1
|
Xipell M, Lledó GM, Egan AC, Tamirou F, Del Castillo CS, Rovira J, Gómez-Puerta JA, García-Herrera A, Cervera R, Kronbichler A, Jayne DRW, Anders HJ, Houssiau F, Espinosa G, Quintana LF. From systemic lupus erythematosus to lupus nephritis: The evolving road to targeted therapies. Autoimmun Rev 2023; 22:103404. [PMID: 37543287 DOI: 10.1016/j.autrev.2023.103404] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
Systemic lupus erythematosus is a chronic autoimmune disease characterized by loss of tolerance against nuclear and cytoplasmic self-antigens, induction of immunity and tissue inflammation. Lupus nephritis (LN), the most important predictor of morbidity in SLE, develops in almost 30% of SLE patients at disease onset and in up to 50-60% within the first 10 years. Firstly, in this review, we put the pathogenic mechanisms of the disease into a conceptual frame, giving emphasis to the role of the innate immune system in this loss of self-tolerance and the induction of the adaptive immune response. In this aspect, many mechanisms have been described such as dysregulation and acceleration of cell-death pathways, an aberrant clearance and overload of immunogenic acid-nucleic-containing debris and IC, and the involvement of antigen-presenting cells and other innate immune cells in the induction of this adaptive immune response. This result in a clonal expansion of autoreactive lymphocytes with generation of effector T-cells, memory B-cells and plasma cells that produce autoantibodies that will cause kidney damage. Secondly, we review the immunological pathways of damage in the kidney parenchyma, initiated by autoantibody binding and immune complex deposition, and followed by complement-mediated microvascular injury, activation of kidney stromal cells and the recruitment of leukocytes. Finally, we summarize the rationale for the treatment of LN, from conventional to new targeted therapies, focusing on their systemic immunologic effects and the minimization of podocytary damage.
Collapse
Affiliation(s)
- Marc Xipell
- Department of Nephrology and Renal Transplantation, Clinic Barcelona, Spain; Reference Center for Complex Glomerular Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gema M Lledó
- Department of Autoimmune Diseases, Clínic Barcelona, Spain; Reference Center for Systemic Autoimmune Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Allyson C Egan
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, United Kingdom
| | - Farah Tamirou
- Rheumatology Department, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium; Pôle de Pathologies Rhumatismales Inflammatoires et Systémiques, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Belgium
| | | | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - José A Gómez-Puerta
- Department of Rheumatology, Clínic Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Catalonia, Spain
| | - Adriana García-Herrera
- Department of Pathology, Clínic Barcelona, Spain; Reference Center for Complex Glomerular Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ricard Cervera
- Department of Autoimmune Diseases, Clínic Barcelona, Spain
| | - Andreas Kronbichler
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - David R W Jayne
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Frédéric Houssiau
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, United Kingdom
| | - Gerard Espinosa
- Department of Autoimmune Diseases, Clínic Barcelona, Spain; Reference Center for Systemic Autoimmune Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.
| | - Luis F Quintana
- Department of Nephrology and Renal Transplantation, Clinic Barcelona, Spain; Reference Center for Complex Glomerular Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
2
|
Olson LB, Hunter NI, Rempel RE, Sullenger BA. Targeting DAMPs with nucleic acid scavengers to treat lupus. Transl Res 2022; 245:30-40. [PMID: 35245691 PMCID: PMC9167234 DOI: 10.1016/j.trsl.2022.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic and often progressive autoimmune disorder marked clinically by a variable constellation of symptoms including fatigue, rash, joint pains, and kidney damage. The lungs, heart, gastrointestinal system, and brain can also be impacted, and individuals with lupus are at higher risk for atherosclerosis, thrombosis, thyroid disease, and other disorders associated with chronic inflammation . Autoimmune diseases are marked by erroneous immune responses in which the target of the immune response is a "self"-antigen, or autoantigen, driven by the development of antigen-specific B or T cells that have overcome the normal systems of self-tolerance built into the development of B and T cells. SLE is specifically characterized by the production of autoantibodies against nucleic acids and their binding proteins, including anti-double stranded DNA, anti-Smith (an RNA binding protein), and many others . These antibodies bind their nuclear-derived antigens to form immune complexes that cause injury and scarring through direct deposition in tissues and activation of innate immune cells . In over 50% of SLE patients, immune complex aggregation in the kidneys drives intrarenal inflammation and injury and leads to lupus nephritis, a progressive destruction of the glomeruli that is one of the most common causes of lupus-related death . To counter this pathology increasing attention has turned to developing approaches to reduce the development and continued generation of such autoantibodies. In particular, the molecular and cellular events that lead to long term, continuous activation of such autoimmune responses have become the focus of new therapeutic strategies to limit renal and other pathologies in lupus patients. The focus of this review is to consider how the innate immune system is involved in the development and progression of lupus nephritis and how a novel approach to inhibit innate immune activation by neutralizing the activators of this response, called Damage Associated Molecular Patterns, may represent a promising approach to treat this and other autoimmune disorders.
Collapse
Affiliation(s)
- Lyra B Olson
- Department of Surgery, Duke University, Durham, North Carolina; Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Nicole I Hunter
- Department of Surgery, Duke University, Durham, North Carolina; Department of Chemistry, Duke University, Durham, North Carolina
| | - Rachel E Rempel
- Department of Surgery, Duke University, Durham, North Carolina
| | - Bruce A Sullenger
- Department of Surgery, Duke University, Durham, North Carolina; Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina; Department of Biomedical Engineering, Duke University, Durham, North Carolina.
| |
Collapse
|
3
|
Saito Y, Miyajima M, Yamamoto S, Sato T, Miura N, Fujimiya M, Chikenji TS. Accumulation of Senescent Neural Cells in Murine Lupus With Depression-Like Behavior. Front Immunol 2021; 12:692321. [PMID: 34804003 PMCID: PMC8597709 DOI: 10.3389/fimmu.2021.692321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/19/2021] [Indexed: 11/18/2022] Open
Abstract
Neuropsychiatric manifestations targeting the central, peripheral, and autonomic nervous system are common in systemic lupus erythematosus (SLE); collectively, these symptoms are termed neuropsychiatric SLE (NPSLE). Among a wide variety of neuropsychiatric symptoms, depression is observed in about 24-39% of SLE patients. Several cytokines and chemokines have been identified as biomarkers or therapeutic targets of NPSLE; in particular, the levels of type 1 interferons, TNFs, and IL-6 are elevated in SLE patient's cerebrospinal fluid (CSF), and these factors contribute to the pathology of depression. Here, we show that senescent neural cells accumulate in the hippocampal cornu ammonis 3 (CA3) region in MRL/lpr SLE model mice with depressive behavior. Furthermore, oral administration of fisetin, a senolytic drug, reduced the number of senescent neural cells and reduced depressive behavior in the MRL/lpr mice. In addition, transcription of several senescence and senescence-associated secretory phenotype (SASP) factors in the hippocampal region also decreased after fisetin treatment in the MRL/lpr mice. These results indicate that the accumulation of senescent neural cells in the hippocampus plays a role in NPSLE pathogenesis, and therapies targeting senescent cells may represent a candidate approach to treat NPSLE.
Collapse
Affiliation(s)
- Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Health Sciences, School of medicine, Hokkaido University, Sapporo, Japan
| | - Maki Miyajima
- Department of Health Sciences, School of medicine, Hokkaido University, Sapporo, Japan
| | - Sena Yamamoto
- Department of Health Sciences, School of medicine, Hokkaido University, Sapporo, Japan
| | - Tsukasa Sato
- Department of Health Sciences, School of medicine, Hokkaido University, Sapporo, Japan
| | - Norihiro Miura
- Department of Health Sciences, School of medicine, Hokkaido University, Sapporo, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako S Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Health Sciences, School of medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
4
|
Wilson NR, Bover L, Konopleva M, Han L, Neelapu S, Pemmaraju N. CD303 (BDCA-2) - a potential novel target for therapy in hematologic malignancies. Leuk Lymphoma 2021; 63:19-30. [PMID: 34486917 DOI: 10.1080/10428194.2021.1975192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) serve as immunoregulatory antigen-presenting cells that play a role in various inflammatory, viral, and malignant conditions. Malignant proliferation of pDCs is implicated in the pathogenesis of certain hematologic cancers, specifically blastic plasmacytoid dendritic cell neoplasm (BPDCN) and acute myelogenous leukemia with clonal expansion of pDC (pDC-AML). In recent years, BPDCN and pDC-AML have been successfully treated with targeted therapy of pDC-specific surface marker, CD123. However, relapsed and refractory BPDCN remains an elusive cancer, with limited therapeutic options. CD303 is another specific surface marker of human pDCs, centrally involved in antigen presentation and immune tolerance. Monoclonal antibodies directed against CD303 have been studied in preclinical models and have achieved disease control in patients with cutaneous lupus erythematosus. We performed a comprehensive review of benign and malignant disorders in which CD303 have been studied, as there may be a potential future CD303-directed therapy for many of these conditions.
Collapse
Affiliation(s)
- Nathaniel R Wilson
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Laura Bover
- Departments of Genomic Medicine and Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Lina Han
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Sattva Neelapu
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
5
|
Ukadike KC, Mustelin T. Implications of Endogenous Retroelements in the Etiopathogenesis of Systemic Lupus Erythematosus. J Clin Med 2021; 10:856. [PMID: 33669709 PMCID: PMC7922054 DOI: 10.3390/jcm10040856] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/09/2021] [Accepted: 02/13/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease. While its etiology remains elusive, current understanding suggests a multifactorial process with contributions by genetic, immunologic, hormonal, and environmental factors. A hypothesis that combines several of these factors proposes that genomic elements, the L1 retrotransposons, are instrumental in SLE pathogenesis. L1 retroelements are transcriptionally activated in SLE and produce two proteins, ORF1p and ORF2p, which are immunogenic and can drive type I interferon (IFN) production by producing DNA species that activate cytosolic DNA sensors. In addition, these two proteins reside in RNA-rich macromolecular assemblies that also contain well-known SLE autoantigens like Ro60. We surmise that cells expressing L1 will exhibit all the hallmarks of cells infected by a virus, resulting in a cellular and humoral immune response similar to those in chronic viral infections. However, unlike exogenous viruses, L1 retroelements cannot be eliminated from the host genome. Hence, dysregulated L1 will cause a chronic, but perhaps episodic, challenge for the immune system. The clinical and immunological features of SLE can be at least partly explained by this model. Here we review the support for, and the gaps in, this hypothesis of SLE and its potential for new diagnostic, prognostic, and therapeutic options in SLE.
Collapse
Affiliation(s)
| | - Tomas Mustelin
- Division of Rheumatology, Department of Medicine, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA;
| |
Collapse
|
6
|
Mustelin T, Ukadike KC. How Retroviruses and Retrotransposons in Our Genome May Contribute to Autoimmunity in Rheumatological Conditions. Front Immunol 2020; 11:593891. [PMID: 33281822 PMCID: PMC7691656 DOI: 10.3389/fimmu.2020.593891] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
More than 200 human disorders include various manifestations of autoimmunity. The molecular events that lead to these diseases are still incompletely understood and their causes remain largely unknown. Numerous potential triggers of autoimmunity have been proposed over the years, but very few of them have been conclusively confirmed or firmly refuted. Viruses have topped the lists of suspects for decades, and it seems that many viruses, including those of the Herpesviridae family, indeed can influence disease initiation and/or promote exacerbations by a number of mechanisms that include prolonged anti-viral immunity, immune subverting factors, and mechanisms, and perhaps “molecular mimicry”. However, no specific virus has yet been established as being truly causative. Here, we discuss a different, but perhaps mechanistically related possibility, namely that retrotransposons or retroviruses that infected us in the past and left a lasting copy of themselves in our genome still can provoke an escalating immune response that leads to autoimmune disease. Many of these loci still encode for retroviral proteins that have retained some, or all, of their original functions. Importantly, these endogenous proviruses cannot be eliminated by the immune system the way it can eliminate exogenous viruses. Hence, if not properly controlled, they may drive a frustrated and escalating chronic, or episodic, immune response to the point of a frank autoimmune disorder. Here, we discuss the evidence and the proposed mechanisms, and assess the therapeutic options that emerge from the current understanding of this field.
