Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
PLoS Negl Trop Dis ; 16(1): e0010149, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35100271

ABSTRACT

Chikungunya virus (CHIKV) is an emerging mosquito-transmitted alphavirus that leads to acute fever and chronic debilitating polyarthralgia. To date, the mechanism underlying chronic recurrent arthralgia is unknown. In the present study, newborn wild-type C57BL/6 mice were infected with CHIKV, and the virological and pathological features of CHIKV infection were analyzed over a period of 50 days. Acute viral infection was readily established by footpad inoculation of CHIKV at doses ranging from 10 plaque forming unit (PFU) to 106 PFU, during which inoculation dose-dependent viral RNA and skeletal muscle damage were detected in the foot tissues. However, persistent CHIKV was observed only when the mice were infected with a high dose of 106 PFU of CHIKV, in which low copy numbers (103-104) of viral positive strand RNA were continuously detectable in the feet from 29 to 50 dpi, along with a low level and progressive reduction in virus-specific CD8+ T cell responses. In contrast, viral negative strand RNA was detected at 50 dpi but not at 29 dpi and was accompanied by significant local skeletal muscle damage at 50 dpi when mild synovial hyperplasia appeared in the foot joints, although the damage was briefly repaired at 29 dpi. These results demonstrated that a high viral inoculation dose leads to viral persistence and progression to chronic tissue damage after recovery from acute infection. Taken together, these results provide a useful tool for elucidating the pathogenesis of persistent CHIKV infection and viral relapse-associated chronic arthritis.


Subject(s)
Arthralgia/virology , Arthritis/virology , Chikungunya Fever/pathology , Chikungunya virus/immunology , Myositis/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Arthralgia/pathology , Arthritis/pathology , CD8-Positive T-Lymphocytes/immunology , Chikungunya virus/genetics , DNA-Binding Proteins/immunology , Disease Models, Animal , Joints/pathology , Joints/virology , Mice , Mice, Inbred C57BL , Myositis/pathology , RNA, Viral/genetics , RNA, Viral/isolation & purification , Viral Load
2.
Sci Rep ; 11(1): 18578, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34535727

ABSTRACT

Chronic rheumatological manifestations similar to those of rheumatoid arthritis (RA) are described after chikungunya virus infection. We aimed to compare the relevance of joint counts and symptoms to clinical outcomes in RA and chronic chikungunya disease. Forty patients with chronic chikungunya arthralgia and 40 patients with RA were enrolled in a cross-sectional study. The association of tenderness and swelling, clinically assessed in 28 joints, and patient evaluations of pain and musculoskeletal stiffness with modified Health Assessment Questionnaire (HAQ) and quality of life (QoL) assessments were investigated. Tender and swollen joint counts, pain and stiffness scores were all associated with the HAQ disability index in RA (all r > 0.55, p ≤ 0.0002), but only stiffness was significantly associated with disability in chikungunya (r = 0.38, p = 0.02). Joint counts, pain and stiffness were also associated with most QoL domains in RA patients. In contrast, in chikungunya disease, tender joint counts were associated only with one QoL domain and swollen joints for none, while pain and stiffness were associated with several domains. Our results confirm the relevance of joint counts in RA, but suggest that in chronic chikungunya disease, joint counts have more limited value. Stiffness and pain score may be more important to quantify chikungunya arthritis impact.


Subject(s)
Arthritis, Rheumatoid/pathology , Arthritis/pathology , Chikungunya Fever/complications , Joints/pathology , Adult , Arthritis/virology , Chikungunya Fever/pathology , Chikungunya Fever/virology , Chikungunya virus/isolation & purification , Cross-Sectional Studies , Disability Evaluation , Disabled Persons , Female , Humans , Joints/virology , Middle Aged , Pain/etiology , Pain/pathology , Pain/virology , Quality of Life , Severity of Illness Index
3.
J Virol ; 94(9)2020 04 16.
Article in English | MEDLINE | ID: mdl-32102875

