1
|
Laragione T, Harris C, Gulko PS. Magnesium Supplementation Modifies Arthritis Synovial and Splenic Transcriptomic Signatures Including Ferroptosis and Cell Senescence Biological Pathways. Nutrients 2024; 16:4247. [PMID: 39683640 DOI: 10.3390/nu16234247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common systemic autoimmune inflammatory disease that can cause joint damage. We have recently reported that oral magnesium supplementation significantly reduces disease severity and joint damage in models of RA. METHODS In the present study, we analyzed the transcriptome of spleens and synovial tissues obtained from mice with KRN serum-induced arthritis (KSIA) consuming either a high Mg supplemented diet (Mg2800; n = 7) or a normal diet (Mg500; n = 7). Tissues were collected at the end of a 15-day KSIA experiment. RNA was extracted and used for sequencing and analyses. RESULTS There was an enrichment of differentially expressed genes (DEGs) belonging to Reactome and Gene Ontology (GO) pathways implicated in RA pathogenesis such as RHO GTPases, the RUNX1 pathway, oxidative stress-induced senescence, and the senescence-associated secretory phenotype. Actc1 and Nr4a3 were among the genes with the highest expression, while Krt79 and Ffar2 were among the genes with the lowest expression in synovial tissues of the Mg2800 group compared with the Mg500 group. Spleens had an enrichment for the metabolism of folate and pterines and the HSP90 chaperone cycle for the steroid hormone receptor. CONCLUSIONS We describe the tissue transcriptomic consequences of arthritis-protecting Mg supplementation in KSIA mice. These results show that oral Mg supplementation may interfere with the response to oxidative stress and senescence and other processes known to participate in RA pathogenesis. We provide new evidence supporting the disease-suppressing effect of increased Mg intake in arthritis and its potential to become a new addition to the therapeutic options for RA and other autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carolyn Harris
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pércio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
2
|
Burg N, Malpass R, Alex L, Tran M, Englebrecht E, Kuo A, Pannelini T, Minett M, Athukorala K, Worgall T, Faust HJ, Goodman S, Mehta B, Brenner M, Vestweber D, Wei K, Blobel C, Hla T, Salmon JE. Endothelial cell sphingosine 1-phosphate receptor 1 restrains VE-cadherin cleavage and attenuates experimental inflammatory arthritis. JCI Insight 2024; 9:e171467. [PMID: 38855867 PMCID: PMC11382883 DOI: 10.1172/jci.insight.171467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 05/01/2024] [Indexed: 06/11/2024] Open
Abstract
In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.
Collapse
Affiliation(s)
- Nathalie Burg
- Hospital for Special Surgery, New York, New York, USA
| | - Ryan Malpass
- Hospital for Special Surgery, New York, New York, USA
| | - Linda Alex
- Hospital for Special Surgery, New York, New York, USA
| | - Miles Tran
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric Englebrecht
- School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | - Tilla Worgall
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Heather J. Faust
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Susan Goodman
- Hospital for Special Surgery, New York, New York, USA
| | - Bella Mehta
- Hospital for Special Surgery, New York, New York, USA
| | - Michael Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Carl Blobel
- Hospital for Special Surgery, New York, New York, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
3
|
Martínez-Ramos S, García S. An update of murine models and their methodologies in immune-mediated joint damage and pain research. Int Immunopharmacol 2024; 128:111440. [PMID: 38176343 DOI: 10.1016/j.intimp.2023.111440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Murine models have played an indispensable role in the understanding of rheumatic and musculoskeletal disorders (RMD), elucidating the genetic, endocrine and biomechanical pathways involved in joint pathology and associated pain. To date, the available models in RMD can be classified as induced or spontaneous, both incorporating transgenic alternatives that improve specific insights. It is worth noting that the selection of the most appropriate model together with the evaluation of their specific characteristics and technical capabilities are crucial when designing the experiments. Furthermore, it is also imperative to consistently adhere to the ethical standards concerning animal experimentation. Recognizing the inherent limitation that any model can entirely encapsulates the complexity of the pathophysiology of these conditions, the aim of this review is to provide an updated overview on the methodology of current murine models in major arthropathies and their immune-mediated pathways, addressing to basic, translational and pharmacological research in joint damage and pain.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain.
| | - Samuel García
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| |
Collapse
|
4
|
Laragione T, Harris C, Azizgolshani N, Beeton C, Bongers G, Gulko PS. Magnesium increases numbers of Foxp3+ Treg cells and reduces arthritis severity and joint damage in an IL-10-dependent manner mediated by the intestinal microbiome. EBioMedicine 2023; 92:104603. [PMID: 37201335 PMCID: PMC10203746 DOI: 10.1016/j.ebiom.2023.104603] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/10/2023] [Accepted: 04/19/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common autoimmune disease with emerging environmental and microbiome risk factors. The western diet is typically deficient in magnesium (Mg), and there is some evidence suggesting that Mg may have anti-inflammatory properties. But the actual role of Mg supplementation in arthritis or in T cell subsets has not been explored. METHODS We investigated the role of a high Mg diet in two different mouse models of RA induced with the KRN serum, and collagen-induced arthritis. We also characterized the phenotypes of splenocytes, gene expression, and an extensive intestinal microbiome analyses including fecal material transplantation (FMT). FINDINGS The high Mg diet group was significantly protected with reduced arthritis severity and joint damage, and reduced expression of IL-1β, IL-6, and TNFα. The high Mg group also had increased numbers of Foxp3+ Treg cells and IL-10-producing T cells. The high Mg protective effect disappeared in IL-10 knockout mice. FMT from the high Mg diet mice recreated the phenotypes seen in the diet-treated mice, with reduced arthritis severity, increased Foxp3+ Treg, and increased IL-10-producing T cells. Intestinal microbiome analyses using 16S rDNA sequencing revealed diet-specific changes, including reduced levels of RA-associated Prevotella in the high Mg group, while increasing levels of Bacteroides and other bacteria associated with increased production of short-chain fatty acids. Metagenomic analyses implicated additional pathways including L-tryptophan biosynthesis and arginine deiminase. INTERPRETATION We describe a new role for Mg in suppressing arthritis, in expanding Foxp3+ T reg cells and in the production of IL-10, and show that these effects are mediated by the intestinal microbiome. Our discoveries suggest a novel strategy for modifying the intestinal microbiome to treat RA and other autoimmune and inflammatory diseases. FUNDING None.
Collapse
Affiliation(s)
- Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Carolyn Harris
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Nasim Azizgolshani
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, United States
| | - Gerold Bongers
- Microbiome Translational Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Percio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States.
| |
Collapse
|
5
|
Rheumatoid arthritis and non-coding RNAs; how to trigger inflammation. Life Sci 2023; 315:121367. [PMID: 36639050 DOI: 10.1016/j.lfs.2023.121367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/31/2022] [Accepted: 12/31/2022] [Indexed: 01/12/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic and chronic inflammatory disease categorized by continuous synovitis in the joints and systemic inflammatory responses that can cause lifelong disability. The major cause of RA is the dysregulation of the immune response. The development of RA disease includes multiplex association of several interleukins and cells, which leads to synovial cell growth, cartilage and bone damage. The primary stage of RA disease is related to the modification of both the innate and adaptive immune systems, which leads to the formation of autoantibodies. This process results in many damaged molecules and epitope spreading. Both the innate (e.g., dendritic cells, macrophages, and neutrophils) and acquired immune cells (e.g., T and B lymphocytes) will increase and continue the chronic inflammatory condition in the next stages of the RA disease. In recent years, non-coding RNAs have been proved as significant controllers of biological functions, especially immune cell expansion and reactions. Non-coding RNAs were primarily containing microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA). Various studies confirmed non-coding RNAs as hopeful markers for diagnosing and curing RA. This review will describe and cover existing knowledge about RA pathogenesis, which might be favorable for discovering possible ncRNA markers for RA.
Collapse
|
6
|
Jeong D, Kim HS, Kim HY, Kang MJ, Jung H, Oh Y, Kim D, Koh J, Cho SY, Jeon YK, Lee EB, Lee SH, Shin EC, Kim HM, Yi EC, Chung DH. Soluble Fas ligand drives autoantibody-induced arthritis by binding to DR5/TRAIL-R2. eLife 2021; 10:48840. [PMID: 34223817 PMCID: PMC8257255 DOI: 10.7554/elife.48840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 06/23/2021] [Indexed: 11/13/2022] Open
Abstract
To date, no study has demonstrated that soluble Fas ligand (sFasL)-mediated inflammation is regulated via interaction with Fas in vivo. We found that FasL interacts specifically with tumor necrosis factor receptor superfamily (TNFRSF)10B, also known as death receptor (DR)5. Autoantibody-induced arthritis (AIA) was attenuated in FasL (Faslgld/gld)- and soluble FasL (FaslΔs/Δs)-deficient mice, but not in Fas (Faslpr/lpr and Fas–/–)- or membrane FasL (FaslΔm/Δm)-deficient mice, suggesting sFasL promotes inflammation by binding to a Fas-independent receptor. Affinity purification mass spectrometry analysis using human (h) fibroblast-like synovial cells (FLSCs) identified DR5 as one of several proteins that could be the elusive Fas-independent FasL receptor. Subsequent cellular and biochemical analyses revealed that DR5 interacted specifically with recombinant FasL–Fc protein, although the strength of this interaction was approximately 60-fold lower than the affinity between TRAIL and DR5. A microarray assay using joint tissues from mice with arthritis implied that the chemokine CX3CL1 may play an important downstream role of the interaction. The interaction enhanced Cx3cl1 transcription and increased sCX3CL1 production in FLSCs, possibly in an NF-κB-dependent manner. Moreover, the sFasL–DR5 interaction-mediated CX3CL1–CX3CR1 axis initiated and amplified inflammation by enhancing inflammatory cell influx and aggravating inflammation via secondary chemokine production. Blockade of FasL or CX3CR1 attenuated AIA. Therefore, the sFasL–DR5 interaction promotes inflammation and is a potential therapeutic target.