Collapse
Affiliation(s)
- Tomas Mustelin
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Kennedy C Ukadike
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
7
|
Hayden L, Semenoff T, Schultz V, Merz SF, Chapple KJ, Rodriguez M, Warrington AE, Shi X, McKimmie CS, Edgar JM, Thümmler K, Linington C, Pingen M. Lipid-specific IgMs induce antiviral responses in the CNS: implications for progressive multifocal leukoencephalopathy in multiple sclerosis. Acta Neuropathol Commun 2020; 8:135. [PMID: 32792006 PMCID: PMC7427287 DOI: 10.1186/s40478-020-01011-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/01/2020] [Indexed: 01/07/2023] Open
Abstract
Progressive multi-focal leukoencephalopathy (PML) is a potentially fatal encephalitis caused by JC polyomavirus (JCV). PML principally affects people with a compromised immune system, such as patients with multiple sclerosis (MS) receiving treatment with natalizumab. However, intrathecal synthesis of lipid-reactive IgM in MS patients is associated with a markedly lower incidence of natalizumab-associated PML compared to those without this antibody repertoire. Here we demonstrate that a subset of lipid-reactive human and murine IgMs induce a functional anti-viral response that inhibits replication of encephalitic Alpha and Orthobunyaviruses in multi-cellular central nervous system cultures. These lipid-specific IgMs trigger microglia to produce IFN-β in a cGAS-STING-dependent manner, which induces an IFN-α/β-receptor 1-dependent antiviral response in glia and neurons. These data identify lipid-reactive IgM as a mediator of anti-viral activity in the nervous system and provide a rational explanation why intrathecal synthesis of lipid-reactive IgM correlates with a reduced incidence of iatrogenic PML in MS.
Collapse
|
8
|
Miyagawa F, Tagaya Y, Ozato K, Horie K, Asada H. Inflammatory monocyte-derived dendritic cells mediate autoimmunity in murine model of systemic lupus erythematosus. J Transl Autoimmun 2020; 3:100060. [PMID: 32743540 PMCID: PMC7388367 DOI: 10.1016/j.jtauto.2020.100060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/18/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Using a mouse model of systemic lupus erythematosus (SLE), we recently demonstrated that the two major manifestations of SLE are mechanistically independent because the type I IFN pathway leads to the autoantibody production whereas the NF-κB activation is sufficient for the development of glomerulonephritis. To further advance our understandings on the molecular pathways regulating the development of SLE, we studied the role of IRF8 because it controls both type I IFN and NF-κB pathways and saw that IRF8-deficient mice failed to develop either glomerulonephritis or the autoantibody production. Furthermore, these genetically engineered mice prompted us to realize the important role of Ly6Chigh inflammatory monocytes in the development of SLE. These monocytes migrate to the peritoneal cavity in WT and IRF7-deficient mice but not in IRF8-deficient mice, and there they produce both type I IFN and proinflammatory cytokines in WT mice, while in IRF7-deficient mice they only produce proinflammatory cytokines. Upon migration to the spleen, Ly6Chigh inflammatory monocytes differentiate into dendritic cells (DCs) which are capable of producing proinflammatory cytokines in response to dsDNA autoantigen. Collectively, type I IFN produced from inflammatory monocytes/monocyte-derived DCs might be essential for autoantibody production whereas proinflammatory cytokines produced from them might mediate tissue damages in this model. Our study reveals a specialized role for monocyte-derived antigen presenting cells in autoimmunity. Plasticity of monocyte might play an important role not only in the pathogenesis of the disease but also in flare-ups of the disease.
Collapse
Affiliation(s)
- Fumi Miyagawa
- Department of Dermatology, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Yutaka Tagaya
- Cell Biology Lab, Division of Infectious Agents and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Keiko Ozato
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kyoji Horie
- Department of Physiology II, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Hideo Asada
- Department of Dermatology, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| |
Collapse
|
9
|
Parodis I, Åkerström E, Sjöwall C, Sohrabian A, Jönsen A, Gomez A, Frodlund M, Zickert A, Bengtsson AA, Rönnelid J, Gunnarsson I. Autoantibody and Cytokine Profiles during Treatment with Belimumab in Patients with Systemic Lupus Erythematosus. Int J Mol Sci 2020; 21:E3463. [PMID: 32422945 PMCID: PMC7278961 DOI: 10.3390/ijms21103463] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/02/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022] Open
Abstract
We investigated whether belimumab treatment impacts on levels of autoantibodies and cytokines of interest in systemic lupus erythematosus (SLE). Longitudinally collected serum samples from 78 belimumab-treated Swedish SLE patients were analysed. Serum cytokine levels were determined using Luminex xMAP technology, and nuclear antigen autoantibody specificities using addressable laser bead immunoassay. In patients with detectable levels at baseline, interferon (IFN)-α2 levels were lower at month 6 (median; interquartile range (IQR): 8.9; 1.5-54.9 pg/mL) versus baseline (28.4; 20.9-100.3 pg/mL; p = 0.043). Interleukin (IL)-6 (baseline: 7.1; 2.9-16.1 pg/mL) decreased from month 6 (0.5; 0.5-6.3 pg/mL; p = 0.018) and throughout a 24 month follow-up. IL-10 (baseline: 12.6; 2.8-29.7 pg/mL) showed more rapid decreases from month 3 (1.8; 0.6-9.1 pg/mL; p = 0.003). Levels of anti-dsDNA (p < 0.001), anti-Smith antigen (Sm) (p = 0.002), anti-U1 small nuclear ribonucleoprotein (U1RNP) (p < 0.001), anti-Sm-U1RNP complex (p = 0.028), and anti-ribosomal P (p = 0.012) antibodies decreased from month 3 and remained decreased. Anti-Sm positivity at baseline was associated with higher probability and/or shorter time to achieve sustained SLE responder index-4 response (hazard ratio (HR): 2.52; 95% CI: 1.20-5.29; p = 0.015), independently of other factors. Decline of IL-6 levels through month 3 was greater in responders. In summary, belimumab treatment lowered IFN-α2, IL-6, and IL-10 levels, as well as levels of multiple autoantibodies, however after different time spans. Notably, anti-Sm positivity and early decline in IL-6 levels were associated with favorable treatment outcome.
Collapse
Affiliation(s)
- Ioannis Parodis
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (E.A.); (A.G.); (A.Z.); (I.G.)
- Rheumatology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Emil Åkerström
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (E.A.); (A.G.); (A.Z.); (I.G.)
- Rheumatology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Christopher Sjöwall
- Rheumatology/Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden; (C.S.); (M.F.)
| | - Azita Sohrabian
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (A.S.); (J.R.)
| | - Andreas Jönsen
- Rheumatology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, SE-222 42 Lund, Sweden; (A.J.); (A.A.B.)
| | - Alvaro Gomez
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (E.A.); (A.G.); (A.Z.); (I.G.)
- Rheumatology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Martina Frodlund
- Rheumatology/Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85 Linköping, Sweden; (C.S.); (M.F.)
| | - Agneta Zickert
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (E.A.); (A.G.); (A.Z.); (I.G.)
- Rheumatology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Anders A Bengtsson
- Rheumatology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, SE-222 42 Lund, Sweden; (A.J.); (A.A.B.)
| | - Johan Rönnelid
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden; (A.S.); (J.R.)
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden; (E.A.); (A.G.); (A.Z.); (I.G.)
- Rheumatology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
10
|
Schwartz N, Stock AD, Putterman C. Neuropsychiatric lupus: new mechanistic insights and future treatment directions. Nat Rev Rheumatol 2020; 15:137-152. [PMID: 30659245 DOI: 10.1038/s41584-018-0156-8] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Patients with systemic lupus erythematosus (SLE) frequently show symptoms of central nervous system (CNS) involvement, termed neuropsychiatric SLE (NPSLE). The CNS manifestations of SLE are diverse and have a broad spectrum of severity and prognostic implications. Patients with NPSLE typically present with nonspecific symptoms, such as headache and cognitive impairment, but might also experience devastating features, such as memory loss, seizures and stroke. Some features of NPSLE, in particular those related to coagulopathy, have been characterized and an evidence-based treatment algorithm is available. The cognitive and affective manifestations of NPSLE, however, remain poorly understood. Various immune effectors have been evaluated as contributors to its pathogenesis, including brain-reactive autoantibodies, cytokines and cell-mediated inflammation. Additional brain-intrinsic elements (such as resident microglia, the blood-brain barrier and other neurovascular interfaces) are important facilitators of NPSLE. As yet, however, no unifying model has been found to underlie the pathogenesis of NPSLE, suggesting that this disease has multiple contributors and perhaps several distinct aetiologies. This heterogeneity presents a challenge for clinicians who have traditionally relied on empirical judgement in choosing treatment modalities for patients with NPSLE. Improved understanding of this manifestation of SLE might yield further options for managing this disease.
Collapse
Affiliation(s)
- Noa Schwartz
- Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA
| | - Ariel D Stock
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chaim Putterman
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA. .,Division of Rheumatology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
11
|
Verbeek JS, Hirose S, Nishimura H. The Complex Association of FcγRIIb With Autoimmune Susceptibility. Front Immunol 2019; 10:2061. [PMID: 31681256 PMCID: PMC6803437 DOI: 10.3389/fimmu.2019.02061] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Abstract
FcγRIIb is the only inhibitory Fc receptor and controls many aspects of immune and inflammatory responses. The observation 19 years ago that Fc γ RIIb -/- mice generated by gene targeting in 129 derived ES cells developed severe lupus like disease when backcrossed more than 7 generations into C57BL/6 background initiated extensive research on the functional understanding of this strong autoimmune phenotype. The genomic region in the distal part of Chr1 both in human and mice in which the Fc γ R gene cluster is located shows a high level of complexity in relation to the susceptibility to SLE. Specific haplotypes of closely linked genes including the Fc γ RIIb and Slamf genes are associated with increased susceptibility to SLE both in mice and human. Using forward and reverse genetic approaches including in human GWAS and in mice congenic strains, KO mice (germline and cell type specific, on different genetic background), knockin mice, overexpressing transgenic mice combined with immunological models such as adoptive transfer of B cells from Ig transgenic mice the involved genes and the causal mutations and their associated functional alterations were analyzed. In this review the results of this 19 years extensive research are discussed with a focus on (genetically modified) mouse models.
