0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Characterization of cxorf21 Provides Molecular Insight Into Female-Bias Immune Response in SLE Pathogenesis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background: Ninety percent of systemic lupus erythematosus (SLE) patients are women. X chromosome-dosage increases susceptibility to SLE and primary Sjögren's syndrome (pSS). Chromosome X open reading frame 21 (CXorf21) escapes X-inactivation and is an SLE risk gene of previously unknown function. We undertook the present study to delineate the function of CXorf21 in the immune system as well as investigate a potential role in the sex bias of SLE and pSS.

          Methods: Western blot protein analysis, qPCR, BioPlex cytokine immunoassay, pHrodo™ assays, as well as in vitro CRISPR-Cas9 knockdown experiments were employed to delineate the role of CXorf21 in relevant immunocytes.

          Results: Expressed in monocytes and B cells, CXorf21 basal Mrna, and protein expression levels are elevated in female primary monocytes, B cells, and EBV-transformed B cells compared to male cells. We also found CXorf21 mRNA and protein expression is higher in both male and female cells from SLE patients compared to control subjects. TLR7 ligation increased CXorf21 protein expression and CXorf21 knockdown abrogated TLR7-driven increased IFNA1 mRNA expression, and reduced secretion of both TNF-alpha and IL-6 in healthy female monocytes. Similarly, we found increased pH in the lysosomes of CXorf21-deficient female monocytes.

          Conclusion: CXorf21 is more highly expressed in female compared to male cells and is involved in a sexually dimorphic response to TLR7 activation. In addition, CXorf21 expression regulates lysosomal pH in a sexually dimorphic manner. Thus, sexually dimorphic expression of CXorf21 skews cellular immune responses in manner consistent with expected properties of a mediator of the X chromosome dose risk in SLE and pSS.

          Related collections

          Most cited references37

          • Record: found
          • Abstract: found
          • Article: not found

          TRANSFAC: transcriptional regulation, from patterns to profiles.

          The TRANSFAC database on eukaryotic transcriptional regulation, comprising data on transcription factors, their target genes and regulatory binding sites, has been extended and further developed, both in number of entries and in the scope and structure of the collected data. Structured fields for expression patterns have been introduced for transcription factors from human and mouse, using the CYTOMER database on anatomical structures and developmental stages. The functionality of Match, a tool for matrix-based search of transcription factor binding sites, has been enhanced. For instance, the program now comes along with a number of tissue-(or state-)specific profiles and new profiles can be created and modified with Match Profiler. The GENE table was extended and gained in importance, containing amongst others links to LocusLink, RefSeq and OMIM now. Further, (direct) links between factor and target gene on one hand and between gene and encoded factor on the other hand were introduced. The TRANSFAC public release is available at http://www.gene-regulation.com. For yeast an additional release including the latest data was made available separately as TRANSFAC Saccharomyces Module (TSM) at http://transfac.gbf.de. For CYTOMER free download versions are available at http://www.biobase.de:8080/index.html.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus.

            Systemic lupus erythematosus (SLE) is a complex, inflammatory autoimmune disease that affects multiple organ systems. We used global gene expression profiling of peripheral blood mononuclear cells to identify distinct patterns of gene expression that distinguish most SLE patients from healthy controls. Strikingly, about half of the patients studied showed dysregulated expression of genes in the IFN pathway. Furthermore, this IFN gene expression "signature" served as a marker for more severe disease involving the kidneys, hematopoetic cells, and/or the central nervous system. These results provide insights into the genetic pathways underlying SLE, and identify a subgroup of patients who may benefit from therapies targeting the IFN pathway.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              CRISPR interference (CRISPRi) for sequence-specific control of gene expression.

              Sequence-specific control of gene expression on a genome-wide scale is an important approach for understanding gene functions and for engineering genetic regulatory systems. We have recently described an RNA-based method, CRISPR interference (CRISPRi), for targeted silencing of transcription in bacteria and human cells. The CRISPRi system is derived from the Streptococcus pyogenes CRISPR (clustered regularly interspaced palindromic repeats) pathway, requiring only the coexpression of a catalytically inactive Cas9 protein and a customizable single guide RNA (sgRNA). The Cas9-sgRNA complex binds to DNA elements complementary to the sgRNA and causes a steric block that halts transcript elongation by RNA polymerase, resulting in the repression of the target gene. Here we provide a protocol for the design, construction and expression of customized sgRNAs for transcriptional repression of any gene of interest. We also provide details for testing the repression activity of CRISPRi using quantitative fluorescence assays and native elongating transcript sequencing. CRISPRi provides a simplified approach for rapid gene repression within 1-2 weeks. The method can also be adapted for high-throughput interrogation of genome-wide gene functions and genetic interactions, thus providing a complementary approach to RNA interference, which can be used in a wider variety of organisms.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                21 October 2019
                2019
                : 10
                : 2160
                Affiliations
                [1] 1Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation , Oklahoma City, OK, United States
                [2] 2Departments of Pathology and Medicine, College of Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, OK, United States
                [3] 3Division of Rheumatology, School of Medicine, University of Colorado , Aurora, CO, United States
                [4] 4Department of Immunology and Microbiology, School of Medicine, University of Colorado , Aurora, CO, United States
                [5] 5Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, United States
                [6] 6Department of Pediatrics, University of Cincinnati College of Medicine , Cincinnati, OH, United States
                [7] 7United States Department of Veterans Affairs Medical Center , Cincinnati, OH, United States
                [8] 8Medical and Research Services, Oklahoma City Department of Veterans Affairs Health Care Center , Oklahoma City, OK, United States
                Author notes

                Edited by: Amr Sawalha, University of Pittsburgh, United States

                Reviewed by: Qianjin Lu, Second Xiangya Hospital, Central South University, China; J. Michelle Kahlenberg, University of Michigan, United States

                *Correspondence: R. Hal Scofield hal-scofield@ 123456omrf.ouhsc.edu

                This article was submitted to Autoimmune and Autoinflammatory Disorders, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2019.02160
                6816314
                31695690
                e298fc4b-ab7c-490f-b635-4b5399f670d1
                Copyright © 2019 Harris, Koelsch, Kurien, Harley, Wren, Harley and Scofield.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 27 June 2019
                : 28 August 2019
                Page count
                Figures: 6, Tables: 1, Equations: 0, References: 58, Pages: 11, Words: 7504
                Funding
                Funded by: Lupus Research Institute 10.13039/100001218
                Funded by: U.S. Department of Veterans Affairs 10.13039/100000738
                Award ID: BX001451
                Award ID: BX001834
                Funded by: National Institute of Arthritis and Musculoskeletal and Skin Diseases 10.13039/100000069
                Award ID: AR007534-32
                Funded by: NIH Office of the Director 10.13039/100000052
                Award ID: AI082714
                Award ID: AI130830
                Award ID: AR053483
                Award ID: AR053734
                Award ID: AR070549
                Award ID: GM104938
                Award ID: HG00866
                Categories
                Immunology
                Original Research

                Immunology
                sex-bias,systemic lupus erythematosus,innate immunity,x-chromosome,interferon-inducible
                Immunology
                sex-bias, systemic lupus erythematosus, innate immunity, x-chromosome, interferon-inducible

                Comments

                Comment on this article