Collapse
Affiliation(s)
- Dongjin Jeong
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Immune Regulation in Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye Sung Kim
- Laboratory of Immune Regulation in Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hye Young Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min Jueng Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science, Seoul, Republic of Korea.,Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyeryeon Jung
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science, Seoul, Republic of Korea.,Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yumi Oh
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Donghyun Kim
- Laboratory of Immune Regulation in Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Yup Cho
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Bong Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyo Lee
- Graduate School of Medical Science and Engineering, Korean Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korean Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ho Min Kim
- Graduate School of Medical Science and Engineering, Korean Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science, Seoul, Republic of Korea.,Technology and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Doo Hyun Chung
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Immune Regulation in Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Galindo-Izquierdo M, Pablos Alvarez JL. Complement as a Therapeutic Target in Systemic Autoimmune Diseases. Cells 2021; 10:cells10010148. [PMID: 33451011 PMCID: PMC7828564 DOI: 10.3390/cells10010148] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/16/2022] Open
Abstract
The complement system (CS) includes more than 50 proteins and its main function is to recognize and protect against foreign or damaged molecular components. Other homeostatic functions of CS are the elimination of apoptotic debris, neurological development, and the control of adaptive immune responses. Pathological activation plays prominent roles in the pathogenesis of most autoimmune diseases such as systemic lupus erythematosus, antiphospholipid syndrome, rheumatoid arthritis, dermatomyositis, and ANCA-associated vasculitis. In this review, we will review the main rheumatologic autoimmune processes in which complement plays a pathogenic role and its potential relevance as a therapeutic target.
Collapse
|
8
|
Lewis BJ, Branch DR. Mouse Models of Rheumatoid Arthritis for Studies on Immunopathogenesis and Preclinical Testing of Fc Receptor-Targeting Biologics. Pharmacology 2020; 105:618-629. [DOI: 10.1159/000508239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/27/2020] [Indexed: 11/19/2022]
Abstract
<b><i>Background:</i></b> Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation, swelling, and pain in the joints and involves systemic complications. Mouse models of RA have been extensively used to model the pathogenesis of RA and to develop effective therapies. Although many components of the immune system have been studied in these models, the role of crystallizable fragment (Fc) gamma receptors (FcγRs) in RA has been sorely neglected. The aim of this review was to introduce the different mouse models of RA and to describe the different drug development strategies that have been tested in these models to target FcγR function, with the focus being on drugs that have been made from the Fc of immunoglobulin G (IgG). <b><i>Summary:</i></b> Evidence suggests that FcγRs play a major role in immune complex-induced inflammation in autoimmune diseases, such as RA. However, there is limited knowledge on the importance of FcγRs in the human disease even though there has been extensive work in mouse models of RA. Numerous mouse models of RA are available, with each model depicting certain aspects of the disease. Induced models of RA have nonspecific immune activation with cartilage-directed autoimmunity, whereas spontaneous models of RA develop without immunization, which results in a more chronic form of arthritis. These models have been used to test FcγR-targeting monoclonal antibodies, intravenous immunoglobulin (IVIg), subcutaneously administered IVIg, and recombinant Fcs for their ability to interact with and modify FcγR function. Recombinant Fcs avidly bind FcγRs and exhibit enhanced therapeutic efficacy in mouse models of RA. <b><i>Key Message:</i></b> The therapeutic utility of targeting FcγRs with recombinant Fcs is great and should be explored in human clinical trials for autoimmune diseases, such as RA.
Collapse
|
9
|
Apel F, Zychlinsky A, Kenny EF. The role of neutrophil extracellular traps in rheumatic diseases. Nat Rev Rheumatol 2019; 14:467-475. [PMID: 29930301 DOI: 10.1038/s41584-018-0039-z] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rheumatic diseases are a collection of disorders defined by the presence of inflammation and destruction of joints and internal organs. A common feature of these diseases is the presence of autoantibodies targeting molecules commonly expressed in neutrophils. These preformed mediators are released by neutrophils but not by other immune cells such as macrophages. Neutrophils, major players in the host innate immune response, initiate a cell death mechanism termed neutrophil extracellular trap (NET) formation as a way to ensnare pathogens. NETs are also a source of released self-molecules found in rheumatic diseases. Subsequently, research on the role of NETs in the onset, progression and resolution of inflammation in rheumatic diseases has intensified. This Review has two aims. First, it aims to highlight the mechanisms required for the generation of NETs, the research landscape of which is rapidly changing. Second, it aims to discuss the role of neutrophils and NETs in systemic lupus erythematosus, vasculitis (specifically anti-neutrophil cytoplasmic autoantibody-associated vasculitis), rheumatoid arthritis and gout. Our goal is to clarify the field of NET research in rheumatic diseases in the hope of improving the therapeutic approaches utilized for these diseases.
Collapse
Affiliation(s)
- Falko Apel
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Arturo Zychlinsky
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany.
| | - Elaine F Kenny
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Berlin, Germany.
| |
Collapse
|
10
|
|
11
|
Ito R, Maruoka S, Gon Y, Katano I, Takahashi T, Ito M, Izuhara K, Nunomura S. Recent Advances in Allergy Research Using Humanized Mice. Int J Mol Sci 2019; 20:ijms20112740. [PMID: 31167385 PMCID: PMC6600417 DOI: 10.3390/ijms20112740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/31/2019] [Accepted: 06/01/2019] [Indexed: 12/18/2022] Open
Abstract
The prevalence rates of allergic diseases are increasing worldwide, particularly in industrial countries. To date, many mouse models have been generated for allergy research; studies conducted using these models have suggested the importance of cross-talk between immune cells and tissue-resident non-immune cells in the onset of allergic diseases. However, there are several differences between the immune systems of rodents and humans, and human studies are limited. Thus, mice reconstituted with human immune cells are a novel tool for the preclinical evaluation of the efficacy and safety of developing drugs. Genetic technologies for generating humanized mice have improved markedly in recent years. In this review, we will discuss recent progress in allergy research using humanized mice and introduce our recent humanized mouse model of airway inflammation in human immune cells.
Collapse
Affiliation(s)
- Ryoji Ito
- Central Institute for Experimental Animals (CIEA), Kawasaki 210-0821, Japan.
| | - Shuichiro Maruoka
- Division of Respiratory Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Yasuhiro Gon
- Division of Respiratory Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Ikumi Katano
- Central Institute for Experimental Animals (CIEA), Kawasaki 210-0821, Japan.
| | - Takeshi Takahashi
- Central Institute for Experimental Animals (CIEA), Kawasaki 210-0821, Japan.
| | - Mamoru Ito
- Central Institute for Experimental Animals (CIEA), Kawasaki 210-0821, Japan.
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga 849-0937, Japan.
| | - Satoshi Nunomura
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga 849-0937, Japan.
| |
Collapse
|
12
|
Schinnerling K, Rosas C, Soto L, Thomas R, Aguillón JC. Humanized Mouse Models of Rheumatoid Arthritis for Studies on Immunopathogenesis and Preclinical Testing of Cell-Based Therapies. Front Immunol 2019; 10:203. [PMID: 30837986 PMCID: PMC6389733 DOI: 10.3389/fimmu.2019.00203] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/23/2019] [Indexed: 01/12/2023] Open
Abstract
Rodent models of rheumatoid arthritis (RA) have been used over decades to study the immunopathogenesis of the disease and to explore intervention strategies. Nevertheless, mouse models of RA reach their limit when it comes to testing of new therapeutic approaches such as cell-based therapies. Differences between the human and the murine immune system make it difficult to draw reliable conclusions about the success of immunotherapies. To overcome this issue, humanized mouse models have been established that mimic components of the human immune system in mice. Two main strategies have been pursued for humanization: the introduction of human transgenes such as human leukocyte antigen molecules or specific T cell receptors, and the generation of mouse/human chimera by transferring human cells or tissues into immunodeficient mice. Recently, both approaches have been combined to achieve more sophisticated humanized models of autoimmune diseases. This review discusses limitations of conventional mouse models of RA-like disease and provides a closer look into studies in humanized mice exploring their usefulness and necessity as preclinical models for testing of cell-based therapies in autoimmune diseases such as RA.
Collapse
Affiliation(s)
- Katina Schinnerling
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Carlos Rosas
- Departamento de Ciencias Morfológicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Lilian Soto
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile.,Unidad de Dolor, Departamento de Medicina, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Ranjeny Thomas
- Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, University of Queensland, Brisbane, QLD, Australia
| | - Juan Carlos Aguillón
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
13
|
Laragione T, Brenner M, Lahiri A, Gao E, Harris C, Gulko PS. Huntingtin-interacting protein 1 (HIP1) regulates arthritis severity and synovial fibroblast invasiveness by altering PDGFR and Rac1 signalling. Ann Rheum Dis 2018; 77:1627-1635. [PMID: 30049830 DOI: 10.1136/annrheumdis-2018-213498] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 11/03/2022]
Abstract
OBJECTIVES While new treatments for rheumatoid arthritis (RA) have markedly improved disease control by targeting immune/inflammatory pathways, current treatments rarely induce remission, underscoring the need for therapies that target other aspects of the disease. Little is known about the regulation of disease severity and joint damage, which are major predictors of disease outcome, and might be better or complementary targets for therapy. In this study, we aimed to discover and characterise a new arthritis severity gene. METHODS An unbiased and phenotype-driven strategy including studies of unique congenic rat strains was used to identify new arthritis severity and joint damage genes. Fibroblast-like synoviocytes (FLS) from rats and patients with RA expressing or not Huntingtin-interacting protein 1 (HIP1) were studied for invasiveness, morphology and cell signalling. HIP1 knockout mice were used in in vivo confirmatory studies. Paired t-test was used. RESULTS DNA sequencing and subcongenic strains studied in pristane-induced arthritis identified a new amino acid changing functional variant in HIP1. HIP1 was required for the increased invasiveness of FLS from arthritic rats and from patients with RA. Knocking down HIP1 expression reduced receptor tyrosine kinase-mediated responses in RA FLS, including RAC1 activation, affecting actin cytoskeleton and cell morphology and interfering with the formation of lamellipodia, consistent with reduced invasiveness. HIP1 knockout mice were protected in KRN serum-induced arthritis and developed milder disease. CONCLUSION HIP1 is a new arthritis severity gene and a potential novel prognostic biomarker and target for therapy in RA.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Biomarkers/metabolism
- Cell Movement/physiology
- Cells, Cultured
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Fibroblasts/physiology
- Humans
- Mice, Knockout
- Polymorphism, Single Nucleotide
- Prognosis
- RNA, Small Interfering/genetics
- Rats
- Receptors, Platelet-Derived Growth Factor/physiology
- Signal Transduction
- Synovial Membrane/pathology
- Synoviocytes/metabolism
- Synoviocytes/pathology
- Synoviocytes/physiology
- rac1 GTP-Binding Protein/physiology
Collapse
Affiliation(s)
- Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Max Brenner
- Center of Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Amit Lahiri
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Erjing Gao
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Carolyn Harris
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Percio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
14
|
Holers VM, Banda NK. Complement in the Initiation and Evolution of Rheumatoid Arthritis. Front Immunol 2018; 9:1057. [PMID: 29892280 PMCID: PMC5985368 DOI: 10.3389/fimmu.2018.01057] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/27/2018] [Indexed: 01/03/2023] Open
Abstract
The complement system is a major component of the immune system and plays a central role in many protective immune processes, including circulating immune complex processing and clearance, recognition of foreign antigens, modulation of humoral and cellular immunity, removal of apoptotic and dead cells, and engagement of injury resolving and tissue regeneration processes. In stark contrast to these beneficial roles, however, inadequately controlled complement activation underlies the pathogenesis of human inflammatory and autoimmune diseases, including rheumatoid arthritis (RA) where the cartilage, bone, and synovium are targeted. Recent studies of this disease have demonstrated that the autoimmune response evolves over time in an asymptomatic preclinical phase that is associated with mucosal inflammation. Notably, experimental models of this disease have demonstrated that each of the three major complement activation pathways plays an important role in recognition of injured joint tissue, although the lectin and amplification pathways exhibit particularly impactful roles in the initiation and amplification of damage. Herein, we review the complement system and focus on its multi-factorial role in human patients with RA and experimental murine models. This understanding will be important to the successful integration of the emerging complement therapeutics pipeline into clinical care for patients with RA.