Collapse
Affiliation(s)
- J Sjef Verbeek
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Sachiko Hirose
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Hiroyuki Nishimura
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| |
Collapse
|
12
|
Qiu CC, Atencio AE, Gallucci S. Inhibition of fatty acid metabolism by etomoxir or TOFA suppresses murine dendritic cell activation without affecting viability. Immunopharmacol Immunotoxicol 2019; 41:361-369. [PMID: 31155984 PMCID: PMC10724852 DOI: 10.1080/08923973.2019.1616754] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/24/2019] [Accepted: 05/01/2019] [Indexed: 02/08/2023]
Abstract
Objective: Dendritic cells (DCs) are important players in immunity against pathogens, but overactive DCs have been implicated in autoimmune diseases, like lupus, in which a paucity of targeted therapies remains. Recent research shows that DCs upregulate their immunometabolism when activating. We explored whether modulating fatty acid (FA) metabolism needed for oxidative phosphorylation can affect the activation of two main DC subsets. Material and methods: Sorted murine plasmacytoid DCs (pDCs) and conventional DCs (cDCs), generated in FLT3-L medium, were treated with etomoxir, an inhibitor of FA oxidation, or TOFA, an inhibitor of FA synthesis, then stimulated with TLR9 agonist CpGA. Surface activation markers and viability were analyzed by flow cytometry, cytokine, and chemokine production and were measured by ELISA. Results: Modulation of FA metabolism suppressed the upregulation of costimulatory molecules and the production of proinflammatory cytokine IL-6 and type I Interferon-dependent chemokine CXCL10 by both subsets of DCs, without affecting DC viability, neither of resting DCs or upon activation. Etomoxir inhibited pDCs at lower doses than cDCs, suggesting that pDCs may be more susceptible to FA metabolic modulation. Conclusions: Both cDCs, the primary antigen presenting cell, and pDCs, the primary type I IFN producer, exhibit a suppressed ability to activate but normal viability when their FA metabolism is inhibited by etomoxir or TOFA. Our findings indicate that FA metabolism plays an important role in the activation of both pDCs and cDCs and suggest that its modulation is an exploitable therapeutic target to suppress DC activation in inflammation or autoimmunity.
Collapse
Affiliation(s)
- Connie C Qiu
- a Laboratory of Dendritic Cell Biology, Department of Microbiology & Immunology , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| | - Atilio E Atencio
- a Laboratory of Dendritic Cell Biology, Department of Microbiology & Immunology , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| | - Stefania Gallucci
- a Laboratory of Dendritic Cell Biology, Department of Microbiology & Immunology , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| |
Collapse
|
13
|
Crow MK, Ronnblom L. Type I interferons in host defence and inflammatory diseases. Lupus Sci Med 2019; 6:e000336. [PMID: 31205729 PMCID: PMC6541752 DOI: 10.1136/lupus-2019-000336] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/21/2022]
Abstract
Type I interferons (IFN) can have dual and opposing roles in immunity, with effects that are beneficial or detrimental to the individual depending on whether IFN pathway activation is transient or sustained. Determinants of IFN production and its functional consequences include the nature of the microbial or nucleic acid stimulus, the type of nucleic acid sensor involved in inducing IFN, the predominant subtype of type I IFN produced and the immune ecology of the tissue at the time of IFN expression. When dysregulated, the type I IFN system drives many autoimmune and non-autoimmune inflammatory diseases, including SLE and the tissue inflammation associated with chronic infection. The type I IFN system may also contribute to outcomes for patients affected by solid cancers or myocardial infarction. Significantly more research is needed to discern the mechanisms of induction and response to type I IFNs across these diseases, and patient endophenotyping may help determine whether the cytokine is acting as 'friend' or 'foe', within a particular patient, and at the time of treatment. This review summarises key concepts and discussions from the second International Summit on Interferons in Inflammatory Diseases, during which expert clinicians and scientists evaluated the evidence for the role of type I IFNs in autoimmune and other inflammatory diseases.
Collapse
Affiliation(s)
- Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, Weill Cornell Medical College, New York City, New York, USA
| | - Lars Ronnblom
- Section of Rheumatology, Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
14
|
Gorji AE, Roudbari Z, Alizadeh A, Sadeghi B. Investigation of systemic lupus erythematosus (SLE) with integrating transcriptomics and genome wide association information. Gene 2019; 706:181-187. [PMID: 31082500 DOI: 10.1016/j.gene.2019.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/10/2019] [Accepted: 05/03/2019] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematous (SEL) is a heterogeneous, systemic autoimmune disorder which is defined by its autoantibody pattern. Transcriptomic data analysis has shown pathways and immune system responses associated with SLE. Eight up-regulated genes (SOCE, MMP9, CXCL8, JUN, IL1B, NFKBIA, TNF and FOS) have been examined with four interactions among different pathways. These genes are associated with SNPs which have been identified through two datasets from SLE genome-wide association studies (GWAS). In this investigation, the GWAS results were integrated with pathway analysis of transcriptomes and several genes were detected with known SLE-related variations (TYK2, C5, SH2B, IRF5, IL2RA, STAT4, FCGR2A, IL7R, LYN, HLA-DRB and TNFAIP3). Pathway-based analysis on the Wikipathway Human Collection allowed the identification of prioritized variants in the relevant pathways, such as thymic stromal lymphopoietin (TSLP) signaling pathway linked to LYN, IL7R, STAT4 and rs7574865. Analysis of existing transcriptomes and GWAS data identified eight up-regulated candidate genes with more than four relationships among the different pathways associated with SNPs to pinpoint the relevant loci linked to SLE. The results of this investigation have expanded the number of candidate genes related to SLE and have highlighted possible pathways and GWAS-based methods for gene detection. Identification of the fundamental genes would assist in revealing the mechanisms responsible for SLE.
Collapse
Affiliation(s)
- Abdolvahab Ebrahimpour Gorji
- Department of Fisheries, Faculty of Animal Sciences and Fisheries, Sari Agricultural and Natural Resources University, Sari, Iran
| | - Zahra Roudbari
- Department of Animal Science, Faculty of Agriculture, University of Jiroft, Jiroft, Iran.
| | - Akram Alizadeh
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Balal Sadeghi
- Food Hygiene and Public Health Department, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran.
| |
Collapse
|
15
|
Zaro BW, Vinogradova EV, Lazar DC, Blewett MM, Suciu RM, Takaya J, Studer S, de la Torre JC, Casanova JL, Cravatt BF, Teijaro JR. Dimethyl Fumarate Disrupts Human Innate Immune Signaling by Targeting the IRAK4-MyD88 Complex. THE JOURNAL OF IMMUNOLOGY 2019; 202:2737-2746. [PMID: 30885957 DOI: 10.4049/jimmunol.1801627] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 02/26/2019] [Indexed: 12/14/2022]
Abstract
Dimethyl fumarate (DMF) is a prescribed treatment for multiple sclerosis and has also been used to treat psoriasis. The electrophilicity of DMF suggests that its immunosuppressive activity is related to the covalent modification of cysteine residues in the human proteome. Nonetheless, our understanding of the proteins modified by DMF in human immune cells and the functional consequences of these reactions remains incomplete. In this study, we report that DMF inhibits human plasmacytoid dendritic cell function through a mechanism of action that is independent of the major electrophile sensor NRF2. Using chemical proteomics, we instead identify cysteine 13 of the innate immune kinase IRAK4 as a principal cellular target of DMF. We show that DMF blocks IRAK4-MyD88 interactions and IRAK4-mediated cytokine production in a cysteine 13-dependent manner. Our studies thus identify a proteomic hotspot for DMF action that constitutes a druggable protein-protein interface crucial for initiating innate immune responses.
Collapse
Affiliation(s)
- Balyn W Zaro
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Daniel C Lazar
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Megan M Blewett
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Radu M Suciu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Junichiro Takaya
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Sean Studer
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Juan Carlos de la Torre
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037;
| | - John R Teijaro
- Department of Immunology and Infectious Disease, The Scripps Research Institute, La Jolla, CA 92037; and
| |
Collapse
|
16
|
Louis C, Ngo D, D'Silva DB, Hansen J, Phillipson L, Jousset H, Novello P, Segal D, Lawlor KE, Burns CJ, Wicks IP. Therapeutic Effects of a
TANK
‐Binding Kinase 1 Inhibitor in Germinal Center–Driven Collagen‐Induced Arthritis. Arthritis Rheumatol 2018; 71:50-62. [DOI: 10.1002/art.40670] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/12/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Cynthia Louis
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Devi Ngo
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Damian B. D'Silva
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Jacinta Hansen
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Louisa Phillipson
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Helene Jousset
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Patrizia Novello
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - David Segal
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Kate E. Lawlor
- The Walter and Eliza Hall Institute of Medical Research and the University of Melbourne Parkville Victoria Australia
| | - Christopher J. Burns
- The Walter and Eliza Hall Institute of Medical Research, the University of Melbourne and the Bio21 Institute Parkville Victoria Australia
| | - Ian P. Wicks
- The Walter and Eliza Hall Institute of Medical Research, the University of Melbourne and Royal Melbourne Hospital Parkville Victoria Australia
| |
Collapse
|
17
|
Fournier N, Jacque E, Fontayne A, Derache D, Dupont G, Verhaeghe L, Baptista L, Dehenne A, Dezetter AS, Terrier A, Longue A, Pochet-Beghin V, Beghin C, Chtourou S, de Romeuf C. Improved in vitro and in vivo activity against CD303-expressing targets of the chimeric 122A2 antibody selected for specific glycosylation pattern. MAbs 2018; 10:651-663. [PMID: 29553870 PMCID: PMC5973763 DOI: 10.1080/19420862.2018.1451283] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Plasmacytoid dendritic cells (pDCs) play a central role for both innate and adaptive antiviral responses, as they direct immune responses through their unique ability to produce substantial concentrations of type I interferon (IFNs) upon viral encounter while also activating multiple immune cells, including macrophages, DCs, B, natural killer and T cells. Recent evidence clearly indicates that pDCs also play a crucial role in some cancers and several auto-immune diseases. Although treatments are currently available to patients with such pathologies, many are not fully efficient. We are proposing here, as a new targeted-based therapy, a novel chimeric monoclonal antibody (mAb) that mediates a strong cellular cytotoxicity directed against a specific human pDC marker, CD303. This antibody, ch122A2 mAb, is characterized by low fucose content in its human IgG1 constant (Fc) region, which induces strong in vitro and in vivo activity against human pDCs. We demonstrated that this effect relates in part to its specific Fc region glycosylation pattern, which increased affinity for CD16/FcγRIIIa. Importantly, ch122A2 mAb induces the down-modulation of CpG-induced IFN-α secretion by pDCs. Additionally, ch122A2 mAb shows in vitro high pDC depletion mediated by antibody-dependent cell-mediated cytotoxicity and antibody-dependent cellular phagocytosis. Remarkably, in vivo ch122A2 mAb efficacy is also demonstrated in humanized mice, resulting in significant pDC depletion in bloodstream and secondary lymphoid organs such as spleen. Together, our data indicates that ch122A2 mAb could represent a promising cytotoxic mAb candidate for pathologies in which decreasing type I IFNs or pDCs depleting may improve patient prognosis.
