAs and epigenetics in LN and highlights potential therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- Ning Xu
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jie Liu
- School of Clinical Medicine, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiangling Li
- Department of Nephrology, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Xiangling Li,
| |
Collapse
|
6
|
Zhou M, Kang Y, Li J, Li R, Lu L. Omics-based integrated analysis identified IKZF2 as a biomarker associated with lupus nephritis. Sci Rep 2022; 12:9612. [PMID: 35688845 PMCID: PMC9187727 DOI: 10.1038/s41598-022-13336-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/23/2022] [Indexed: 11/09/2022] Open
Abstract
Lupus nephritis (LN) is a crucial complication of systemic lupus erythematosus (SLE). IKZF2 was identified as a lupus susceptibility locus, while its exact molecular function in LN is unknown. We aimed to explore the relationship between IKZF2 and LN based on multi-omics data. In our study, we carried out a meta-analysis of publicly available data, including not only tubulointerstitium, but also glomerulus tissue samples from LN patients and controls. Based on the common differentially expressed genes (co-DEGs) and previous researches, we selected IKZF2 for further analysis. Then, we analyzed potential molecular mechanisms of co-DEGs and IKZF2 in LN. To explore the possible targets of IKZF2, protein-protein interaction network (PPI) network and ceRNA network of IKZF2 were also constructed. Moreover, we performed immune infiltration analysis and evaluated clinical value of IKZF2. A total of 26 co-DEGs were observed in the integration of the above DEGs coming from the four sets of data, of which IKZF2 was selected for further analysis. Functional enrichment analysis from IKZF2 and related PPI network confirmed the tight relationship between IKZF2 and the immune reaction. Moreover, immune filtration analysis revealed the significant correlation between IKZF2 and naïve B cell, NK cell activation, NK cell rest and other immune cells. Receiver operating characteristic (ROC) analysis showed that the areas under the ROC curves were 0.721, 0.80, 0.682, and 0.859 for IKZF2 in four datasets, which demonstrated the clinical value of IKZF2. Our study revealed that IKZF2 may play an essential role in the molecular function and development of LN, and might be a potential biomarker for distinguishing LN patients and healthy ones.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, 200001, China
| | - Yuening Kang
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, 200001, China
| | - Jun Li
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, 200001, China
| | - Rongxiu Li
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, 200240, China.
| | - Liangjing Lu
- Department of Rheumatology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, 200001, China.
| |
Collapse
|
7
|
Papageorgiou L, Alkenaris H, Zervou MI, Vlachakis D, Matalliotakis I, Spandidos DA, Bertsias G, Goulielmos GN, Eliopoulos E. Epione application: An integrated web‑toolkit of clinical genomics and personalized medicine in systemic lupus erythematosus. Int J Mol Med 2021; 49:8. [PMID: 34791504 PMCID: PMC8612305 DOI: 10.3892/ijmm.2021.5063] [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: 08/29/2021] [Accepted: 11/02/2021] [Indexed: 12/16/2022] Open
Abstract
Genome wide association studies (GWAS) have identified autoimmune disease-associated loci, a number of which are involved in numerous disease-associated pathways. However, much of the underlying genetic and pathophysiological mechanisms remain to be elucidated. Systemic lupus erythematosus (SLE) is a chronic, highly heterogeneous auto-immune disease, characterized by differences in autoantibody profile, serum cytokines and a multi-system involvement. This study presents the Epione application, an integrated bioinformatics web-toolkit, designed to assist medical experts and researchers in more accurately diagnosing SLE. The application aims to identify the most credible gene variants and single nucleotide polymorphisms (SNPs) associated with SLE susceptibility, by using patient's genomic data to aid the medical expert in SLE diagnosis. The application contains useful knowledge of >70,000 SLE-related publications that have been analyzed, using data mining and semantic techniques, towards extracting the SLE-related genes and the corresponding SNPs. Probable genes associated with the patient's genomic profile are visualized with several graphs, including chromosome ideograms, statistic bars and regulatory networks through data mining studies with relative publications, to obtain a representative number of the most credible candidate genes and biological pathways associated with the SLE. Furthermore, an evaluation study was performed on a patient diagnosed with SLE and is presented herein. Epione has also been expanded in family-related candidate patients to evaluate its predictive power. All the recognized gene variants that were previously considered to be associated with SLE were accurately identified in the output profile of the patient, and by comparing the results, novel findings have emerged. The Epione application may assist and facilitate in early stage diagnosis by using the patients' genomic profile to compare against the list of the most predictable candidate gene variants related to SLE. Its diagnosis-oriented output presents the user with a structured set of results on variant association, position in genome and links to specific bibliography and gene network associations. The overall aim of the present study was to provide a reliable tool for the most effective study of SLE. This novel and accessible webserver tool of SLE is available at http://geneticslab.aua.gr/epione/.
Collapse
Affiliation(s)
- Louis Papageorgiou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Haris Alkenaris
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Maria I Zervou
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dimitriοs Vlachakis
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Ioannis Matalliotakis
- Department of Obstetrics and Gynecology, Venizeleio and Pananio General Hospital of Heraklion, 71409 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - George Bertsias
- Department of Rheumatology and Clinical Immunology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - George N Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Elias Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| |
Collapse
|
8
|
Yang M, Yang B, Deng D. Targeting of EIF4EBP1 by miR-99a-3p affects the functions of B lymphocytes via autophagy and aggravates SLE disease progression. J Cell Mol Med 2021; 25:10291-10305. [PMID: 34668631 PMCID: PMC8572797 DOI: 10.1111/jcmm.16991] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/22/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Excessive activation of immune cells plays a key role in the pathogenesis of systemic lupus erythematosus (SLE). The regulation of immune cells by miRNAs is a research hotspot. In this study, second-generation high-throughput sequencing revealed a reduction in miR-99a-3p expression in patients with SLE; however, the specific mechanism underlying this phenomenon remains unclear. After transfection with an miR-99a-3p agomir, the proliferation of Ball-1 cells decreased and the levels of their apoptosis increased. The opposite effects were observed in cells transfected with the miR-99a-3p antagomir. Luciferase reporter assay indicated that miR-99a-3p directly targeted EIF4EBP1. Rescue experiments confirmed the proposed interaction between miR-99a-3p and EIF4EBP1. In vitro, in vivo and clinical investigations further confirmed that the miR-99a-3p agomir reduced the expression of EIF4EBP1, LC3B and LAMP-2A. In the in vivo experiments, serum levels of anti-nuclear antibodies, double-stranded DNA, IgE, IgM, IL-6, IL-10 and B lymphocyte stimulator were higher in mice from the antagomir group than those in mice from the MRL/lpr group. Furthermore, the protein and mRNA levels of EIF4EBP1, LC3B and LAMP-2A, the intensity of immunohistochemical staining of EIF4EBP1, LC3B and LAMP-2A, the urinary protein levels, and the C3 immunofluorescence deposition increased in mice from the antagomir group. The upregulation of miR-99a-3p expression protected B cells from EIF4EBP1-mediated autophagy, whilst the downregulation of miR-99a-3p expression induced autophagy via the EIF4EBP1-mediated regulation of the autophagy signalling pathway in B cells isolated from individuals with SLE. Based on these results, miR-99a-3p and EIF4EBP1 may be considered potential targets for SLE treatment.
Collapse
Affiliation(s)
- Meng Yang
- Department of DermatologyThe Second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
- Department of DermatologyThe Third Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Binbin Yang
- Department of DermatologyThe Second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Danqi Deng
- Department of DermatologyThe Second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
9
|
You Y, Zhao X, Wu Y, Mao J, Ge L, Guo J, Zhao C, Chen D, Song Z. Integrated Transcriptome Profiling Revealed That Elevated Long Non-Coding RNA- AC007278.2 Expression Repressed CCR7 Transcription in Systemic Lupus Erythematosus. Front Immunol 2021; 12:615859. [PMID: 34220794 PMCID: PMC8242351 DOI: 10.3389/fimmu.2021.615859] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose Systemic lupus erythematosus (SLE) is a serious autoimmune disease. Its molecular pathogenesis, especially the long non-coding RNA (lncRNA) function, remains unclear. We want to investigate the lncRNA dysregulation profile and their molecular mechanisms in SLE. Methods In this study, we analyzed the transcriptome profiles (RNA-seq) of peripheral blood mononuclear cells (PBMCs) from SLE patients and two published transcriptome datasets to explore lncRNA profiles. The differentially expressed lncRNAs were confirmed by quantitative real-time PCR in another set of female patients. We constructed the lncRNA-mRNA regulatory networks by performing weighted gene co-expression network analysis (WGCNA). Dysregulated lncRNA AC007278.2 was repressed by short hairpin RNA (shRNA) in Jurkat cells. Dual-luciferase reporter gene assay was performed to investigate the regulatory mechanism of AC007278.2 on target gene CCR7. Results We observed dominant up-regulation of transcripts, including mRNAs and lncRNAs, in SLE patients. By WGCNA method, we identified three modules that were highly related to SLE. We then focused on one lncRNA, AC007278.2, with a T-helper 1 lineage-specific expression pattern. We observed consistently higher AC007278.2 expression in SLE patients. Co-expression network revealed that AC007278.2 participated in the innate immune response and inflammatory bowel disease pathways. By knocking down AC007278.2 expression, we found that AC007278.2 could regulate the expression of inflammatory and cytokine stimulus response-related genes, including CCR7, AZU1, and TNIP3. AC007278.2 inhibits the functional CCR7 promoter to repress its transcription, thereby regulating autoimmunity and follicular T-helper cell differentiation. Conclusion In summary, our study indicated the important regulatory role of lncRNAs in SLE. AC007278.2 may be treated as a novel biomarker for SLE diagnosis and treatment.