Collapse
Affiliation(s)
| | - Nirmal K. Banda
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
15
|
Abstract
Autoimmune disorders are characterized by a loss of immune tolerance and consequent autoimmunity-mediated disease manifestation. Experimental models are invaluable research tools helping us to understand disease pathogenesis and to search for novel therapeutics. Animal models of autoimmune diseases consist of two groups, spontaneous and induced models. In this review article, we focus on the induced models of autoimmune diseases. Due to the complex nature of autoimmune disorders, many strategies have been applied for the induction of corresponding experimental models in animals like monkeys, rabbits, rats, and mice. Methodologically, these strategies can be categorized into three categories, namely immunization with autoantigen, transfer of autoimmunity, and induction by environmental factors. In this review article, we aim to provide a comprehensive overview of the field of induced experimental autoimmune diseases. On the one hand, we describe and summarize the different strategies used for induction of experimental autoimmune disease. On the other hand, we discuss how to select a strategy for modeling human disease, including the choice of an appropriate species and method for such an approach.
Collapse
Affiliation(s)
- Xinhua Yu
- Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany; Xiamen-Borstel Joint Laboratory of Autoimmunity, Medical College of Xiamen University, Xiamen, 361102, China.
| | - Frank Petersen
- Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany
| |
Collapse
|
16
|
|
17
|
Atkinson SM, Nansen A. Pharmacological Value of Murine Delayed-type Hypersensitivity Arthritis: A Robust Mouse Model of Rheumatoid Arthritis in C57BL/6 Mice. Basic Clin Pharmacol Toxicol 2016; 120:108-114. [PMID: 27553641 DOI: 10.1111/bcpt.12657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022]
Abstract
In this MiniReview, we summarize the body of knowledge on the delayed-type hypersensitivity arthritis (DTHA) model, a recently developed arthritis model with 100% incidence, low variation and synchronized onset in C57BL/6 (B6) mice, and compare it to other murine arthritis models. It is desirable to have robust arthritis models in B6 mice, as many transgene strains are bred on this background. However, several of the most widely used mouse model of arthritis cannot be induced in B6 mice without the drawback of lower incidence, reduced severity and higher variation, if at all. DTHA is induced by modifying a classical methylated bovine serum albumin (mBSA)-induced DTH response by administering a cocktail of anti-type II collagen antibodies (anti-CII) between immunization and challenge. Arthritis affects one, predefined paw in which acute inflammation and severe arthritis rapidly develop and peak after 4-7 days. Disease is self-resolving over the course of around 3 weeks. Disease manifestations resemble those seen in other arthritis models and include bone erosion, cartilage destruction, oedema, pannus and new bone formation. Induction of DTHA is dependent on CD4+ T cells while B cells are dispensable. The DTHA model is set apart from other murine arthritis models in that it can be induced in B6 mice with 100% incidence and with high and consistent severity. This is the clearest advantage of the model, as the mechanisms of disease and clinical manifestations can be found in other arthritis models. The model holds potential for future modifications that may improve the lack of chronicity.
Collapse
Affiliation(s)
- Sara Marie Atkinson
- Novo Nordisk & LIFE In Vivo Pharmacology Centre & the Danish In Vivo Pharmacology PhD Program, University of Copenhagen, Frederiksberg C, Denmark.,Diabetes Complications Research, Novo Nordisk A/S, Maaloev, Denmark
| | | |
Collapse
|
18
|
Horiuchi T, Tsukamoto H. Complement-targeted therapy: development of C5- and C5a-targeted inhibition. Inflamm Regen 2016; 36:11. [PMID: 29259684 PMCID: PMC5725830 DOI: 10.1186/s41232-016-0013-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/10/2016] [Indexed: 01/27/2023] Open
Abstract
The complement system is a major effector of humoral immunity and natural immunity. The complement system has three independent pathways of complement activation: a classical pathway, an alternative pathway, and a lectin pathway. These pathways converge to a common pathway that activates C3. This pathway also leads to the formation of various bioactive molecules such as C5a and the formation of membrane attack complex on the surface of target cells. In the past, the only preparations with anti-complementary action were C1 inhibitors (C1-INH), but an anti-C5 monoclonal antibody (eculizumab) appeared a few years ago, and this antibody has yielded encouraging results. In addition, a C5a receptor (C5aR) antagonist is in the clinical trial phase, and this antagonist should also prove efficacious. Anti-complement agents have garnered attention as a new treatment strategy for refractory inflammatory diseases.
Collapse
Affiliation(s)
| | - Hiroshi Tsukamoto
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
19
|
Christensen AD, Haase C, Cook AD, Hamilton JA. K/BxN Serum-Transfer Arthritis as a Model for Human Inflammatory Arthritis. Front Immunol 2016; 7:213. [PMID: 27313578 PMCID: PMC4889615 DOI: 10.3389/fimmu.2016.00213] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/17/2016] [Indexed: 12/29/2022] Open
Abstract
The K/BxN serum-transfer arthritis (STA) model is a murine model in which the immunological mechanisms occurring in rheumatoid arthritis (RA) and other arthritides can be studied. To induce K/BxN STA, serum from arthritic transgenic K/BxN mice is transferred to naive mice and manifestations of arthritis occur a few days later. The inflammatory response in the model is driven by autoantibodies against the ubiquitously expressed self-antigen, glucose-6-phosphate isomerase (G6PI), leading to the formation of immune complexes that drive the activation of different innate immune cells such as neutrophils, macrophages, and possibly mast cells. The pathogenesis further involves a range of immune mediators including cytokines, chemokines, complement factors, Toll-like receptors, Fc receptors, and integrins, as well as factors involved in pain and bone erosion. Hence, even though the K/BxN STA model mimics only the effector phase of RA, it still involves a wide range of relevant disease mediators. Additionally, as a murine model for arthritis, the K/BxN STA model has some obvious advantages. First, it has a rapid and robust onset of arthritis with 100% incidence in genetically identical animals. Second, it can be induced in a wide range of strain backgrounds and can therefore also be induced in gene-deficient strains to study the specific importance of disease mediators. Even though G6PI might not be an essential autoantigen, for example, in RA, the K/BxN STA model is a useful tool to understand how autoantibodies, in general, drive the progression of arthritis by interacting with downstream components of the innate immune system. Finally, the model has also proven useful as a model wherein arthritic pain can be studied. Taken together, these features make the K/BxN STA model a relevant one for RA, and it is a potentially valuable tool, especially for the preclinical screening of new therapeutic targets for RA and perhaps other forms of inflammatory arthritis. Here, we describe the molecular and cellular pathways in the development of K/BxN STA focusing on the recent advances in the understanding of the important mechanisms. Additionally, this review provides a comparison of the K/BxN STA model to some other arthritis models.
Collapse
Affiliation(s)
- Anne D Christensen
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia; Novo Nordisk A/S, Måløv, Denmark
| | | | - Andrew D Cook
- Department of Medicine, University of Melbourne , Parkville, VIC , Australia
| | - John A Hamilton
- Department of Medicine, University of Melbourne , Parkville, VIC , Australia
| |
Collapse
|
20
|
Bas DB, Su J, Wigerblad G, Svensson CI. Pain in rheumatoid arthritis: models and mechanisms. Pain Manag 2016; 6:265-84. [PMID: 27086843 DOI: 10.2217/pmt.16.4] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pain is one of the most challenging symptoms for patients with rheumatoid arthritis (RA). RA-related pain is frequently considered to be solely a consequence of inflammation in the joints; however, recent studies show that multiple mechanisms are involved. Indeed, RA pain may start even before the disease manifests, and frequently does not correlate with the degree of inflammation or pharmacological management. In this aspect, animal studies have the potential to provide new insights into the pathology that initiate and maintain pain in RA. The focus of this review is to describe the most commonly used animal models for studies of RA pathology, which have also been utilized in pain research, and to summarize findings providing potential clues to the mechanisms involved in the regulation of RA-induced pain.
Collapse
Affiliation(s)
- Duygu B Bas
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Jie Su
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Gustaf Wigerblad
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Camilla I Svensson
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
21
|
Christensen AD, Haase C, Cook AD, Hamilton JA. Granulocyte colony-stimulating factor (G-CSF) plays an important role in immune complex-mediated arthritis. Eur J Immunol 2016; 46:1235-45. [DOI: 10.1002/eji.201546185] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/22/2015] [Accepted: 01/29/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Anne D. Christensen
- Department of Medicine; University of Melbourne; Parkville Victoria Australia
- Novo Nordisk A/S; Måløv Denmark
| | | | - Andrew D. Cook
- Department of Medicine; University of Melbourne; Parkville Victoria Australia
| | - John A. Hamilton
- Department of Medicine; University of Melbourne; Parkville Victoria Australia
| |
Collapse
|
22
|
Xu Y, Huang Y, Cai D, Liu J, Cao X. Analysis of differences in the molecular mechanism of rheumatoid arthritis and osteoarthritis based on integration of gene expression profiles. Immunol Lett 2015; 168:246-53. [PMID: 26404854 DOI: 10.1016/j.imlet.2015.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND We aimed to elucidate the molecular mechanisms underlying rheumatoid arthritis (RA) and osteoarthritis (OA) and analyze the mechanism differences between them. METHODS The gene expression profile of GSE1919, GSE12021, GSE21959 and GSE48780 were downloaded from Gene Expression Omnibus. Total 165 samples of synovial fibroblasts (118 RA samples, 15 OA samples and 32 normal controls) were used. The differentially expressed genes (DEGs) in RA samples but no differences in OA samples (RA.DEGs) and genes in OA samples but no differences in RA samples (OA.DEGs) were screened using limma package. Functional enrichment analysis was performed using DAVID. Moreover, transcriptional regulatory network (TRN) and microRNA regulatory network were constructed. RESULTS Total 211 RA.DEGs (96 up- and 115 down-regulate) and 497 OA.DEGs (224 up- and 273 down-regulated) were identified. TRN analysis showed that C-ETS-1 and P53 were important transcription factors. C-ETS-1 could interact with matrix metallopeptidase 1 (MMP1) and CD53 while P53 could interact with epidermal growth factor receptor (EGFR) and dual specificity phosphatase 1 (DUSP1). Besides, v-myc avian myelocytomatosis viral oncogene homolog (MYC) and interleukin 1, beta (IL1B) could be regulated by the most microRNAs in microRNA regulatory network. Our study indicates that ETS-1 may contribute to RA progression by up-regulation of MMP1 and result in OA progression via up-regulating CD53. CONCLUSIONS P53 may be involved in the progression of RA and OA via targeting downstream EGFR and DUSP1 respectively. Besides, MYC and IL1B may play an important role in OA progression via the regulation of microRNAs.
