1
|
L’Estrange-Stranieri E, Gottschalk TA, Wright MD, Hibbs ML. The dualistic role of Lyn tyrosine kinase in immune cell signaling: implications for systemic lupus erythematosus. Front Immunol 2024; 15:1395427. [PMID: 39007135 PMCID: PMC11239442 DOI: 10.3389/fimmu.2024.1395427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Systemic lupus erythematosus (SLE, lupus) is a debilitating, multisystem autoimmune disease that can affect any organ in the body. The disease is characterized by circulating autoantibodies that accumulate in organs and tissues, which triggers an inflammatory response that can cause permanent damage leading to significant morbidity and mortality. Lyn, a member of the Src family of non-receptor protein tyrosine kinases, is highly implicated in SLE as remarkably both mice lacking Lyn or expressing a gain-of-function mutation in Lyn develop spontaneous lupus-like disease due to altered signaling in B lymphocytes and myeloid cells, suggesting its expression or activation state plays a critical role in maintaining tolerance. The past 30 years of research has begun to elucidate the role of Lyn in a duplicitous signaling network of activating and inhibitory immunoreceptors and related targets, including interactions with the interferon regulatory factor family in the toll-like receptor pathway. Gain-of-function mutations in Lyn have now been identified in human cases and like mouse models, cause severe systemic autoinflammation. Studies of Lyn in SLE patients have presented mixed findings, which may reflect the heterogeneity of disease processes in SLE, with impairment or enhancement in Lyn function affecting subsets of SLE patients that may be a means of stratification. In this review, we present an overview of the phosphorylation and protein-binding targets of Lyn in B lymphocytes and myeloid cells, highlighting the structural domains of the protein that are involved in its function, and provide an update on studies of Lyn in SLE patients.
Collapse
Affiliation(s)
- Elan L’Estrange-Stranieri
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Timothy A. Gottschalk
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Mark D. Wright
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Fiske BE, Wemlinger SM, Crute BW, Getahun A. The Src-family kinase Lyn plays a critical role in establishing and maintaining B cell anergy by suppressing PI3K-dependent signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595208. [PMID: 38826354 PMCID: PMC11142063 DOI: 10.1101/2024.05.21.595208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Although the Src family kinase (SFK) Lyn is known to be involved in induction and maintenance of peripheral B cell tolerance, the molecular basis of its action in this context remains unclear. This question has been approached using conventional as well as B cell-targeted knockouts of Lyn, with varied conclusions likely confused by collateral loss of Lyn functions in B cell and myeloid cell development and activation. Here we utilized a system in which Lyn gene deletion is tamoxifen inducible and B cell restricted. This system allows acute elimination of Lyn in B cells without off-target effects. This genetic tool was employed in conjunction with immunoglobulin transgenic mice in which peripheral B cells are autoreactive. DNA reactive Ars/A1 B cells require continuous inhibitory signaling, mediated by the inositol phosphatase SHIP-1 and the tyrosine phosphatase SHP-1, to maintain an unresponsive (anergic) state. Here we show that Ars/A1 B cells require Lyn to establish and maintain B cell unresponsiveness. Lyn primarily functions by restricting PI3K-dependent signaling pathways. This Lyn-dependent mechanism complements the impact of reduced mIgM BCR expression to restrict BCR signaling in Ars/A1 B cells. Our findings suggest that a subset of autoreactive B cells requires Lyn to become anergic and that the autoimmunity associated with dysregulated Lyn function may, in part, be due to an inability of these autoreactive B cells to become tolerized.
Collapse
|
3
|
Ottens K, Schneider J, Satterthwaite AB. B-1a Cells, but Not Marginal Zone B Cells, Are Implicated in the Accumulation of Autoreactive Plasma Cells in Lyn-/- Mice. Immunohorizons 2024; 8:47-56. [PMID: 38189742 PMCID: PMC10835670 DOI: 10.4049/immunohorizons.2300089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/22/2023] [Indexed: 01/09/2024] Open
Abstract
Mice deficient in Lyn, a tyrosine kinase that limits B cell activation, develop a lupus-like autoimmune disease characterized by the accumulation of splenic plasma cells and the production of autoantibodies. Lyn-/- mice have reduced numbers of marginal zone (MZ) B cells, a B cell subset that is enriched in autoreactivity and prone to plasma cell differentiation. We hypothesized that this is due to unchecked terminal differentiation of this potentially pathogenic B cell subpopulation. However, impairing MZ B cell development in Lyn-/- mice did not reduce plasma cell accumulation or autoantibodies, and preventing plasma cell differentiation did not restore MZ B cell numbers. Instead, Lyn-/- mice accumulated B-1a cells when plasma cell differentiation was impaired. Similar to MZ B cells, B-1a cells tend to be polyreactive or weakly autoreactive and are primed for terminal differentiation. Our results implicate B-1a cells, but not MZ B cells, as contributors to the autoreactive plasma cell pool in Lyn-/- mice.
