Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731877

ABSTRACT

Epstein-Barr virus (EBV) DNA is known to be shed upon reactivation of latent EBV. Based on our previous findings linking Toll-like receptor-9 (TLR9) to an EBV DNA-driven surge in IL-17A production, we aimed to examine the therapeutic potential of TLR9 inhibition in EBV DNA-exacerbated arthritis in a collagen-induced arthritis (CIA) mouse model. C57BL/6J mice were administered either collagen, EBV DNA + collagen, EBV DNA + collagen + TLR9 inhibitor, or only the TLR9 inhibitor. After 70 days, paw thicknesses, clinical scores, and gripping strength were recorded. Moreover, affected joints, footpads, and colons were histologically scored. Furthermore, the number of cells co-expressing IL-17A, IFN-γ, and FOXP3 in joint sections was determined by immunofluorescence assays. Significantly decreased paw thicknesses, clinical scores, and histological scores with a significantly increased gripping strength were observed in the group receiving EBV DNA + collagen + TLR9 inhibitor, compared to those receiving EBV DNA + collagen. Similarly, this group showed decreased IL-17A+ IFN-γ+, IL-17A+ FOXP3+, and IL-17A+ IFN-γ+ FOXP3+ foci counts in joints. We show that inhibiting TLR9 limits the exacerbation of arthritis induced by EBV DNA in a CIA mouse model, suggesting that TLR9 could be a potential therapeutic target for rheumatoid arthritis management in EBV-infected individuals.


Subject(s)
Arthritis, Experimental , DNA, Viral , Disease Models, Animal , Herpesvirus 4, Human , Toll-Like Receptor 9 , Animals , Mice , Arthritis, Experimental/virology , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/virology , DNA, Viral/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/physiology , Interleukin-17/metabolism , Mice, Inbred C57BL , Toll-Like Receptor 9/metabolism
2.
Elife ; 102021 06 03.
Article in English | MEDLINE | ID: mdl-34080972

ABSTRACT

Epstein-Barr virus (EBV) infection is associated with rheumatoid arthritis (RA) in adults, though the nature of the relationship remains unknown. Herein, we have examined the contribution of viral infection to the severity of arthritis in mice. We have provided the first evidence that latent gammaherpesvirus infection enhances clinical arthritis, modeling EBV's role in RA. Mice latently infected with a murine analog of EBV, gammaherpesvirus 68 (γHV68), develop more severe collagen-induced arthritis and a Th1-skewed immune profile reminiscent of human disease. We demonstrate that disease enhancement requires viral latency and is not due to active virus stimulation of the immune response. Age-associated B cells (ABCs) are associated with several human autoimmune diseases, including arthritis, though their contribution to disease is not well understood. Using ABC knockout mice, we have provided the first evidence that ABCs are mechanistically required for viral enhancement of disease, thereby establishing that ABCs are impacted by latent gammaherpesvirus infection and provoke arthritis.


Rheumatoid arthritis is one of the most common autoimmune diseases, leaving patients in pain as their immune system mistakenly attacks the lining of their joints. The precise cause is unknown, but research suggests a link to the Epstein-Barr virus, the agent responsible for mononucleosis (also known as glandular fever). After infection and recovery, the virus remains in the body, lying dormant inside immune 'B cells' which are often responsible for autoimmune diseases. Of particular interest are a sub-group known as 'age-associated B-cells', which are mostly cells left over from fighting past infections such as mononucleosis. Yet, the link between Epstein-Barr virus and rheumatoid arthritis remains hard to investigate because of the long gap between the two diseases: the virus mostly affects children and young people, while rheumatoid arthritis tends to develop in middle age. To investigate how exactly the two conditions are connected, Mouat et al. created a new animal model: they infected young mice with the murine equivalent of the Epstein-Barr virus, and then used a collagen injection to trigger rheumatoid arthritis-like disease once the animals were older. Next, Mouat et al. monitored the paws of the mice, revealing that viral infection early in life worsened arthritis later on. These animals also had more age-associated B cells than normal, and the cells showed signs of participating in inflammation. On the other hand, early viral infection did not make arthritis worse in mice unable to produce age-associated B cells. Taken together, these results suggest that the immune cells are required to enhance the effect of the viral infection on rheumatoid arthritis. This new insight may help to refine current treatments that work by reducing the overall number of B cells. Ultimately, the animal model developed by Mouat et al. could be useful to identify better ways to diagnose, monitor and treat this debilitating disease.