Collapse
Affiliation(s)
- YiSheng Xu
- Orthopedics Department, Guangdong Provincial Hospital of Traditional Chinese Medicine, 111 Dade Road, Guangzhou 510120, Guangdong, China.
| | - YongMing Huang
- Orthopedics Department, Guangdong Provincial Hospital of Traditional Chinese Medicine, 55 Neihuanxi Road, Guangzhou 510006, Guangdong, China
| | - DaKe Cai
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangdong, Guangzhou 510095, China
| | - JinWen Liu
- Orthopedics Department, Guangdong Provincial Hospital of Traditional Chinese Medicine, 111 Dade Road, Guangzhou 510120, Guangdong, China
| | - XueWei Cao
- Orthopedics Department, Guangdong Provincial Hospital of Traditional Chinese Medicine, 111 Dade Road, Guangzhou 510120, Guangdong, China
| |
Collapse
|
23
|
Iwata H, Witte M, Samavedam UKSRL, Gupta Y, Shimizu A, Ishiko A, Schröder T, Seeger K, Dahlke M, Rades D, Zillikens D, Ludwig RJ. Radiosensitive Hematopoietic Cells Determine the Extent of Skin Inflammation in Experimental Epidermolysis Bullosa Acquisita. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26202985 DOI: 10.4049/jimmunol.1501003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Animal models have enhanced our understanding of the pathogenesis of autoimmune diseases. For these models, genetically identical, inbred mice have commonly been used. Different inbred mouse strains, however, show a high variability in disease manifestation. Identifying the factors that influence this disease variability could provide unrecognized insights into pathogenesis. We established a novel Ab transfer-induced model of epidermolysis bullosa acquisita (EBA), an autoimmune disease characterized by (muco)-cutaneous blistering caused by anti-type VII collagen (COL7) autoantibodies. Blistering after anti-COL7 IgG (directed against the von Willebrand factor A-like domain 2) transfer showed clear variability among inbred mouse strains, that is, severe cutaneous blistering and inflammation in C57BL/6J and absence of skin lesions in MRL/MpJ mice. The transfer of anti-COL7 IgG into irradiated, EBA-resistant MRL/MpJ mice, rescued by transplantation with bone marrow from EBA-susceptible B6.AK-H2k mice, induced blistering. To the contrary, irradiated EBA-susceptible B6.AK-H2k mice that were rescued using MRL/MpJ bone marrow were devoid of blistering. In vitro, immune complex activation of neutrophils from C57BL/6J or MRL/MpJ mice showed an impaired reactive oxygen species release from the latter, whereas no differences were observed after PMA activation. This finding was paralleled by divergent expression profiles of immune complex-activated neutrophils from either C57BL/6J or MRL/MpJ mice. Collectively, we demonstrate that radiosensitive cells determine the varying extent of skin inflammation and blistering in the end-stage effector phase of EBA.
Collapse
Affiliation(s)
- Hiroaki Iwata
- Department of Dermatology, University of Lübeck, D-23538 Lübeck, Germany
| | - Mareike Witte
- Lübeck Institute of Experimental Dermatology, University of Lübeck, D-23538 Lübeck, Germany
| | | | - Yask Gupta
- Lübeck Institute of Experimental Dermatology, University of Lübeck, D-23538 Lübeck, Germany
| | - Atsushi Shimizu
- First Department of Dermatology, School of Medicine, Faculty of Medicine Toho University, Tokyo 143-8540, Japan
| | - Akira Ishiko
- First Department of Dermatology, School of Medicine, Faculty of Medicine Toho University, Tokyo 143-8540, Japan
| | - Tobias Schröder
- Lübeck Institute of Experimental Dermatology, University of Lübeck, D-23538 Lübeck, Germany
| | - Karsten Seeger
- Department of Chemistry, University of Lübeck, D-23538 Lübeck, Germany; and
| | - Markus Dahlke
- Department of Radiation Oncology, University of Lübeck, D-23538 Lübeck, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University of Lübeck, D-23538 Lübeck, Germany
| | - Detlef Zillikens
- Department of Dermatology, University of Lübeck, D-23538 Lübeck, Germany; Lübeck Institute of Experimental Dermatology, University of Lübeck, D-23538 Lübeck, Germany
| | - Ralf J Ludwig
- Department of Dermatology, University of Lübeck, D-23538 Lübeck, Germany; Lübeck Institute of Experimental Dermatology, University of Lübeck, D-23538 Lübeck, Germany;
| |
Collapse
|
24
|
Sadeghi H, Gupta Y, Möller S, Samavedam UK, Behnen M, Kasprick A, Bieber K, Müller S, Kalies K, de Castro Marques A, Recke A, Schmidt E, Zillikens D, Laskay T, Mariani J, Ibrahim SM, Ludwig RJ. The retinoid-related orphan receptor alpha is essential for the end-stage effector phase of experimental epidermolysis bullosa acquisita. J Pathol 2015; 237:111-22. [DOI: 10.1002/path.4556] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 04/13/2015] [Accepted: 04/23/2015] [Indexed: 12/25/2022]
Affiliation(s)
| | - Yask Gupta
- Department of Dermatology; University of Lübeck; Germany
| | - Steffen Möller
- Department of Dermatology; University of Lübeck; Germany
| | | | - Martina Behnen
- Institute for Medical Microbiology and Hygiene; University of Lübeck; Germany
| | - Anika Kasprick
- Department of Dermatology; University of Lübeck; Germany
| | - Katja Bieber
- Department of Dermatology; University of Lübeck; Germany
| | - Susen Müller
- Department of Dermatology; University of Lübeck; Germany
| | | | | | - Andreas Recke
- Department of Dermatology; University of Lübeck; Germany
| | - Enno Schmidt
- Department of Dermatology; University of Lübeck; Germany
| | | | - Tamás Laskay
- Institute for Medical Microbiology and Hygiene; University of Lübeck; Germany
| | - Jean Mariani
- Sorbonne Universités; UPMC Univ Paris 06, UMR 8256 B2A Biological Adaptation and Ageing Paris France
- CNRS; UMR 8256 B2A Biological Adaptation and Ageing Paris France
| | | | - Ralf J Ludwig
- Department of Dermatology; University of Lübeck; Germany
| |
Collapse
|
25
|
Laragione T, Cheng KF, Tanner MR, He M, Beeton C, Al-Abed Y, Gulko PS. The cation channel Trpv2 is a new suppressor of arthritis severity, joint damage, and synovial fibroblast invasion. Clin Immunol 2015; 158:183-92. [PMID: 25869297 DOI: 10.1016/j.clim.2015.04.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 03/04/2015] [Accepted: 04/02/2015] [Indexed: 12/23/2022]
Abstract
Little is known about the regulation of arthritis severity and joint damage in rheumatoid arthritis (RA). Fibroblast-like synoviocytes (FLS) have a central role in joint damage and express increased levels of the cation channel Trpv2. We aimed at determining the role of Trpv2 in arthritis. Treatment with Trpv2-specific agonists decreased the in vitro invasiveness of FLS from RA patients and arthritic rats and mice. Trpv2 stimulation suppressed IL-1β-induced expression of MMP-2 and MMP-3. Trpv2 agonists, including the new and more potent LER13, significantly reduced disease severity in KRN serum- and collagen-induced arthritis, and reduced histologic joint damage, synovial inflammation, and synovial blood vessel numbers suggesting anti-angiogenic activity. In this first in vivo use of Trpv2 agonists we discovered a new central role for Trpv2 in arthritis. These new compounds have the potential to become new therapies for RA and other diseases associated with inflammation, invasion, and angiogenesis.