Collapse
Affiliation(s)
- Nathalie Fournier
- a Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Emilie Jacque
- a Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Alexandre Fontayne
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Delphine Derache
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Gilles Dupont
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Lucie Verhaeghe
- a Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Linda Baptista
- a Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Aurélie Dehenne
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Anne-Sophie Dezetter
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Aurélie Terrier
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Alain Longue
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Virginie Pochet-Beghin
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Cecile Beghin
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Sami Chtourou
- b Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| | - Christophe de Romeuf
- c Therapeutic Innovation Department , LFB Biotechnologies , Lille Cedex , Nord Pas de calais , France
| |
Collapse
|
18
|
Louis C, Burns C, Wicks I. TANK-Binding Kinase 1-Dependent Responses in Health and Autoimmunity. Front Immunol 2018; 9:434. [PMID: 29559975 PMCID: PMC5845716 DOI: 10.3389/fimmu.2018.00434] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/19/2018] [Indexed: 01/05/2023] Open
Abstract
The pathogenesis of autoimmune diseases, such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) is driven by genetic predisposition and environmental triggers that lead to dysregulated immune responses. These include the generation of pathogenic autoantibodies and aberrant production of inflammatory cytokines. Current therapies for RA and other autoimmune diseases reduce inflammation by targeting inflammatory mediators, most of which are innate response cytokines, resulting in generalized immunosuppression. Overall, this strategy has been very successful, but not all patients respond, responses can diminish over time and numerous side effects can occur. Therapies that target the germinal center (GC) reaction and/or antibody-secreting plasma cells (PC) potentially provide a novel approach. TANK-binding kinase 1 (TBK1) is an IKK-related serine/threonine kinase best characterized for its involvement in innate antiviral responses through the induction of type I interferons. TBK1 is also gaining attention for its roles in humoral immune responses. In this review, we discuss the role of TBK1 in immunological pathways involved in the development and maintenance of antibody responses, with particular emphasis on its potential relevance in the pathogenesis of humoral autoimmunity. First, we review the role of TBK1 in the induction of type I IFNs. Second, we highlight how TBK1 mediates inducible T cell co-stimulator signaling to the GC T follicular B helper population. Third, we discuss emerging evidence on the contribution of TBK1 to autophagic pathways and the potential implications for immune cell function. Finally, we discuss the therapeutic potential of TBK1 inhibition in autoimmunity.
Collapse
Affiliation(s)
- Cynthia Louis
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Chris Burns
- Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Ian Wicks
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Rheumatology Unit, Royal Melbourne Hospital, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
19
|
Increased Abundance of Plasmacytoid Dendritic Cells and Interferon-Alpha Induces Plasma Cell Differentiation in Patients of IgA Nephropathy. Mediators Inflamm 2017; 2017:4532409. [PMID: 29403161 PMCID: PMC5748321 DOI: 10.1155/2017/4532409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/30/2017] [Accepted: 09/24/2017] [Indexed: 11/22/2022] Open
Abstract
The roles of pDC and IFN-α have not been well defined in IgA nephropathy (IgAN). In this study, we investigated the abundance of pDCs and IFN-α in IgAN patients and the response of peripheral blood mononuclear cells (PBMCs) after stimulation of the pDC-preferred TLR9 ligand CpG2216. The effects of IFN-α on plasma cell differentiation and leukocyte migration were also investigated. Here, we found that the percentages of pDCs were increased in PBMCs of IgAN patients, than in those of healthy controls. Plasma levels of IFN-α proteins and abundance of plasma cells were higher in IgAN patients than in healthy donors. Plasma IFN-α levels were positively associated with proteinuria, renal IgM deposition, and renal tubular atrophy/interstitial fibrosis grade in IgAN patients. Ex vivo activation of TLR9 on pDCs resulted in increased IFN-α production and enhanced plasma cell differentiation in IgAN patients as compared with healthy donors. IFN-α treatment led to increased plasma cell differentiation in vitro. IFN-α also significantly promoted expression of chemokines IP-10 and MCP-1 in human mesangial cells, which subsequently facilitated the transendothelial migration of human CD4+ and CD14+ cells. In conclusion, pDC and its secreted cytokine IFN-α may play important roles in pathological changes of IgA nephropathy.
Collapse
|
20
|
Li H, Reksten TR, Ice JA, Kelly JA, Adrianto I, Rasmussen A, Wang S, He B, Grundahl KM, Glenn SB, Miceli-Richard C, Bowman S, Lester S, Eriksson P, Eloranta ML, Brun JG, Gøransson LG, Harboe E, Guthridge JM, Kaufman KM, Kvarnström M, Cunninghame Graham DS, Patel K, Adler AJ, Farris AD, Brennan MT, Chodosh J, Gopalakrishnan R, Weisman MH, Venuturupalli S, Wallace DJ, Hefner KS, Houston GD, Huang AJW, Hughes PJ, Lewis DM, Radfar L, Vista ES, Edgar CE, Rohrer MD, Stone DU, Vyse TJ, Harley JB, Gaffney PM, James JA, Turner S, Alevizos I, Anaya JM, Rhodus NL, Segal BM, Montgomery CG, Scofield RH, Kovats S, Mariette X, Rönnblom L, Witte T, Rischmueller M, Wahren-Herlenius M, Omdal R, Jonsson R, Ng WF, for UK Primary Sjögren's Syndrome Registry, Nordmark G, Lessard CJ, Sivils KL. Identification of a Sjögren's syndrome susceptibility locus at OAS1 that influences isoform switching, protein expression, and responsiveness to type I interferons. PLoS Genet 2017. [PMID: 28640813 PMCID: PMC5501660 DOI: 10.1371/journal.pgen.1006820] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Sjögren's syndrome (SS) is a common, autoimmune exocrinopathy distinguished by keratoconjunctivitis sicca and xerostomia. Patients frequently develop serious complications including lymphoma, pulmonary dysfunction, neuropathy, vasculitis, and debilitating fatigue. Dysregulation of type I interferon (IFN) pathway is a prominent feature of SS and is correlated with increased autoantibody titers and disease severity. To identify genetic determinants of IFN pathway dysregulation in SS, we performed cis-expression quantitative trait locus (eQTL) analyses focusing on differentially expressed type I IFN-inducible transcripts identified through a transcriptome profiling study. Multiple cis-eQTLs were associated with transcript levels of 2'-5'-oligoadenylate synthetase 1 (OAS1) peaking at rs10774671 (PeQTL = 6.05 × 10-14). Association of rs10774671 with SS susceptibility was identified and confirmed through meta-analysis of two independent cohorts (Pmeta = 2.59 × 10-9; odds ratio = 0.75; 95% confidence interval = 0.66-0.86). The risk allele of rs10774671 shifts splicing of OAS1 from production of the p46 isoform to multiple alternative transcripts, including p42, p48, and p44. We found that the isoforms were differentially expressed within each genotype in controls and patients with and without autoantibodies. Furthermore, our results showed that the three alternatively spliced isoforms lacked translational response to type I IFN stimulation. The p48 and p44 isoforms also had impaired protein expression governed by the 3' end of the transcripts. The SS risk allele of rs10774671 has been shown by others to be associated with reduced OAS1 enzymatic activity and ability to clear viral infections, as well as reduced responsiveness to IFN treatment. Our results establish OAS1 as a risk locus for SS and support a potential role for defective viral clearance due to altered IFN response as a genetic pathophysiological basis of this complex autoimmune disease.
Collapse
Affiliation(s)
- He Li
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Tove Ragna Reksten
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - John A. Ice
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Jennifer A. Kelly
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Indra Adrianto
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Astrid Rasmussen
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Shaofeng Wang
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Bo He
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Kiely M. Grundahl
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Stuart B. Glenn
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Corinne Miceli-Richard
- Université Paris-Sud, AP-HP, Hôpitaux Universitaires Paris-Sud, INSERM U1012, Le Kremlin Bicêtre, France
| | - Simon Bowman
- Rheumatology Department, University Hospital Birmingham, Birmingham, United Kingdom
| | - Sue Lester
- The Queen Elizabeth Hospital, Adelaide, South Australia, Australia
| | - Per Eriksson
- Department of Rheumatology, Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Maija-Leena Eloranta
- Department of Medical Sciences, Rheumatology, SciLIfeLab, Uppsala University, Uppsala, Sweden
| | - Johan G. Brun
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - Lasse G. Gøransson
- Clinical Immunology Unit, Department of Internal Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Erna Harboe
- Clinical Immunology Unit, Department of Internal Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Joel M. Guthridge
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Kenneth M. Kaufman
- Division of Rheumatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, United States of America
| | | | | | - Ketan Patel
- Division of Oral and Maxillofacial Surgery, Department of Developmental and Surgical Science, University of Minnesota School of Dentistry, Minneapolis, Minnesota, United States of America
- Department of Oral and Maxillofacial Surgery, North Memorial Medical Center, Robbinsdale, Minnesota, United States of America
| | - Adam J. Adler
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - A. Darise Farris
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Michael T. Brennan
- Department of Oral Medicine, Carolinas Medical Center, Charlotte, North Carolina, United States of America
| | - James Chodosh
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rajaram Gopalakrishnan
- Division of Oral Pathology, Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, United States of America
| | - Michael H. Weisman
- Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Swamy Venuturupalli
- Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Daniel J. Wallace
- Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Kimberly S. Hefner
- Hefner Eye Care and Optical Center, Oklahoma City, Oklahoma, United States of America
| | - Glen D. Houston
- Department of Oral and Maxillofacial Pathology, University of Oklahoma College of Dentistry, Oklahoma City, Oklahoma, United States of America
- Heartland Pathology Consultants, Edmond, Oklahoma, United States of America
| | - Andrew J. W. Huang
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, Missouri, United States of America
| | - Pamela J. Hughes
- Division of Oral and Maxillofacial Surgery, Department of Developmental and Surgical Science, University of Minnesota School of Dentistry, Minneapolis, Minnesota, United States of America
| | - David M. Lewis
- Department of Oral and Maxillofacial Pathology, University of Oklahoma College of Dentistry, Oklahoma City, Oklahoma, United States of America
| | - Lida Radfar
- Oral Diagnosis and Radiology Department, University of Oklahoma College of Dentistry, Oklahoma City, Oklahoma, United States of America
| | - Evan S. Vista
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- University of Santo Tomas Hospital, Manila, The Philippines
| | - Contessa E. Edgar
- The Biology Department, Oklahoma Baptist University, Oklahoma City, Oklahoma, United States of America
| | - Michael D. Rohrer
- Hard Tissue Research Laboratory, University of Minnesota School of Dentistry, Minneapolis, Minnesota, United States of America
| | - Donald U. Stone
- Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Timothy J. Vyse
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - John B. Harley
- Division of Rheumatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- US Department of Veterans Affairs Medical Center, Cincinnati, Ohio, United States of America
| | - Patrick M. Gaffney
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Judith A. James
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Sean Turner
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Ilias Alevizos
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, United States of America
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research, Universidad del Rosario, Bogotá, Colombia
| | - Nelson L. Rhodus
- Department of Oral Surgery, University of Minnesota School of Dentistry, Minneapolis, Minnesota, United States of America
| | - Barbara M. Segal
- Division of Rheumatology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Courtney G. Montgomery
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - R. Hal Scofield
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- US Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma, United States of America
| | - Susan Kovats
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Xavier Mariette
- Université Paris-Sud, AP-HP, Hôpitaux Universitaires Paris-Sud, INSERM U1012, Le Kremlin Bicêtre, France
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology, SciLIfeLab, Uppsala University, Uppsala, Sweden
| | - Torsten Witte
- Clinic for Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Maureen Rischmueller
- The Queen Elizabeth Hospital, Adelaide, South Australia, Australia
- The University of Adelaide, Adelaide, South Australia, Australia
| | | | - Roald Omdal
- Clinical Immunology Unit, Department of Internal Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Roland Jonsson
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - Wan-Fai Ng
- Institute of Cellular Medicine & NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Gunnel Nordmark
- Department of Medical Sciences, Rheumatology, SciLIfeLab, Uppsala University, Uppsala, Sweden
| | - Christopher J. Lessard
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Kathy L. Sivils
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
21
|
Xue S, Liu C, Sun X, Li W, Zhang C, Zhou X, Lu Y, Xiao J, Li C, Xu X, Sun B, Xu G, Wang H. TET3 Inhibits Type I IFN Production Independent of DNA Demethylation. Cell Rep 2016; 16:1096-1105. [PMID: 27425624 DOI: 10.1016/j.celrep.2016.06.068] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/11/2016] [Accepted: 06/15/2016] [Indexed: 12/12/2022] Open
Abstract
Type I interferons (IFNs) play both beneficial and harmful roles in antiviral responses. Precise regulation of host type I IFNs is thus needed to prevent immune dysregulation. Here, we find that the DNA demethylase TET3 is a negative regulator of IFN-β in response to poly(I:C) stimulation or viral infection. Deletion of TET3 enhances antiviral responses, with elevated expression of IFN-β and IFN-stimulated genes. The catalytic domain of TET3 was critical for the suppression of IFN-β production, but TET3 enzymatic activity was dispensable. Instead, the catalytic domain of TET3 interacts with HDAC1 and SIN3A, thus enhancing their binding to the Ifnb1 promoter. Our study demonstrates that TET3 negatively regulates type I IFN production independent of DNA demethylation. This not only sheds light on TET3 as a signaling protein in immune cells for gene regulation but also will help to develop strategies to prevent type I IFN-related disease.