Subject(s)
Arthritis, Experimental/virology , B-Lymphocytes/virology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Latent Infection/virology , Virus Latency , Age Factors , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Case-Control Studies , Cytokines/metabolism , Disease Progression , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/metabolism , Female , Herpesvirus 4, Human/immunology , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Latent Infection/immunology , Latent Infection/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Severity of Illness Index , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/virology
3.
Front Immunol ; 12: 672752, 2021.
Article in English | MEDLINE | ID: mdl-34040613

ABSTRACT

Objective: We recently demonstrated that EBV DNA is correlated with proinflammatory responses in mice and in rheumatoid arthritis (RA) patients; hence, we utilized an RA mouse model to examine whether EBV DNA enhances the risk and severity of arthritis and to assess its immunomodulatory effects. Methods: C57BL/6J mice were treated with collagen (arthritis-inducing agent), EBV DNA 6 days before collagen, EBV DNA 15 days after collagen, Staphylococcus epidermidis DNA 6 days before collagen, EBV DNA alone, or water. Mice were then monitored for clinical signs and affected joints/footpads were histologically analysed. The relative concentration of IgG anti- chicken collagen antibodies and serum cytokine levels of IL-17A and IFNϒ were determined by ELISA. The number of cells co-expressing IL-17A and IFNϒ in joint histological sections was determined by immunofluorescence. Results: The incidence of arthritis was significantly higher in mice that received EBV DNA prior to collagen compared to mice that only received collagen. Similarly, increased clinical scores, histological scores and paw thicknesses with a decreased gripping strength were observed in groups treated with EBV DNA and collagen. The relative concentration of IgG anti-chicken collagen antibodies was significantly increased in the group that received EBV DNA 6 days prior to collagen in comparison to the collagen receiving group. On the other hand, the highest number of cells co-expressing IFNϒ and IL-17A was observed in joints from mice that received both collagen and EBV DNA. Conclusion: EBV DNA increases the incidence and severity of arthritis in a RA mouse model. Targeting mediators triggered by viral DNA may hence be a potential therapeutic avenue.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , DNA, Viral/immunology , Epstein-Barr Virus Infections/immunology , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/virology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/virology , Epstein-Barr Virus Infections/complications , Female , Herpesvirus 4, Human , Incidence , Mice , Mice, Inbred C57BL
4.
PLoS Pathog ; 15(8): e1007993, 2019 08.
Article in English | MEDLINE | ID: mdl-31465513

ABSTRACT

Chikungunya virus (CHIKV) is an arthritogenic alphavirus that acutely causes fever as well as severe joint and muscle pain. Chronic musculoskeletal pain persists in a substantial fraction of patients for months to years after the initial infection, yet we still have a poor understanding of what promotes chronic disease. While replicating virus has not been detected in joint-associated tissues of patients with persistent arthritis nor in various animal models at convalescent time points, viral RNA is detected months after acute infection. To identify the cells that might contribute to pathogenesis during this chronic phase, we developed a recombinant CHIKV that expresses Cre recombinase (CHIKV-3'-Cre). CHIKV-3'-Cre replicated in myoblasts and fibroblasts, and it induced arthritis during the acute phase in mice. Importantly, it also induced chronic disease, including persistent viral RNA and chronic myositis and synovitis similar to wild-type virus. CHIKV-3'-Cre infection of tdTomato reporter mice resulted in a population of tdTomato+ cells that persisted for at least 112 days. Immunofluorescence and flow cytometric profiling revealed that these tdTomato+ cells predominantly were myofibers and dermal and muscle fibroblasts. Treatment with an antibody against Mxra8, a recently defined host receptor for CHIKV, reduced the number of tdTomato+ cells in the chronic phase and diminished the levels of chronic viral RNA, implicating these tdTomato+ cells as the reservoir of chronic viral RNA. Finally, isolation and flow cytometry-based sorting of the tdTomato+ fibroblasts from the skin and ankle and analysis for viral RNA revealed that the tdTomato+ cells harbor most of the persistent CHIKV RNA at chronic time points. Therefore, this CHIKV-3'-Cre and tdTomato reporter mouse system identifies the cells that survive CHIKV infection in vivo and are enriched for persistent CHIKV RNA. This model represents a useful tool for studying CHIKV pathogenesis in the acute and chronic stages of disease.