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes explosive epidemics of a febrile illness characterized by debilitating arthralgia and arthritis that can endure for months to years following infection. In mouse models, CHIKV persists in joint tissues for weeks to months and is associated with chronic synovitis. Using a recombinant CHIKV strain encoding a CD8+ T cell receptor epitope from ovalbumin, as well as a viral peptide-specific major histocompatibility complex class I tetramer, we interrogated CD8+ T cell responses during CHIKV infection. Epitope-specific CD8+ T cells, which were reduced in Batf3-/- and Wdfy4-/- mice with known defects in antigen cross-presentation, accumulated in joint tissue and the spleen. Antigen-specific ex vivo restimulation assays and in vivo killing assays demonstrated that CD8+ T cells produce cytokine and have cytolytic activity. Despite the induction of a virus-specific CD8+ T cell response, the CHIKV burden in joint-associated tissues and the spleen were equivalent in wild-type (WT) and CD8α-/- mice during both the acute and the chronic phases of infection. In comparison, CD8+ T cells were essential for the control of acute and chronic lymphocytic choriomeningitis virus infection in the joint and spleen. Moreover, adoptive transfer of virus-specific effector CD8+ T cells or immunization with a vaccine that induces virus-specific effector CD8+ T cells prior to infection enhanced the clearance of CHIKV infection in the spleen but had a minimal impact on CHIKV infection in the joint. Collectively, these data suggest that CHIKV establishes and maintains a persistent infection in joint-associated tissue in part by evading CD8+ T cell immunity.IMPORTANCE CHIKV is a reemerging mosquito-transmitted virus that in the last decade has spread into Europe, Asia, the Pacific Region, and the Americas. Joint pain, swelling, and stiffness can endure for months to years after CHIKV infection, and epidemics have a severe economic impact. Elucidating the mechanisms by which CHIKV subverts antiviral immunity to establish and maintain a persistent infection may lead to the development of new therapeutic strategies against chronic CHIKV disease. In this study, we found that CHIKV establishes and maintains a persistent infection in joint-associated tissue in part by evading antiviral CD8+ T cell immunity. Thus, immunomodulatory therapies that improve CD8+ T cell immune surveillance and clearance of CHIKV infection could be a strategy for mitigating chronic CHIKV disease.


Subject(s)
Chikungunya Fever/immunology , Chikungunya virus/metabolism , Joints/virology , Adaptive Immunity/immunology , Adoptive Transfer/methods , Animals , Antibodies, Viral/immunology , Antiviral Agents/therapeutic use , Arthritis/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Chikungunya Fever/metabolism , Chikungunya virus/pathogenicity , Chikungunya virus/physiology , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Immunization , Joints/immunology , Lectins, C-Type , Male , Mice , Receptors, Mitogen
4.
Vet Pathol ; 54(3): 413-424, 2017 05.
Article in English | MEDLINE | ID: mdl-28113037

ABSTRACT

The objective of this work was to comparatively study the tissue tropism and the associated pathology of 2 autochthonous small ruminant lentivirus (SRLV) field strains using an experimental infection in sheep through the bone marrow. Fifteen male, SRLV-free lambs of the Rasa Aragonesa breed were inoculated with strain 697 (nervous tissue origin, animals A1-A6), with strain 496 (articular origin, animals B1-B6), or with uninfected culture medium (C1-C3). Clinical, serologic, and polymerase chain reaction (PCR) evaluations were performed periodically. Two lambs from each infected group and a control animal were euthanized at 134, 273, and 319 days postinfection. Tissues were analyzed by gross and histopathologic evaluation; immunohistochemistry for CD3, CD4, CD8, CD68, and FoxP3 cell markers; lung morphometric evaluation; and tissue proviral quantification by PCR. All infected animals became positive either by enzyme-linked immunosorbent assay and/or PCR, with group B lambs showing the highest serologic values and more consistently positive PCR reactions. Group A lambs showed representative lung lesions but only mild histopathologic changes in the central nervous system (CNS) or in carpal joints. Contrarily, group B lambs demonstrated intense carpal arthritis and interstitial pneumonia but an absence of lesions in the CNS. Proviral copies in tissues were detected only in group B lambs. Experimental infection with these SRLV strains indicates that strain 496 is more virulent than strain 697 and more prone to induce arthritis, whereas strain 697 is more likely to reproduce encephalitis in Rasa Aragonesa lambs. Host factors as well as viral factors are responsible for the final clinicopathologic picture during SRLV infections.