Collapse
Affiliation(s)
- Kristina Ottens
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Jalyn Schneider
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Anne B. Satterthwaite
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX
| |
Collapse
|
4
|
Chu E, Mychasiuk R, Tsantikos E, Raftery AL, L’Estrange-Stranieri E, Dill LK, Semple BD, Hibbs ML. Regulation of Microglial Signaling by Lyn and SHIP-1 in the Steady-State Adult Mouse Brain. Cells 2023; 12:2378. [PMID: 37830592 PMCID: PMC10571795 DOI: 10.3390/cells12192378] [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: 09/06/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023] Open
Abstract
Chronic neuroinflammation and glial activation are associated with the development of many neurodegenerative diseases and neuropsychological disorders. Recent evidence suggests that the protein tyrosine kinase Lyn and the lipid phosphatase SH2 domain-containing inositol 5' phosphatase-1 (SHIP-1) regulate neuroimmunological responses, but their homeostatic roles remain unclear. The current study investigated the roles of Lyn and SHIP-1 in microglial responses in the steady-state adult mouse brain. Young adult Lyn-/- and SHIP-1-/- mice underwent a series of neurobehavior tests and postmortem brain analyses. The microglial phenotype and activation state were examined by immunofluorescence and flow cytometry, and neuroimmune responses were assessed using gene expression analysis. Lyn-/- mice had an unaltered behavioral phenotype, neuroimmune response, and microglial phenotype, while SHIP-1-/- mice demonstrated reduced explorative activity and exhibited microglia with elevated activation markers but reduced granularity. In addition, expression of several neuroinflammatory genes was increased in SHIP-1-/- mice. In response to LPS stimulation ex vivo, the microglia from both Lyn-/- and SHIP-1-/- showed evidence of hyper-activity with augmented TNF-α production. Together, these findings demonstrate that both Lyn and SHIP-1 have the propensity to control microglial responses, but only SHIP-1 regulates neuroinflammation and microglial activation in the steady-state adult brain, while Lyn activity appears dispensable for maintaining brain homeostasis.
Collapse
Affiliation(s)
- Erskine Chu
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (E.T.); (A.L.R.); (E.L.-S.)
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Evelyn Tsantikos
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (E.T.); (A.L.R.); (E.L.-S.)
| | - April L. Raftery
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (E.T.); (A.L.R.); (E.L.-S.)
| | - Elan L’Estrange-Stranieri
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (E.T.); (A.L.R.); (E.L.-S.)
| | - Larissa K. Dill
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Margaret L. Hibbs
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (E.T.); (A.L.R.); (E.L.-S.)