Collapse
Affiliation(s)
- Yi You
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xingwang Zhao
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yaguang Wu
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiangming Mao
- Center for Genome Analysis, ABLife Inc., Wuhan, China
| | - Lan Ge
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Junkai Guo
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chenglei Zhao
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dong Chen
- Center for Genome Analysis, ABLife Inc., Wuhan, China.,Science Department, ABLife BioBigData Institute, Wuhan, China
| | - Zhiqiang Song
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
10
|
Gene Expression as a Guide to the Development of Novel Therapies in Primary Glomerular Diseases. J Clin Med 2021; 10:jcm10112262. [PMID: 34073694 PMCID: PMC8197155 DOI: 10.3390/jcm10112262] [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: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/19/2021] [Indexed: 11/17/2022] Open
Abstract
Despite improvements in understanding the pathogenic mechanisms of primary glomerular diseases, therapy still remains nonspecific. We sought to identify novel therapies targeting kidney-intrinsic injury of distinct primary glomerulonephritides through computational systems biology approaches. We defined the unique transcriptional landscape within kidneys from patients with focal segmental glomerulosclerosis (FSGS), minimal change disease (MCD), immunoglobulin A nephropathy (IgAN), membranous nephropathy (MN) and thin basement membrane nephropathy (TBMN). Differentially expressed genes were functionally annotated with enrichment analysis, and distinct biological processes and pathways implicated in each primary glomerular disease were uncovered. Finally, we identified novel drugs and small-molecule compounds that may reverse each glomerulonephritis phenotype, suggesting they should be further tested as precise therapy in primary glomerular diseases.
Collapse
|
11
|
Rothamel K, Arcos S, Kim B, Reasoner C, Lisy S, Mukherjee N, Ascano M. ELAVL1 primarily couples mRNA stability with the 3' UTRs of interferon-stimulated genes. Cell Rep 2021; 35:109178. [PMID: 34038724 PMCID: PMC8225249 DOI: 10.1016/j.celrep.2021.109178] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/13/2021] [Accepted: 05/04/2021] [Indexed: 12/17/2022] Open
Abstract
Upon pathogen detection, the innate immune system triggers signaling events leading to upregulation of pro-inflammatory and anti-microbial mRNA transcripts. RNA-binding proteins (RBPs) interact with these critical mRNAs and regulate their fates at the post-transcriptional level. One such RBP is ELAVL1. Although significant progress has been made in understanding how embryonic lethal vision-like protein 1 (ELAVL1) regulates mRNAs, its target repertoire and binding distribution within an immunological context remain poorly understood. We overlap four high-throughput approaches to define its context-dependent targets and determine its regulatory impact during immune activation. ELAVL1 transitions from binding overwhelmingly intronic sites to 3′ UTR sites upon immune stimulation of cells, binding previously and newly expressed mRNAs. We find that ELAVL1 mediates the RNA stability of genes that regulate pathways essential to pathogen sensing and cytokine production. Our findings reveal the importance of examining RBP regulatory impact under dynamic transcriptomic events to understand their post-transcriptional regulatory roles within specific biological circuitries. Rothamel et al. show that upon immune activation, the RNA-binding protein ELAVL1 accumulates in the cytoplasm and redistributes from introns to mRNA 3′ UTRs. 3′ UTR binding confers enrichment and transcript stability. Many top-ranking transcripts are interferon-stimulated genes (ISGs), indicating that ELAVL1 is a positive regulator of an innate immune response.
Collapse
Affiliation(s)
- Katherine Rothamel
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sarah Arcos
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Byungil Kim
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Clara Reasoner
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Samantha Lisy
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Neelanjan Mukherjee
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Manuel Ascano
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
12
|
Wu L, Jiang X, Qi C, Zhang C, Qu B, Shen N. EZH2 Inhibition Interferes With the Activation of Type I Interferon Signaling Pathway and Ameliorates Lupus Nephritis in NZB/NZW F1 Mice. Front Immunol 2021; 12:653989. [PMID: 33868295 PMCID: PMC8044841 DOI: 10.3389/fimmu.2021.653989] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/08/2021] [Indexed: 12/29/2022] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a histone-lysine N-methyltransferase mediating trimethylation of H3K27, which represses gene expression and is critical to immune regulation. Inhibition of EZH2 is proved to have the potential of treating many diseases. However, whether inhibition of EZH2 affects type I interferon (IFN-I) signaling pathway, the abnormality of which is an important pathogenic mechanism for SLE, is still elusive. Here, we report, unexpectedly, a positive regulatory function of EZH2 in IFN-I signaling pathway, which contributes to the overactivation of IFN-I signaling pathway in SLE. We show that the expression of EZH2 was upregulated and positively correlated with the overexpression of interferon stimulated genes (ISGs) in both peripheral blood mononuclear cells and renal tissues of SLE patients. In vitro inhibition of EZH2 by either siRNAs or chemical inhibitors reduced the phosphorylation of STAT1 and the induction of ISGs stimulated by IFN-I. Additionally, inhibition of EZH2 interfered with the in vivo and ex vivo activation of IFN-I signaling pathway elicited by intravenous injection of adenovirus vector expressing mouse IFN-α5 and exogeneous stimulation with IFN-α, respectively. We evaluated the therapeutic effects of EZH2 inhibitor in NZB/NZW F1 mice which depend on IFN-I signaling pathway for the lupus-like disease development. Administration of EZH2 inhibitor prolonged the survival, reduced the levels of anti-dsDNA autoantibodies, and improved lupus nephritis of the mice. What’s more, EZH2 inhibitor attenuated the expression of ISGs in the kidneys of these mice. In summary, we show that excessive EZH2 contributes to the overactivation of IFN-I signaling pathway in SLE. EZH2 inhibitor has the potential to inhibit IFN-I signaling pathway and alleviate lupus nephritis. Additionally, diverse disease driving pathways exist among systemic lupus erythematosus (SLE) patient, and even in the same patients. Common regulators of different pathogenic pathways can be multivalent therapeutic targets. Together with previous studies showing EZH2 is involved in T-cell and B-cell mediated immune responses, EZH2 could be a potent multivalent therapeutic target for SLE.
Collapse
Affiliation(s)
- Lingling Wu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyue Jiang
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaojun Qi
- Department of Nephrology, Molecular Cell Lab for Kidney Disease, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunyan Zhang
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Qu
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Shen
- Department of Rheumatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China.,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
13
|
Zhou S, Zhang D, Guo J, Chen Z, Chen Y, Zhang J. Long non‐coding
RNA
NORAD functions as a
microRNA‐204‐5p
sponge to repress the progression of Parkinson's disease in vitro by increasing the solute carrier family 5 member 3 expression. IUBMB Life 2020; 72:2045-2055. [PMID: 32687247 DOI: 10.1002/iub.2344] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Shufang Zhou
- Department of NeurologyHuaihe Hospital of Henan University Kaifeng China
| | - Dan Zhang
- Department of DentistryThe First Affiliated Hospital of Zhengzhou University Zhengzhou China
| | - Junnan Guo
- Department of NeurologyHuaihe Hospital of Henan University Kaifeng China
| | - Zhenzhen Chen
- Department of Rehabilitation MedicineHuaihe Hospital of Henan University Kaifeng China
| | - Yong Chen
- Department of NeurologyHuaihe Hospital of Henan University Kaifeng China
| | - Junshi Zhang
- Department of NeurologyHuaihe Hospital of Henan University Kaifeng China
| |
Collapse
|
14
|
Update on the cellular and molecular aspects of lupus nephritis. Clin Immunol 2020; 216:108445. [PMID: 32344016 DOI: 10.1016/j.clim.2020.108445] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/26/2020] [Accepted: 04/23/2020] [Indexed: 12/17/2022]
Abstract
Recent progress has highlighted the involvement of a variety of innate and adaptive immune cells in lupus nephritis. These include activated neutrophils producing extracellular chromatin traps that induce type I interferon production and endothelial injury, metabolically-rewired IL-17-producing T-cells causing tissue inflammation, follicular and extra-follicular helper T-cells promoting the maturation of autoantibody-producing B-cells that may also sustain the formation of germinal centers, and alternatively activated monocytes/macrophages participating in tissue repair and remodeling. The role of resident cells such as podocytes and tubular epithelial cells is increasingly recognized in regulating the local immune responses and determining the kidney function and integrity. These findings are corroborated by advanced, high-throughput genomic studies, which have revealed an unprecedented amount of data highlighting the molecular heterogeneity of immune and non-immune cells implicated in lupus kidney disease. Importantly, this research has led to the discovery of putative pathogenic pathways, enabling the rationale design of novel treatments.