Subject(s)
Bone Marrow/virology , Lentivirus Infections/veterinary , Lentiviruses, Ovine-Caprine/pathogenicity , Viral Tropism , Animals , Bone Marrow/pathology , Central Nervous System/pathology , Central Nervous System/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Joints/pathology , Joints/virology , Lentivirus Infections/pathology , Lentivirus Infections/virology , Lung/pathology , Lung/virology , Male , Real-Time Polymerase Chain Reaction/veterinary , Sheep/virology , Viral Tropism/physiology
5.
Sci Rep ; 6: 37603, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27886209

ABSTRACT

The use of IL-2/JES6-1 Ab complex (IL-2 Ab Cx) has been considered as a potential therapeutic for inflammatory diseases due to its selective expansion of regulatory T cells (Tregs) in mice. Here, IL-2 Ab Cx was explored as a therapeutic agent to reduce joint inflammation induced by chikungunya virus, an alphavirus causing debilitating joint disease globally. Virus-infected mice treated with IL-2 Ab Cx exhibited exacerbated joint inflammation due to infiltration of highly activated CD4+ effector T cells (Teffs). Virus infection led to upregulation of CD25 on the Teffs, rendering them sensitive towards IL2 Ab Cx. Ready responsiveness of Teffs to IL-2 was further demonstrated in healthy human donors, suggesting that the use of IL-2 Ab Cx in humans is not suitable. Changes in IL-2 sensitivity during active virus infection could change the responsive pattern towards the IL-2 Ab Cx, resulting in the expansion of pro-inflammatory rather than anti-inflammatory responses.


Subject(s)
Antibodies/metabolism , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/physiology , Inflammation Mediators/metabolism , Interleukin-2/metabolism , Animals , Antigens, CD/metabolism , Female , Forkhead Transcription Factors/metabolism , Interleukin-2/administration & dosage , Interleukin-2/pharmacology , Joints/pathology , Joints/virology , Lymph Nodes/pathology , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
J Virol ; 89(15): 7955-69, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25995257

ABSTRACT

UNLABELLED: Chikungunya virus (CHIKV) is a mosquito-borne arthralgic alphavirus that has garnered international attention as an important emerging pathogen since 2005. More recently, it invaded the Caribbean islands and the Western Hemisphere. Intriguingly, the current CHIKV outbreak in the Caribbean is caused by the Asian CHIKV genotype, which differs from the La Réunion LR2006 OPY1 isolate belonging to the Indian Ocean lineage. Here, we adopted a systematic and comparative approach against LR2006 OPY1 to characterize the pathogenicity of the Caribbean CNR20235 isolate and consequential host immune responses in mice. Ex vivo infection using primary mouse tail fibroblasts revealed a weaker replication efficiency by CNR20235 isolate. In the CHIKV mouse model, CNR20235 infection induced an enervated joint pathology characterized by moderate edema and swelling, independent of mononuclear cell infiltration. Based on systemic cytokine analysis, localized immunophenotyping, and gene expression profiles in the popliteal lymph node and inflamed joints, two pathogenic phases were defined for CHIKV infection: early acute (2 to 3 days postinfection [dpi]) and late acute (6 to 8 dpi). Reduced joint pathology during early acute phase of CNR20235 infection was associated with a weaker proinflammatory Th1 response and natural killer (NK) cell activity. The pathological role of NK cells was further demonstrated as depletion of NK cells reduced joint pathology in LR2006 OPY1. Taken together, this study provides evidence that the Caribbean CNR20235 isolate has an enfeebled replication and induces a less pathogenic response in the mammalian host. IMPORTANCE: The introduction of CHIKV in the Americas has heightened the risk of large-scale outbreaks due to the close proximity between the United States and the Caribbean. The immunopathogenicity of the circulating Caribbean CHIKV isolate was explored, where it was demonstrated to exhibit reduced infectivity resulting in a weakened joint pathology. Analysis of serum cytokine levels, localized immunophenotyping, and gene expression profiles in the organs revealed that a limited Th1 response and reduced NK cells activity could underlie the reduced pathology in the host. Interestingly, higher asymptomatic infections were observed in the Caribbean compared to the La Réunion outbreaks in 2005 and 2006. This is the first study that showed an association between key proinflammatory factors and pathology-mediating leukocytes with a less severe pathological outcome in Caribbean CHIKV infection. Given the limited information regarding the sequela of Caribbean CHIKV infection, our study is timely and will aid the understanding of this increasingly important disease.


Subject(s)
Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/isolation & purification , Joints/immunology , Killer Cells, Natural/immunology , Th1 Cells/immunology , Animals , Caribbean Region/epidemiology , Chikungunya Fever/epidemiology , Chikungunya Fever/pathology , Chikungunya virus/genetics , Chikungunya virus/physiology , Cytokines/immunology , Disease Models, Animal , Female , Humans , Joints/pathology , Joints/virology , Killer Cells, Natural/virology , Mice , Mice, Inbred C57BL , Reunion/epidemiology , Th1 Cells/virology
7.
J Virol ; 87(24): 13878-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24131709