| |
Collapse
|
5
|
Brian BF, Sauer ML, Greene JT, Senevirathne SE, Lindstedt AJ, Funk OL, Ruis BL, Ramirez LA, Auger JL, Swanson WL, Nunez MG, Moriarity BS, Lowell CA, Binstadt BA, Freedman TS. A dominant function of LynB kinase in preventing autoimmunity. SCIENCE ADVANCES 2022; 8:eabj5227. [PMID: 35452291 PMCID: PMC9032976 DOI: 10.1126/sciadv.abj5227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
Here, we report that the LynB splice variant of the Src-family kinase Lyn exerts a dominant immunosuppressive function in vivo, whereas the LynA isoform is uniquely required to restrain autoimmunity in female mice. We used CRISPR-Cas9 gene editing to constrain lyn splicing and expression, generating single-isoform LynA knockout (LynAKO) or LynBKO mice. Autoimmune disease in total LynKO mice is characterized by production of antinuclear antibodies, glomerulonephritis, impaired B cell development, and overabundance of activated B cells and proinflammatory myeloid cells. Expression of LynA or LynB alone uncoupled the developmental phenotype from the autoimmune disease: B cell transitional populations were restored, but myeloid cells and differentiated B cells were dysregulated. These changes were isoform-specific, sexually dimorphic, and distinct from the complete LynKO. Despite the apparent differences in disease etiology and penetrance, loss of either LynA or LynB had the potential to induce severe autoimmune disease with parallels to human systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- Ben F. Brian
- Graduate Program in Molecular Pharmacology and Therapeutics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Monica L. Sauer
- Graduate Program in Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph T. Greene
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - S. Erandika Senevirathne
- Graduate Program in Molecular Pharmacology and Therapeutics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anders J. Lindstedt
- Graduate Program in Microbiology, Immunology, and Cancer Biology, University of Minnesota, Minneapolis, MN 55455, USA
- Medical Scientist Training Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Olivia L. Funk
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Brian L. Ruis
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Luis A. Ramirez
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jennifer L. Auger
- Department of Pediatrics, Division of Rheumatology, Allergy and Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Whitney L. Swanson
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Myra G. Nunez
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Branden S. Moriarity
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Clifford A. Lowell
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bryce A. Binstadt
- Department of Pediatrics, Division of Rheumatology, Allergy and Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tanya S. Freedman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Autoimmune Diseases Research, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
6
|
Getahun A. Role of inhibitory signaling in peripheral B cell tolerance*. Immunol Rev 2022; 307:27-42. [PMID: 35128676 PMCID: PMC8986582 DOI: 10.1111/imr.13070] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022]
Abstract
At least 20% of B cells in the periphery expresses an antigen receptor with a degree of self-reactivity. If activated, these autoreactive B cells pose a risk as they can contribute to the development of autoimmune diseases. To prevent their activation, both B cell-intrinsic and extrinsic tolerance mechanisms are in place in healthy individuals. In this review article, I will focus on B cell-intrinsic mechanisms that prevent the activation of autoreactive B cells in the periphery. I will discuss how inhibitory signaling circuits are established in autoreactive B cells, focusing on the Lyn-SHIP-1-SHP-1 axis, how they contribute to peripheral immune tolerance, and how disruptions of these circuits can contribute to the development of autoimmunity.
Collapse
Affiliation(s)
- Andrew Getahun
- Department of Immunology and Microbiology University of Colorado SOM Aurora Colorado USA
- Department of Immunology and Genomic Medicine National Jewish Health Denver Colorado USA
| |
Collapse
|
7
|
Hibbs ML, Raftery AL, Tsantikos E. Regulation of hematopoietic cell signaling by SHIP-1 inositol phosphatase: growth factors and beyond. Growth Factors 2018; 36:213-231. [PMID: 30764683 DOI: 10.1080/08977194.2019.1569649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SHIP-1 is a hematopoietic-specific inositol phosphatase activated downstream of a multitude of receptors including those for growth factors, cytokines, antigen, immunoglobulin and toll-like receptor agonists where it exerts inhibitory control. While it is constitutively expressed in all immune cells, SHIP-1 expression is negatively regulated by the inflammatory and oncogenic micro-RNA miR-155. Knockout mouse studies have shown the importance of SHIP-1 in various immune cell subsets and have revealed a range of immune-mediated pathologies that are engendered due to loss of SHIP-1's regulatory activity, impelling investigations into the role of SHIP-1 in human disease. In this review, we provide an overview of the literature relating to the role of SHIP-1 in hematopoietic cell signaling and function, we summarize recent reports that highlight the dysregulation of the SHIP-1 pathway in cancers, autoimmune disorders and inflammatory diseases, and lastly we discuss the importance of SHIP-1 in restraining myeloid growth factor signaling.