Subject(s)
Arthritis, Experimental/virology , Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Dermis/pathology , Fibroblasts/pathology , Muscle, Skeletal/pathology , RNA, Viral/metabolism , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Chikungunya Fever/metabolism , Chikungunya virus/genetics , Dermis/metabolism , Dermis/virology , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/virology , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/virology , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , RNA, Viral/genetics , Virus Replication
5.
Sci Immunol ; 4(32)2019 02 22.
Article in English | MEDLINE | ID: mdl-30796092

ABSTRACT

Chikungunya virus (CHIKV) is an emerging mosquito-borne virus that has caused explosive outbreaks worldwide. Although neutralizing monoclonal antibodies (mAbs) against CHIKV inhibit infection in animals, the contribution of Fc effector functions to protection remains unknown. Here, we evaluated the activity of therapeutic mAbs that had or lacked the ability to engage complement and Fcγ receptors (FcγR). When administered as post-exposure therapy in mice, the Fc effector functions of mAbs promoted virus clearance from infected cells and reduced joint swelling-results that were corroborated in antibody-treated transgenic animals lacking activating FcγR. The control of CHIKV infection by antibody-FcγR engagement was associated with an accelerated influx of monocytes. A series of immune cell depletions revealed that therapeutic mAbs required monocytes for efficient clearance of CHIKV infection. Overall, our study suggests that in mice, FcγR expression on monocytes is required for optimal therapeutic activity of antibodies against CHIKV and likely other related viruses.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antibodies, Viral/therapeutic use , Arthritis, Experimental/therapy , Chikungunya virus/immunology , Immunoglobulin Fc Fragments/immunology , Immunologic Factors/therapeutic use , Monocytes/immunology , Receptors, IgG/immunology , Animals , Antibodies, Neutralizing/therapeutic use , Arthritis, Experimental/virology , Chikungunya Fever/virology , Complement Activation/immunology , Complement C1q/immunology , Disease Models, Animal , Epitopes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, Interferon alpha-beta/genetics
6.
Eur J Immunol ; 48(2): 250-257, 2018 02.
Article in English | MEDLINE | ID: mdl-28980305

ABSTRACT

Viruses, particularly the Epstein-Barr virus (EBV) has long been suspected to exacerbate acute arthritic symptoms. However, the cell populations that contribute to import viruses into the inflamed tissues remain to be identified. In the present study, we have investigated the role of monocytes in the transport of Murid herpesvirus 68 (MHV-68), a mouse virus closely related to EBV, using the serum transfer-induced arthritis (STIA) model. We found compelling evidence that MHV-68 infection markedly increased disease severity in NR4A1-/- mice, which are deficient for Ly6Clow monocytes. In contrast, the MHV-68-induced enhancement of joint inflammation was lessened in CCR2-/- mice, suggesting the involvement of inflammatory Ly6Chigh monocytes in viral transport. We also observed that following selective depletion of monocyte subsets by administration of clodronate, MHV-68 transport into the synovium occurs only in the presence of Ly6Chigh monocytes. Tracking of adoptively transferred Ly6Chigh GFP infected monocytes into arthritic CCR2-/- mice by two-photon intravital microscopy showed that this monocyte subset has the capacity to deliver viruses to inflamed AR joints, as confirmed by the detection of viral DNA in inflamed tissues of recipient mice. We thus conclude that Ly6Chigh monocytes import MHV-68 when they are mobilized to the inflamed arthritic joint.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Epstein-Barr Virus Infections/immunology , Herpesviridae Infections/immunology , Herpesvirus 4, Human/physiology , Monocytes/immunology , Rhadinovirus/physiology , Tumor Virus Infections/immunology , Adoptive Transfer , Animals , Antigens, Ly/metabolism , Arthritis, Experimental/virology , Arthritis, Rheumatoid/virology , Cells, Cultured , DNA, Viral/analysis , Disease Models, Animal , Epstein-Barr Virus Infections/virology , Female , Herpesviridae Infections/virology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/transplantation , Muridae , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Receptors, CCR2/genetics , Rhadinovirus/pathogenicity , Tumor Virus Infections/virology
7.
PLoS Pathog ; 13(12): e1006788, 2017 12.
Article in English | MEDLINE | ID: mdl-29281739

