Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
PLoS Pathog ; 18(3): e1010365, 2022 03.
Article in English | MEDLINE | ID: mdl-35324997

ABSTRACT

Type I interferon (IFN) has been identified in patients with Lyme disease, and its abundant expression in joint tissues of C3H mice precedes development of Lyme arthritis. Forward genetics using C3H mice with severe Lyme arthritis and C57BL/6 (B6) mice with mild Lyme arthritis identified the Borrelia burgdorferi arthritis-associated locus 1 (Bbaa1) on chromosome 4 (Chr4) as a regulator of B. burgdorferi-induced IFNß expression and Lyme arthritis severity. B6 mice introgressed with the C3H allele for Bbaa1 (B6.C3-Bbaa1 mice) displayed increased severity of arthritis, which is initiated by myeloid lineage cells in joints. Using advanced congenic lines, the physical size of the Bbaa1 interval has been reduced to 2 Mbp, allowing for identification of potential genetic regulators. Small interfering RNA (siRNA)-mediated silencing identified Cdkn2a as the gene responsible for Bbaa1 allele-regulated induction of IFNß and IFN-stimulated genes (ISGs) in bone marrow-derived macrophages (BMDMs). The Cdkn2a-encoded p19 alternative reading frame (p19ARF) protein regulates IFNß induction in BMDMs as shown by siRNA silencing and overexpression of ARF. In vivo studies demonstrated that p19ARF contributes to joint-specific induction of IFNß and arthritis severity in B. burgdorferi-infected mice. p19ARF regulates B. burgdorferi-induced IFNß in BMDMs by stabilizing the tumor suppressor p53 and sequestering the transcriptional repressor BCL6. Our findings link p19ARF regulation of p53 and BCL6 to the severity of IFNß-induced Lyme arthritis in vivo and indicate potential novel roles for p19ARF, p53, and BCL6 in Lyme disease and other IFN hyperproduction syndromes.


Subject(s)
Arthritis , Cyclin-Dependent Kinase Inhibitor p16 , Lyme Disease , Animals , Arthritis/genetics , Borrelia burgdorferi , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genes, p16 , Interferon-beta/genetics , Interferon-beta/metabolism , Lyme Disease/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Small Interfering , Reading Frames , Tumor Suppressor Protein p53/genetics
2.
J Immunol ; 200(4): 1457-1470, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29330323

ABSTRACT

T cells predominate the immune responses in the synovial fluid of patients with persistent Lyme arthritis; however, their role in Lyme disease remains poorly defined. Using a murine model of persistent Lyme arthritis, we observed that bystander activation of CD4+ and CD8+ T cells leads to arthritis-promoting IFN-γ, similar to the inflammatory environment seen in the synovial tissue of patients with posttreatment Lyme disease. TCR transgenic mice containing monoclonal specificity toward non-Borrelia epitopes confirmed that bystander T cell activation was responsible for disease development. The microbial pattern recognition receptor TLR2 was upregulated on T cells following infection, implicating it as marker of bystander T cell activation. In fact, T cell-intrinsic expression of TLR2 contributed to IFN-γ production and arthritis, providing a mechanism for microbial-induced bystander T cell activation during infection. The IL-10-deficient mouse reveals a novel TLR2-intrinsic role for T cells in Lyme arthritis, with potentially broad application to immune pathogenesis.


Subject(s)
Interleukin-10/deficiency , Lyme Disease/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 2/immunology , Animals , Female , Interleukin-10/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
J Immunol ; 199(10): 3525-3534, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28986440

ABSTRACT

Previously, using a forward genetic approach, we identified differential expression of type I IFN as a positional candidate for an expression quantitative trait locus underlying Borrelia burgdorferi arthritis-associated locus 1 (Bbaa1). In this study, we show that mAb blockade revealed a unique role for IFN-ß in Lyme arthritis development in B6.C3-Bbaa1 mice. Genetic control of IFN-ß expression was also identified in bone marrow-derived macrophages stimulated with B. burgdorferi, and it was responsible for feed-forward amplification of IFN-stimulated genes. Reciprocal radiation chimeras between B6.C3-Bbaa1 and C57BL/6 mice revealed that arthritis is initiated by radiation-sensitive cells, but orchestrated by radiation-resistant components of joint tissue. Advanced congenic lines were developed to reduce the physical size of the Bbaa1 interval, and confirmed the contribution of type I IFN genes to Lyme arthritis. RNA sequencing of resident CD45- joint cells from advanced interval-specific recombinant congenic lines identified myostatin as uniquely upregulated in association with Bbaa1 arthritis development, and myostatin expression was linked to IFN-ß production. Inhibition of myostatin in vivo suppressed Lyme arthritis in the reduced interval Bbaa1 congenic mice, formally implicating myostatin as a novel downstream mediator of the joint-specific inflammatory response to B. burgdorferi.