Collapse
|
15
|
Barral-Arca R, Gómez-Carballa A, Cebey-López M, Bello X, Martinón-Torres F, Salas A. A Meta-Analysis of Multiple Whole Blood Gene Expression Data Unveils a Diagnostic Host-Response Transcript Signature for Respiratory Syncytial Virus. Int J Mol Sci 2020; 21:E1831. [PMID: 32155831 PMCID: PMC7084441 DOI: 10.3390/ijms21051831] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/30/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the major causes of acute lower respiratory tract infection worldwide. The absence of a commercial vaccine and the limited success of current therapeutic strategies against RSV make further research necessary. We used a multi-cohort analysis approach to investigate host transcriptomic biomarkers and shed further light on the molecular mechanism underlying RSV-host interactions. We meta-analyzed seven transcriptome microarray studies from the public Gene Expression Omnibus (GEO) repository containing a total of 922 samples, including RSV, healthy controls, coronaviruses, enteroviruses, influenzas, rhinoviruses, and coinfections, from both adult and pediatric patients. We identified > 1500 genes differentially expressed when comparing the transcriptomes of RSV-infected patients against healthy controls. Functional enrichment analysis showed several pathways significantly altered, including immunologic response mediated by RSV infection, pattern recognition receptors, cell cycle, and olfactory signaling. In addition, we identified a minimal 17-transcript host signature specific for RSV infection by comparing transcriptomic profiles against other respiratory viruses. These multi-genic signatures might help to investigate future drug targets against RSV infection.
Collapse
Affiliation(s)
- Ruth Barral-Arca
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigaciones Sanitarias (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), 15706 Galicia, Spain; (R.B.-A.); (A.G.-C.); (M.C.-L.); (X.B.)
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, 15706 Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Alberto Gómez-Carballa
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigaciones Sanitarias (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), 15706 Galicia, Spain; (R.B.-A.); (A.G.-C.); (M.C.-L.); (X.B.)
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, 15706 Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Miriam Cebey-López
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigaciones Sanitarias (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), 15706 Galicia, Spain; (R.B.-A.); (A.G.-C.); (M.C.-L.); (X.B.)
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, 15706 Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Xabier Bello
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigaciones Sanitarias (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), 15706 Galicia, Spain; (R.B.-A.); (A.G.-C.); (M.C.-L.); (X.B.)
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, 15706 Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, 15706 Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Antonio Salas
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and GenPoB Research Group, Instituto de Investigaciones Sanitarias (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), 15706 Galicia, Spain; (R.B.-A.); (A.G.-C.); (M.C.-L.); (X.B.)
- Genetics, Vaccines and Infections Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| |
Collapse
|
16
|
Wang TY, Wang YF, Zhang Y, Shen JJ, Guo M, Yang J, Lau YL, Yang W. Identification of Regulatory Modules That Stratify Lupus Disease Mechanism through Integrating Multi-Omics Data. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:318-329. [PMID: 31877408 PMCID: PMC6938958 DOI: 10.1016/j.omtn.2019.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 11/05/2022]
Abstract
Although recent advances in genetic studies have shed light on systemic lupus erythematosus (SLE), its detailed mechanisms remain elusive. In this study, using datasets on SLE transcriptomic profiles, we identified 750 differentially expressed genes (DEGs) in T and B lymphocytes and peripheral blood cells. Using transcription factor (TF) binding data derived from chromatin immunoprecipitation sequencing (ChIP-seq) experiments from the Encyclopedia of DNA Elements (ENCODE) project, we inferred networks of co-regulated genes (NcRGs) based on binding profiles of the upregulated DEGs by significantly enriched TFs. Modularization analysis of NcRGs identified co-regulatory modules among the DEGs and master TFs vital for each module. Remarkably, the co-regulatory modules stratified the common SLE interferon (IFN) signature and revealed SLE pathogenesis pathways, including the complement cascade, cell cycle regulation, NETosis, and epigenetic regulation. By integrative analyses of disease-associated genes (DAGs), DEGs, and enriched TFs, as well as proteins interacting with them, we identified a hierarchical regulatory cascade with TFs regulated by DAGs, which in turn regulates gene expression. Integrative analysis of multi-omics data provided valuable molecular insights into the molecular mechanisms of SLE.
Collapse
Affiliation(s)
- Ting-You Wang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yong-Fei Wang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yan Zhang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jiangshan Jane Shen
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong; Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China; Lupus Research Institute, Affiliated Hospital of Jining Medical University, Jining, China
| | - Mengbiao Guo
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jing Yang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| | - Wanling Yang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
17
|
Tian S, Liu X, Fan Q, Ma J, Yao L, Li Y. Microarray expression and functional analysis of circular RNAs in the glomeruli of NZB/W F1 mice with lupus nephritis. Exp Ther Med 2019; 18:2813-2824. [PMID: 31555374 PMCID: PMC6755417 DOI: 10.3892/etm.2019.7901] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 06/20/2019] [Indexed: 02/06/2023] Open
Abstract
The present study applied a circular RNA (circRNA) microarray to examine the circRNA expression profiles in the glomeruli of NZB/W F1 mice with lupus nephritis (LN) during the pathogenesis of the disease. Glomeruli from two groups of female NZB/W F1 mice of the same age with either severe or mild LN were isolated by perfusion with dynabeads. A microarray analysis was then performed to evaluate the differentially expressed circRNAs of the glomeruli in the two groups, which were then confirmed by reverse transcription-quantitative PCR (RT-qPCR) assays. In addition, using a biomathematical strategy, the differentially-expressed circRNAs were identified in severe LN when compared with mild LN, and the commonly expressed circRNA species among these profiles were optimized via competing endogenous RNA (ceRNA) analysis. The predicted microRNAs (miRNAs/miRs) as downstream targets of circRNAs and upstream regulators of mRNAs were verified by RT-qPCR and the final circRNA-miRNA-mRNA network was constructed to identify the circRNA that was a pathogenic link in LN. The present study obtained 116 differentially expressed circRNAs, including 41 up- and 75 downregulated circRNAs, in severe LN when compared with mild LN, and 12 circRNAs were confirmed by RT-qPCR. The most significant difference was in the expression of mmu_circRNA_34428 (P<0.001) when comparing severe and mild LN glomeruli. A network of mmu_circRNA_34428-targeted miRNA-gene interactions was subsequently constructed, including miR-338-3p, miR-670-3p, miR-3066-5p, miR-210-5p and their corresponding mRNA targets. To the best of our knowledge, the present study elucidated, for the first time, circRNA profiling and the circRNA-miRNA interactions in the development of LN in female NZB/W F1 mice. The results revealed that mmu_circRNA_34428 could serve an important role in LN progression; however, the present study did not elucidate the functions of this circRNA or others in LN progression.
Collapse
Affiliation(s)
- Shuyan Tian
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xue Liu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Qiuling Fan
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jianfei Ma
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yanqiu Li
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
18
|
Frangou E, Vassilopoulos D, Boletis J, Boumpas DT. An emerging role of neutrophils and NETosis in chronic inflammation and fibrosis in systemic lupus erythematosus (SLE) and ANCA-associated vasculitides (AAV): Implications for the pathogenesis and treatment. Autoimmun Rev 2019; 18:751-760. [DOI: 10.1016/j.autrev.2019.06.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/02/2019] [Indexed: 02/08/2023]
|
19
|
Frangou E, Chrysanthopoulou A, Mitsios A, Kambas K, Arelaki S, Angelidou I, Arampatzioglou A, Gakiopoulou H, Bertsias GK, Verginis P, Ritis K, Boumpas DT. REDD1/autophagy pathway promotes thromboinflammation and fibrosis in human systemic lupus erythematosus (SLE) through NETs decorated with tissue factor (TF) and interleukin-17A (IL-17A). Ann Rheum Dis 2019; 78:238-248. [PMID: 30563869 PMCID: PMC6352428 DOI: 10.1136/annrheumdis-2018-213181] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 10/28/2018] [Accepted: 11/01/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The release of neutrophil extracellular traps (NETs) represents a novel neutrophil effector function in systemic lupus erythematosus (SLE) pathogenesis. However, the molecular mechanism underlying NET release and how NETs mediate end-organ injury in SLE remain elusive. METHODS NET formation and NET-related proteins were assessed in the peripheral blood and biopsies from discoid lupus and proliferative nephritis, using immunofluorescence, immunoblotting, quantitative PCR and ELISA. Autophagy was assessed by immunofluorescence and immunoblotting. The functional effects of NETs in vitro were assessed in a primary fibroblast culture. RESULTS Neutrophils from patients with active SLE exhibited increased basal autophagy levels leading to enhanced NET release, which was inhibited in vitro by hydroxychloroquine. NETosis in SLE neutrophils correlated with increased expression of the stress-response protein REDD1. Endothelin-1 (ET-1) and hypoxia-inducible factor-1α (HIF-1α) were key mediators of REDD1-driven NETs as demonstrated by their inhibition with bosentan and L-ascorbic acid, respectively. SLE NETs were decorated with tissue factor (TF) and interleukin-17A (IL-17A), which promoted thrombin generation and the fibrotic potential of cultured skin fibroblasts. Notably, TF-bearing and IL-17A-bearing NETs were abundant in discoid skin lesions and in the glomerular and tubulointerstitial compartment of proliferative nephritis biopsy specimens. CONCLUSIONS Our data suggest the involvement of REDD1/autophagy/NET axis in end-organ injury and fibrosis in SLE, a likely candidate for repositioning of existing drugs for SLE therapy. Autophagy-mediated release of TF-bearing and IL-17A-bearing NETs provides a link between thromboinflammation and fibrosis in SLE and may account for the salutary effects of hydroxychloroquine.