ABSTRACT

Chikungunya virus (CHIKV) belongs to a group of mosquito-borne alphaviruses associated with acute and chronic arthropathy, with peripheral and limb joints most commonly affected. Using a mouse model of CHIKV infection and arthritic disease, we show that CHIKV replication and the ensuing foot arthropathy were dramatically reduced when mice were housed at 30°C, rather than the conventional 22°C. The effect was not associated with a detectable fever, but was dependent on type I interferon responses. Bioinformatics analyses of RNA-Seq data after injection of poly(I:C)/jetPEI suggested the unfolded protein response and certain type I interferon responses are promoted when feet are slightly warmer. The ambient temperature thus appears able profoundly to effect anti-viral activity in the periphery, with clear consequences for alphaviral replication and the ensuing arthropathy. These observations may provide an explanation for why alphaviral arthropathies are largely restricted to joints of the limbs and the extremities.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/virology , Arthritis, Experimental/immunology , Arthritis, Experimental/virology , Arthritis, Infectious/immunology , Arthritis, Infectious/virology , Interferon Type I/metabolism , Alphavirus Infections/pathology , Animals , Arthritis, Experimental/pathology , Arthritis, Infectious/pathology , Chikungunya Fever/immunology , Chikungunya Fever/pathology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Chikungunya virus/physiology , Female , Foot , Host-Pathogen Interactions/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Ross River virus/immunology , Ross River virus/pathogenicity , Ross River virus/physiology , Temperature , Viral Load , Virus Replication/immunology , Virus Replication/physiology
8.
Arthritis Rheumatol ; 69(2): 398-409, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27696782

ABSTRACT

OBJECTIVE: Human endogenous retroviruses (HERVs) are remnants of past retroviral infections in the human genome and have been implicated in different aspects of human biology. The aim of this study was to identify HERVs that are associated with the pathogenesis of rheumatic diseases such as systemic lupus erythematosus (SLE). METHODS: The study subjects included 45 female patients with SLE and 50 healthy controls matched for geographic area, age, and sex. Real-time reverse transcription-polymerase chain reaction analysis was used to examine the transcription levels of 11 genes with coding capacity for complete envelope (Env) protein in these individuals. In this way, 1 HERV locus was identified as a potential modulator of autoimmunity. The env gene encoded by this HERV locus was cloned and examined for the ability to express a functional protein with immunosuppressive potential. RESULTS: Expression of the env59 gene was negatively correlated with pathogenetic factors of human autoimmune rheumatic diseases, including such factors as the levels of interleukin-6 (IL-6) and Toll-like receptor 7. This gene was capable of encoding a fully functional Env glycoprotein that was found to contain a domain, the immunosuppressive (ISU) domain, that, when evaluated ex vivo in patients with SLE and those with rheumatoid arthritis as well as in animal models, showed strong antiinflammatory activity, including the ability to lower IL-6 levels. CONCLUSION: The env59 gene has been adapted by the immune system as a control mechanism in autoimmunity. The peptides derived from the ISU domain contained in the Env59 protein may be useful as potentially new biologic treatments in rheumatic diseases such as SLE.