ABSTRACT

Chikungunya virus (CHIKV) is a reemerging mosquito-borne pathogen that causes incapacitating disease in humans characterized by intense joint pain that can persist for weeks, months, or even years. Although there is some evidence of persistent CHIKV infection in humans suffering from chronic rheumatologic disease symptoms, little is known about chronic disease pathogenesis, and no specific therapies exist for acute or chronic CHIKV disease. To investigate mechanisms of chronic CHIKV-induced disease, we utilized a mouse model and defined the duration of CHIKV infection in tissues and the associated histopathological changes. Although CHIKV RNA was readily detectable in a variety of tissues very early after infection, CHIKV RNA persisted specifically in joint-associated tissues for at least 16 weeks. Inoculation of Rag1(-/-) mice, which lack T and B cells, resulted in higher viral levels in a variety of tissues, suggesting that adaptive immunity controls the tissue specificity and persistence of CHIKV infection. The presence of CHIKV RNA in tissues of wild-type and Rag1(-/-) mice was associated with histopathological evidence of synovitis, arthritis, and tendonitis; thus, CHIKV-induced persistent arthritis is not mediated primarily by adaptive immune responses. Finally, we show that prophylactic administration of CHIKV-specific monoclonal antibodies prevented the establishment of CHIKV persistence, whereas therapeutic administration had tissue-specific efficacy. These findings suggest that chronic musculoskeletal tissue pathology is caused by persistent CHIKV infection and controlled by adaptive immune responses. Our results have significant implications for the development of strategies to mitigate the disease burden associated with CHIKV infection in humans.


Subject(s)
Adaptive Immunity , Alphavirus Infections/immunology , Arthralgia/immunology , Chikungunya virus/physiology , Alphavirus Infections/drug therapy , Alphavirus Infections/pathology , Alphavirus Infections/virology , Animals , Antibodies, Viral/therapeutic use , Arthralgia/drug therapy , Arthralgia/pathology , Arthralgia/virology , Chikungunya Fever , Chronic Disease , Disease Models, Animal , Female , Humans , Joints/immunology , Joints/virology , Male , Mice , Mice, Inbred C57BL
8.
Clin Exp Rheumatol ; 31(3 Suppl 77): 47-53, 2013.
Article in English | MEDLINE | ID: mdl-23739526

ABSTRACT

OBJECTIVES: The major role of herpes simplex virus (HSV) type 1 infection in Behçet's disease (BD) immunopathogenesis has been demonstrated and inoculating the earlobes of ICR mice with HSV produced a BD-like mouse model. (18)Ffluorodeoxyglucose positron emission tomography (FDG PET) is widely used for diagnosing numerous human diseases other than malignancies. The aim of our study was to evaluate the inflammatory activities of BD-like symptoms in a HSV type 1-induced BD-like mouse model by small-animal FDG PET. METHODS: Five HSV-infected ICR mice with BD-like lesions, two asymptomatic HSV-infected mice, and two untreated mice were scanned with microPET, and autopsy specimens were histopathologically assessed to evaluate for infiltration by mixed inflammatory cells. RESULTS: The histopathological evaluation of the inflammatory process in knee and elbow joints significantly correlated with the quantitative assessment of FDG accumulation in the same joints in BD-like ICR mice, HSV-infected asymptomatic mice, and untreated control mice. Small-animal FDG PET clearly detected asymptomatic joint inflammatory processes in both BD-like mice and HSV-infected asymptomatic mice. In addition, genital ulcers and skin ulcers with associated perilesional lymphadenopathies in BD-like models were detected by microPET. However, biodistributed PET-positive images from the stasis of secreted FDG into the bowel lumen could not be distinguished from the inflammatory bowel lesions of BD when compared to FDG uptake in control mice. CONCLUSIONS: Our data indicate that FDG PET can non-invasively and quantitatively detect the inflammatory process in an HSV-induced BD-like mouse model.


Subject(s)
Behcet Syndrome/diagnostic imaging , Fluorodeoxyglucose F18 , Herpes Simplex/diagnostic imaging , Inflammation/diagnostic imaging , Joints/diagnostic imaging , Positron-Emission Tomography , Radiopharmaceuticals , Animals , Behcet Syndrome/immunology , Behcet Syndrome/pathology , Behcet Syndrome/virology , Disease Models, Animal , Herpes Simplex/immunology , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Joints/immunology , Joints/pathology , Joints/virology , Male , Mice , Mice, Inbred ICR , Predictive Value of Tests
9.
Rheumatol Int ; 33(5): 1149-57, 2013 May.
Article in English | MEDLINE | ID: mdl-22955798