Collapse
Affiliation(s)
- Eleni Frangou
- Laboratory of Immune Regulation and Tolerance, Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Internal Medicine, Medical School, University of Cyprus, Nicosia, Cyprus
- Department of Nephrology and Transplantation, Nicosia General Hospital, Nicosia, Cyprus
| | - Akrivi Chrysanthopoulou
- Department of Internal Medicine, Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Alexandros Mitsios
- Department of Internal Medicine, Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Konstantinos Kambas
- Department of Internal Medicine, Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Stella Arelaki
- Department of Pathology, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Iliana Angelidou
- Department of Internal Medicine, Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Athanasios Arampatzioglou
- Department of Internal Medicine, Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Hariklia Gakiopoulou
- 1st Department of Pathology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - George K Bertsias
- Rheumatology, Clinical Immunology and Allergy, University of Crete School of Medicine, Heraklion, Greece
| | - Panayotis Verginis
- Laboratory of Immune Regulation and Tolerance, Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Konstantinos Ritis
- Department of Internal Medicine, Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Dimitrios T Boumpas
- Laboratory of Immune Regulation and Tolerance, Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Internal Medicine, Medical School, University of Cyprus, Nicosia, Cyprus
- 4th Department of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
20
|
Khan S, Dar SA, Mandal RK, Jawed A, Wahid M, Panda AK, Lohani M, Mishra BN, Akhter N, Haque S. Angiotensin-Converting Enzyme Gene I/D Polymorphism Is Associated With Systemic Lupus Erythematosus Susceptibility: An Updated Meta-Analysis and Trial Sequential Analysis. Front Physiol 2018; 9:1793. [PMID: 30618805 PMCID: PMC6305102 DOI: 10.3389/fphys.2018.01793] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 11/28/2018] [Indexed: 01/08/2023] Open
Abstract
Angiotensin-converting enzyme (ACE) gene is indispensable for endothelial control and vascular tone regulatory systems, usually affected in Systemic Lupus Erythematosus (SLE). ACE insertion/deletion (I/D) polymorphism may influence the progress of SLE. Earlier studies have investigated this association without any consistency in results. We performed this meta-analysis to evaluate the precise association between ACE I/D polymorphism and SLE susceptibility. The relevant studies were searched until December, 2017 using Medline (PubMed), Google-Scholar and EMBASE search engines. Twenty-five published studies involving 3,308 cases and 4,235 controls were included in this meta-analysis. Statistically significant increased risk was found for allelic (D vs. I: p = 0.007; OR = 1.202, 95% CI = 1.052–1.374), homozygous (DD vs. II: p = 0.025; OR = 1.347, 95% CI = 1.038–1.748), dominant (DD+ID vs. II: p = 0.002; OR = 1.195, 95% CI = 1.070–1.334), and recessive (DD vs. ID+II: p = 0.023; OR = 1.338, 95% CI = 1.042–1.718) genetic models. Subgroup analysis stratified by Asian ethnicity revealed significant risk of SLE in allelic (D vs. I: p = 0.045; OR = 1.238, 95% CI = 1.005–1.525) and marginal risk in dominant (DD+ID vs. II: p = 0.056; OR = 1.192, 95% CI = 0.995–1.428) models; whereas, no association was observed for Caucasian and African population. Publication bias was absent. In conclusion, ACE I/D polymorphism has significant role in overall SLE risk and it can be exploited as a prognostic marker for early SLE predisposition.
Collapse
Affiliation(s)
- Saif Khan
- Department of Basic Science, College of Dental Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Sajad A Dar
- The University College of Medical Sciences and GTB, Guru Teg Bahadur Hospital (University of Delhi), New Delhi, India.,Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Raju K Mandal
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Arshad Jawed
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Mohd Wahid
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Aditya K Panda
- Department of Bioscience & Bioinformatics, Khallikote University, Berhampur, India
| | - Mohtashim Lohani
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - B N Mishra
- Department of Biotechnology, Institute of Engineering and Technology, Lucknow, India
| | - Naseem Akhter
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing & Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
21
|
Wright VJ, Herberg JA, Kaforou M, Shimizu C, Eleftherohorinou H, Shailes H, Barendregt AM, Menikou S, Gormley S, Berk M, Hoang LT, Tremoulet AH, Kanegaye JT, Coin LJM, Glodé MP, Hibberd M, Kuijpers TW, Hoggart CJ, Burns JC, Levin M. Diagnosis of Kawasaki Disease Using a Minimal Whole-Blood Gene Expression Signature. JAMA Pediatr 2018; 172:e182293. [PMID: 30083721 PMCID: PMC6233768 DOI: 10.1001/jamapediatrics.2018.2293] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
IMPORTANCE To date, there is no diagnostic test for Kawasaki disease (KD). Diagnosis is based on clinical features shared with other febrile conditions, frequently resulting in delayed or missed treatment and an increased risk of coronary artery aneurysms. OBJECTIVE To identify a whole-blood gene expression signature that distinguishes children with KD in the first week of illness from other febrile conditions. DESIGN, SETTING, AND PARTICIPANTS The case-control study comprised a discovery group that included a training and test set and a validation group of children with KD or comparator febrile illness. The setting was pediatric centers in the United Kingdom, Spain, the Netherlands, and the United States. The training and test discovery group comprised 404 children with infectious and inflammatory conditions (78 KD, 84 other inflammatory diseases, and 242 bacterial or viral infections) and 55 healthy controls. The independent validation group comprised 102 patients with KD, including 72 in the first 7 days of illness, and 130 febrile controls. The study dates were March 1, 2009, to November 14, 2013, and data analysis took place from January 1, 2015, to December 31, 2017. MAIN OUTCOMES AND MEASURES Whole-blood gene expression was evaluated using microarrays, and minimal transcript sets distinguishing KD were identified using a novel variable selection method (parallel regularized regression model search). The ability of transcript signatures (implemented as disease risk scores) to discriminate KD cases from controls was assessed by area under the curve (AUC), sensitivity, and specificity at the optimal cut point according to the Youden index. RESULTS Among 404 patients in the discovery set, there were 78 with KD (median age, 27 months; 55.1% male) and 326 febrile controls (median age, 37 months; 56.4% male). Among 202 patients in the validation set, there were 72 with KD (median age, 34 months; 62.5% male) and 130 febrile controls (median age, 17 months; 56.9% male). A 13-transcript signature identified in the discovery training set distinguished KD from other infectious and inflammatory conditions in the discovery test set, with AUC of 96.2% (95% CI, 92.5%-99.9%), sensitivity of 81.7% (95% CI, 60.0%-94.8%), and specificity of 92.1% (95% CI, 84.0%-97.0%). In the validation set, the signature distinguished KD from febrile controls, with AUC of 94.6% (95% CI, 91.3%-98.0%), sensitivity of 85.9% (95% CI, 76.8%-92.6%), and specificity of 89.1% (95% CI, 83.0%-93.7%). The signature was applied to clinically defined categories of definite, highly probable, and possible KD, resulting in AUCs of 98.1% (95% CI, 94.5%-100%), 96.3% (95% CI, 93.3%-99.4%), and 70.0% (95% CI, 53.4%-86.6%), respectively, mirroring certainty of clinical diagnosis. CONCLUSIONS AND RELEVANCE In this study, a 13-transcript blood gene expression signature distinguished KD from other febrile conditions. Diagnostic accuracy increased with certainty of clinical diagnosis. A test incorporating the 13-transcript disease risk score may enable earlier diagnosis and treatment of KD and reduce inappropriate treatment in those with other diagnoses.
Collapse
Affiliation(s)
| | - Jethro A. Herberg
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | - Myrsini Kaforou
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | - Chisato Shimizu
- Department of Pediatrics, University of California San Diego, La Jolla,Rady Children’s Hospital–San Diego, San Diego, California
| | | | - Hannah Shailes
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | - Anouk M. Barendregt
- Department of Pediatric Hematology, Immunology & Infectious Diseases, Emma Children’s Hospital, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Stephanie Menikou
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | - Stuart Gormley
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | - Maurice Berk
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | | | - Adriana H. Tremoulet
- Department of Pediatrics, University of California San Diego, La Jolla,Rady Children’s Hospital–San Diego, San Diego, California
| | - John T. Kanegaye
- Department of Pediatrics, University of California San Diego, La Jolla,Rady Children’s Hospital–San Diego, San Diego, California
| | - Lachlan J. M. Coin
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, United Kingdom,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Australia
| | - Mary P. Glodé
- Section of Infectious Diseases, Department of Pediatrics, University of Colorado Denver School of Medicine Anschutz Medical Campus, Aurora,Children’s Hospital Colorado, Aurora
| | - Martin Hibberd
- Infectious Diseases, Genome Institute of Singapore, Singapore
| | - Taco W. Kuijpers
- Department of Pediatric Hematology, Immunology & Infectious Diseases, Emma Children’s Hospital, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands,Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Clive J. Hoggart
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | - Jane C. Burns
- Department of Pediatrics, University of California San Diego, La Jolla,Rady Children’s Hospital–San Diego, San Diego, California
| | - Michael Levin
- Section of Paediatrics, Imperial College London, London, United Kingdom
| | | |
Collapse
|
22
|
A DNA-Methylated Sight on Autoimmune Inflammation Network across RA, pSS, and SLE. J Immunol Res 2018; 2018:4390789. [PMID: 30159339 PMCID: PMC6109517 DOI: 10.1155/2018/4390789] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/08/2018] [Indexed: 12/14/2022] Open
Abstract
Methylation variabilities of inflammatory cytokines play important roles in the development of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and primary Sjögren's syndrome (pSS). With heightened focus on personalized and precise medicine, it is necessary to compare and contrast the difference and similarity of cytokine methylation status between the 3 most classic autoimmune diseases (AIDs). In this study, we integrated 5 Cytokine-Chips from genome-wide DNA methylation datasets of the 3 kind of AIDs, delta-beta value was calculated for intergroup difference, and comprehensive bioinformatics analyses of cytokine genes with aberrant methylations were performed. 125 shared differential methylation variabilities (DMVs) were identified. There were 102 shared DMVs with similar methylation status; 3 hypomethylated differential methylation regions (DMRs) across the AIDs were found, and all 3 DMRs were hypomethylated. DMRs (AZU1, LTBR, and RTEL1) were likely to serve as activator in the inflammatory process. Particularly, AZU1 and LTBR with hypomethylated TSS and first exon located in the promoter regions were able to trigger inflammation signaling cascades and play critical roles in autoimmune tautology. Moreover, functional epigenetic module (FEM) algorithm showed that different inflammatory networks are involved in different AIDs; 5 hotspots were identified as biologically plausible pathways in inducing or perpetuating of inflammation which are epigenetically deregulated in AIDs. We concluded methylation variabilities among the same cytokines can greatly impact the perpetuation of inflammatory process or signal pathway of AIDs. Differentiating the cytokine methylation status will serve as valuable resource for researchers alike to gain better understanding of the epigenetic mechanisms of the three AIDs. Even more importantly, better understanding of cytokine methylation variability existing between the three classic AIDs will aid in identification of potential epigenetic biomarkers and therapeutic targets. This trial is registered with ChiCTR-INR-16010290, a clinical trial for the treatment of rheumatoid arthritis with Warming yang and Smoothening Meridians.