Subject(s)
Arthritis, Experimental/virology , Arthritis, Rheumatoid/virology , Autoimmune Diseases/virology , Endogenous Retroviruses/genetics , Immune Tolerance/genetics , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/virology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Female , Humans , Interleukin-6/biosynthesis , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice
9.
Avian Pathol ; 43(4): 371-8, 2014.
Article in English | MEDLINE | ID: mdl-24980420

ABSTRACT

Turkey arthritis reovirus (TARV) has been isolated from the gastrocnemius tendons and tibiotarsal joint fluid of lame male turkeys >12 weeks old in the Midwest. Two experiments were conducted to compare the pathogenicity in turkeys of three TARVs (TARV-MN2, TARV-MN4 and TARV-O'Neil), one turkey enteric reovirus (TERV strain MN1) and one chicken arthritis reovirus (CARV strain MN1). Two hundred microlitres of virus were inoculated by the oral, intratracheal, or footpad route into 6-day-old poults placed in isolator units. Poults were necropsied at 1 and 4 weeks post infection in Experiment 1, and at 2 and 4 weeks post infection in Experiment 2. Reovirus was detected by reverse transcription-polymerase chain reaction and virus isolation in tendons of TARV-inoculated poults at 1, 2 and 4 weeks post infection. TARV-O'Neil and TARV-MN2 were detected in tendons of sentinal birds at 1 and 4 weeks and 1 week p.i., respectively. In general, TARVs produced lymphocytic tenosynovitis of the gastrocnemius and digital flexor tendon sheaths without inflammation of the tendons proper. In Experiment 1, poults inoculated with TARV-MN2 and TARV-O'Neil had significantly higher gastrocnemius tendon inflammation scores, as determined by histology, than those inoculated with TERV-MN1 or CARV-MN1. In Experiment 2, poults inoculated with TARV-MN2 and TARV-O'Neil had significantly higher gastrocnemius tendon inflammation scores than those inoculated with TARV-MN4 and virus-free medium (negative control group). Koch's postulates was fulfilled when TARV-MN2 and TARV-O'Neil were re-isolated from tendons of poults that had originally been challenged with either of these viruses. Results of these experiments indicate that TARVs have a unique ability to induce gastrocnemius tenosynovitis in turkeys and that administration of TARV-O'Neil through the oral or intratracheal route is a reproducible model to study pathogenesis of TARV infection.


Subject(s)
Antibodies, Viral/blood , Chickens , Orthoreovirus, Avian/pathogenicity , Poultry Diseases/pathology , Reoviridae Infections/veterinary , Turkeys , Animals , Arthritis, Experimental/mortality , Arthritis, Experimental/pathology , Arthritis, Experimental/veterinary , Arthritis, Experimental/virology , Disease Models, Animal , Joints/pathology , Male , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/immunology , Orthoreovirus, Avian/isolation & purification , Poultry Diseases/mortality , Poultry Diseases/virology , RNA, Viral/genetics , Reoviridae Infections/mortality , Reoviridae Infections/pathology , Reoviridae Infections/virology , Tendons/pathology , Tenosynovitis/mortality , Tenosynovitis/pathology , Tenosynovitis/veterinary , Tenosynovitis/virology
10.
Clin Immunol ; 149(3): 487-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24239837

ABSTRACT

Chikungunya virus (CHIKV) recently caused the largest epidemic ever recorded for this virus involving an estimated 1.4-6.5million cases, with imported cased reported in over 40 countries. The number of monoclonal antibodies specific for this re-emerging alphavirus is currently limited. Herein we describe the generation and characterisation of five monoclonal antibodies specific for the E2 glycoprotein of CHIKV. The antibodies detected a range of CHIKV isolates in several assays including ELISA, Western blot, immunofluorescence assay (IFA) and immunohistochemistry (IHC) without evidence of cross-reactivity with other alphaviruses. Four antibodies also neutralised CHIKV in vitro, two of which provided complete protection against arthritis in a CHIKV mouse model when administered prior to infection. Given the current shortage of widely available reagents for CHIKV, these specific antibodies will be useful not only in research, but may also provide the basis for new diagnostics and treatments.