ABSTRACT

Antibody to Epstein-Barr virus (EBV) early antigen diffuse (anti-EA-D) is associated with viral replication. However, their possible associations with clinical/therapeutic features in primary Sjögren's syndrome (pSS) were not established. We evaluated 100 pSS patients (American-European Criteria) and 89 age/gender/ethnicity-matched healthy controls. Disease activity was measured by EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI). Antibodies to EBV (anti-VCA IgG/IgM, anti-EBNA-1 IgG, anti-EA-D IgG) were determined by ELISA. Patients and controls had comparable frequencies and mean levels of anti-VCA IgG (90 vs. 86.5 %, p = 0.501; 2.6 ± 1.1 vs. 2.5 ± 1.1 AU/mL, p = 0.737) and anti-EBNA-1 IgG (92 vs. 94.4 %, p = 0.576; 141.3 ± 69.8 vs. 135.6 ± 67.5 RU/mL, p = 0.464). Anti-VCA IgM was negative in all cases. Noteworthy, higher frequency and increased mean levels of anti-EA-D were observed in patients than controls (36 vs. 4.5 %, p < 0.0001; 38.6 ± 57.4 vs. 7.9 ± 26.3 RU/mL, p < 0.0001). Further analysis of patients with (n = 36) and without (n = 64) anti-EA-D revealed comparable age/gender/ethnicity (p ≥ 0.551), current prednisone dose (4.8 ± 6.9 vs. 5.1 ± 10.4 mg/day, p = 0.319), and current uses of prednisone (52.8 vs. 37.5 %, p = 0.148) and immunosuppressants (44.4 vs. 31.3 %, p = 0.201). ESSDAI values were comparable (p = 0.102), but joint activity was more frequent (25 vs. 9.4 %, p = 0.045) in anti-EA-D positive patients. Anti-EA-D antibodies were not associated with anti-Ro/SSA (p = 1.000), anti-La/SSB (p = 0.652), rheumatoid factor (p = 1.000), anti-α-fodrin (p = 0.390) or antiphospholipid antibodies (p = 0.573), not suggesting cross-reactivity. The higher anti-EA-D frequency associated with joint activity raises the possibility that a subclinical EBV reactivation may trigger or perpetuate the articular involvement in pSS.


Subject(s)
Antigens, Viral/immunology , Herpesvirus 4, Human/immunology , Joints/immunology , Joints/virology , Sjogren's Syndrome/immunology , Sjogren's Syndrome/virology , Virus Activation , Adult , Antibodies, Antinuclear/blood , Antibodies, Viral/blood , Biomarkers/blood , Case-Control Studies , Chi-Square Distribution , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Glucocorticoids/therapeutic use , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunosuppressive Agents/therapeutic use , Joints/drug effects , Male , Middle Aged , Prednisone/therapeutic use , Risk Factors , Severity of Illness Index , Sjogren's Syndrome/blood , Sjogren's Syndrome/diagnosis , Sjogren's Syndrome/drug therapy
10.
Gene Ther ; 20(6): 670-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23151520

ABSTRACT

With the long-term goal of developing a gene-based treatment for osteoarthritis (OA), we performed studies to evaluate the equine joint as a model for adeno-associated virus (AAV)-mediated gene transfer to large, weight-bearing human joints. A self-complementary AAV2 vector containing the coding regions for human interleukin-1-receptor antagonist (hIL-1Ra) or green fluorescent protein was packaged in AAV capsid serotypes 1, 2, 5, 8 and 9. Following infection of human and equine synovial fibroblasts in culture, we found that both were only receptive to transduction with AAV1, 2 and 5. For these serotypes, however, transgene expression from the equine cells was consistently at least 10-fold higher. Analyses of AAV surface receptor molecules and intracellular trafficking of vector genomes implicate enhanced viral uptake by the equine cells. Following delivery of 1 × 10(11) vector genomes of serotypes 2, 5 and 8 into the forelimb joints of the horse, all three enabled hIL-1Ra expression at biologically relevant levels and effectively transduced the same cell types, primarily synovial fibroblasts and, to a lesser degree, chondrocytes in articular cartilage. These results provide optimism that AAV vectors can be effectively adapted for gene delivery to large human joints affected by OA.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Interleukin 1 Receptor Antagonist Protein/genetics , Osteoarthritis/genetics , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/virology , Genetic Vectors , Green Fluorescent Proteins/genetics , Horses , Humans , Interleukin-1/genetics , Joints/metabolism , Joints/pathology , Joints/virology , Osteoarthritis/therapy , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovial Membrane/virology
11.
PLoS One ; 6(10): e26630, 2011.
Article in English | MEDLINE | ID: mdl-22028930