Collapse
Affiliation(s)
- Margaret L Hibbs
- a Department of Immunology and Pathology , Alfred Medical Research and Education Precinct Monash University , Melbourne , Victoria , Australia
| | - April L Raftery
- a Department of Immunology and Pathology , Alfred Medical Research and Education Precinct Monash University , Melbourne , Victoria , Australia
| | - Evelyn Tsantikos
- a Department of Immunology and Pathology , Alfred Medical Research and Education Precinct Monash University , Melbourne , Victoria , Australia
| |
Collapse
|
8
|
Brodie EJ, Infantino S, Low MSY, Tarlinton DM. Lyn, Lupus, and (B) Lymphocytes, a Lesson on the Critical Balance of Kinase Signaling in Immunity. Front Immunol 2018; 9:401. [PMID: 29545808 PMCID: PMC5837976 DOI: 10.3389/fimmu.2018.00401] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/13/2018] [Indexed: 01/23/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a progressive autoimmune disease characterized by increased sensitivity to self-antigens, auto-antibody production, and systemic inflammation. B cells have been implicated in disease progression and as such represent an attractive therapeutic target. Lyn is a Src family tyrosine kinase that plays a major role in regulating signaling pathways within B cells as well as other hematopoietic cells. Its role in initiating negative signaling cascades is especially critical as exemplified by Lyn-/- mice developing an SLE-like disease with plasma cell hyperplasia, underscoring the importance of tightly regulating signaling within B cells. This review highlights recent advances in our understanding of the function of the Src family tyrosine kinase Lyn in B lymphocytes and its contribution to positive and negative signaling pathways that are dysregulated in autoimmunity.
Collapse
Affiliation(s)
- Erica J. Brodie
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Simona Infantino
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Michael S. Y. Low
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Parkville, VIC, Australia
- Department of Haematology, Monash Health, Monash Hospital, Clayton, VIC, Australia
| | - David M. Tarlinton
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Abram CL, Lowell CA. Shp1 function in myeloid cells. J Leukoc Biol 2017; 102:657-675. [PMID: 28606940 DOI: 10.1189/jlb.2mr0317-105r] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/28/2023] Open
Abstract
The motheaten mouse was first described in 1975 as a model of systemic inflammation and autoimmunity, as a result of immune system dysregulation. The phenotype was later ascribed to mutations in the cytoplasmic tyrosine phosphatase Shp1. This phosphatase is expressed widely throughout the hematopoietic system and has been shown to impact a multitude of cell signaling pathways. The determination of which cell types contribute to the different aspects of the phenotype caused by global Shp1 loss or mutation and which pathways within these cell types are regulated by Shp1 is important to further our understanding of immune system regulation. In this review, we focus on the role of Shp1 in myeloid cells and how its dysregulation affects immune function, which can impact human disease.
Collapse
Affiliation(s)
- Clare L Abram
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| | - Clifford A Lowell
- Department of Laboratory Medicine and Immunology Program, University of California, San Francisco, California, USA
| |
Collapse
|
10
|
Huang R, Fang P, Hao Z, Kay BK. Directed Evolution of a Highly Specific FN3 Monobody to the SH3 Domain of Human Lyn Tyrosine Kinase. PLoS One 2016; 11:e0145872. [PMID: 26731115 PMCID: PMC4701441 DOI: 10.1371/journal.pone.0145872] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 12/09/2015] [Indexed: 11/26/2022] Open
Abstract
Affinity reagents of high affinity and specificity are very useful for studying the subcellular locations and quantities of individual proteins. To generate high-quality affinity reagents for human Lyn tyrosine kinase, a phage display library of fibronectin type III (FN3) monobodies was affinity selected with a recombinant form of the Lyn SH3 domain. While a highly specific monobody, TA8, was initially isolated, we chose to improve its affinity through directed evolution. A secondary library of 1.2 × 109 variants was constructed and screened by affinity selection, yielding three variants, two of which have affinities of ~ 40 nM, a 130-fold increase over the original TA8 monobody. One of the variants, 2H7, displayed high specificity to the Lyn SH3 domain, as shown by ELISA and probing arrays of 150 SH3 domains. Furthermore, the 2H7 monobody was able to pull down endogenous Lyn from a lysate of Burkitt's lymphoma cells, thereby demonstrating its utility as an affinity reagent for detecting Lyn in a complex biological mixture.