Collapse
Affiliation(s)
- Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Kai F Cheng
- Laboratory of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Mark R Tanner
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA; Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mingzhu He
- Laboratory of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Yousef Al-Abed
- Laboratory of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Pércio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
26
|
Zhou HF, Yan H, Hu Y, Springer LE, Yang X, Wickline SA, Pan D, Lanza GM, Pham CTN. Fumagillin prodrug nanotherapy suppresses macrophage inflammatory response via endothelial nitric oxide. ACS NANO 2014; 8:7305-17. [PMID: 24941020 PMCID: PMC4108210 DOI: 10.1021/nn502372n] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/18/2014] [Indexed: 05/19/2023]
Abstract
Antiangiogenesis has been extensively explored for the treatment of a variety of cancers and certain inflammatory processes. Fumagillin, a mycotoxin produced by Aspergillus fumigatus that binds methionine aminopeptidase 2 (MetAP-2), is a potent antiangiogenic agent. Native fumagillin, however, is poorly soluble and extremely unstable. We have developed a lipase-labile fumagillin prodrug (Fum-PD) that eliminated the photoinstability of the compound. Using αvβ3-integrin-targeted perfluorocarbon nanocarriers to deliver Fum-PD specifically to angiogenic vessels, we effectively suppressed clinical disease in an experimental model of rheumatoid arthritis (RA). The exact mechanism by which Fum-PD-loaded targeted nanoparticles suppressed inflammation in experimental RA, however, remained unexplained. We herein present evidence that Fum-PD nanotherapy indirectly suppresses inflammation in experimental RA through the local production of endothelial nitric oxide (NO). Fum-PD-induced NO activates AMP-activated protein kinase (AMPK), which subsequently modulates macrophage inflammatory response. In vivo, NO-induced AMPK activation inhibits mammalian target of rapamycin (mTOR) activity and enhances autophagic flux, as evidenced by p62 depletion and increased autolysosome formation. Autophagy in turn mediates the degradation of IkappaB kinase (IKK), suppressing the NF-κB p65 signaling pathway and inflammatory cytokine release. Inhibition of NO production by N(G)-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, reverses the suppression of NF-κB-mediated inflammatory response induced by Fum-PD nanotherapy. These unexpected results uncover an activity of Fum-PD nanotherapy that may be further explored in the treatment of angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Hui-fang Zhou
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
| | - Huimin Yan
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
| | - Ying Hu
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
| | - Luke E. Springer
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
| | - Xiaoxia Yang
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
| | - Samuel A. Wickline
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
| | - Dipanjan Pan
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
| | - Gregory M. Lanza
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
- Address correspondence to (G. Lanza) , (C. Pham)
| | - Christine T. N. Pham
- Division of Rheumatology and Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States
- Address correspondence to (G. Lanza) , (C. Pham)
| |
Collapse
|
27
|
Kessel C, Nandakumar KS, Peters FB, Gauba V, Schultz PG, Holmdahl R. A single functional group substitution in c5a breaks B cell and T cell tolerance and protects against experimental arthritis. Arthritis Rheumatol 2014; 66:610-21. [PMID: 24574221 DOI: 10.1002/art.38237] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 10/15/2013] [Indexed: 12/26/2022]
Abstract
OBJECTIVE A deficiency in C5 protects against arthritis development. However, there is currently no approach successfully translating these findings into arthritis therapy, as by targeting the key component, C5a. The aim of this study was to develop a vaccination strategy targeting C5a as therapy for patients with rheumatoid arthritis. METHODS An anti-C5a vaccine was generated by incorporating the unnatural amino acid p-nitrophenylalanine (4NPA) into selected sites in the murine C5a molecule. C5a-4NPA variants were screened for their immunogenicity in mice on different arthritis-susceptible class II major histocompatibility complex (MHC) backgrounds. A candidate vaccine was tested for its impact on disease in a murine model of collagen-induced arthritis (CIA). Immunity toward endogenous C5a as well as type II collagen was monitored and characterized. RESULTS Replacing a single tyrosine residue in position 35 (Y(35) ) with 4NPA allowed the generation of an anti-C5a vaccine, which partly protected mice against the development of CIA while strongly ameliorating the severity of clinical disease. Although differing in just 3 atoms from wild-type C5a (wtC5a), C5aY(35) 4NPA induced loss of T cell and B cell tolerance toward the endogenous protein in mice expressing class II MHC H-2(q) molecules. Despite differential B cell epitope recognition, antibodies induced by both wtC5a and C5aY(35) 4NPA neutralized C5a. Thus, anti-wtC5a IgG titers during arthritis priming were potentially of critical importance for disease protection, because high titers of C5a-neutralizing antibodies after disease onset were unable to reverse the course of arthritis. CONCLUSION The results of this study suggest that the most effective anti-C5a treatment in arthritis can be accomplished using a preventive vaccination strategy, and that treatment using conventional biologic or small molecule strategies targeting the C5a/C5aR axis may miss the optimal window for therapeutic intervention during the subclinical priming phase of the disease.
Collapse
|
28
|
Bükülmez H, Khan F, Bartels CF, Murakami S, Ortiz-Lopez A, Sattar A, Haqqi TM, Warman ML. Protective effects of C-type natriuretic peptide on linear growth and articular cartilage integrity in a mouse model of inflammatory arthritis. Arthritis Rheumatol 2014; 66:78-89. [PMID: 24449577 PMCID: PMC4034591 DOI: 10.1002/art.38199] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 09/15/2013] [Indexed: 01/15/2023]
Abstract
Objective The C-type natriuretic peptide (CNP) signaling pathway is a major contributor to postnatal skeletal growth in humans. This study was undertaken to investigate whether CNP signaling could prevent growth delay and cartilage damage in an animal model of inflammatory arthritis. Methods We generated transgenic mice that overexpress CNP (B6.SJL-Col2a1-NPPC) in chondrocytes. We introduced the CNP transgene into mice with experimental systemic inflammatory arthritis (K/BxN T cell receptor [TCR]) and determined the effect of CNP overexpression in chondrocytes on the severity of arthritis, cartilage damage, and linear growth. We also examined primary chondrocyte cultures for changes in gene and protein expression resulting from CNP overexpression. Results K/BxN TCR mice exhibited linear growth delay (P < 0.01) compared to controls, and this growth delay was correlated with the severity of arthritis. Diminished chondrocyte proliferation and matrix production was also seen in K/BxN TCR mice. Compared to non–CNP-transgenic mice, K/BxN TCR mice with overexpressed CNP had milder arthritis, no growth delay, and less cartilage damage. Primary chondrocytes from mice overexpressing CNP were less sensitive to inflammatory cytokines than wild-type mouse chondrocytes. Conclusion CNP overexpression in chondrocytes can prevent endochondral growth delay and protect against cartilage damage in a mouse model of inflammatory arthritis. Pharmacologic or biologic modulation of the CNP signaling pathway may prevent growth retardation and protect cartilage in patients with inflammatory joint diseases, such as juvenile idiopathic arthritis.
Collapse
Affiliation(s)
- Hülya Bükülmez
- MetroHealth Medical Center and Case Western Reserve University, Cleveland, Ohio
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Merlo LMF, Pigott E, DuHadaway JB, Grabler S, Metz R, Prendergast GC, Mandik-Nayak L. IDO2 is a critical mediator of autoantibody production and inflammatory pathogenesis in a mouse model of autoimmune arthritis. THE JOURNAL OF IMMUNOLOGY 2014; 192:2082-2090. [PMID: 24489090 DOI: 10.4049/jimmunol.1303012] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Rheumatoid arthritis and other autoimmune disorders are associated with altered activity of the immunomodulatory enzyme IDO. However, the precise contributions of IDO function to autoimmunity remain unclear. In this article, we examine the effect of two different IDO enzymes, IDO1 and IDO2, on the development of autoimmune arthritis in the KRN preclinical model of rheumatoid arthritis. We find that IDO2, not IDO1, is critical for arthritis development, providing direct evidence of separate in vivo functions for IDO1 and IDO2. Mice null for Ido2 display decreased joint inflammation relative to wild-type mice owing to a reduction in pathogenic autoantibodies and Ab-secreting cells. Notably, IDO2 appears to specifically mediate autoreactive responses, but not normal B cell responses, as total serum Ig levels are not altered and IDO2 knockout mice are able to mount productive Ab responses to model Ags in vitro and in vivo. Reciprocal adoptive transfer studies confirm that autoantibody production and arthritis are modulated by IDO2 expression in a cell type extrinsic to the T cell. Taken together, our results, provide important insights into IDO2 function by defining its pathogenic contributions to autoantibody-mediated autoimmunity.
Collapse
Affiliation(s)
| | | | | | | | | | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood PA USA.,Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia PA USA.,Kimmel Cancer Center, Thomas Jefferson University, Philadelphia PA USA
| | - Laura Mandik-Nayak
- Lankenau Institute for Medical Research, Wynnewood PA USA.,Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia PA USA
| |
Collapse
|
30
|
A Glucose-6-Phosphate Isomerase Peptide Induces T and B Cell–Dependent Chronic Arthritis in C57BL/10 Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1144-1155. [DOI: 10.1016/j.ajpath.2013.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 06/18/2013] [Accepted: 06/19/2013] [Indexed: 01/24/2023]
|
31
|
Clinical presentation, pathogenesis, diagnosis, and treatment of epidermolysis bullosa acquisita. ISRN DERMATOLOGY 2013; 2013:812029. [PMID: 23956869 PMCID: PMC3727188 DOI: 10.1155/2013/812029] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 06/02/2013] [Indexed: 11/18/2022]
Abstract
Epidermolysis bullosa acquisita (EBA) is a chronic mucocutaneous autoimmune skin blistering disease. The pathogenic relevance of autoantibodies targeting type VII collagen (COL7) has been well-documented. Therefore, EBA is a prototypical autoimmune disease with a well-characterized pathogenic relevance of autoantibody binding to the target antigen. EBA is a rare disease with an incidence of 0.2 new cases per million and per year. The current treatment of EBA relies on general immunosuppressive therapy, which does not lead to remission in all cases. Therefore, there is a high, so far unmet medical need for the development of novel therapeutic options. During the last 10 years, several novel in vitro and in vivo models of EBA have been established. These models demonstrated a critical role of the genetic background, T cells, and cytokines for mediating the loss of tolerance towards COL7. Neutrophils, complement activation, Fc gamma receptor engagement, cytokines, several molecules involved in cell signaling, release of reactive oxygen species, and matrix metalloproteinases are crucial for autoantibody-induced tissue injury in EBA. Based on this growing understanding of the diseases' pathogenesis, several potential novel therapeutic targets have emerged. In this review, the clinical presentation, pathogenesis, diagnosis, and current treatment options for EBA are discussed in detail.
Collapse
|
32
|
Shilling RA, Williams JW, Perera J, Berry E, Wu Q, Cummings OW, Sperling AI, Huang H. Autoreactive T and B cells induce the development of bronchus-associated lymphoid tissue in the lung. Am J Respir Cell Mol Biol 2013; 48:406-14. [PMID: 23371062 DOI: 10.1165/rcmb.2012-0065oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis-related interstitial lung disease (RA-ILD) is associated with significant morbidity and mortality. Studies in humans have found that the incidence of bronchus-associated lymphoid tissue (BALT) correlates with the severity of lung injury. However, the mechanisms underlying the development of BALT during systemic autoimmunity remain unknown. We have determined whether systemic autoimmunity in a murine model of autoimmune arthritis can promote the development of BALT by generating a novel murine model derived from K/BxN mice. Transgenic mice with the KRN T-cell receptor specific for the autoantigen, glucose-6-phosphate isomerase (GPI), were crossed with GPI-specific immunoglobulin heavy and light chain knock-in mice, producing mice with a majority of T and B cells specific for the same autoantigen. We found that 67% of these mice demonstrated lymphocytic infiltration in the lungs, localized to either the perivascular or peribronchial regions. Fifty percent of the mice with lymphocytic infiltration manifested lymphoid-like lesions resembling BALT, with distinct T and B cell follicles. The lungs from mice with lymphoid infiltrates had increased numbers of cytokine-producing T cells, including IL-17A(+) T cells and increased major histocompatibility complex Class II expression on B cells. Interestingly, challenge with bleomycin failed to elicit a significant fibrotic response, compared with wild-type control mice. Our data suggest that systemic autoreactivity promotes ectopic lymphoid tissue development in the lung through the cooperation of autoreactive T and B cells. However, these BALT-like lesions may not be sufficient to promote fibrotic lung disease at steady state or after inflammatory challenge.