Collapse
Affiliation(s)
- Shengjie Xue
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chang Liu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Xiujie Sun
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Weiyun Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chi Zhang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Xin Zhou
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Yao Lu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jun Xiao
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Chunyang Li
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Xiaoyan Xu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Bing Sun
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoliang Xu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongyan Wang
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China.
| |
Collapse
|
22
|
Wang F, Qiao L, Lv X, Trivett A, Yang R, Oppenheim JJ, Yang D, Zhang N. Alarmin human α defensin HNP1 activates plasmacytoid dendritic cells by triggering NF-κB and IRF1 signaling pathways. Cytokine 2016; 83:53-60. [PMID: 27031443 PMCID: PMC7822553 DOI: 10.1016/j.cyto.2016.03.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/14/2016] [Accepted: 03/21/2016] [Indexed: 01/13/2023]
Abstract
Human neutrophil peptide 1 (HNP1), a predominant α defensin in the azurophilic granules of human neutrophils, is an alarmin capable of inducing the migration and maturation of human myeloid/conventional dendritic cells. However, it is not determined whether it can activate plasmacytoid dendritic cells (pDCs). Herein, we found that both human pDCs and CAL-1 cells, a pDC-like cell line, produced IFNα upon treatment with HNP1. Additionally, HNP1 could promote CpG ODN-induced pDC production of proinflammatory cytokines including IFNα. HNP1 triggered activation of NF-κB and nuclear translocation of interferon regulatory factor 1 (IRF1) in CAL-1 cells. HNP1 upregulation of cytokine expression in pDCs was inhibited by blockade of NF-κB activation or knockdown of IRF1, demonstrating the importance of these two signaling events in HNP1-induced pDC activation. Using a human pDC-nude mouse model, HNP1 was shown to induce IFNα production by human pDCs in vivo. Thus, HNP1 can activate human pDCs using NF-κB and IRF signaling pathways, and HNP-induced IFN production may participate in the inflammatory pathogenesis in certain authoimmune diseases such as rheumatoid arthritis.
Collapse
Affiliation(s)
- Fang Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Research Center of Basic Medical Science, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, People's Republic of China
| | - Linan Qiao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Research Center of Basic Medical Science, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, People's Republic of China
| | - Xing Lv
- Department of Rheumatism and Immunology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, People's Republic of China
| | - Anna Trivett
- Basic Science Program, Leidos Biomedical Research Inc., and Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, Frederick National Laboratory for Cancer Research (FNLCR), 1050 Boyles Street, Frederick, MD 21702, USA
| | - Rui Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Research Center of Basic Medical Science, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, People's Republic of China
| | - Joost J Oppenheim
- Basic Science Program, Leidos Biomedical Research Inc., and Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, Frederick National Laboratory for Cancer Research (FNLCR), 1050 Boyles Street, Frederick, MD 21702, USA
| | - De Yang
- Basic Science Program, Leidos Biomedical Research Inc., and Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, Frederick National Laboratory for Cancer Research (FNLCR), 1050 Boyles Street, Frederick, MD 21702, USA.
| | - Ning Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Research Center of Basic Medical Science, Tianjin Medical University, 22 Qixiangtai Road, Heping District, Tianjin 300070, People's Republic of China.
| |
Collapse
|
23
|
Multiplex giant magnetoresistive biosensor microarrays identify interferon-associated autoantibodies in systemic lupus erythematosus. Sci Rep 2016; 6:27623. [PMID: 27279139 PMCID: PMC4899742 DOI: 10.1038/srep27623] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/20/2016] [Indexed: 01/05/2023] Open
Abstract
High titer, class-switched autoantibodies are a hallmark of systemic lupus erythematosus (SLE). Dysregulation of the interferon (IFN) pathway is observed in individuals with active SLE, although the association of specific autoantibodies with chemokine score, a combined measurement of three IFN-regulated chemokines, is not known. To identify autoantibodies associated with chemokine score, we developed giant magnetoresistive (GMR) biosensor microarrays, which allow the parallel measurement of multiple serum antibodies to autoantigens and peptides. We used the microarrays to analyze serum samples from SLE patients and found individuals with high chemokine scores had significantly greater reactivity to 13 autoantigens than individuals with low chemokine scores. Our findings demonstrate that multiple autoantibodies, including antibodies to U1-70K and modified histone H2B tails, are associated with IFN dysregulation in SLE. Further, they show the microarrays are capable of identifying autoantibodies associated with relevant clinical manifestations of SLE, with potential for use as biomarkers in clinical practice.
Collapse
|
24
|
Wolf C, Rapp A, Berndt N, Staroske W, Schuster M, Dobrick-Mattheuer M, Kretschmer S, König N, Kurth T, Wieczorek D, Kast K, Cardoso MC, Günther C, Lee-Kirsch MA. RPA and Rad51 constitute a cell intrinsic mechanism to protect the cytosol from self DNA. Nat Commun 2016; 7:11752. [PMID: 27230542 PMCID: PMC4895045 DOI: 10.1038/ncomms11752] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/26/2016] [Indexed: 11/15/2022] Open
Abstract
Immune recognition of cytosolic DNA represents a central antiviral defence mechanism. Within the host, short single-stranded DNA (ssDNA) continuously arises during the repair of DNA damage induced by endogenous and environmental genotoxic stress. Here we show that short ssDNA traverses the nuclear membrane, but is drawn into the nucleus by binding to the DNA replication and repair factors RPA and Rad51. Knockdown of RPA and Rad51 enhances cytosolic leakage of ssDNA resulting in cGAS-dependent type I IFN activation. Mutations in the exonuclease TREX1 cause type I IFN-dependent autoinflammation and autoimmunity. We demonstrate that TREX1 is anchored within the outer nuclear membrane to ensure immediate degradation of ssDNA leaking into the cytosol. In TREX1-deficient fibroblasts, accumulating ssDNA causes exhaustion of RPA and Rad51 resulting in replication stress and activation of p53 and type I IFN. Thus, the ssDNA-binding capacity of RPA and Rad51 constitutes a cell intrinsic mechanism to protect the cytosol from self DNA. A central antiviral defence is immune recognition of cystolic DNA. Here the authors show that RPA and RAD51, in cooperation with TREX1, function to protect the cytosol from self-DNA.
Collapse
Affiliation(s)
- Christine Wolf
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Alexander Rapp
- Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Nicole Berndt
- Department of Dermatology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Wolfgang Staroske
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Max Schuster
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Manuela Dobrick-Mattheuer
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Stefanie Kretschmer
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Nadja König
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Thomas Kurth
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany.,Center for Regenerative Therapies, Technische Universität Dresden, 01307 Dresden, Germany
| | - Dagmar Wieczorek
- Institute of Human Genetics, Heinrich-Heine-University, Medical Faculty, 40225 Düsseldorf, Germany
| | - Karin Kast
- Department of Gynecology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - M Cristina Cardoso
- Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Claudia Günther
- Department of Dermatology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
25
|
Tamura Y, Yoneda A, Takei N, Sawada K. Spatiotemporal Regulation of Hsp90-Ligand Complex Leads to Immune Activation. Front Immunol 2016; 7:201. [PMID: 27252703 PMCID: PMC4877505 DOI: 10.3389/fimmu.2016.00201] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 05/09/2016] [Indexed: 12/19/2022] Open
Abstract
Although heat shock proteins (HSPs) primarily play a pivotal role in the maintenance of cellular homeostasis while reducing extracellular as well as intracellular stresses, their role in immunologically relevant scenarios, including activation of innate immunity as danger signals, antitumor immunity, and autoimmune diseases, is now gaining much attention. The most prominent feature of HSPs is that they function both in their own and as an HSP–ligand complex. We here show as a unique feature of extracellular HSPs that they target chaperoned molecules into a particular endosomal compartment of dendritic cells, thereby inducing innate and adaptive immune responses via spatiotemporal regulation.
Collapse
Affiliation(s)
- Yasuaki Tamura
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| | - Akihiro Yoneda
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| | - Norio Takei
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| | - Kaori Sawada
- Department of Molecular Therapeutics, Center for Food and Medical Innovation, Institute for Innovation and Business Promotion, Hokkaido University , Sapporo , Japan
| |
Collapse
|
26
|
Touzot M, Dahirel A, Cappuccio A, Segura E, Hupé P, Soumelis V. Using Transcriptional Signatures to Assess Immune Cell Function: From Basic Mechanisms to Immune-Related Disease. J Mol Biol 2015; 427:3356-67. [PMID: 25986308 DOI: 10.1016/j.jmb.2015.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 12/24/2022]
Abstract
Assessing human immune response remains a challenge as it involves multiple cell types in specific tissues. The use of microarray-based expression profiling as a tool for assessing the immune response has grown increasingly over the past decade. Transcriptome analyses provide investigators with a global perspective of the complex molecular and cellular events that unfold during the development of an immune response. In this review, we will detail the broad use of gene expression profiling to decipher the complexity of immune responses from disease biomarkers identification to cell activation, polarisation or functional specialisation. We will also describe how such data-driven strategies revealed the flexibility of immune function with common and specific transcriptional programme under multiple stimuli.