Collapse
|
23
|
Chetina EV, Markova GA. [Upcoming value of gene expression analysis in rheumatology]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2018; 64:221-232. [PMID: 29964257 DOI: 10.18097/pbmc20186403221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease of unknown etiology, which involves disturbance in immune system signaling pathway functions, damage of other tissues, pain and joint destruction. Modern treatment attempts to improve pathophysiological and biochemical mechanisms damaged by the disease. However, due to the RA patient heterogeneity personalized approach to treatment is required; the choice of personalized treatment is complicated by the variability of patient's response to treatment. Gene expression analysis might serve a tool for the disease control and therapy personification for inhibition of inflammation and pain as well as for prevention of joint destruction.
Collapse
Affiliation(s)
- E V Chetina
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - G A Markova
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| |
Collapse
|
24
|
Patra P, Izawa T, Pena-Castillo L. REPA: Applying Pathway Analysis to Genome-Wide Transcription Factor Binding Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:1270-1283. [PMID: 27019499 DOI: 10.1109/tcbb.2015.2453948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Pathway analysis has been extensively applied to aid in the interpretation of the results of genome-wide transcription profiling studies, and has been shown to successfully find associations between the biological phenomena under study and biological pathways. There are two widely used approaches of pathway analysis: over-representation analysis, and gene set analysis. Recently genome-wide transcription factor binding data has become widely available allowing for the application of pathway analysis to this type of data. In this work, we developed regulatory enrichment pathway analysis (REPA) to apply gene set analysis to genome-wide transcription factor binding data to infer associations between transcription factors and biological pathways. We used the transcription factor binding data generated by the ENCODE project, and gene sets from the Molecular Signatures and KEGG databases. Our results showed that 54 percent of the predictions examined have literature support and that REPA's recall is roughly 54 percent. This level of precision is promising as several of REPA's predictions are expected to be novel and can be used to guide new research avenues. In addition, the results of our case studies showed that REPA enhances the interpretation of genome-wide transcription profiling studies by suggesting putative regulators behind the observed transcriptional responses.
Collapse
|
25
|
The Involvement of MicroRNAs in Modulation of Innate and Adaptive Immunity in Systemic Lupus Erythematosus and Lupus Nephritis. J Immunol Res 2018; 2018:4126106. [PMID: 29854836 PMCID: PMC5964414 DOI: 10.1155/2018/4126106] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 04/03/2018] [Indexed: 12/20/2022] Open
Abstract
Noncoding RNAs (ncRNAs), including microRNAs (miRNAs), represent a family of RNA molecules that do not translate into protein. Nevertheless, they have the ability to regulate gene expression and play an essential role in immune cell differentiation and function. MicroRNAs were found to be differentially expressed in various tissues, and changes in their expression have been associated with several pathological processes. Yet, their roles in systemic lupus erythematosus (SLE) and lupus nephritis (LN) remain to be elucidated. Both SLE and LN are characterized by a complex dysfunction of the innate and adaptive immunity. Recently, significant findings have been made in understanding SLE through the use of genetic variant identification and expression pattern analysis and mouse models, as well as epigenetic analyses. Abnormalities in immune cell responses, cytokine and chemokine production, cell activation, and apoptosis have been linked to a unique expression pattern of a number of miRNAs that have been implicated in the immune pathogenesis of this autoimmune disease. The recent evidence that significantly increased the understanding of the pathogenesis of SLE drives a renewed interest in efficient therapy targets. This review aims at providing an overview of the current state of research on the expression and role of miRNAs in the immune pathogenesis of SLE and LN.
Collapse
|
26
|
Anaya JM, Leon KJ, Rojas M, Rodriguez Y, Pacheco Y, Acosta-Ampudia Y, Monsalve DM, Ramirez-Santana C. Progress towards precision medicine for lupus: the role of genetic biomarkers. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018. [DOI: 10.1080/23808993.2018.1448266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Kelly J. Leon
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yhojan Rodriguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yovana Pacheco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Diana M. Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Carolina Ramirez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
27
|
Tchetina E, Markova G. The clinical utility of gene expression examination in rheumatology. Mediterr J Rheumatol 2017; 28:116-126. [PMID: 32185269 PMCID: PMC7046055 DOI: 10.31138/mjr.28.3.116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/24/2017] [Indexed: 01/09/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease with unknown etiology that affects various pathways within the immune system, involves many other tissues and is associated with pain and joint destruction. Current treatments fail to address pathophysiological and biochemical mechanisms involved in joint degeneration and the induction of pain. Moreover, RA patients are extremely heterogeneous and require specific treatments, the choice of which is complicated by the fact that not all patients equally respond to therapy. Gene expression analysis offer tools for patient management and personalization of patient’s care to meet individual needs in controlling inflammation and pain and delaying joint destruction.
Collapse
Affiliation(s)
- Elena Tchetina
- Immunology and Molecular Biology Laboratory, Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - Galina Markova
- Immunology and Molecular Biology Laboratory, Nasonova Research Institute of Rheumatology, Moscow, Russia
| |
Collapse
|
28
|
Defective DNA repair and chromatin organization in patients with quiescent systemic lupus erythematosus. Arthritis Res Ther 2016; 18:182. [PMID: 27492607 PMCID: PMC4973109 DOI: 10.1186/s13075-016-1081-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/21/2016] [Indexed: 11/21/2022] Open
Abstract
Background Excessive autoantibody production characterizing systemic lupus erythematosus (SLE) occurs irrespective of the disease’s clinical status and is linked to increased lymphocyte apoptosis. Herein, we tested the hypothesis that defective DNA damage repair contributes to increased apoptosis in SLE. Methods We evaluated nucleotide excision repair at the N-ras locus, DNA double-strand breaks repair and apoptosis rates in peripheral blood mononuclear cells from anti-dsDNA autoantibody-positive patients (six with quiescent disease and six with proliferative nephritis) and matched healthy controls following ex vivo treatment with melphalan. Chromatin organization and expression levels of DNA repair- and apoptosis-associated genes were also studied in quiescent SLE. Results Defective nucleotide excision repair and DNA double-strand breaks repair were found in SLE, with lupus nephritis patients showing higher DNA damage levels than those with quiescent disease. Melphalan-induced apoptosis rates were higher in SLE than control cells and correlated inversely with DNA repair efficiency. Chromatin at the N-ras locus was more condensed in SLE than controls, while treatment with the histone deacetylase inhibitor vorinostat resulted in hyperacetylation of histone H4, chromatin decondensation, amelioration of DNA repair efficiency and decreased apoptosis. Accordingly, genes involved in DNA damage repair and signaling pathways, such as DDB1, ERCC2, XPA, XPC, MRE11A, RAD50, PARP1, MLH1, MLH3, and ATM were significantly underexpressed in SLE versus controls, whereas PPP1R15A, BARD1 and BBC3 genes implicated in apoptosis were significantly overexpressed. Conclusions Epigenetically regulated functional abnormalities of DNA repair machinery occur in SLE, regardless of clinical disease activity, and may promote lymphocyte apoptosis. Approaches to correct these abnormalities may be of therapeutic value in SLE. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1081-3) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Zhu H, Mi W, Luo H, Chen T, Liu S, Raman I, Zuo X, Li QZ. Whole-genome transcription and DNA methylation analysis of peripheral blood mononuclear cells identified aberrant gene regulation pathways in systemic lupus erythematosus. Arthritis Res Ther 2016; 18:162. [PMID: 27412348 PMCID: PMC4942934 DOI: 10.1186/s13075-016-1050-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/20/2016] [Indexed: 12/13/2022] Open
Abstract
Background Recent achievement in genetics and epigenetics has led to the exploration of the pathogenesis of systemic lupus erythematosus (SLE). Identification of differentially expressed genes and their regulatory mechanism(s) at whole-genome level will provide a comprehensive understanding of the development of SLE and its devastating complications, lupus nephritis (LN). Methods We performed whole-genome transcription and DNA methylation analysis in PBMC of 30 SLE patients, including 15 with LN (SLE LN+) and 15 without LN (SLE LN−), and 25 normal controls (NC) using HumanHT-12 Beadchips and Illumina Human Methy450 chips. The serum proinflammatory cytokines were quantified using Bio-plex Human Cytokine 27-plex assay. Differentially expressed genes and differentially methylated CpG were analyzed with GenomeStudio, R, and SAM software. The association between DNA methylation and gene expression were tested. Gene interaction pathways of the differentially expressed genes were analyzed by IPA software. Results We identified 552 upregulated genes and 550 downregulated genes in PBMC of SLE. Integration of DNA methylation and gene expression profiling showed that 334 upregulated genes were hypomethylated, and 479 downregulated genes were hypermethylated. Pathway analysis on the differential genes in SLE revealed significant enrichment in interferon (IFN) signaling and toll-like receptor (TLR) signaling pathways. Nine IFN- and seven TLR-related genes were identified and displayed step-wise increase in SLE LN− and SLE LN+. Hypomethylated CpG sites were detected on these genes. The gene expressions for MX1, GPR84, and E2F2 were increased in SLE LN+ as compared to SLE LN− patients. The serum levels of inflammatory cytokines, including IL17A, IP-10, bFGF, TNF-α, IL-6, IL-15, GM-CSF, IL-1RA, IL-5, and IL-12p70, were significantly elevated in SLE compared with NC. The levels of IL-15 and IL1RA correlated with their mRNA expression. The upregulation of IL-15 may be regulated by hypomethylated CpG sites in the promotor region of the gene. Conclusions Our study has demonstrated that significant number of differential genes in SLE were involved in IFN, TLR signaling pathways, and inflammatory cytokines. The enrichment of differential genes has been associated with aberrant DNA methylation, which may be relevant to the pathogenesis of SLE. Our observations have laid the groundwork for further diagnostic and mechanistic studies of SLE and LN. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-1050-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Honglin Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China.,Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Wentao Mi
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Hui Luo
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China.,Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Tao Chen
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Shengxi Liu
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Indu Raman
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xiaoxia Zuo
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China.