ABSTRACT

Epstein-Barr virus (EBV) has been implicated in the pathogenesis of rheumatoid arthritis (RA) on the basis of indirect evidence, such as its presence in affected joint tissues, antigenic cross reactions between EBV and human proteins, and elevated humoral and cellular anti-EBV immune responses in patients. Here we report development of erosive arthritis closely resembling RA in humanized mice inoculated with EBV. Human immune system components were reconstituted in mice of the NOD/Shi-scid/IL-2Rγ(null) (NOG) strain by transplantation with CD34(+) hematopoietic stem cells isolated from cord blood. These humanized mice were then inoculated with EBV and examined pathologically for the signs of arthritis. Erosive arthritis accompanied by synovial membrane proliferation, pannus formation, and bone marrow edema developed in fifteen of twenty-three NOG mice transplanted with human HSC and inoculated with EBV, but not in the nine NOG mice that were transplanted with HSC but not inoculated with EBV. This is the first report of an animal model of EBV-induced arthritis and strongly suggest a causative role of the virus in RA.


Subject(s)
Arthritis/pathology , Arthritis/virology , Herpesvirus 4, Human/pathogenicity , Animals , Bone Marrow/pathology , Bone Marrow/virology , Disease Models, Animal , Female , Humans , Joints/pathology , Joints/virology , Mice
12.
Expert Rev Anti Infect Ther ; 9(5): 545-54, 2011 May.
Article in English | MEDLINE | ID: mdl-21609266

ABSTRACT

Viral infections may manifest as acute or chronic arthritis. Joint involvement arises from either direct infection of the joint, through an immunological response directed towards the virus or autoimmunity. Epidemiological clues to the diagnosis include geographic location and exposure to vector-borne, blood-borne or sexually transmitted viruses. Although not always possible, it is important to diagnose the pathogenic virus, usually by serology, nucleic acid tests or rarely, viral culture. In general, viral arthritides are self-limiting and treatment is targeted at symptomatic relief. This article focuses on the causes, clinical features, diagnosis and treatment of viral arthritides.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Infectious/drug therapy , Arthritis, Infectious/virology , Joints/drug effects , Joints/virology , Animals , Anti-Inflammatory Agents/administration & dosage , Arthritis, Infectious/blood , Arthritis, Infectious/diagnosis , Arthritis, Infectious/epidemiology , Culicidae/virology , Flaviviridae/growth & development , Hepadnaviridae/growth & development , Humans , Insect Vectors/virology , Joints/physiopathology , Parvoviridae/growth & development , Retroviridae/growth & development , Serologic Tests , Togaviridae/growth & development
13.
J Clin Invest ; 120(3): 657-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20179348

ABSTRACT

Chikungunya disease is a severely debilitating, mosquito-borne, viral illness that has reached epidemic proportions in Africa, Asia, and the islands of the Indian Ocean. A mutation enhancing the ability of the chikungunya virus (CHIKV) to infect and be transmitted by Aedes albopictus has increased the geographical range at risk for infection due to the continuing global spread of this mosquito. Research into disease pathogenesis, vaccine development, and therapeutic design has been hindered by the lack of appropriate animal models of this disease. The meticulous study reported in this issue of the JCI by Labadie et al. is one of the first reports describing CHIKV infection of adult immunocompetent nonhuman primates. Using traditional and modern molecular and immunological approaches, the authors demonstrate that macaques infected with CHIKV are a good model of human CHIKV infection and also show that persistent arthralgia in humans may be caused by persistent CHIKV infection of macrophages.


Subject(s)
Alphavirus Infections/pathology , Chikungunya virus , Alphavirus Infections/virology , Animals , Disease Models, Animal , Humans , Joints/pathology , Joints/virology , Liver/pathology , Liver/virology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macaca fascicularis , Macrophages/pathology , Macrophages/virology , Mice , Muscles/pathology , Muscles/virology
14.
J Clin Invest ; 120(3): 894-906, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20179353

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that induces in humans a disease characterized by fever, rash, and pain in muscles and joints. The recent emergence or reemergence of CHIKV in the Indian Ocean Islands and India has stressed the need to better understand the pathogenesis of this disease. Previous CHIKV disease models have used young or immunodeficient mice, but these do not recapitulate human disease patterns and are unsuitable for testing immune-based therapies. Herein, we describe what we believe to be a new model for CHIKV infection in adult, immunocompetent cynomolgus macaques. CHIKV infection in these animals recapitulated the viral, clinical, and pathological features observed in human disease. In the macaques, long-term CHIKV infection was observed in joints, muscles, lymphoid organs, and liver, which could explain the long-lasting CHIKV disease symptoms observed in humans. In addition, the study identified macrophages as the main cellular reservoirs during the late stages of CHIKV infection in vivo. This model of CHIKV physiopathology should allow the development of new therapeutic and/or prophylactic strategies.