Collapse
Affiliation(s)
- Renhua Huang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail: (RH); (BK)
| | - Pete Fang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Zengping Hao
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Brian K. Kay
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail: (RH); (BK)
| |
Collapse
|
11
|
Gao R, Ma Z, Ma M, Yu J, Chen J, Li Z, Shetty S, Fu J. Deletion of Src family kinase Lyn aggravates endotoxin-induced lung inflammation. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1376-81. [DOI: 10.1152/ajplung.00219.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022] Open
Abstract
Overwhelming acute inflammation often leads to tissue damage during endotoxemia. In the present study, we investigated the role of Lyn, a member of the Src family tyrosine kinases, in modulating inflammatory responses in a murine model of endotoxemia. We examined lung inflammatory signaling in Lyn knockout (Lyn−/−) mice and wild-type littermates (Lyn+/+) during endotoxemia. Our data indicate that Lyn deletion aggravates endotoxin-induced pulmonary inflammation and proinflammatory signaling. We found increased activation of proinflammatory transcription factor NF-κB in the lung tissues of Lyn−/− mice after endotoxin challenge. Furthermore, during endotoxemia, the lung tissues of Lyn−/− mice showed increased inflammasome activation indicated by augmented caspase-1 and IL-1β cleavage and activation. The aggravated lung inflammatory signaling in Lyn−/− mice was associated with increased production of proinflammatory mediators and elevated matrix metallopeptidase 9 and reduced VE-cadherin levels. Our results suggest that Lyn kinase modulates inhibitory signaling to suppress endotoxin-induced lung inflammation.
Collapse
Affiliation(s)
- Rong Gao
- The Second Hospital of Jilin University, Changchun, China
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky
- Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky
| | - Zhongsen Ma
- The Second Hospital of Jilin University, Changchun, China
| | - Mengshi Ma
- The Second Hospital of Jilin University, Changchun, China
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky
| | - Jinyan Yu
- The Second Hospital of Jilin University, Changchun, China
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky
| | - Jiao Chen
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky
| | - Zhenyu Li
- Division of Cardiovascular Medicine, College of Medicine, University of Kentucky, Lexington, Kentucky; and
| | - Sreerama Shetty
- Center for Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Jian Fu
- Center for Research on Environmental Disease, University of Kentucky, Lexington, Kentucky
- Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
12
|
B cells biology in systemic lupus erythematosus—from bench to bedside. SCIENCE CHINA-LIFE SCIENCES 2015; 58:1111-25. [DOI: 10.1007/s11427-015-4953-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/09/2015] [Indexed: 12/20/2022]
|
13
|
Gottschalk TA, Tsantikos E, Hibbs ML. Pathogenic Inflammation and Its Therapeutic Targeting in Systemic Lupus Erythematosus. Front Immunol 2015; 6:550. [PMID: 26579125 PMCID: PMC4623412 DOI: 10.3389/fimmu.2015.00550] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/14/2015] [Indexed: 12/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE, lupus) is a highly complex and heterogeneous autoimmune disease that most often afflicts women in their child-bearing years. It is characterized by circulating self-reactive antibodies that deposit in tissues, including skin, kidneys, and brain, and the ensuing inflammatory response can lead to irreparable tissue damage. Over many years, clinical trials in SLE have focused on agents that control B- and T-lymphocyte activation, and, with the single exception of an agent known as belimumab which targets the B-cell survival factor BAFF, they have been disappointing. At present, standard therapy for SLE with mild disease is the agent hydroxychloroquine. During disease flares, steroids are often used, while the more severe manifestations with major organ involvement warrant potent, broad-spectrum immunosuppression with cyclophosphamide or mycophenolate. Current treatments have severe and dose-limiting toxicities and thus a more specific therapy targeting a causative factor or signaling pathway would be greatly beneficial in SLE treatment. Moreover, the ability to control inflammation alongside B-cell activation may be a superior approach for disease control. There has been a recent focus on the innate immune system and associated inflammation, which has uncovered key players in driving the pathogenesis of SLE. Delineating some of these intricate inflammatory mechanisms has been possible with studies using spontaneous mouse mutants and genetically engineered mice. These strains, to varying degrees, exhibit hallmarks of the human disease and therefore have been utilized to model human SLE and to test new drugs. Developing a better understanding of the initiation and perpetuation of disease in SLE may uncover suitable novel targets for therapeutic intervention. Here, we discuss the involvement of inflammation in SLE disease pathogenesis, with a focus on several key proinflammatory cytokines and myeloid growth factors, and review the known outcomes or the potential for targeting these factors in SLE.