| | - Quan-Zhen Li
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China. .,Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
| |
Collapse
|
30
|
Ho RC, Ong H, Thiaghu C, Lu Y, Ho CS, Zhang MW. Genetic Variants That Are Associated with Neuropsychiatric Systemic Lupus Erythematosus. J Rheumatol 2016; 43:541-551. [PMID: 26773105 DOI: 10.3899/jrheum.150884] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE While genetic risks have been implicated in systemic lupus erythematosus (SLE), the involvement of various genotypes in neuropsychiatric SLE (NPSLE) remains uncertain. The present metaanalysis aimed to combine data from different studies and evaluate the association between each genotype and the risk of developing NPSLE. METHODS Studies were searched and retrieved from online databases (PubMed, EMBASE, BIOSIS, and ScienceDirect). Case-control studies were chosen if they reported genotype frequencies of the γ Fc region (FCγR) receptors II-A, III-A, and III-B; tumor necrosis factor-α (TNF-α); mannan-binding lectin (MBL); integrin alpha M (ITGAM); interleukin (IL) 1, IL-1β, and IL-6; IL-10 promoter; and vitamin D genes. The OR were used to assess the strength of this association between patients with NPSLE and SLE. RESULTS A total of 33 studies were considered in this metaanalysis. The results suggest that these genotypes demonstrated a significant association with NPSLE: the homozygous FCγR IIIa 158 FF genotype (OR 1.89, p = 0.03 for FF vs VV + FV), heterozygous FCγR IIIb NA1/2 genotype (OR 2.14, p = 0.03 for NA1/2 vs NA1/1; OR 1.81, p = 0.04 for NA1/2 vs NA1/1 + NA2/2), and homozygous ITGAM rs1143679 HH genotype (OR 3.39, p = 0.04 for HH vs RH; OR 3.11, p = 0.048 for HH vs RR + RH). Polymorphisms of the TNF-α, MBL2, IL-1, IL-1β, IL-6, IL-10 promoter, and vitamin D receptor genes did not show a statistically significant association with the risk of developing NPSLE (p > 0.05). CONCLUSION This metaanalysis indicates that polymorphisms in the pathways of immune complex clearance, such as the FcγRIIIa, FcγRIIIb, and ITGAM genotypes, are potential susceptibility genes for NPSLE.
Collapse
Affiliation(s)
- Roger C Ho
- From the Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Institute of Mental Health, Singapore; Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China.R.C. Ho, FRCPC, Assistant Professor, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; H. Ong, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; C. Thiaghu, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Y. Lu, PhD, Assistant Professor, Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine; C.S. Ho, MRCPsych, Specialist Registrar, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; M.W. Zhang, MRCPsych, Specialist Registrar, Institute of Mental Health
| | - Huiyi Ong
- From the Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Institute of Mental Health, Singapore; Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China.R.C. Ho, FRCPC, Assistant Professor, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; H. Ong, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; C. Thiaghu, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Y. Lu, PhD, Assistant Professor, Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine; C.S. Ho, MRCPsych, Specialist Registrar, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; M.W. Zhang, MRCPsych, Specialist Registrar, Institute of Mental Health
| | - Chandra Thiaghu
- From the Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Institute of Mental Health, Singapore; Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China.R.C. Ho, FRCPC, Assistant Professor, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; H. Ong, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; C. Thiaghu, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Y. Lu, PhD, Assistant Professor, Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine; C.S. Ho, MRCPsych, Specialist Registrar, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; M.W. Zhang, MRCPsych, Specialist Registrar, Institute of Mental Health
| | - Yanxia Lu
- From the Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Institute of Mental Health, Singapore; Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China.R.C. Ho, FRCPC, Assistant Professor, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; H. Ong, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; C. Thiaghu, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Y. Lu, PhD, Assistant Professor, Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine; C.S. Ho, MRCPsych, Specialist Registrar, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; M.W. Zhang, MRCPsych, Specialist Registrar, Institute of Mental Health.
| | - Cyrus S Ho
- From the Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Institute of Mental Health, Singapore; Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China.R.C. Ho, FRCPC, Assistant Professor, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; H. Ong, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; C. Thiaghu, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Y. Lu, PhD, Assistant Professor, Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine; C.S. Ho, MRCPsych, Specialist Registrar, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; M.W. Zhang, MRCPsych, Specialist Registrar, Institute of Mental Health
| | - Melvyn W Zhang
- From the Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Institute of Mental Health, Singapore; Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China.R.C. Ho, FRCPC, Assistant Professor, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; H. Ong, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; C. Thiaghu, MBBS, Medical Officer, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; Y. Lu, PhD, Assistant Professor, Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine; C.S. Ho, MRCPsych, Specialist Registrar, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore; M.W. Zhang, MRCPsych, Specialist Registrar, Institute of Mental Health
| |
Collapse
|
31
|
Perez-Hernandez J, Olivares MD, Forner MJ, Chaves FJ, Cortes R, Redon J. Urinary dedifferentiated podocytes as a non-invasive biomarker of lupus nephritis. Nephrol Dial Transplant 2016; 31:780-9. [PMID: 26932688 DOI: 10.1093/ndt/gfw002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/03/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Currently, renal biopsy remains the gold standard for the diagnosis and prognosis of lupus nephritis (LN). However, it is an invasive method, and new non-invasive laboratory tests are needed to identify renal involvement without renal biopsy. Podocyte damage plays an important role in the pathogenesis and progression of systemic lupus erythematosus (SLE). We characterize whether the phenotype of urinary podocytes (viability, apoptosis, mRNA and protein levels of the podocyte-associated molecules) is a novel marker of clinical and histological features in SLE patients with or without LN. METHODS We quantified in urinary sediments of 32 SLE patients and 20 controls, mRNA and protein levels of podocalyxin, synapotopodin, podocin, nephrin and WT-1 by quantitative real-time polymerase chain reaction and western blot analysis and correlated these with clinical and histological parameters. The viability of detached urine podocytes was analysed by flow cytometry with podocalyxin and annexin V/7-AAD double staining and immunofluorescence of urine podocyte cultures. RESULTS The degree of a poptotic podocytes from urine samples was significantly decreased in patients with LN, especially in the active state (33% compared with 75% in controls, P < 0.001), and the majority of the detached podocytes in the urine of patients with active LN were viable (70% grew in culture). Furthermore, urinary mRNA of podocyte-associated molecules was significantly lower in patients with active LN (P < 0.05) compared with healthy controls, and protein levels of podocyte markers were significantly increased in SLE patients, especially with LN compared with SLE without LN (P < 0.05) and the healthy control group (P < 0.01). Finally, urinary protein levels of podocyte-related markers were associated with proteinuria and histological features (P < 0.05 and P < 0.01), and receiver operating characteristics curves of protein levels discriminate between LN and healthy controls with an area under the curve (AUC) between 0.91 and 0.77 (P < 0.001). CONCLUSIONS Urinary dedifferentiated podocytes were shown in active LN, and their protein levels correlated with proteinuria and histological features in LN. These preliminary results suggest that it could be a potentially useful non-invasive marker for evaluating the progression of glomerular disease in SLE.