Collapse
Affiliation(s)
- Maxime Touzot
- INSERM U932, 26 rue d'Ulm, 75005 Paris, France; Institut Curie, Section Recherche, 26 rue d'Ulm, 75005 Paris, France; Laboratoire d'Immunologie Clinique, Institut Curie, 26 rue d'Ulm, 75005 Paris, France
| | - Alix Dahirel
- INSERM U932, 26 rue d'Ulm, 75005 Paris, France; Institut Curie, Section Recherche, 26 rue d'Ulm, 75005 Paris, France; Laboratoire d'Immunologie Clinique, Institut Curie, 26 rue d'Ulm, 75005 Paris, France
| | - Antonio Cappuccio
- INSERM U932, 26 rue d'Ulm, 75005 Paris, France; Institut Curie, Section Recherche, 26 rue d'Ulm, 75005 Paris, France; Service de Bioinformatique, INSERM U900, Institut Curie, 26 rue d'Ulm, 75248 Paris, France; Laboratoire d'Immunologie Clinique, Institut Curie, 26 rue d'Ulm, 75005 Paris, France
| | - Elodie Segura
- INSERM U932, 26 rue d'Ulm, 75005 Paris, France; Institut Curie, Section Recherche, 26 rue d'Ulm, 75005 Paris, France
| | - Philippe Hupé
- Institut Curie, Section Recherche, 26 rue d'Ulm, 75005 Paris, France; Service de Bioinformatique, INSERM U900, Institut Curie, 26 rue d'Ulm, 75248 Paris, France; CNRS UMR 144
| | - Vassili Soumelis
- INSERM U932, 26 rue d'Ulm, 75005 Paris, France; Institut Curie, Section Recherche, 26 rue d'Ulm, 75005 Paris, France; Laboratoire d'Immunologie Clinique, Institut Curie, 26 rue d'Ulm, 75005 Paris, France.
| |
Collapse
|
27
|
Touzot M, Cacoub P, Bodaghi B, Soumelis V, Saadoun D. IFN-α induces IL-10 production and tilt the balance between Th1 and Th17 in Behçet disease. Autoimmun Rev 2015; 14:370-5. [DOI: 10.1016/j.autrev.2014.12.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/14/2014] [Indexed: 12/30/2022]
|
28
|
Günther C, Kind B, Reijns MAM, Berndt N, Martinez-Bueno M, Wolf C, Tüngler V, Chara O, Lee YA, Hübner N, Bicknell L, Blum S, Krug C, Schmidt F, Kretschmer S, Koss S, Astell KR, Ramantani G, Bauerfeind A, Morris DL, Cunninghame Graham DS, Bubeck D, Leitch A, Ralston SH, Blackburn EA, Gahr M, Witte T, Vyse TJ, Melchers I, Mangold E, Nöthen MM, Aringer M, Kuhn A, Lüthke K, Unger L, Bley A, Lorenzi A, Isaacs JD, Alexopoulou D, Conrad K, Dahl A, Roers A, Alarcon-Riquelme ME, Jackson AP, Lee-Kirsch MA. Defective removal of ribonucleotides from DNA promotes systemic autoimmunity. J Clin Invest 2014; 125:413-24. [PMID: 25500883 DOI: 10.1172/jci78001] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/09/2014] [Indexed: 01/22/2023] Open
Abstract
Genome integrity is continuously challenged by the DNA damage that arises during normal cell metabolism. Biallelic mutations in the genes encoding the genome surveillance enzyme ribonuclease H2 (RNase H2) cause Aicardi-Goutières syndrome (AGS), a pediatric disorder that shares features with the autoimmune disease systemic lupus erythematosus (SLE). Here we determined that heterozygous parents of AGS patients exhibit an intermediate autoimmune phenotype and demonstrated a genetic association between rare RNASEH2 sequence variants and SLE. Evaluation of patient cells revealed that SLE- and AGS-associated mutations impair RNase H2 function and result in accumulation of ribonucleotides in genomic DNA. The ensuing chronic low level of DNA damage triggered a DNA damage response characterized by constitutive p53 phosphorylation and senescence. Patient fibroblasts exhibited constitutive upregulation of IFN-stimulated genes and an enhanced type I IFN response to the immunostimulatory nucleic acid polyinosinic:polycytidylic acid and UV light irradiation, linking RNase H2 deficiency to potentiation of innate immune signaling. Moreover, UV-induced cyclobutane pyrimidine dimer formation was markedly enhanced in ribonucleotide-containing DNA, providing a mechanism for photosensitivity in RNase H2-associated SLE. Collectively, our findings implicate RNase H2 in the pathogenesis of SLE and suggest a role of DNA damage-associated pathways in the initiation of autoimmunity.
Collapse
|
29
|
Biermann MHC, Veissi S, Maueröder C, Chaurio R, Berens C, Herrmann M, Munoz LE. The role of dead cell clearance in the etiology and pathogenesis of systemic lupus erythematosus: dendritic cells as potential targets. Expert Rev Clin Immunol 2014; 10:1151-64. [DOI: 10.1586/1744666x.2014.944162] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
30
|
The pathogenesis and diagnosis of systemic lupus erythematosus: still not resolved. Semin Immunopathol 2014; 36:301-11. [PMID: 24763531 DOI: 10.1007/s00281-014-0428-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 04/01/2014] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with various clinical manifestations affecting different tissues. A characteristic feature of SLE is the presence of autoantibodies against double-stranded (ds)DNA, histones and nucleosomes, and other chromatin components. SLE is a prototype type III hypersensitivity reaction. Local deposition of anti-nuclear antibodies in complex with released chromatin induces serious inflammatory conditions by activation of the complement system. The severe renal manifestation, lupus nephritis, is classified based on histological findings in renal biopsies. Apoptotic debris, including chromatin, is present in the extracellular matrix and circulation of patients with SLE. This may be due to an aberrant process of apoptosis and/or insufficient clearance of apoptotic cells/chromatin. The non-cleared apoptotic debris may lead to activation of both the innate and adaptive immune systems. In addition, an aberrant presentation of peptides by antigen-presenting cells, disturbed selection processes for lymphocytes, and deregulated lymphocyte responses may be involved in the development of autoimmunity. In the present review, we briefly will summarize current knowledge on the pathogenesis of SLE. We will also critically discuss and challenge central issues that need to be addressed in order to fully understand the pathogenic mechanisms involved in the development of SLE and in order to have an improved diagnosis for SLE. Disappointingly, in our opinion, there are still more questions than answers for the pathogenesis, diagnosis, and treatment of SLE.
Collapse
|
31
|
Eriksson C, Rantapää-Dahlqvist S. Cytokines in relation to autoantibodies before onset of symptoms for systemic lupus erythematosus. Lupus 2014; 23:691-6. [PMID: 24531079 DOI: 10.1177/0961203314523869] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 01/23/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVES A number of cytokines and chemokines were analysed and related to autoantibodies in blood samples pre-dating the onset of symptoms of systemic lupus erythematosus. METHODS Thirty-five patients with systemic lupus erythematosus (American College of Rheumatology criteria) were identified as having donated blood samples, prior to symptom onset, to the Biobank of northern Sweden. Altogether, 140 age- and sex-matched controls were also identified. The concentrations of interferon-α, interleukin-4, interleukin-9, interleukin-10, interferon inducible protein-10 and monocyte chemotactic protein-1 were analysed using multiplex technology and related to autoantibodies (ANA, ENA, anti-dsDNA and anti-histone antibodies) analysed from the same blood sample. RESULTS The interferon-γ inducible protein-10 levels were higher in the pre-symptomatic individuals than in controls (p < 0.05) and correlated with interferon-α (p < 0.01). The interferon-γ inducible protein-10 and interferon-α concentrations were significantly increased in individuals positive for autoantibodies: interferon-γ inducible protein-10 for ANA; anti-SSA/Ro and anti-Jo-1 antibodies; interferon-α with anti-SSB/La antibodies. The levels of interleukin-10, interferon-γ inducible protein-10 and monocyte chemotactic protein-1 increased significantly from the pre-symptomatic individuals to after onset of systemic lupus erythematosus. CONCLUSIONS An increased concentration of interferon-γ inducible protein-10 pre-dated the onset of systemic lupus erythematosus and was related to autoantibodies before the onset of disease. The levels of interferon-γ inducible protein-10 and interferon-α were correlated. These findings support the proposal that the interferon system is important early in the pathogenesis of systemic lupus erythematosus and autoantibody formation.
Collapse
Affiliation(s)
- C Eriksson
- Department of Clinical Immunology/Clinical Microbiology, Umeå University, Umeå, Sweden
| | - S Rantapää-Dahlqvist
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, Umeå, Sweden
| |
Collapse
|
32
|
Lee-Kirsch MA, Wolf C, Günther C. Aicardi-Goutières syndrome: a model disease for systemic autoimmunity. Clin Exp Immunol 2014; 175:17-24. [PMID: 23786362 DOI: 10.1111/cei.12160] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2013] [Indexed: 02/06/2023] Open
Abstract
Systemic autoimmunity is a complex disease process that results from a loss of immunological tolerance characterized by the inability of the immune system to discriminate self from non-self. In patients with the prototypic autoimmune disease systemic lupus erythematosus (SLE), formation of autoantibodies targeting ubiquitous nuclear antigens and subsequent deposition of immune complexes in the vascular bed induces inflammatory tissue injury that can affect virtually any organ system. Given the extraordinary genetic and phenotypic heterogeneity of SLE, one approach to the genetic dissection of complex SLE is to study monogenic diseases, for which a single gene defect is responsible. Considerable success has been achieved from the analysis of the rare monogenic disorder Aicardi-Goutières syndrome (AGS), an inflammatory encephalopathy that clinically resembles in-utero-acquired viral infection and that also shares features with SLE. Progress in understanding the cellular and molecular functions of the AGS causing genes has revealed novel pathways of the metabolism of intracellular nucleic acids, the major targets of the autoimmune attack in patients with SLE. Induction of autoimmunity initiated by immune recognition of endogenous nucleic acids originating from processes such as DNA replication/repair or endogenous retro-elements represents novel paradigms of SLE pathogenesis. These findings illustrate how investigating rare monogenic diseases can also fuel discoveries that advance our understanding of complex disease. This will not only aid the development of improved tools for SLE diagnosis and disease classification, but also the development of novel targeted therapeutic approaches.
Collapse
Affiliation(s)
- M A Lee-Kirsch
- Department of Pediatrics, University Hospital, Technical University Dresden, Dresden, Germany
| | | | | |
Collapse
|
33
|
Kim BY, Park I, Jung YK, Han MS, Kim GW, Han SW. DICAM-mediated Inhibition of Type 1 Interferon System during Macrophage Differentiation of THP-1 Cells. JOURNAL OF RHEUMATIC DISEASES 2014. [DOI: 10.4078/jrd.2014.21.3.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Bo Yeon Kim
- Department of Internal Medicine, Daegu Fatima Hospital, Daegu, Korea
| | - In Park
- Department of Internal Medicine, Daegu Fatima Hospital, Daegu, Korea
| | - Youn Kwan Jung
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu, Korea
| | - Min Su Han
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu, Korea
| | - Gun Woo Kim
- Department of Internal Medicine, Daegu Fatima Hospital, Daegu, Korea
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu, Korea
| | - Seung Woo Han
- Department of Internal Medicine, Daegu Fatima Hospital, Daegu, Korea
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu, Korea
| |
Collapse
|
34
|
Hagberg N, Theorell J, Eloranta ML, Pascal V, Bryceson YT, Rönnblom L. Anti-NKG2A autoantibodies in a patient with systemic lupus erythematosus. Rheumatology (Oxford) 2013; 52:1818-23. [PMID: 23825044 DOI: 10.1093/rheumatology/ket220] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
OBJECTIVES To characterize a novel anti-NKG2A autoantibody detected in a patient with SLE during a severe flare, and in a cross-sectional study investigate the occurrence of such autoantibodies in patients with SLE and primary SS (pSS). METHODS Serum or IgG from patients with SLE, pSS and healthy volunteers were assayed for blocking of anti-NKG2A or HLA-E binding to peripheral blood mononuclear cells or CD94/NKG2A- and CD94/NKG2C-transfected Ba/F3 cells. The anti-NKG2A autoantibodies were evaluated for effect on NK cell degranulation in response to HLA-E-transfected K562 cells. IFN-α was determined by an immunoassay and disease activity by the SLEDAI score. RESULTS Anti-NKG2A autoantibodies, which blocked binding of HLA-E tetramers to CD94/NKG2A-transfected cells and impaired NKG2A-mediated inhibition of NK cell activation, were observed in a patient with SLE. The presence of anti-NKG2A autoantibodies was associated with high SLE disease activity (SLEDAI score 14 and 16) and increased serum IFN-α. Of 94 SLE, 60 pSS and 30 healthy donor sera, only the index patient serum contained anti-NKG2A autoantibodies. CONCLUSION The presence of autoantibodies targeting NKG2A is a rare event, but when such autoantibodies occur they may promote excessive NK cell function. This can contribute to the pathogenesis by increasing the killing of cells and the release of autoantigens. Our findings highlight the possible importance of NK cells in the SLE disease process.