Collapse
Affiliation(s)
- Timothy A Gottschalk
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University , Melbourne, VIC , Australia
| | - Evelyn Tsantikos
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University , Melbourne, VIC , Australia
| | - Margaret L Hibbs
- Leukocyte Signalling Laboratory, Department of Immunology and Pathology, Alfred Medical Research and Education Precinct, Monash University , Melbourne, VIC , Australia
| |
Collapse
|
14
|
BAFF-driven autoimmunity requires CD19 expression. J Autoimmun 2015; 62:1-10. [DOI: 10.1016/j.jaut.2015.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/29/2015] [Accepted: 06/01/2015] [Indexed: 11/19/2022]
|
15
|
Mayeux J, Skaug B, Luo W, Russell LM, John S, Saelee P, Abbasi H, Li QZ, Garrett-Sinha LA, Satterthwaite AB. Genetic Interaction between Lyn, Ets1, and Btk in the Control of Antibody Levels. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26209625 DOI: 10.4049/jimmunol.1500165] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tight control of B cell differentiation into plasma cells (PCs) is critical for proper immune responses and the prevention of autoimmunity. The Ets1 transcription factor acts in B cells to prevent PC differentiation. Ets1(-/-) mice accumulate PCs and produce autoantibodies. Ets1 expression is downregulated upon B cell activation through the BCR and TLRs and is maintained by the inhibitory signaling pathway mediated by Lyn, CD22 and SiglecG, and SHP-1. In the absence of these inhibitory components, Ets1 levels are reduced in B cells in a Btk-dependent manner. This leads to increased PCs, autoantibodies, and an autoimmune phenotype similar to that of Ets1(-/-) mice. Defects in inhibitory signaling molecules, including Lyn and Ets1, are associated with human lupus, although the effects are more subtle than the complete deficiency that occurs in knockout mice. In this study, we explore the effect of partial disruption of the Lyn/Ets1 pathway on B cell tolerance and find that Lyn(+/-)Ets1(+/-) mice demonstrate greater and earlier production of IgM, but not IgG, autoantibodies compared with Lyn(+/-) or Ets1(+/-) mice. We also show that Btk-dependent downregulation of Ets1 is important for normal PC homeostasis when inhibitory signaling is intact. Ets1 deficiency restores the decrease in steady state PCs and Ab levels observed in Btk(-/-) mice. Thus, depending on the balance of activating and inhibitory signals to Ets1, there is a continuum of effects on autoantibody production and PC maintenance. This ranges from full-blown autoimmunity with complete loss of Ets1-maintaining signals to reduced PC and Ab levels with impaired Ets1 downregulation.
Collapse
Affiliation(s)
- Jessica Mayeux
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Brian Skaug
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Wei Luo
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Lisa M Russell
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Shinu John
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Prontip Saelee
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Hansaa Abbasi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lee Ann Garrett-Sinha
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Anne B Satterthwaite
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
16
|
Tsantikos E, Gottschalk TA, Maxwell MJ, Hibbs ML. Role of the Lyn tyrosine kinase in the development of autoimmune disease. ACTA ACUST UNITED AC 2014. [DOI: 10.2217/ijr.14.44] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
17
|
Colombo E, Tentorio P, Musio S, Rajewsky K, Pedotti R, Casola S, Farina C. Skewed B cell differentiation affects lymphoid organogenesis but not T cell-mediated autoimmunity. Clin Exp Immunol 2014; 176:58-65. [PMID: 24325711 DOI: 10.1111/cei.12250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2013] [Indexed: 01/23/2023] Open
Abstract
B cell receptor (BCR) signalling determines B cell differentiation and may potentially alter T cell-mediated immune responses. In this study we used two transgenic strains of BCR-deficient mice expressing Epstein-Barr virus latent membrane protein (LMP)2A in B cells, where either follicular and marginal zone differentiation (D(H)LMP2A mice) or B-1 cell development (V(H)LMP2A mice) were supported, and evaluated the effects of skewed B lymphocyte differentiation on lymphoid organogenesis and T cell responses in vivo. Compared to wild-type animals, both transgenic strains displayed alterations in the composition of lymphoid organs and in the dynamics of distinct immune cell subsets following immunization with the self-antigen PLP₁₈₅₋₂₀₆. However, ex-vivo T cell proliferation to PLP₁₈₅₋₂₀₆ peptide measured in immunized D(H)LMP2A and V(H)LMP2A mice was similar to that detected in immunized control mice. Further, clinical expression of experimental autoimmune encephalitis in both LMP2A strains was identical to that of wild-type mice. In conclusion, mice with skewed B cell differentiation driven by LMP2A expression in BCR-negative B cells do not show changes in the development of a T cell mediated disease model of autoimmunity, suggesting that compensatory mechanisms support the generation of T cell responses.