Collapse
Affiliation(s)
- Rebecca A Shilling
- Center for Immunobiology, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Sato-Hayashizaki A, Ohtsuji M, Lin Q, Hou R, Ohtsuji N, Nishikawa K, Tsurui H, Sudo K, Ono M, Izui S, Shirai T, Takai T, Nishimura H, Hirose S. Presumptive role of 129 strain-derived Sle16 locus in rheumatoid arthritis in a new mouse model with Fcγ receptor type IIb-deficient C57BL/6 genetic background. ACTA ACUST UNITED AC 2013; 63:2930-8. [PMID: 21953083 DOI: 10.1002/art.30485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Fcγ receptor type IIb (FcγRIIb) is a major negative regulator of B cells, and the lack of FcγRIIb expression has been reported to induce systemic lupus erythematosus (SLE) in mice of the C57BL/6 (B6) genetic background. The 129 strain-derived Sle16 locus on the telomeric region of chromosome 1 including polymorphic Fcgr2b confers the predisposition to systemic autoimmunity when present on the B6 background. We undertook this study to examine the effect of the Sle16 locus on autoimmune disease in FcγRIIb-deficient B6 mice. METHODS We established 2 lines of FcγRIIb-deficient B6 congenic mouse strains (KO1 and KO2) by selective backcrossing of the originally constructed FcγRIIb-deficient mice on a hybrid (129×B6) background into a B6 background. Although both lack FcγRIIb expression, the KO1 and KO2 strains carry different lengths of the 129 strain-derived telomeric chromosome 1 segment flanked to the null-mutated Fcgr2b gene; the KO1 strain carries a 129 strain-derived ∼6.3-Mb interval distal from the null-mutated Fcgr2b gene within the Sle16 locus, while this interval in the KO2 strain is of B6 origin. RESULTS Unexpectedly, both strains failed to develop SLE; instead, the KO1 strain, but not the KO2 strain, spontaneously developed severe rheumatoid arthritis (RA) with an incidence reaching >90% at age 12 months. CONCLUSION The current study shows evidence that the epistatic interaction between the Fcgr2b-null mutation and a polymorphic gene(s) in the 129 strain-derived interval located in the distal Sle16 locus contributes to RA susceptibility in a new mouse model with the B6 genetic background, although the participation of other genetic polymorphisms cannot be totally excluded.
Collapse
|
34
|
Lack of association between TRAF1/C5 rs10818488 polymorphism and rheumatoid arthritis in Iranian population. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2013. [DOI: 10.1016/j.ejmhg.2012.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
35
|
Auger JL, Haasken S, Binstadt BA. Autoantibody-mediated arthritis in the absence of C3 and activating Fcγ receptors: C5 is activated by the coagulation cascade. Arthritis Res Ther 2012; 14:R269. [PMID: 23237573 PMCID: PMC3674630 DOI: 10.1186/ar4117] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 12/11/2012] [Indexed: 01/14/2023] Open
Abstract
Introduction The effector functions of immunoglobulin G (IgG) are mediated by interaction of its Fc region with Fc receptors (FcγRs) and/or the complement system. The three main pathways of complement activation converge at C3. However, C3-independent pathways can activate C5 and other downstream complement components during IgG-initiated inflammatory responses. These C3-independent pathways of C5 activation are triggered by activating FcγRs in some systems or can be activated by factors of the coagulation cascade such as thrombin. Here we studied the interplay of C3, C5, and activating FcγRs in a model of spontaneous autoantibody-driven arthritis. Methods We utilized the K/BxN TCR transgenic mouse model of arthritis. We bred K/BxN mice bearing targeted or naturally-occurring mutations in one or more of the genes encoding complement components C3, C5, and FcRγ, the cytoplasmic signaling chain shared by the activating FcγRs. We measured arthritis development, the production of arthritogenic autoantibodies, T cell activation status and cytokine synthesis. In addition, we treated mice with anti-C5 monoclonal antibodies or with the thrombin inhibitor argatroban. Results We have previously shown that genetic deficiency of C5 protects K/BxN mice from the development of arthritis. We found here that C3-deficient K/BxN mice developed arthritis equivalent in severity to C3-sufficient animals. Arthritis also developed normally in K/BxN mice lacking both C3 and FcRγ, but could be ameliorated in these animals by treatment with anti-C5 monoclonal antibody or by treatment with argatroban. Production of arthritogenic autoantibodies, T cell activation, and T cell cytokine production were not affected by the absence of C3, C5, and/or FcRγ. Conclusions In K/BxN mice, C5-dependent autoantibody-driven arthritis can occur in the genetic absence of both complement C3 and activating FcγRs. Our findings suggest that in this setting, thrombin activates C5 to provoke arthritis.
Collapse
|
36
|
Patel HB, Kornerup KN, Sampaio ALF, D'Acquisto F, Seed MP, Girol AP, Gray M, Pitzalis C, Oliani SM, Perretti M. The impact of endogenous annexin A1 on glucocorticoid control of inflammatory arthritis. Ann Rheum Dis 2012; 71:1872-80. [PMID: 22562975 PMCID: PMC3440300 DOI: 10.1136/annrheumdis-2011-201180] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2012] [Indexed: 01/11/2023]
Abstract
OBJECTIVES To establish the role and effect of glucocorticoids and the endogenous annexin A1 (AnxA1) pathway in inflammatory arthritis. METHODS Ankle joint mRNA and protein expression of AnxA1 and its receptors were analysed in naive and arthritic mice by real-time PCR and immunohistochemistry. Inflammatory arthritis was induced with the K/BxN arthritogenic serum in AnxA1(+/+) and AnxA1(-/-) mice; in some experiments, animals were treated with dexamethasone (Dex) or with human recombinant AnxA1 or a protease-resistant mutant (termed SuperAnxA1). Readouts were arthritic score, disease incidence, paw oedema and histopathology, together with pro-inflammatory gene expression. RESULTS All elements of the AnxA1 pathway could be detected in naive joints, with augmentation during ongoing disease, due to the infiltration of immune cells. No difference in arthritis intensity of profile could be observed between AnxA1(+/+) and AnxA1(-/-) mice. Treatment of mice with Dex (10 µg intraperitoneally daily from day 2) afforded potent antiarthritic effects highly attenuated in the knockouts: macroscopic changes were mirrored by histopathological findings and pro-inflammatory gene (eg, Nos2) expression. Presence of proteinase 3 mRNA in the arthritic joints led the authors to test AnxA1 and the mutant SuperAnxA1 (1 µg intraperitoneally daily in both cases from day 2), with the latter one being able to accelerate the resolving phase of the disease. CONCLUSION AnxA1 is an endogenous determinant for the therapeutic efficacy of Dex in inflammatory arthritis. Such an effect can be partially mimicked by application of SuperAnxA1 which may represent the starting point for novel antiarthritic therapeutic strategies.
Collapse
Affiliation(s)
- Hetal B Patel
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| | - Kristin N Kornerup
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| | - Andre' LF Sampaio
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| | - Fulvio D'Acquisto
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| | - Michael P Seed
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| | - Ana Paula Girol
- Department of Biology; Instituto de Biociências, Letras e Ciências Exatas (IBILCE), São Paulo State University, São José do Rio Preto, Brazil
| | - Mohini Gray
- Medical Research Council Centre for Inflammation, University of Edinburgh, Edinburgh, UK Experimental Medicine and Rheumatology, William Harvey Research Institute, London, UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| | - Sonia M Oliani
- Department of Biology; Instituto de Biociências, Letras e Ciências Exatas (IBILCE), São Paulo State University, São José do Rio Preto, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine, London UK
| |
Collapse
|
37
|
Vincent TL, Williams RO, Maciewicz R, Silman A, Garside P. Mapping pathogenesis of arthritis through small animal models. Rheumatology (Oxford) 2012; 51:1931-41. [PMID: 22427408 DOI: 10.1093/rheumatology/kes035] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Animal models have been used for a number of decades to study arthritis and have contributed greatly to unravelling mechanisms of pathogenesis and validating new targets for treatment. All animal models have sets of limitations and over the years there has been natural refinement of existing models as well as creation of new ones. The success of genetic modification in mice has fuelled an exponential increase in the use of murine models for arthritis research and has significantly increased our understanding of disease processes. This review focuses on those rodent models of RA and OA that have current utility and are widely used by the research community. We highlight the subtle but important differences in existing models by positioning them on a pathogenesis map whereby model selection is determined by the specific aspect of disease to be studied. We discuss the evolving challenges in in vivo arthritis studies and our perceived gaps for future new model development. The document includes technical and cost implications of performing the described models, and the ethical considerations of such approaches.
Collapse
Affiliation(s)
- Tonia L Vincent
- Room B526, Institute of Infection, Immunology and Inflammation, Associate Member Wellcome Trust Centre for Molecular Parasitology, College of Medical, Veterinary and Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
In this chapter, we describe the usage of this rheumatoid arthritis model to investigate pain-like behavior in mice, including the assessment of clinical changes and the time-dependent changes in nociceptive behavior during disease progresses.
Collapse
|
39
|
Coutinho AE, Gray M, Brownstein DG, Salter DM, Sawatzky DA, Clay S, Gilmour JS, Seckl JR, Savill JS, Chapman KE. 11β-Hydroxysteroid dehydrogenase type 1, but not type 2, deficiency worsens acute inflammation and experimental arthritis in mice. Endocrinology 2012; 153:234-40. [PMID: 22067318 PMCID: PMC3279737 DOI: 10.1210/en.2011-1398] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glucocorticoids profoundly influence immune responses, and synthetic glucocorticoids are widely used clinically for their potent antiinflammatory effects. Endogenous glucocorticoid action is modulated by the two isozymes of 11β-hydroxysteroid dehydrogenase (11β-HSD). In vivo, 11β-HSD1 catalyzes the reduction of inactive cortisone or 11-dehydrocorticosterone into active cortisol or corticosterone, respectively, thereby increasing intracellular glucocorticoid levels. 11β-HSD2 catalyzes the reverse reaction, inactivating intracellular glucocorticoids. Both enzymes have been postulated to modulate inflammatory responses. In the K/BxN serum transfer model of arthritis, 11β-HSD1-deficient mice showed earlier onset and slower resolution of inflammation than wild-type controls, with greater exostoses in periarticular bone and, uniquely, ganglion cysts, consistent with greater inflammation. In contrast, K/BxN serum arthritis was unaffected by 11β-HSD2 deficiency. In a distinct model of inflammation, thioglycollate-induced sterile peritonitis, 11β-HSD1-deficient mice had more inflammatory cells in the peritoneum, but again 11β-HSD2-deficient mice did not differ from controls. Additionally, compared with control mice, 11β-HSD1-deficient mice showed greater numbers of inflammatory cells in pleural lavages in carrageenan-induced pleurisy with lung pathology consistent with slower resolution. These data suggest that 11β-HSD1 limits acute inflammation. In contrast, 11β-HSD2 plays no role in acute inflammatory responses in mice. Regulation of local 11β-HSD1 expression and/or delivery of substrate may afford a novel approach for antiinflammatory therapy.