Subject(s)
Alphavirus Infections/pathology , Chikungunya virus , Disease Models, Animal , Alphavirus Infections/virology , Animals , Humans , Joints/pathology , Joints/virology , Liver/pathology , Liver/virology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macaca fascicularis , Macrophages/pathology , Macrophages/virology , Mice , Muscles/pathology , Muscles/virology
15.
Pharmacol Ther ; 124(3): 301-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19788897

ABSTRACT

A large range of human viruses are associated with the development of arthritis or arthralgia. Although there are many parallels with autoimmune arthritides, there is little evidence that viral arthritides lead to autoimmune disease. In humans viral arthritides usually last from weeks to months, can be debilitating, and are usually treated with non-steroidal anti-inflammatory drugs, but with variable success. Viral arthritides likely arise from immunopathological inflammatory responses directed at viruses and/or their products residing and/or replicating within joint tissues. Macrophages recruited by monocyte chemoattractant protein-1 (MCP-1/CCL2) and activated by interferon, and proinflammatory mediators like tumour necrosis factor alpha, interferon gamma, interleukin-6 and interleukin-1beta appear to be common elements in this group of diseases. The challenge for new treatments is to target excessive inflammation without compromising anti-viral immunity. Recent evidence from mouse models suggests targeting MCP-1 or complement may emerge as viable new treatment options for viral arthritides.


Subject(s)
Arthritis, Infectious/drug therapy , Arthritis, Infectious/immunology , Macrophages/metabolism , Viruses/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Arthritis, Infectious/etiology , Arthritis, Infectious/genetics , Arthritis, Infectious/pathology , Autoimmunity , Complement System Proteins/metabolism , Disease Models, Animal , Drug Delivery Systems , Humans , Inflammation Mediators/metabolism , Joints/immunology , Joints/metabolism , Joints/virology , Models, Immunological , Virulence , Viruses/genetics , Viruses/pathogenicity
16.
Rheum Dis Clin North Am ; 35(1): 95-110, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19480999

ABSTRACT

B19 infection-associated joint symptoms occur most frequently in adults, usually presenting as a self-limited, acute symmetric polyarthritis affecting the small joints of the hands, wrists, and knees. A small percentage of patients persist with chronic polyarthritis that mimics rheumatoid arthritis raising the question of whether B19 virus may have a role as a concomitant or precipitating factor in the pathogenesis of autoimmune conditions. Comprehensive and updated reviews address different aspects of human parvovirus infection. This article focuses on the evidence supporting the arthritogenic potential of the B19 virus and the proposed mechanisms that underlie it.


Subject(s)
Arthritis, Infectious/virology , Parvoviridae Infections/complications , Parvovirus B19, Human/physiology , Arthritis, Infectious/immunology , Arthritis, Infectious/physiopathology , Host-Pathogen Interactions , Humans , Joints/physiopathology , Joints/virology , Parvoviridae Infections/immunology , Parvovirus B19, Human/pathogenicity
17.
Bull Exp Biol Med ; 148(4): 631-3, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20396758

ABSTRACT

Dissemination of infectious inflammation was studied in experimental influenza and acute and chronic herpesvirus infections. The possibility of articular involvement into the infectious process was evaluated. Pathomorphological signs of changes in the articular tissue confirmed the effects of these viruses on the locomotor pathology. Results of virus infection simulation in experimental animals suggest this model for studies of the pathogenesis of diseases of viral etiology (including those with articular involvement) in humans.