Collapse
Affiliation(s)
- Javier Perez-Hernandez
- Genotyping and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain Cardiometabolic and Renal Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain
| | - Maria D Olivares
- Genotyping and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain Cardiometabolic and Renal Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain
| | - Maria J Forner
- Cardiometabolic and Renal Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Minister of Health, Madrid, Spain
| | - Felipe J Chaves
- Genotyping and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Institute of Health Carlos III, Minister of Health, Barcelona, Spain
| | - Raquel Cortes
- Genotyping and Genetic Diagnosis Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain Cardiometabolic and Renal Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain
| | - Josep Redon
- Cardiometabolic and Renal Unit, INCLIVA Biomedical Research Institute, Avd. Menéndez Pelayo, acceso 4, 46010 Valencia, Spain CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, Minister of Health, Madrid, Spain
| |
Collapse
|
32
|
Abstract
Epigenetic abnormalities are part of the pathogenetic alterations involved in the development of rheumatic disorders. In this context, the main musculoskeletal cell lineages, which are generated from the pool of mesenchymal stromal cells (MSCs), and the immune cells that participate in rheumatic diseases are deregulated. In this Review, we focus on microRNA (miRNA)-mediated regulatory pathways that control cell proliferation, drive the production of proinflammatory mediators and modulate bone remodelling. The main studies that identify miRNAs as regulators of immune cell fate, MSC differentiation and immunomodulatory properties - parameters that are altered in rheumatoid arthritis (RA) and osteoarthritis (OA) - are also discussed, with emphasis on the importance of miRNAs in the regulation of cellular machinery, extracellular matrix remodelling and cytokine release. A deeper understanding of the involvement of miRNAs in rheumatic diseases is needed before these regulatory pathways can be explored as therapeutic approaches for patients with RA or OA.
Collapse
|
33
|
Krasoudaki E, Banos A, Stagakis E, Loupasakis K, Drakos E, Sinatkas V, Zampoulaki A, Papagianni A, Iliopoulos D, Boumpas DT, Bertsias GK. Micro-RNA analysis of renal biopsies in human lupus nephritis demonstrates up-regulated miR-422a driving reduction of kallikrein-related peptidase 4. Nephrol Dial Transplant 2015; 31:1676-86. [DOI: 10.1093/ndt/gfv374] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 10/05/2015] [Indexed: 12/31/2022] Open
|
34
|
Dozmorov MG, Dominguez N, Bean K, Macwana SR, Roberts V, Glass E, James JA, Guthridge JM. B-Cell and Monocyte Contribution to Systemic Lupus Erythematosus Identified by Cell-Type-Specific Differential Expression Analysis in RNA-Seq Data. Bioinform Biol Insights 2015; 9:11-9. [PMID: 26512198 PMCID: PMC4599594 DOI: 10.4137/bbi.s29470] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by complex interplay among immune cell types. SLE activity is experimentally assessed by several blood tests, including gene expression profiling of heterogeneous populations of cells in peripheral blood. To better understand the contribution of different cell types in SLE pathogenesis, we applied the two methods in cell-type-specific differential expression analysis, csSAM and DSection, to identify cell-type-specific gene expression differences in heterogeneous gene expression measures obtained using RNA-seq technology. We identified B-cell-, monocyte-, and neutrophil-specific gene expression differences. Immunoglobulin-coding gene expression was altered in B-cells, while a ribosomal signature was prominent in monocytes. On the contrary, genes differentially expressed in the heterogeneous mixture of cells did not show any functional enrichment. Our results identify antigen binding and structural constituents of ribosomes as functions altered by B-cell- and monocyte-specific gene expression differences, respectively. Finally, these results position both csSAM and DSection methods as viable techniques for cell-type-specific differential expression analysis, which may help uncover pathogenic, cell-type-specific processes in SLE.
Collapse
Affiliation(s)
- Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Nicolas Dominguez
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Krista Bean
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Susan R Macwana
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Virginia Roberts
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Edmund Glass
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Judith A James
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Joel M Guthridge
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| |
Collapse
|
35
|
Alghasham A, Al Salloom AAM, Alghamadi ASS, Rasheed Z. Impact of anti-peroxynitrite-damaged-thymidine-monophosphate antibodies on disease activity in patients with systemic lupus erythematosus. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2015; 34:56-68. [PMID: 25513864 DOI: 10.1080/15257770.2014.958235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Present study probes the role of peroxynitrite (ONOO(-))-modified thymidine-5'-monophosphate (TMP) in SLE patients with different disease activity scores according to the SLE Disease Activity Index (SLEDAI). Serum analysis showed significant increased number of subjects positive for anti-ONOO(-)-TMP-protein antibodies in SLE patients with different SLEDAI scores. Interestingly, the levels of these antibodies were significantly higher among SLE patients, whose SLEDAI scores were ≥20. In addition, a significant correlation was observed between the levels of anti-ONOO(-)-TMP-protein antibodies and the SLEDAI score (r = 0.595, p < 0.0001). In short, this study shows a positive association between anti-ONOO(-)-TMP-protein antibodies and SLEDAI. The stronger response observed in patients with higher SLEDAI scores suggests that anti-ONOO(-)-TMP-protein antibodies may be useful in evaluating the progression of SLE and in elucidating the mechanisms of disease pathogenesis.
Collapse
Affiliation(s)
- Abdullah Alghasham
- a Department of Pharmacology and Therapeutics, College of Medicine , Qassim University , Buraidah , KSA
| | | | | | | |
Collapse
|
36
|
Duroux-Richard I, Cuenca J, Ponsolles C, Piñeiro AB, Gonzalez F, Roubert C, Areny R, Chea R, Pefaur J, Pers YM, Figueroa FE, Jorgensen C, Khoury M, Apparailly F. MicroRNA Profiling of B Cell Subsets from Systemic Lupus Erythematosus Patients Reveals Promising Novel Biomarkers. Int J Mol Sci 2015; 16:16953-16965. [PMID: 26225955 PMCID: PMC4581178 DOI: 10.3390/ijms160816953] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/10/2015] [Accepted: 07/15/2015] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs control the differentiation and function of B cells, which are considered key elements in the pathogenesis of systemic lupus erythematosus (SLE). However, a common micro(mi)RNA signature has not emerged since published data includes patients of variable ethnic background, type of disease, and organ involvement, as well as heterogeneous cell populations. Here, we aimed at identifying a miRNA signature of purified B cells from renal and non-renal severe SLE patients of Latin American background, a population known to express severe disease. Genome-wide miRNA expression analyses were performed on naive and memory B cells and revealed two categories of miRNA signatures. The first signature represents B cell subset-specific miRNAs deregulated in SLE: 11 and six miRNAs discriminating naive and memory B cells of SLE patients from healthy controls (HC), respectively. Whether the miRNA was up or down-regulated in memory B cells as compared with naive B cells in HC, this difference was abolished in SLE patients, and vice versa. The second signature identifies six miRNAs associated with specific pathologic features affecting renal outcome, providing a further understanding for SLE pathogenesis. Overall, the present work provided promising biomarkers in molecular diagnostics for disease severity as well as potential new targets for therapeutic intervention in SLE.
Collapse
Affiliation(s)
- Isabelle Duroux-Richard
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Saint Eloi, Montpellier 34295, France.
- University of Montpellier, Montpellier 34090, France.
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile.
- Cells for Cells, Santiago 7620001, Chile.
| | - Clara Ponsolles
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Saint Eloi, Montpellier 34295, France.
- University of Montpellier, Montpellier 34090, France.
| | - Alejandro Badilla Piñeiro
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile.
| | - Fernando Gonzalez
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile.
| | | | - Roser Areny
- Hospital Félix Bulnes, Santiago 7510021, Chile.
| | - Rosa Chea
- Hospital Barros Luco, Santiago 8900085, Chile.
| | | | - Yves-Marie Pers
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Saint Eloi, Montpellier 34295, France.
- University of Montpellier, Montpellier 34090, France.
- Clinical Department for Osteoarticular Diseases, University Hospital Lapeyronie, Montpellier 34295, France.
| | - Fernando E Figueroa
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile.
| | - Christian Jorgensen
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Saint Eloi, Montpellier 34295, France.
- University of Montpellier, Montpellier 34090, France.
- Clinical Department for Osteoarticular Diseases, University Hospital Lapeyronie, Montpellier 34295, France.
| | - Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago 7620001, Chile.
- Cells for Cells, Santiago 7620001, Chile.
| | - Florence Apparailly
- INSERM, U1183, Institute of Regenerative Medicine and Biotherapy, University Hospital Saint Eloi, Montpellier 34295, France.
- University of Montpellier, Montpellier 34090, France.
- Clinical Department for Osteoarticular Diseases, University Hospital Lapeyronie, Montpellier 34295, France.
| |
Collapse
|
37
|
Yang Y, Tang Q, Zhao M, Liang G, Wu H, Li D, Xie Y, Tan Y, Dai Y, Yung S, Chan TM, Lu Q. The effect of mycophenolic acid on epigenetic modifications in lupus CD4+T cells. Clin Immunol 2015; 158:67-76. [PMID: 25791245 DOI: 10.1016/j.clim.2015.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 01/26/2015] [Accepted: 03/09/2015] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex systemic autoimmune disease involving multiple organs and characterized by overproduction of autoantibodies and T and B cell abnormalities. The treatment for SLE has been restricted to immunosuppressants and corticosteroids. Mycophenolate mofetil (MMF), as a relatively new immunosuppressant, is now widely used in the treatment of SLE patients, particularly those with nephritis. However, it is unclear whether mycophenolic acid (MPA) could modulate the reported disorders of epigenetic status in CD4(+)T cells from SLE patients. In this study, we demonstrated that MPA can upregulate the histone H3/H4 global acetylation status by regulating HATs and HDACs in lupus CD4(+)T cells. Furthermore, we found that MPA also affected the histone H4 acetylation and histone H3K4 tri-methylation levels in CD40L promoter region that inhibited the expression of CD40L. These findings indicate the potential epigenetic mechanism of therapeutic effects of MPA in SLE.