Collapse
Affiliation(s)
- Niklas Hagberg
- Department of Medical Sciences, Section of Rheumatology, Systemic Autoimmunity Group, Clinical Research Department 3, Entrance 85 3rd Floor, 751 85 Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
35
|
Di Domizio J, Cao W. Fueling autoimmunity: type I interferon in autoimmune diseases. Expert Rev Clin Immunol 2013; 9:201-10. [PMID: 23445195 DOI: 10.1586/eci.12.106] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In recent years, active research using genomic, cellular and animal modeling approaches has revealed the fundamental forces driving the development of autoimmune diseases. Type I interferon imprints unique molecular signatures in a list of autoimmune diseases. Interferon is induced by diverse nucleic acid-containing complexes, which trigger innate immune activation of plasmacytoid dendritic cells. Interferon primes, activates or differentiates various leukocyte populations to promote autoimmunity. Accordingly, interferon signaling is essential for the initiation and/or progression of lupus in several experimental models. However, the heterogeneous nature of systemic lupus erythematosus requires better characterization on how interferon pathways are activated and subsequently promote the advancement of autoimmune diseases. Given the central role of type I interferon, various strategies are devised to target these cytokines or related pathways to curtail the progression of autoimmune diseases.
Collapse
Affiliation(s)
- Jeremy Di Domizio
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | | |
Collapse
|
36
|
Franchin G, Son M, Kim SJ, Ben-Zvi I, Zhang J, Diamond B. Anti-DNA antibodies cross-react with C1q. J Autoimmun 2013; 44:34-9. [PMID: 23834843 DOI: 10.1016/j.jaut.2013.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 12/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder that involves multiple organ systems and typically presents as a chronic inflammatory disease. Antibodies to double-stranded (ds) DNA are present in approximately 70% of patients and form nucleic acid containing immune complexes which activate dendritic cells through engagement of toll-like receptors, leading to a pro-inflammatory, pro-immunogenic milieu. In addition, anti-dsDNA antibodies deposit in kidneys to initiate glomerulonephritis. Antibodies to C1q have also been implicated in lupus nephritis and are found in 30-50% of patients. C1q is a known suppressor of immune activation and C1q deficiency is the strongest risk factor for SLE. We previously identified a subset of anti-DNA antibodies that binds the N-methyl-D-aspartate receptor. We now show that both mouse and human anti-DNA antibodies with this specificity bind C1q. These antibodies bind to Clq in glomeruli and exhibit decreased glomerular deposition in the absence of C1q. We propose that this subset of anti-DNA antibodies participates in lupus pathogenesis through direct targeting of C1q on glomeruli and also through removal of soluble C1q thereby limiting the ability of C1q to mediate immune homeostasis.
Collapse
Affiliation(s)
- Giovanni Franchin
- The Center for Autoimmune and Musculoskeletal Disease, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | | | | | | | | | | |
Collapse
|
37
|
Carlsen AL, Schetter AJ, Nielsen CT, Lood C, Knudsen S, Voss A, Harris CC, Hellmark T, Segelmark M, Jacobsen S, Bengtsson AA, Heegaard NHH. Circulating microRNA expression profiles associated with systemic lupus erythematosus. ACTA ACUST UNITED AC 2013; 65:1324-34. [PMID: 23401079 DOI: 10.1002/art.37890] [Citation(s) in RCA: 184] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 01/24/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the specificity of expression patterns of cell-free circulating microRNAs (miRNAs) in systemic lupus erythematosus (SLE). METHODS Total RNA was purified from plasma, and 45 different specific, mature miRNAs were determined using quantitative reverse transcription-polymerase chain reaction assays. A total of 409 plasma samples were obtained from 364 different patients with SLE, healthy control subjects, and control subjects with other autoimmune diseases. The results in the primary cohort of 62 patients with SLE and 29 healthy control subjects were validated in 2 independent cohorts: a validation cohort comprising 68 patients with SLE and 68 healthy control subjects, and a disease control cohort comprising 20 patients with SLE (19 of whom were from the other validation cohort), 46 healthy control subjects, 38 patients with vasculitis, 18 patients with rheumatoid arthritis, and 20 immunosuppressed patients. RESULTS Seven miRNAs were statistically significantly differentially expressed in plasma from patients with SLE. The expression of miRNA-142-3p (miR-142-3p) and miR-181a was increased, and the expression of miR-106a, miR-17, miR-20a, miR-203, and miR-92a was decreased. In addition, the expression of miR-342-3p, miR-223, and miR-20a was significantly decreased in SLE patients with active nephritis. A predictive model for SLE based on 2 or 4 miRNAs differentiated patients with SLE from control subjects (76% accuracy) when validated independently (P < 2 × 10(-9) ). Use of the 4-miRNA model provided highly significant differentiation between the SLE group and disease controls, except for those with vasculitis. CONCLUSION Circulating miRNAs are systematically altered in SLE. A 4-miRNA signature was diagnostic of SLE, and a specific subset of miRNA profiles was associated with nephritis. All of the signature miRNAs target genes in the transforming growth factor β signaling pathways. Other targets include regulation of apoptosis, cytokine-cytokine receptors, T cell development, and cytoskeletal organization. These findings highlight possible dysregulated pathways in SLE and suggest that circulating miRNA patterns distinguish SLE from other immunoinflammatory phenotypes.
Collapse
|
38
|
Deshpande A, Putcha BDK, Kuruganti S, Walter MR. Kinetic analysis of cytokine-mediated receptor assembly using engineered FC heterodimers. Protein Sci 2013; 22:1100-8. [PMID: 23703950 DOI: 10.1002/pro.2285] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 11/06/2022]
Abstract
A method for analyzing ligand-receptor binding kinetics is described, which is based on an engineered FC domain (FChk) that forms a covalent heterodimer. To validate the system, the type I IFN receptors (IFNAR1 and IFNAR2) were expressed as IFNAR1-FChk, IFNAR2-FCkh, and IFNAR1/IFNAR2-FChk fusion proteins. Surface plasmon resonance (SPR) analysis of binary IFNα2a/IFNAR interactions confirmed prior affinity measurements, while the affinity of the IFNα2a/IFNAR1/IFNAR2-FChk interaction reproduced the affinity of IFNα2a binding to living cells. In cellular assays, IFNAR1/IFNAR2-FChk potently neutralized IFNα2a bioactivity with an inhibitory concentration equivalent to the KD measured by SPR. These studies suggest that FChk provides a simple reagent to evaluate the binding kinetics of multiple ligand-receptor signaling systems that control cell growth, development, and immunity.
Collapse
Affiliation(s)
- Ashlesha Deshpande
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | | | | | | |
Collapse
|
39
|
Oke V, Wahren-Herlenius M. Cutaneous lupus erythematosus: clinical aspects and molecular pathogenesis. J Intern Med 2013; 273:544-54. [PMID: 23464352 DOI: 10.1111/joim.12057] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lupus erythematosus (LE) is an autoimmune disease with diverse clinical manifestations ranging from limited cutaneous (CLE) to potentially life-threatening systemic disease (SLE). Susceptibility to LE arises from genetic variation in multiple loci, and disease activity is provoked by exogenous or endogenous trigger(s), the best characterized of which is exposure to ultraviolet radiation (UVR). Amongst patients with LE, a cluster of photosensitive subjects with cutaneous lesions and positivity for anti-Ro/SSA autoantibodies have been described. The Ro52 antigen belongs to the tripartite motif protein family and has E3 ligase activity. New data reveal that Ro52 ubiquitinates interferon regulatory factors and modulates their transcriptional activity, indicating an important role for Ro52 in inflammation as a negative feedback regulator. Our findings indicate that UVR exposure induces upregulation of Ro52 in the CLE target cell, the keratinocyte, and that Ro52 is upregulated in spontaneous and UVR-induced CLE lesions. Recently described functional analysis of Ro52-deficient mice revealed that loss of Ro52 results in uncontrolled inflammation in response to minor skin injury leading to an LE-like condition. In summary, emerging data suggest that abnormal function or regulation of Ro52 contributes to the pathogenesis of UVR-induced CLE in genetically susceptible individuals. Ro52 may thus be an interesting therapeutic target, as its activation could contribute to downregulation of the chronic inflammatory process in LE. Here, we review the available data on the pathogenesis of CLE and, in particular, the role of the Ro52 autoantigen.
Collapse
Affiliation(s)
- V Oke
- Department of Medicine, Rheumatology Unit, Karolinska Institutet, Karolinska University Hospital in Solna, Stockholm, Sweden
| | | |
Collapse
|
40
|
Lu MC, Lai NS, Chen HC, Yu HC, Huang KY, Tung CH, Huang HB, Yu CL. Decreased microRNA(miR)-145 and increased miR-224 expression in T cells from patients with systemic lupus erythematosus involved in lupus immunopathogenesis. Clin Exp Immunol 2013. [PMID: 23199328 DOI: 10.1111/j.1365-2249.2012.04676.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with abnormal T cell immune responses. We hypothesized that aberrant expression of microRNAs (miRNAs) in T cells may contribute to the pathogenesis of SLE. First, we analysed the expression profiles of 270 human miRNAs in T cells from five SLE patients and five healthy controls and then validated those potentially aberrant-expressed miRNAs using real-time polymerase chain reaction (PCR). Then, the expression of mRNAs regulated by these aberrant-expressed miRNAs was detected using real-time PCR. Finally, miRNA transfection into Jurkat T cells was conducted for confirming further the biological functions of these miRNAs. The initial analysis indicated that seven miRNAs, including miR-145, miR-224, miR-513-5p, miR-150, miR-516a-5p, miR-483-5p and miR-629, were found to be potentially abnormally expressed in SLE T cells. After validation, under-expressed miR-145 and over-expressed miR-224 were noted. We further found that STAT1 mRNA targeted by miR-145 was over-expressed and apoptosis inhibitory protein 5 (API5) mRNA targeted by miR-224 was under-expressed in SLE T cells. Transfection of Jurkat cells with miR-145 suppressed STAT1 and miR-224 transfection suppressed API5 protein expression. Over-expression of miR-224 facilitates activation-induced cell death in Jurkat cells. In the clinical setting, the increased transcript levels of STAT1 were associated significantly with lupus nephritis. In conclusion, we first demonstrated that miR-145 and miR-224 were expressed aberrantly in SLE T cells that modulated the protein expression of their target genes, STAT1 and API5, respectively. These miRNA aberrations accelerated T cell activation-induced cell death by suppressing API5 expression and associated with lupus nephritis by enhancing signal transducer and activator of transcription-1 (STAT)-1 expression in patients with SLE.