Subject(s)
Herpesviridae Infections , Influenza, Human , Joint Diseases , Motor Activity/physiology , Orthomyxoviridae Infections , Animals , Disease Models, Animal , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/veterinary , Humans , Influenza, Human/immunology , Influenza, Human/pathology , Joint Diseases/pathology , Joint Diseases/veterinary , Joint Diseases/virology , Joints/pathology , Joints/virology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/veterinary
18.
J Virol ; 82(22): 11263-72, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18787004

ABSTRACT

Alphaviruses such as Ross River virus (RRV) and chikungunya virus are mosquito-transmitted viruses that cause explosive epidemics of debilitating arthritis and myositis affecting millions of humans worldwide. Previous studies using a mouse model of RRV-induced disease demonstrated that viral infection results in a severe inflammatory arthritis and myositis and that complement component 3 (C3) contributes to the destructive phase of the inflammatory disease but not the recruitment of cellular infiltrates to the sites of RRV-induced inflammation. Here, we demonstrate that mice deficient in complement receptor 3 (CR3) (CD11b(-/-)), a signaling receptor activated by multiple ligands including the C3 cleavage fragment iC3b, develop less-severe disease signs and decreased tissue destruction compared to RRV-infected wild-type mice. CR3 deficiency had no effect on viral replication, nor did it diminish the magnitude, kinetics, and composition of the cellular infiltrates at the sites of inflammation. However, the genetic absence of CR3 diminished the expression of specific proinflammatory and cytotoxic effectors, including S100A9/S100A8 and interleukin-6, within the inflamed tissues, suggesting that CR3-dependent signaling at the sites of inflammation contributes to tissue damage and severe disease.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/pathology , Macrophage-1 Antigen/immunology , Ross River virus/immunology , Animals , Calgranulin B/biosynthesis , Interleukin-6/biosynthesis , Joints/pathology , Joints/virology , Macrophage-1 Antigen/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/virology , Severity of Illness Index , Viral Load
19.
J Virol ; 80(2): 737-49, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16378976

ABSTRACT

Mosquito-borne alphaviruses are a significant cause of both encephalitic and arthritic disease in humans worldwide. In contrast to the encephalitic alphaviruses, the pathogenesis of alphavirus-induced arthritic disease is not well understood. Utilizing a mouse model of Ross River virus (RRV) disease, we found that the primary targets of RRV infection are bone, joint, and skeletal muscle tissues of the hind limbs in both outbred CD-1 mice and adult C57BL/6J mice. Moreover, histological analyses demonstrated that RRV infection resulted in severe inflammation of these tissues. Characterization of the inflammatory infiltrate within the skeletal muscle tissue identified inflammatory macrophages, NK cells, and CD4+ and CD8+ T lymphocytes. To determine the contribution of the adaptive immune system, the outcome of RRV-induced disease was examined in C57BL/6J RAG-1(-/-) mice, which lack functional T and B lymphocytes. RAG-1(-/-) and wild-type mice developed similar disease signs, infiltration of inflammatory macrophages and NK cells, and muscle pathology, suggesting that the adaptive immune response does not play a critical role in the development of disease. These results establish the mouse model of RRV disease as a useful system for the identification of viral and host factors that contribute to alphavirus-induced arthritis and myositis.


Subject(s)
Alphavirus Infections/pathology , Alphavirus Infections/virology , Arthritis, Infectious/pathology , Arthritis, Infectious/virology , Inflammation/pathology , Myositis/pathology , Myositis/virology , Ross River virus , Animals , Bone and Bones/pathology , Bone and Bones/virology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Disease Models, Animal , Female , Hindlimb/pathology , Hindlimb/virology , Homeodomain Proteins/genetics , Inflammation/immunology , Joints/pathology , Joints/virology , Killer Cells, Natural , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Muscle, Skeletal/virology , Organ Specificity
20.
J Immunol ; 173(8): 5238-46, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15470069

ABSTRACT

It is not known what is required for successive relapses in autoimmune diseases or evolution to a progressive chronic disease. Autoimmune arthritis caused by passive transfer of autoantibodies against glucose 6-phosphate isomerase is transient and therefore lends itself well to test for what might extend the disease. Herpesviruses have long been suspected of contributing to human autoimmune disease. We infected mice with a murine gamma-herpesvirus (MHV-68). In immunodeficient mice, transient arthritis was followed by a relapse. This was due to lytic viral infection of synovial tissues demonstrated by PCR, immunohistochemistry, and electron microscopy. Latent infection could be reactivated in the synovium of normal mice when treated with Cytoxan and this was associated with increased clinical arthritis. We conclude that herpesviruses may play an ancillary pathogenic role in autoimmune arthritis by infection of the inflammatory target tissue.


Subject(s)
Arthritis, Experimental/etiology , Autoimmune Diseases/etiology , Joints/virology , Rhadinovirus/physiology , Animals , Antigens, Viral/analysis , CD8-Positive T-Lymphocytes/immunology , DNA, Viral/analysis , Mice , Mice, Inbred C57BL , Recurrence , Virus Activation , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...