Collapse
Affiliation(s)
- Agnes E Coutinho
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Johnsen AK, Valdar W, Golden L, Ortiz-Lopez A, Hitzemann R, Flint J, Mathis D, Benoist C. Genome-wide and species-wide dissection of the genetics of arthritis severity in heterogeneous stock mice. ACTA ACUST UNITED AC 2011; 63:2630-40. [PMID: 21560115 DOI: 10.1002/art.30425] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Susceptibility to inflammatory arthritis is determined by a complex set of environmental and genetic factors, but only a portion of the genetic effect can be explained. Conventional genome-wide screens of arthritis models using crosses between inbred mice have been hampered by the low resolution of results and by the restricted range of natural genetic variation sampled. The aim of this study was to address these limitations by performing a genome-wide screen for determinants of arthritis severity using a genetically heterogeneous cohort of mice. METHODS Heterogeneous stock (HS) mice derive from 8 founder inbred strains by serial intercrossing (n>60), resulting in fine-grained genetic variation. With a cohort of 570 HS mice, we performed a genome-wide screen for determinants of arthritis severity in the K/BxN serum-transfer model. RESULTS We mapped regions on chromosomes 1, 2, 4, 6, 7, and 15 that contain quantitative trait loci influencing arthritis severity at a resolution of a few megabases. In several instances, these regions proved to contain 2 quantitative trait loci: the region on chromosome 2 included the C5 fraction of complement known to be required for K/BxN serum-transfer arthritis but also contained a second adjacent quantitative trait locus, for which an intriguing candidate is Ptgs1 (Cox1). Interesting candidates on chromosome 4 included the Padi family, encoding the peptidyl arginine deiminases responsible for citrulline protein modification; suggestively, Padi2 and Padi4 RNA expression was correlated with arthritis severity in HS mice. CONCLUSION These results provide a broad overview of the genetic variation that controls the severity of K/BxN serum-transfer arthritis and suggest intriguing candidate genes for further study.
Collapse
Affiliation(s)
- Alyssa K Johnsen
- Joslin Diabetes Center and Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cook AD, Turner AL, Braine EL, Pobjoy J, Lenzo JC, Hamilton JA. Regulation of systemic and local myeloid cell subpopulations by bone marrow cell-derived granulocyte-macrophage colony-stimulating factor in experimental inflammatory arthritis. ACTA ACUST UNITED AC 2011; 63:2340-51. [PMID: 21809323 DOI: 10.1002/art.30354] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Even though there are clinical trials assessing granulocyte-macrophage colony-stimulating factor (GM-CSF) blockade in rheumatoid arthritis (RA), questions remain as to how GM-CSF acts as a proinflammatory cytokine. The aims of this study on the regulation of arthritis progression by GM-CSF were to determine the source of the GM-CSF, whether there are systemic effects, the changes in synovial tissue leukocyte populations, and the arthritis model dependence on GM-CSF. METHODS Bone marrow chimeras were used to determine the source of GM-CSF required for the development of collagen-induced arthritis (CIA). The K/BxN serum-transfer model of arthritis was tested in GM-CSF(-/-) mice and using anti-GM-CSF monoclonal antibodies. Cell populations from arthritic mice were assessed by differential staining and flow cytometry. RESULTS In the CIA model, GM-CSF produced by bone marrow-derived cells was required for arthritis development. GM-CSF blockade, while ameliorating the development of CIA, was found to have systemic effects, limiting the increase in circulating Ly-6C(high) monocytes and neutrophils. GM-CSF blockade led to fewer synovial macrophages (both Ly-6C(high) and Ly-6C(low)), neutrophils, and lymphocytes. In the absence of GM-CSF, K/BxN serum-transfer arthritis initially developed normally; however, the numbers of Ly-6C(high) monocytes and synovial macrophages (both Ly-6C(high) and Ly-6C(low)) were again reduced, along with the peak disease severity and maintenance. CONCLUSION GM-CSF is a key player in two arthritis models, participating in interactions between hemopoietic cells, both locally and systemically, to control myeloid cell numbers as well as presumably to "activate" them. These results could be useful for the analysis of current clinical trials targeting GM-CSF in patients with RA.
Collapse
Affiliation(s)
- Andrew D Cook
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
42
|
Kudryavtseva E, Forde TS, Pucker AD, Adarichev VA. Wnt signaling genes of murine chromosome 15 are involved in sex-affected pathways of inflammatory arthritis. ACTA ACUST UNITED AC 2011; 64:1057-68. [PMID: 22005949 DOI: 10.1002/art.33414] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Sex disparities in rheumatoid arthritis (RA) are well documented despite the lack of any known major RA susceptibility genes mapped to sex chromosomes. Murine chromosome 15 carries the sex-affected Pgia8 locus that mediates proteoglycan-induced arthritis, and homologous human loci are associated with RA. This study was undertaken to identify genes/mechanisms implicated in sex disparities in arthritis. METHODS Gene expression analysis was performed using RNA isolated from the paws of male and female Pgia8-congenic mice with collagen antibody-induced arthritis. Results were corroborated by reverse transcription-polymerase chain reaction, and mice were also studied prior to disease onset. Ingenuity Pathways Analysis of the expression patterns and gene functions was used to discover locus-specific and sex-affected signature transcripts. RESULTS We found that the Pgia8 locus regulates antibody-mediated inflammatory arthritis differently in males and females. In Pgia8-congenic males, arthritis severity was 30% less (P < 0.005) than in wild-type males, but the antiinflammatory effect was similar in wild-type and congenic females. Transcriptome analysis indicated that 12 genes within the locus were significantly dysregulated in arthritic joints of congenic mice; expression of these genes was also sex specific. The genes that correlated most highly with arthritis severity included those for collagen triple-helix repeat-containing 1 (Cthrc1), metalloproteinase (Adamts12), R-spondin (Rspo2), and syndecan (Sdc2) (r = 0.87-0.91). The level of Cthrc1 message also correlated with that of the genes for the proinflammatory cytokines interleukin-1β and interleukin-6. CONCLUSION These results indicate that sex-specific disparities in RA are linked to transcriptional regulation of genes involved in cartilage degradation (Adamts12) and canonical and noncanonical Wnt signaling (Cthrc1, Rspo2, Sdc2).
Collapse
|
43
|
KRN/I-Ag7 mouse arthritis is independent of complement C3. J Clin Immunol 2011; 31:857-63. [PMID: 21732014 DOI: 10.1007/s10875-011-9562-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 06/17/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND KRN/I-A(g7) (KxB/N) is a mouse model of inflammatory arthritis, which resembles human rheumatoid arthritis. Arthritis in these animals is caused by autoreactivity to a ubiquitously expressed autoantigen, glucose-6 phosphate isomerase. Tolerance is broken at both the T cell and B cell level. The sera from KRN/I-A(g7) mice can induce mouse arthritis in healthy mice. Complement components of the alternative complement pathway, including C3, have been shown to be required in induction of mouse arthritis by serum transfer. METHODS We have bred KRN/I-A(g7) mice onto a C3-deficient background and followed cohorts for the spontaneous appearance of arthritis. We have also transferred KxB/N serum to B6.I-A ( g7 ) recipients. RESULTS C3-deficient KRN/I-A(g7) mice spontaneously developed severe, destructive arthritis, comparable to that seen in C3-intact KRN/I-A(g7) mice. However, serum transfer experiments confirmed the strong requirement for C3 in the passive model. CONCLUSION The pathogenesis of spontaneous KRN/I-A(g7) arthritis can largely proceed by complement-independent pathways and must have pathology effector mechanisms in addition to those seen in the passive serum transfer model.
Collapse
|
44
|
Patel DA, Puig-Canto A, Challa DK, Perez Montoyo H, Ober RJ, Ward ES. Neonatal Fc receptor blockade by Fc engineering ameliorates arthritis in a murine model. THE JOURNAL OF IMMUNOLOGY 2011; 187:1015-22. [PMID: 21690327 DOI: 10.4049/jimmunol.1003780] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multiple autoimmune diseases are characterized by the involvement of autoreactive Abs in pathogenesis. Problems associated with existing therapeutics such as the delivery of intravenous immunoglobulin have led to interest in developing alternative approaches using recombinant or synthetic methods. Toward this aim, in the current study, we demonstrate that the use of Fc-engineered Abs (Abs that enhance IgG degradation [Abdegs]) to block neonatal FcR (FcRn) through high-affinity, Fc region binding is an effective strategy for the treatment of Ab-mediated disease. Specifically, Abdegs can be used at low, single doses to treat disease in the K/B×N serum transfer model of arthritis using BALB/c mice as recipients. Similar therapeutic effects are induced by 25- to 50-fold higher doses of i.v. Ig. Importantly, we show that FcRn blockade is a primary contributing factor toward the observed reduction in disease severity. The levels of albumin, which is also recycled by FcRn, are not affected by Abdeg delivery. Consequently, Abdegs do not alter FcRn expression levels or subcellular trafficking behavior. The engineering of Ab Fc regions to generate potent FcRn blockers therefore holds promise for the therapy of Ab-mediated autoimmunity.