Collapse
Affiliation(s)
- E Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy; Neuroimmunology and Neuromuscular Disorders Unit, Foundation IRCCS-Neurological Institute Carlo Besta, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
18
|
Liossis SNC, Melissaropoulos K. Molecular abnormalities of the B cell in systemic lupus erythematosus are candidates for functional inhibition treatments. Expert Opin Pharmacother 2014; 15:833-40. [PMID: 24588739 DOI: 10.1517/14656566.2014.894976] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION The B cell is a key player in the pathogenesis of systemic lupus erythematosus (SLE). Loss of B cell tolerance resulting in autoantibody production and immune complex formation and deposition are central features of the disease. B cell overactivity is a hallmark of SLE and molecular abnormalities in B cell signaling cascade have been described. AREAS COVERED In this review, we will focus on the aberrant phenotype of B cell signaling in patients with lupus. We will also discuss data stemming from the use of small molecules that have recently been recognized to target important steps of the B cell signal transduction pathways with therapeutic implications for SLE. EXPERT OPINION Attempts to target the B cell in SLE have been made through depletion, blocking of survival factors and co-receptor inhibition. However, the still unmet need for effective therapy of refractory disease makes the necessity for new drugs impelling.
Collapse
Affiliation(s)
- Stamatis-Nick C Liossis
- Associate Professor of Medicine/Rheumatology, University of Patras Medical School, Patras University Hospital, Division of Rheumatology, Department of Internal Medicine , GR 26504, Patras , Greece +30 2613 603 693 ; +30 2610 993 982 ;
| | | |
Collapse
|
19
|
Hua Z, Gross AJ, Lamagna C, Ramos-Hernández N, Scapini P, Ji M, Shao H, Lowell CA, Hou B, DeFranco AL. Requirement for MyD88 signaling in B cells and dendritic cells for germinal center anti-nuclear antibody production in Lyn-deficient mice. THE JOURNAL OF IMMUNOLOGY 2013; 192:875-85. [PMID: 24379120 DOI: 10.4049/jimmunol.1300683] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intracellular tyrosine kinase Lyn mediates inhibitory receptor function in B cells and myeloid cells, and Lyn(-/-) mice spontaneously develop an autoimmune and inflammatory disease that closely resembles human systemic lupus erythematosus. TLR-signaling pathways have been implicated in the production of anti-nuclear Abs in systemic lupus erythematosus and mouse models of it. We used a conditional allele of Myd88 to determine whether the autoimmunity of Lyn(-/-) mice is dependent on TLR/MyD88 signaling in B cells and/or in dendritic cells (DCs). The production of IgG anti-nuclear Abs, as well as the deposition of these Abs in the glomeruli of the kidneys, leading to glomerulonephritis in Lyn(-/-) mice, were completely abolished by selective deletion of Myd88 in B cells, and autoantibody production and glomerulonephritis were delayed or decreased by deletion of Myd88 in DCs. The reduced autoantibody production in mice lacking MyD88 in B cells or DCs was accompanied by a dramatic decrease in the spontaneous germinal center (GC) response, suggesting that autoantibodies in Lyn(-/-) mice may depend on GC responses. Consistent with this view, IgG anti-nuclear Abs were absent if T cells were deleted (TCRβ(-/-) TCRδ(-/-) mice) or if T cells were unable to contribute to GC responses as the result of mutation of the adaptor molecule SAP. Thus, the autoimmunity of Lyn(-/-) mice was dependent on T cells and on TLR/MyD88 signaling in B cells and in DCs, supporting a model in which DC hyperactivity combines with defects in tolerance in B cells to lead to a T cell-dependent systemic autoimmunity in Lyn(-/-) mice.