Subject(s)
Alphavirus Infections/prevention & control , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , Arthritis, Experimental/prevention & control , Viral Envelope Proteins/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/biosynthesis , Antibodies, Viral/isolation & purification , Arthritis, Experimental/immunology , Arthritis, Experimental/virology , COS Cells , Chikungunya virus/drug effects , Chikungunya virus/immunology , Chlorocebus aethiops , Female , Humans , Hybridomas/immunology , Mice , Mice, Inbred BALB C , Vero Cells , Viral Envelope Proteins/genetics
11.
J Immunol ; 190(1): 259-69, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23209328

ABSTRACT

Chikungunya virus (CHIKV) is an alphavirus that causes chronic and incapacitating arthralgia in humans. Injury to the joint is believed to occur because of viral and host immune-mediated effects. However, the exact involvement of the different immune mediators in CHIKV-induced pathogenesis is unknown. In this study, we assessed the roles of T cells in primary CHIKV infection, virus replication and dissemination, and virus persistence, as well as in the mediation of disease severity in adult RAG2(-/-), CD4(-/-), CD8(-/-), and wild-type CHIKV C57BL/6J mice and in wild-type mice depleted of CD4(+) or CD8(+) T cells after Ab treatment. CHIKV-specific T cells in the spleen and footpad were investigated using IFN-γ ELISPOT. Interestingly, our results indicated that CHIKV-specific CD4(+), but not CD8(+), T cells are essential for the development of joint swelling without any effect on virus replication and dissemination. Infection in IFN-γ(-/-) mice demonstrated that pathogenic CD4(+) T cells do not mediate inflammation via an IFN-γ-mediated pathway. Taken together, these observations strongly indicate that mechanisms of joint pathology induced by CHIKV in mice resemble those in humans and differ from infections caused by other arthritogenic viruses, such as Ross River virus.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Chikungunya virus/immunology , Adaptive Immunity/genetics , Alphavirus Infections/genetics , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/virology , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/pathology , Cell Movement/genetics , Cell Movement/immunology , Chikungunya Fever , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Vero Cells
12.
Arthritis Rheum ; 63(2): 488-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21280003

ABSTRACT

OBJECTIVE: Mosquito-borne alphaviruses such as chikungunya virus, o'nyong-nyong virus, and Ross River virus (RRV) cause sporadic, sometimes large, outbreaks of rheumatic disease worldwide. This study was designed to test the effect of treating RRV-induced arthritis using the anti-tumor necrosis factor (anti-TNF) drug etanercept in a mouse model of rheumatic disease. METHODS: Mice were infected with RRV and treated with etanercept. Weight gain was measured, tissue viral titers were determined, and histologic changes in muscle and joint tissues were assessed. RESULTS: RRV-infected mice treated with etanercept showed decreased weight gain, higher viral titers in muscle, joints, and blood, and more tissue damage and inflammatory cell recruitment than RRV-infected mice without treatment. CONCLUSION: Anti-TNF therapy is unlikely to be useful in treating alphaviral arthritides. During alphaviral epidemics, careful monitoring of patients being treated with anti-TNF agents may be warranted.


Subject(s)
Alphavirus Infections/drug therapy , Arthritis, Experimental/drug therapy , Immunoglobulin G/toxicity , Immunosuppressive Agents/toxicity , Myositis/drug therapy , Alphavirus/immunology , Alphavirus Infections/complications , Alphavirus Infections/pathology , Animals , Ankle Joint/drug effects , Ankle Joint/pathology , Ankle Joint/virology , Arthritis, Experimental/pathology , Arthritis, Experimental/virology , Disease Models, Animal , Etanercept , Host-Pathogen Interactions/immunology , Longevity/drug effects , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/virology , Myositis/pathology , Myositis/virology , Receptors, Tumor Necrosis Factor , Treatment Outcome , Viral Load , Weight Gain/drug effects
13.
Arthritis Rheum ; 60(8): 2513-23, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19644852