Collapse
Affiliation(s)
- Dipesh A Patel
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9093, USA
| | | | | | | | | | | |
Collapse
|
45
|
Murray IA, Flaveny CA, Chiaro CR, Sharma AK, Tanos RS, Schroeder JC, Amin SG, Bisson WH, Kolluri SK, Perdew GH. Suppression of cytokine-mediated complement factor gene expression through selective activation of the Ah receptor with 3',4'-dimethoxy-α-naphthoflavone. Mol Pharmacol 2011; 79:508-19. [PMID: 21127131 PMCID: PMC3061363 DOI: 10.1124/mol.110.069369] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 12/01/2010] [Indexed: 01/06/2023] Open
Abstract
We have characterized previously a class of aryl hydrocarbon receptor (AHR) ligand termed selective AHR modulators (SAhRMs). SAhRMs exhibit anti-inflammatory properties, including suppression of cytokine-mediated acute phase genes (e.g., Saa1), through dissociation of non-dioxin-response element (DRE) AHR activity from DRE-dependent xenobiotic gene expression. The partial AHR agonist α-naphthoflavone (αNF) mediates the suppressive, non-DRE dependent effects on SAA1 expression and partial DRE-mediated CYP1A1 induction. These observations suggest that αNF may be structurally modified to a derivative exhibiting only SAhRM activity. A screen of αNF derivatives identifies 3',4'-dimethoxy-αNF (DiMNF) as a candidate SAhRM. Competitive ligand binding validates DiMNF as an AHR ligand, and DRE-dependent reporter assays with quantitative mRNA analysis of AHR target genes reveal minimal agonist activity associated with AHR binding. Consistent with loss of agonist activity, DiMNF fails to promote AHR binding to DRE probes as determined through electromobility shift assay. Importantly, mRNA analysis indicates that DiMNF retains the suppressive capacity of αNF regarding cytokine-mediated SAA1 expression in Huh7 cells. Interestingly, predictive docking modeling suggests that DiMNF adopts a unique orientation within the AHR ligand binding pocket relative to αNF and may facilitate the rational design of additional SAhRMs. Microarray studies with a non-DRE binding but otherwise functional AHR mutant identified complement factor C3 as a potential SAhRM target. We confirmed this observation in Huh7 cells using 10 μM DiMNF, which significantly repressed C3 mRNA and protein. These data expand the classes of AHR ligands exerting DRE-independent anti-inflammatory SAhRM activity, suggesting SAhRMs may have application in the amelioration of inflammatory disorders.
Collapse
Affiliation(s)
- Iain A Murray
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kim JH, Chung DH. CD1d-restricted IFN-γ-secreting NKT cells promote immune complex-induced acute lung injury by regulating macrophage-inflammatory protein-1α production and activation of macrophages and dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 186:1432-41. [PMID: 21191075 DOI: 10.4049/jimmunol.1003140] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immune complex-induced acute lung injury (IC-ALI) has been implicated in various pulmonary disease states. However, the role of NKT cells in IC-ALI remains unknown. Therefore, we explored NKT cell functions in IC-ALI using chicken egg albumin and anti-chicken egg albumin IgG. The bronchoalveolar lavage fluid of CD1d(-/-) and Jα18(-/-) mice contained few Ly6G(+)CD11b(+) granulocytes, whereas levels in B6 mice were greater and were increased further by α-galactosyl ceramide. IFN-γ and MIP-1α production in the lungs was greater in B6 than CD1d(-/-) mice. Adoptive transfer of wild type (WT) but not IFN-γ-, MIP-1α-, or FcγR-deficient NKT cells into CD1d(-/-) mice caused recruitment of inflammatory cells to the lungs. Moreover, adoptive transfer of IFN-γR-deficient NKT cells enhanced MIP-1α production and cell recruitment in the lungs of CD1d(-/-) or CD1d(-/-)IFN-γ(-/-) mice, but to a lesser extent than WT NKT cells. This suggests that IFN-γ-producing NKT cells enhance MIP-1α production in both an autocrine and a paracrine manner. IFN-γ-deficient NKT cells induced less IL-1β and TNF-α production by alveolar macrophages and dendritic cells in CD1d(-/-) mice than did WT NKT cells. Taken together, these data suggest that CD1d-restricted IFN-γ-producing NKT cells promote IC-ALI by producing MIP-1α and enhancing proinflammatory cytokine production by alveolar macrophages and dendritic cells.
Collapse
Affiliation(s)
- Ji Hyung Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | | |
Collapse
|
47
|
JNK1 controls mast cell degranulation and IL-1{beta} production in inflammatory arthritis. Proc Natl Acad Sci U S A 2010; 107:22122-7. [PMID: 21135226 DOI: 10.1073/pnas.1016401107] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease marked by bone and cartilage destruction. Current biologic therapies are beneficial in only a portion of patients; hence small molecules targeting key pathogenic signaling cascades represent alternative therapeutic strategies. Here we show that c-Jun N-terminal kinase (JNK) 1, but not JNK2, is critical for joint swelling and destruction in a serum transfer model of arthritis. The proinflammatory function of JNK1 requires bone marrow-derived cells, particularly mast cells. Without JNK1, mast cells fail to degranulate efficiently and release less IL-1β after stimulation via Fcγ receptors (FcγRs). Pharmacologic JNK inhibition effectively prevents arthritis onset and abrogates joint swelling in established disease. Hence, JNK1 controls mast cell degranulation and FcγR-triggered IL-1β production, in addition to regulating cytokine and matrix metalloproteinase biosynthesis, and is an attractive therapeutic target in inflammatory arthritis.
Collapse
|
48
|
Kimura Y, Zhou L, Miwa T, Song WC. Genetic and therapeutic targeting of properdin in mice prevents complement-mediated tissue injury. J Clin Invest 2010; 120:3545-54. [PMID: 20941861 DOI: 10.1172/jci41782] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The alternative pathway (AP) of complement activation is constitutively active and must be regulated by host proteins to prevent autologous tissue injury. Dysfunction of AP regulatory proteins has been linked to several human inflammatory disorders. Properdin is a positive regulator of AP complement activation that has been shown to extend the half-life of cell surface–bound C3 convertase C3bBb; it may also initiate AP complement activation. Here, we demonstrate a critical role for properdin in autologous tissue injury mediated by AP complement activation. We identified myeloid lineage cells as the principal source of plasma properdin by generating mice with global and tissue-specific knockout of Cfp (which encodes properdin) and by generating BM chimeric mice. Properdin deficiency rescued mice from AP complement–mediated embryonic lethality caused by deficiency of the membrane complement regulator Crry and markedly reduced disease severity in the K/BxN model of arthritis. Ab neutralization of properdin in WT mice similarly ameliorated arthritis development, whereas reconstitution of properdin-null mice with exogenous properdin restored arthritis sensitivity. These data implicate systemic properdin as a key contributor to AP complement–mediated injury and support its therapeutic targeting in complement-dependent human diseases.
Collapse
Affiliation(s)
- Yuko Kimura
- Institute for Translational Medicine and Therapeutics and Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | |
Collapse
|
49
|
Ungethuem U, Haeupl T, Witt H, Koczan D, Krenn V, Huber H, von Helversen TM, Drungowski M, Seyfert C, Zacher J, Pruss A, Neidel J, Lehrach H, Thiesen HJ, Ruiz P, Bläss S. Molecular signatures and new candidates to target the pathogenesis of rheumatoid arthritis. Physiol Genomics 2010; 42A:267-82. [PMID: 20858714 DOI: 10.1152/physiolgenomics.00004.2010] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory joint disease of unknown etiology and pronounced interpatient heterogeneity. To characterize RA at the molecular level and to uncover pathomechanisms, we performed genome-wide gene expression analysis. We identified a set of 1,054 genes significantly deregulated in pair-wise comparisons between RA and osteoarthritis (OA) patients, RA and normal donors (ND), or OA and ND. Correlation analysis revealed gene sets regulated identically in all three groups. As a prominent example secreted phosphoprotein 1 (SPP1) was identified to be significantly upregulated in RA compared with both OA and ND. SPP1 expression was found to correlate with genes expressed during an inflammatory response, T-cell activation and apoptosis, suggesting common underlying regulatory networks. A subclassification of RA patients was achieved on the basis of proteoglycan 4 (PRG4) expression, distinguishing PRG4 high and low expressors and reflecting the heterogeneity of the disease. In addition, we found that low PRG4 expression was associated with a more aggressive disease stage, which is in accordance with PRG4 loss-of-function mutations causing camptodactyly-arthropathy-coxa vara-pericarditis syndrome. Altogether we provide evidence for molecular signatures of RA and RA subclasses, sets of new candidate genes as well as for candidate gene networks, which extend our understanding of disease mechanisms and may lead to an improved diagnosis.
Collapse
Affiliation(s)
- U Ungethuem
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, CharitéUniversitätsmedizin Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mensah KA, Mathian A, Ma L, Xing L, Ritchlin CT, Schwarz EM. Mediation of nonerosive arthritis in a mouse model of lupus by interferon-alpha-stimulated monocyte differentiation that is nonpermissive of osteoclastogenesis. ACTA ACUST UNITED AC 2010; 62:1127-37. [PMID: 20131244 DOI: 10.1002/art.27312] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE In contrast to rheumatoid arthritis (RA), the joint inflammation referred to as Jaccoud's arthritis that occurs in systemic lupus erythematosus (SLE) is nonerosive. Although the mechanism responsible is unknown, the antiosteoclastogenic cytokine interferon-alpha (IFNalpha), whose transcriptome is present in SLE monocytes, may be responsible. This study was undertaken to examine the effects of IFNalpha and lupus on osteoclasts and erosion in the (NZB x NZW)F(1) mouse model of SLE with K/BxN serum-induced arthritis. METHODS Systemic IFNalpha levels in (NZB x NZW)F(1) mice were elevated by administration of AdIFNalpha. SLE disease was marked by anti-double-stranded DNA (anti-dsDNA) antibody titer and proteinuria, and Ifi202 and Mx1 expression represented the IFNalpha transcriptome. Microfocal computed tomography was used to evaluate bone erosions. Flow cytometry for CD11b and CD11c was used to evaluate the frequency of circulating osteoclast precursors (OCPs) and myeloid dendritic cells (DCs) in blood. RESULTS Administration of AdIFNalpha to (NZB x NZW)F(1) mice induced osteopetrosis. (NZB x NZW)F(1) mice without autoimmune disease were fully susceptible to focal erosions in the setting of serum-induced arthritis. However, (NZB x NZW)F(1) mice with high anti-dsDNA antibody titers and the IFNalpha transcriptome were protected against bone erosions. AdIFNalpha pretreatment of NZW mice before K/BxN serum administration also resulted in protection against bone erosion (r(2) = 0.4720, P < 0.01), which was associated with a decrease in the frequency of circulating CD11b+CD11c- OCPs and a concomitant increase in the percentage of CD11b+CD11c+ cells (r(2) = 0.6330, P < 0.05), which are phenotypic of myeloid DCs. CONCLUSION These findings suggest that IFNalpha in SLE shifts monocyte development toward myeloid DCs at the expense of osteoclastogenesis, thereby resulting in decreased bone erosion.
Collapse
Affiliation(s)
- Kofi A Mensah
- University of Rochester Medical Center and University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | | | | | | |
Collapse
|