Collapse
Affiliation(s)
- M-C Lu
- Division of Allergy, Immunology and Rheumatology, Buddhist Dalin Tzu Chi General Hospital, Chia-Yi, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Over the past 15 years protein kinases have become the pharmaceutical industry's most important class of drug target in the field of cancer. Some 20 drugs that target kinases have been approved for clinical use over the past decade, and hundreds more are undergoing clinical trials. However, the recent approval of the first protein kinase inhibitors for the treatment of inflammatory diseases, coupled with an enhanced understanding of the signaling networks that control the immune system, suggests that there will be a surge of interest in this area over the next 10 years. In this connection, we discuss opportunities for targeting protein kinases in the MyD88 signaling network for the development of drugs to treat chronic inflammatory and autoimmune diseases. Activating mutations in protein kinases underlie many other diseases and conditions, and we also discuss why the protein kinases SPAK/OSR1 and LRRK2 have recently become interesting targets for the treatment of hypertension and Parkinson's disease, respectively, and the progress that has been made in developing LRRK2 inhibitors. Finally we suggest that more focus on the identification of inhibitors of kinase activation, rather than kinase activity, may pay dividends in identifying exquisitely specific inhibitors of signal transduction cascades, and we also highlight "pseudo-kinases" as an attractive and unexplored area for drug development that merits much more attention in the years to come.
Collapse
Affiliation(s)
- Philip Cohen
- MRC Protein Phosphorylation Unit, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Dario R. Alessi
- MRC Protein Phosphorylation Unit, University of Dundee, Dundee DD1 5EH, United Kingdom
| |
Collapse
|
42
|
The function of hematopoietic stem cells is altered by both genetic and inflammatory factors in lupus mice. Blood 2013; 121:1986-94. [PMID: 23315165 DOI: 10.1182/blood-2012-05-433755] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are protected in a metabolically dormant state within the bone marrow stem cell niche. Inflammation has been shown to disrupt HSC dormancy and cause multiple functional changes. Here, we investigated whether HSC functions were altered in systemic lupus erythematosus (SLE)-prone mice and whether this contributed to clinical manifestations of SLE. We found that HSCs were significantly expanded in lupus mice. The increase in HSC cellularity was caused by both genetic lupus risk factors and inflammatory cytokines in lupus mice. In addition, the inflammatory conditions of lupus led to HSC mobilization and lineage-biased hematopoiesis. Strikingly, these functionally altered HSCs possessed robust self-renewal capacity and exhibited repopulating advantages over wild-type HSCs. A single-nucleotide polymorphism in the cdkn2c gene encoding p18(INK4c) within a SLE susceptibility locus was found to account for reduced p18(INK4c) expression and the increase in HSC self-renewal capacity in lupus mice. Lupus HSCs with enhanced self-renewal capacity and resistance to stress may compete out transplanted healthy HSCs, thereby leading to relapses after HSC transplantation.
Collapse
|
43
|
Rauch I, Müller M, Decker T. The regulation of inflammation by interferons and their STATs. JAKSTAT 2013; 2:e23820. [PMID: 24058799 PMCID: PMC3670275 DOI: 10.4161/jkst.23820] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 01/29/2013] [Accepted: 01/29/2013] [Indexed: 12/30/2022] Open
Abstract
Interferons (IFN) are subdivided into type I IFN (IFN-I, here synonymous with IFN-α/β), type II (IFN-γ) and type III IFN (IFN-III/IFN-λ) that reprogram nuclear gene expression through STATs 1 and 2 by forming STAT1 dimers (mainly IFN-γ) or the ISGF3 complex, a STAT1-STAT2-IRF9 heterotrimer (IFN-I and IFN-III). Dominant IFN activities in the immune system are to protect cells from viral replication and to activate macrophages for enhanced effector function. However, the impact of IFN and their STATs on the immune system stretches far beyond these activities and includes the control of inflammation. The goal of this review is to give an overview of the different facets of the inflammatory process that show regulatory input by IFN/STAT.
Collapse
Affiliation(s)
- Isabella Rauch
- Max F. Perutz Laboratories; University of Vienna; Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics and Biomodels Austria; University of Veterinary Medicine Vienna; Vienna, Austria
| | - Thomas Decker
- Max F. Perutz Laboratories; University of Vienna; Vienna, Austria
| |
Collapse
|
44
|
Sun Z, Zhang R, Wang H, Jiang P, Zhang J, Zhang M, Gu L, Yang X, Zhang M, Ji X. Serum IL-10 from systemic lupus erythematosus patients suppresses the differentiation and function of monocyte-derived dendritic cells. J Biomed Res 2012; 26:456-66. [PMID: 23554785 PMCID: PMC3597043 DOI: 10.7555/jbr.26.20120115] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 12/21/2011] [Accepted: 08/29/2012] [Indexed: 11/03/2022] Open
Abstract
The role played by cytokines, other than interferon (IFN)-α, in the differentiation and function of dendritic cells (DCs) in systemic lupus erythematosus (SLE), remains unclear. Serum interleukin-10 (IL-10) levels are generally elevated in SLE patients, which might modulate the differentiation of DCs. In this study, DCs were induced from monocytes either by transendothelial trafficking or by culture with granulocyte-macrophage colony-stimulating factor (GM-CSF) + IL-4 + tumor necrosis factor (TNF)-α. Both systems were used to investigate the effects of elevated serum IL-10 level on DC differentiation in SLE patients. The results showed that monocyte-derived DCs induced by either SLE serum or exogenous IL-10 reduced the expression of human leukocyte antigen (HLA)-DR and CD80, decreased IL-12p40 level, and increased IL-10 level, and exhibited an impaired capacity to stimulate allogenic T-cell proliferation. These results indicate that serum IL-10 may be involved in the pathogenesis of SLE by modulating the differentiation and function of DCs.
Collapse
Affiliation(s)
- Zhida Sun
- Department of Oral Mucosal Diseases, the Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ludigs K, Parfenov V, Du Pasquier RA, Guarda G. Type I IFN-mediated regulation of IL-1 production in inflammatory disorders. Cell Mol Life Sci 2012; 69:3395-418. [PMID: 22527721 PMCID: PMC11115130 DOI: 10.1007/s00018-012-0989-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 03/14/2012] [Accepted: 04/03/2012] [Indexed: 02/07/2023]
Abstract
Although contributing to inflammatory responses and to the development of certain autoimmune pathologies, type I interferons (IFNs) are used for the treatment of viral, malignant, and even inflammatory diseases. Interleukin-1 (IL-1) is a strongly pyrogenic cytokine and its importance in the development of several inflammatory diseases is clearly established. While the therapeutic use of IL-1 blocking agents is particularly successful in the treatment of innate-driven inflammatory disorders, IFN treatment has mostly been appreciated in the management of multiple sclerosis. Interestingly, type I IFNs exert multifaceted immunomodulatory effects, including the reduction of IL-1 production, an outcome that could contribute to its efficacy in the treatment of inflammatory diseases. In this review, we summarize the current knowledge on IL-1 and IFN effects in different inflammatory disorders, the influence of IFNs on IL-1 production, and discuss possible therapeutic avenues based on these observations.
Collapse
Affiliation(s)
- Kristina Ludigs
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland.
| | | | | | | |
Collapse
|
46
|
Tolerogenic versus inflammatory activity of peripheral blood monocytes and dendritic cells subpopulations in systemic lupus erythematosus. Clin Dev Immunol 2012; 2012:934161. [PMID: 22969819 PMCID: PMC3437291 DOI: 10.1155/2012/934161] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 07/10/2012] [Accepted: 07/17/2012] [Indexed: 01/02/2023]
Abstract
Abnormalities in monocytes and in peripheral blood dendritic cells (DC) subsets have been reported in systemic lupus erythematosus (SLE). We aim to clarify the tolerogenic or inflammatory role of these cells based on ICOSL or IFN-α and chemokine mRNA expression, respectively, after cell purification. The study included 18 SLE patients with active disease (ASLE), 25 with inactive disease (ISLE), and 30 healthy controls (HG). In purified plasmacytoid DC (pDC) was observed a lower ICOSL mRNA expression in ASLE and an increase in ISLE; similarly, a lower ICOSL mRNA expression in monocytes of ALSE patients was found. However, a higher ICOSL mRNA expression was observed in ASLE compared to HG in myeloid DCs. Interestingly, clinical parameters seem to be related with ICOSL mRNA expression.
Regarding the inflammatory activity it was observed in purified monocytes and CD14−/low
CD16+ DCs an increase of CCL2, CXCL9, and CXCL10 mRNA expression in ASLE compared to HG. In myeloid DC no differences were observed regarding chemokines, and IFN-α mRNA expression. In pDC, a higher IFN-α mRNA expression was observed in ASLE.
Deviations in ICOSL, chemokine, and IFN-α mRNA expression in peripheral blood monocytes and dendritic cells subpopulations in SLE appear to be related to disease activity.
Collapse
|
47
|
Zhou XJ, Zhang H. Autophagy in immunity: implications in etiology of autoimmune/autoinflammatory diseases. Autophagy 2012; 8:1286-99. [PMID: 22878595 DOI: 10.4161/auto.21212] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Autophagy is now emerging as a spotlight in trafficking events that activate innate and adaptive immunity. It facilitates innate pathogen detection and antigen presentation, as well as pathogen clearance and lymphocyte homeostasis. In this review, we first summarize new insights into its functions in immunity, which underlie its associations with autoimmunity. As some lines of evidence are emerging to support its role in autoimmune and autoinflammatory diseases, we further discuss whether and how it affects autoimmune diseases including systemic lupus erythematosus, rheumatoid arthritis, diabetes mellitus and multiple sclerosis, as well as autoinflammatory diseases, such as Crohn disease and vitiligo.
Collapse
Affiliation(s)
- Xu-Jie Zhou
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | | |
Collapse
|
48
|
Oke V, Wahren-Herlenius M. The immunobiology of Ro52 (TRIM21) in autoimmunity: A critical review. J Autoimmun 2012; 39:77-82. [DOI: 10.1016/j.jaut.2012.01.014] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 01/22/2012] [Indexed: 12/20/2022]
|
49
|
Fairweather D, Petri MA, Coronado MJ, Cooper LT. Autoimmune heart disease: role of sex hormones and autoantibodies in disease pathogenesis. Expert Rev Clin Immunol 2012; 8:269-84. [PMID: 22390491 DOI: 10.1586/eci.12.10] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) and autoimmune diseases (ADs) are the first and third highest causes of death in the USA, respectively. Men have an increased incidence of the majority of CVDs, including atherosclerosis, myocarditis, dilated cardiomyopathy and heart failure. By contrast, nearly 80% of all ADs occur in women. However, in one category of ADs, rheumatic diseases, CVD is the main cause of death. Factors that link rheumatic ADs to CVD are inflammation and the presence of autoantibodies. In this review we will examine recent findings regarding sex differences in the immunopathogenesis of CVD and ADs, explore possible reasons for the increased occurrence of CVD within rheumatic ADs and discuss whether autoantibodies, including rheumatoid factor, could be involved in disease pathogenesis.
Collapse
Affiliation(s)
- DeLisa Fairweather
- Johns Hopkins University Bloomberg School of Public Health, Department of Environmental Health Sciences, 615 N. Wolfe Street, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
50
|
Structural insights into a human anti-IFN antibody exerting therapeutic potential for systemic lupus erythematosus. J Mol Med (Berl) 2012; 90:837-46. [DOI: 10.1007/s00109-012-0866-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 01/09/2012] [Accepted: 01/12/2012] [Indexed: 01/19/2023]
|