ABSTRACT

OBJECTIVE: Alphaviruses such as chikungunya virus, Sindbis virus, o'nyong-nyong virus, Mayaro virus, and Ross River virus (RRV), are commonly associated with arthralgias and overt arthritides worldwide. Understanding the processes by which arthritogenic viruses cause disease is a prerequisite in the quest for better treatments. In this regard, we have recently established that monocyte/macrophages are mediators of alphavirus-induced arthritis in mice. We hypothesized that chemokines associated with monocyte/macrophage recruitment may play an important role in disease. The aim of the present investigations was to determine whether bindarit, an inhibitor of monocyte chemotactic protein (MCP) synthesis, could ameliorate alphavirus-induced rheumatic disease in mice. METHODS: Using our recently developed mouse model of RRV-induced arthritis, which has many characteristics of RRV disease (RRVD) in humans, the effects of bindarit treatment on RRVD in mice were determined via histologic analyses, immunohistochemistry, flow cytometry, real-time polymerase chain reaction analysis, enzyme-linked immunosorbent assay, and electrophoretic mobility shift assay. RESULTS: Bindarit-treated RRV-infected mice developed mild disease and had substantially reduced tissue destruction and inflammatory cell recruitment as compared with untreated RRV-infected mice. The virus load in the tissues was not affected by bindarit treatment. Bindarit exhibited its activity by down-regulating MCPs, which in turn led to inhibition of cell infiltration and lower production of NF-kappaB and tumor necrosis factor alpha, which are involved in mediating tissue damage. CONCLUSION: Our data support the use of inhibitors of MCP production in the treatment of arthritogenic alphavirus syndromes and suggest that bindarit may be useful in treating RRVD and other alphavirus-induced arthritides in humans.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Indazoles/therapeutic use , Monocyte Chemoattractant Proteins/antagonists & inhibitors , Myositis/drug therapy , Propionates/therapeutic use , Alphavirus/immunology , Alphavirus Infections/complications , Alphavirus Infections/drug therapy , Alphavirus Infections/pathology , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/virology , Cell Line , Disease Models, Animal , Down-Regulation/drug effects , Humans , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Monocyte Chemoattractant Proteins/drug effects , Monocyte Chemoattractant Proteins/genetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myositis/pathology , Myositis/virology , RNA, Messenger/metabolism
14.
J Immunol ; 173(7): 4675-83, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15383603

ABSTRACT

Human parvovirus B19 (B19) often causes acute polyarthritis in adults. In this paper, we analyzed nucleotide sequences of the B19 genome of patients with rheumatoid arthritis (RA), and then introduced the nonstructural protein 1 (NS1) gene of B19 into C57BL/6 mice that had a genetic origin not susceptible to arthritis. The transgenic mice developed no lesions spontaneously, but were susceptible to type II collagen (CII)-induced arthritis. B19 NS1 was expressed in synovial cells on the articular lesions that were histologically characteristic of granulomatous synovitis and pannus formation in cartilage and bone. Serum levels of anti-CII Abs and TNF-alpha increased in NS1 transgenic mice to the same levels as those of DBA/1 mice, which were susceptible to polyarthritis. Stimulation with CII increased secretion of Th1-type- and Th2-type cytokines in NS1 transgenic mice, indicating that a nonpermissive H-2(b) haplotype in the wild type of C57BL/6 mice can be made susceptible to polyarthritis through the expression of NS1. This study is the first to show that a viral agent from the joints in humans can cause CII-induced arthritis resembling RA.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Experimental/virology , Genetic Predisposition to Disease , Mice, Transgenic/virology , Parvovirus B19, Human/genetics , Viral Nonstructural Proteins/genetics , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/virology , Autoantibodies/blood , Base Sequence , Collagen Type II/administration & dosage , Collagen Type II/immunology , Crosses, Genetic , Female , Humans , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymph Nodes/virology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Parvovirus B19, Human/immunology , Synovial Membrane/pathology , Synovial Membrane/virology , Tumor Necrosis Factor-alpha/metabolism , Viral Nonstructural Proteins/administration & dosage
15.
J Immunol ; 172(9): 5656-63, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15100310