Collapse
Affiliation(s)
- Yang Yang
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Tang
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhao
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gongping Liang
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Duo Li
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yubing Xie
- Changsha Blood Center, Changsha, Hunan, China
| | - Yixin Tan
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Dai
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
| | - Susan Yung
- Division of Nephrology, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Tak Mao Chan
- Division of Nephrology, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Qianjin Lu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
38
|
Wang H, Jiang Y, Peng H, Chen Y, Zhu P, Huang Y. Recent progress in microRNA delivery for cancer therapy by non-viral synthetic vectors. Adv Drug Deliv Rev 2015; 81:142-60. [PMID: 25450259 DOI: 10.1016/j.addr.2014.10.031] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 10/26/2014] [Accepted: 10/30/2014] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression. Because of significant changes in their expression in cancer, miRNAs are believed to be key factors in cancer genetics and to have potential as anticancer drugs. However, the delivery of miRNAs is limited by many barriers, such as low cellular uptake, immunogenicity, renal clearance, degradation by nucleases, elimination by phagocytic immune cells, poor endosomal release, and untoward side effects. Nonviral delivery systems have been developed to overcome these obstacles. In this review, we provide insights into the development of non-viral synthetic miRNA vectors and the promise of miRNA-based anticancer therapies, including therapeutic applications of miRNAs, challenges of vector design to overcome the delivery obstacles, and the development of miRNA delivery systems for cancer therapy. Additionally, we highlight some representative examples that give a glimpse into the current trends into the design and application of efficient synthetic systems for miRNA delivery. Overall, a better understanding of the rational design of miRNA delivery systems will promote their translation into effective clinical treatments.
Collapse
|
39
|
Souliotis VL, Sfikakis PP. Increased DNA double-strand breaks and enhanced apoptosis in patients with lupus nephritis. Lupus 2014; 24:804-15. [PMID: 25542905 DOI: 10.1177/0961203314565413] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 11/25/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE DNA double-strand breaks (DSBs) lead to mutations, genomic instability and apoptotic death, whereas accumulation of apoptotic cells results in excessive autoantigen presentation and autoantibody formation. We aimed to measure DSB levels in lupus nephritis, a severe complication of the prototypic systemic autoimmune disease. METHODS The intrinsic DNA damage and the apoptosis induction/DSB levels were evaluated in peripheral blood mononuclear cells of six patients and 10 healthy controls following exposure to genotoxic agents (melphalan, cisplatin) ex vivo. DSBs were assessed using immunofluorescence quantification of γH2AX foci and comet assay. RESULTS Intrinsic DNA damage was increased in lupus versus control cells in both assays (Olive Tail Moment units of 15.8 ± 2.3 versus 3.0 ± 1.4 in comet, p < 0.01; % γH2AX-positive cells: 13.6 ± 1.8 versus 4.6 ± 0.9, p < 0.01, respectively). Melphalan or cisplatin doses as low as 9.9 ± 4.8 or 29.8 ± 8.3 µg/ml, respectively, were sufficient to induce apoptosis in lupus cells; control cells required doses of 32.3 ± 7.7 and 67.7 ± 5.5 µg/ml, respectively. Drug-induced DSB levels were increased in lupus versus control cells, with the area under the curve (AUC) for melphalan-induced DSBs being 3050 ± 610 (% γH2AX-positive staining cells) × (drug dose) in patients and 1580 ± 350 in controls (p < 0.05); the corresponding values for cisplatin-induced AUC were 13900 ± 1800 for lupus and 4500 ± 750 for controls (p < 0.01). Interestingly, within either lupus patients or controls examined, the accumulation of DSBs correlated with apoptosis degrees (all p < 0.01). Results in lupus cells were not associated with individual disease activity level or treatment modalities at the time of the study. CONCLUSION These findings suggest a novel mechanism by which increased accumulation of DSBs may render cells more sensitive to apoptosis, thus contributing to the induction of systemic autoimmunity.
Collapse
Affiliation(s)
- V L Souliotis
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - P P Sfikakis
- Rheumatology Unit, First Department of Propedeutic Internal Medicine, Athens University Medical School, Athens, Greece
| |
Collapse
|
40
|
Piotrowski P, Grobelna MK, Wudarski M, Olesińska M, Jagodziński PP. Genetic variants of DNMT3A and systemic lupus erythematosus susceptibility. Mod Rheumatol 2014; 25:96-9. [PMID: 24716599 DOI: 10.3109/14397595.2014.902296] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES A significant increase in DNA methyltransferase 3A (DNMT3A) transcript levels has recently been demonstrated in peripheral blood mononuclear cells from systemic lupus erythematosus (SLE) patients as compared to healthy individuals. METHODS Employing high resolution melting curve analysis (HRM) and PCR-restriction fragment length polymorphism analysis, we assessed the frequency of five single nucleotide polymorphisms (SNPs) of this gene: rs2289195, rs7590760, rs13401241, rs749131 and rs1550117, situated in different linkage disequilibrium blocks of the DNMT3A gene in two hundred and fifty seven women with SLE and six hundred and twenty five controls. RESULTS The lowest p values of the trend test were observed for the DNMT3A -448A> G (rs1550117) SNP (ptrend = 0.0111). We also found that, in a dominant inheritance model, the DNMT3A -448A> G SNP may protect from SLE development [odds ratio (OR) = 0.494 (0.294-0.830), p = 0.0068, pcorr = 0.034]. Furthermore, we observed that the DNMT3A -448A > G SNP in dominant inheritance models may protect from immunologic manifestations of SLE [OR = 0.1753 (95% CI = 0.04976-0.6176, p = 0.0026, pcorr = 0.0468). CONCLUSIONS Our study demonstrates that the DNMT3A -448A> G SNP might protect from SLE and its immunologic manifestations in a sample from the Polish population.
Collapse
Affiliation(s)
- Piotr Piotrowski
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences , Poznań , Poland
| | | | | | | | | |
Collapse
|
41
|
Ratliff ML, Templeton TD, Ward JM, Webb CF. The Bright Side of Hematopoiesis: Regulatory Roles of ARID3a/Bright in Human and Mouse Hematopoiesis. Front Immunol 2014; 5:113. [PMID: 24678314 PMCID: PMC3958700 DOI: 10.3389/fimmu.2014.00113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/04/2014] [Indexed: 12/12/2022] Open
Abstract
ARID3a/Bright is a DNA-binding protein that was originally discovered for its ability to increase immunoglobulin transcription in antigen-activated B cells. It interacts with DNA as a dimer through its ARID, or A/T-rich interacting domain. In association with other proteins, ARID3a increased transcription of the immunoglobulin heavy chain and led to improved chromatin accessibility of the heavy chain enhancer. Constitutive expression of ARID3a in B lineage cells resulted in autoantibody production, suggesting its regulation is important. Abnormal ARID3a expression has also been associated with increased proliferative capacity and malignancy. Roles for ARID3a in addition to interactions with the immunoglobulin locus were suggested by transgenic and knockout mouse models. Over-expression of ARID3a resulted in skewing of mature B cell subsets and altered gene expression patterns of follicular B cells, whereas loss of function resulted in loss of B1 lineage B cells and defects in hematopoiesis. More recent studies showed that loss of ARID3a in adult somatic cells promoted developmental plasticity, alterations in gene expression patterns, and lineage fate decisions. Together, these data suggest new regulatory roles for ARID3a. The genes influenced by ARID3a are likely to play pivotal roles in lineage decisions, highlighting the importance of this understudied transcription factor.
Collapse
Affiliation(s)
- Michelle L Ratliff
- Immunobiology and Cancer Research, Oklahoma Medical Research Foundation , Oklahoma City, OK , USA
| | - Troy D Templeton
- Department of Cell Biology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| | - Julie M Ward
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| | - Carol F Webb
- Immunobiology and Cancer Research, Oklahoma Medical Research Foundation , Oklahoma City, OK , USA ; Department of Cell Biology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA ; Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| |
Collapse
|
42
|
Silent Burdens in Disease: Fatigue and Depression in SLE. Autoimmune Dis 2014; 2014:790724. [PMID: 24592329 PMCID: PMC3926392 DOI: 10.1155/2014/790724] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/07/2013] [Accepted: 11/13/2013] [Indexed: 11/24/2022] Open
Abstract
At a time when health is being recognized as more than just avoiding death, age and comorbidity are becoming increasingly important aspects of chronic disease. Systemic Lupus Erythematous (SLE) is probably one of the best paradigms of modern chronic disease, sitting at the crossroads of numerous somatic health problems, immune activation, depression, pain, and fatigue. One hundred forty-eight female participants were enrolled in the present study: 50 diagnosed with SLE, 45 with major depressive disorder (MDD), and 53 age-matched controls. Statistically significant lower scores in quality-of-life dimensions related to physical impairment were found in SLE. Patients with MDD presented significant levels of pain, reduced physical summary component (PSC), and general health scores different from healthy controls. Fatigue was reported in 90% of women with SLE and 77.8% of the MDD patients in contrast with 39.6% in the control group. Significant correlations were seen among fatigue severity, age, and educational level in SLE. From our own previous work and more recent work on the association of immune activation and depression, unexplained fatigue in SLE may signify an early sign of immune activation flare-up. The search for cytokine markers should perhaps be extended to fatigue in SLE.
Collapse
|