Collapse
Affiliation(s)
- Zhaolin Hua
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zikherman J, Parameswaran R, Hermiston M, Weiss A. The structural wedge domain of the receptor-like tyrosine phosphatase CD45 enforces B cell tolerance by regulating substrate specificity. THE JOURNAL OF IMMUNOLOGY 2013; 190:2527-35. [PMID: 23396948 DOI: 10.4049/jimmunol.1202928] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
CD45 is a receptor-like tyrosine phosphatase that positively regulates BCR signaling by dephosphorylating the inhibitory tyrosine of the Src family kinases. We showed previously that a single point mutation, E613R, introduced into the cytoplasmic membrane-proximal "wedge" domain of CD45 is sufficient to drive a lupus-like autoimmune disease on a susceptible genetic background. To clarify the molecular mechanism of this disease, we took advantage of a unique allelic series of mice in which the expression of CD45 is varied across a broad range. Although both E613R B cells and those with supraphysiologic CD45 expression exhibited hyperresponsive BCR signaling, they did so by opposite regulation of the Src family kinase Lyn. We demonstrated that the E613R allele of CD45 does not function as a hyper- or hypomorphic allele but rather alters the substrate specificity of CD45 for Lyn. Despite similarly enhancing BCR signaling, only B cells with supraphysiologic CD45 expression became anergic, whereas only mice harboring the E613R mutation developed frank autoimmunity on a susceptible genetic background. We showed that selective impairment of a Lyn-dependent negative-regulatory circuit in E613R B cells drove autoimmunity in E613R mice. This demonstrates that relaxing negative regulation of BCR signaling, rather than enhancing positive regulation, is critical for driving autoimmunity in this system.
Collapse
Affiliation(s)
- Julie Zikherman
- Division of Rheumatology, Department of Medicine, Rosalind Russell Medical Research Center for Arthritis, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | |
Collapse
|
21
|
Lau M, Tsantikos E, Maxwell MJ, Tarlinton DM, Anderson GP, Hibbs ML. Loss of STAT6 promotes autoimmune disease and atopy on a susceptible genetic background. J Autoimmun 2012; 39:388-97. [PMID: 22867713 DOI: 10.1016/j.jaut.2012.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 06/22/2012] [Accepted: 06/27/2012] [Indexed: 01/22/2023]
Abstract
Atopy and autoimmunity are usually considered opposed immunological manifestations. Lyn(-/-) mice develop lupus-like autoimmune disease yet have coexistent intrinsic allergic traits and are prone to severe, persistent asthma induced exogenously. Recently it has been proposed that the Th2 environment and IgE auto-Abs promotes autoimmune disease in Lyn(-/-) mice. To examine this apparent contradiction, we derived Lyn(-/-) mice with a null mutation in STAT6, a regulator of Th2 immunity that integrates signaling from the IL-4/IL-13 receptor complex. Atopy and spontaneous peritoneal eosinophilia, characteristic of Lyn(-/-) mice, were lost in young Lyn(-/-)STAT6(-/-) mice; however, autoimmune disease was markedly exacerbated. At a time-point where Lyn(-/-) mice showed only mild autoimmune disease, Lyn(-/-)STAT6(-/-) mice had maximal titres of IgG and IgA auto-Abs, impaired renal function, myeloid expansion and a highly activated T cell compartment. Remarkably, low level IgE auto-Abs but not IgG1 auto-Abs were a feature of some aged Lyn(-/-)STAT6(-/-) mice. Furthermore, aged Lyn(-/-)STAT6(-/-) mice showed dramatically increased levels of serum IgE but minimal IgG1, suggesting that class-switching to IgE can occur in the absence of an IgG1 intermediate. The results show that Lyn-deficient mice can overcome the effects of disabling Th2 immunity, highlighting the importance of Lyn in controlling Th2 responses. Our data also indicates that, under certain conditions, STAT6-independent factors can promote IgE class-switching. This work has important clinical implications as many experimental therapies designed for the treatment of asthma or atopy are based on targeting the STAT6 axis, which could potentially reveal life endangering autoimmunity or promote atopy in susceptible individuals.
Collapse
Affiliation(s)
- Maverick Lau
- Leukocyte Signaling Laboratory, Department of Immunology, Monash University, Central Clinical School, Alfred Medical Research and Education Precinct, Commercial Road, Melbourne, Victoria 3004, Australia.
| | | | | | | | | | | |
Collapse
|