ABSTRACT

Viral infections often lead to arthralgias and overt arthritic states. The inflammatogenic compound of the viruses giving rise to such an outcome has to date not been identified. Because expression of dsRNA is a common feature of all viruses, we decided to analyze whether this property leads to the induction of arthritis. Histological signs of arthritis were evident already on day 3 following intra-articular administration of dsRNA. Arthritis was characterized by infiltration of macrophages into synovial tissue. It was not dependent on acquired immune responses because SCID mice also raised joint inflammation. NF-kappa B was activated upon in vitro exposure to dsRNA, indicating its role in the induction/progression of arthritis. Importantly, we found that dsRNA arthritis was triggered through IL-1R signaling because mice being deficient for this molecule were unable to develop joint inflammation. Although dsRNA is typically recognized by Toll-like receptor 3, Toll-like receptor 3 knockout mice developed arthritis, indicating that some other receptors are instrumental in the inducing of inflammation. Our results from in vitro experiments indicate that proinflammatory cytokines and chemokines stimulating monocyte influx were readily triggered in response to stimulation with dsRNA. These findings demonstrate that viral dsRNA is clearly arthritogenic. Importantly, macrophages and their products play an important role in the development of arthritis triggered by dsRNA.


Subject(s)
Arthritis, Experimental/virology , RNA, Double-Stranded/toxicity , RNA, Viral/toxicity , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Female , Injections, Intra-Articular , Interleukin-6/blood , Leukopenia/chemically induced , Leukopenia/immunology , Macrophages/drug effects , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Monocytes/drug effects , NF-kappa B/physiology , Poly I-C/administration & dosage , Poly I-C/toxicity , RNA, Double-Stranded/administration & dosage , RNA, Double-Stranded/chemical synthesis , RNA, Viral/administration & dosage , RNA, Viral/chemical synthesis , Receptors, Cell Surface/physiology , Rotavirus/chemistry , Toll-Like Receptor 3 , Toll-Like Receptors
16.
J Rheumatol ; 25(6): 1118-25, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9632074

ABSTRACT

OBJECTIVE: To evaluate the feasibility of direct in vivo gene transfer in an animal model of arthritis using a retroviral vector. METHODS: The timing and dose of retroviral vector was examined using very high titer retroviral vector (> or = 10(9) CFU) in rat adjuvant arthritis. Retroviral vector expressing beta-galactosidase (beta-gal) or vehicle alone was injected into the right ankle of rats with adjuvant arthritis. Ankles were injected either on Day 7 (pre-arthritis), Day 10 (early arthritis), Day 15 (accelerating arthritis), or Day 28 (chronic arthritis) after adjuvant immunization. Joints were harvested 3 days later and extracts were assayed for beta-gal activity. RESULTS: Synovial beta-gal expression was minimal in the Day 7 group and elevated in the Day 10, Day 15, and Day 28 groups. Gene transfer with retroviral vector did not exacerbate the local inflammatory response. Minimal or no beta-gal expression was observed in the contralateral uninjected paw or in the spleen, lung, liver, and kidneys. Frozen sections of retroviral vector injected joints were stained with X-gal and revealed transduced cells in the lining and superficial sublining layers. To determine the longevity of gene expression, ankle joints were injected with vector on Day 15 post-adjuvant, harvested, and assayed for beta-gal activity for up to 49 days after injection. Expression of the enzyme peaked from Day 3 to 7 and was still readily detected up to 49 days after retrovirus infection. CONCLUSION: This is the first report of successful direct in vivo gene transfer in the rat adjuvant arthritis model using a retroviral vector. Appropriate timing of administration and very high titer retroviral vector preparations are key determinants of adequate gene transduction.


Subject(s)
Arthritis, Experimental/enzymology , Gene Transfer Techniques , Genetic Vectors , Retroviridae/genetics , Synovial Membrane/virology , Animals , Ankle Joint/pathology , Ankle Joint/virology , Arthritis, Experimental/pathology , Arthritis, Experimental/virology , Cells, Cultured , Humans , Lac Operon , Male , Rats , Rats, Inbred Lew , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...