Subject(s)
Borrelia burgdorferi/immunology , Inflammation/immunology , Interferon-beta/metabolism , Lyme Disease/immunology , Macrophages/immunology , Myostatin/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Genetic Loci/genetics , Inflammation/genetics , Lyme Disease/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myostatin/genetics , Radiation Chimera , Up-Regulation
4.
PLoS One ; 10(8): e0135142, 2015.
Article in English | MEDLINE | ID: mdl-26252010

ABSTRACT

MicroRNA-155 has been shown to play a role in immune activation and inflammation, and is suppressed by IL-10, an important anti-inflammatory cytokine. The established involvement of IL-10 in the murine model of Borrelia burgdorferi-induced Lyme arthritis and carditis allowed us to assess the interplay between IL-10 and miR-155 in vivo. As reported previously, Mir155 was highly upregulated in joints from infected severely arthritic B6 Il10-/- mice, but not in mildly arthritic B6 mice. In infected hearts, Mir155 was upregulated in both strains, suggesting a role of miR-155 in Lyme carditis. Using B. burgdorferi-infected B6, Mir155-/-, Il10-/-, and Mir155-/- Il10-/- double-knockout (DKO) mice, we found that anti-inflammatory IL-10 and pro-inflammatory miR-155 have opposite and somewhat compensatory effects on myeloid cell activity, cytokine production, and antibody response. Both IL-10 and miR-155 were required for suppression of Lyme carditis. Infected Mir155-/- mice developed moderate/severe carditis, had higher B. burgdorferi numbers, and had reduced Th1 cytokine expression in hearts. In contrast, while Il10-/- and DKO mice also developed severe carditis, hearts had reduced bacterial numbers and elevated Th1 and innate cytokine expression. Surprisingly, miR-155 had little effect on Lyme arthritis. These results show that antagonistic interplay between IL-10 and miR-155 is required to balance host defense and immune activation in vivo, and this balance is particularly important for suppression of Lyme carditis. These results also highlight tissue-specific differences in Lyme arthritis and carditis pathogenesis, and reveal the importance of IL-10-mediated regulation of miR-155 in maintaining healthy immunity.


Subject(s)
Arthritis/metabolism , Interleukin-10/metabolism , Lyme Disease/metabolism , MicroRNAs/metabolism , Myocarditis/metabolism , Animals , Arthritis/microbiology , Bone Marrow Cells/cytology , Borrelia burgdorferi , Disease Models, Animal , Female , Gene Expression Regulation , Genotype , Immune System , Immunity, Innate , Lyme Disease/microbiology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/microbiology , Phagocytosis , Protein Binding , Th1 Cells/cytology
5.
Genome Biol ; 16: 28, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25886764

ABSTRACT

BACKGROUND: The prevalence of some autoimmune diseases is greater in females compared with males, although disease severity is often greater in males. The reason for this sexual dimorphism is unknown, but it may reflect negative selection of Y chromosome-bearing sperm during spermatogenesis or male fetuses early in the course of conception/pregnancy. Previously, we showed that the sexual dimorphism in experimental autoimmune encephalomyelitis (EAE) is associated with copy number variation (CNV) in Y chromosome multicopy genes. Here, we test the hypothesis that CNV in Y chromosome multicopy genes influences the paternal parent-of-origin effect on EAE susceptibility in female mice. RESULTS: We show that C57BL/6 J consomic strains of mice possessing an identical X chromosome and CNV in Y chromosome multicopy genes exhibit sperm head abnormalities and female-biased sex ratio. This is consistent with X-Y intragenomic conflict arising from an imbalance in CNV between homologous X:Y chromosome multicopy genes. These males also display paternal transmission of EAE to female offspring and differential loading of microRNAs within the sperm nucleus. Furthermore, in humans, families of probands with multiple sclerosis similarly exhibit a female-biased sex ratio, whereas families of probands affected with non-sexually dimorphic autoimmune diseases exhibit unbiased sex ratios. CONCLUSIONS: These findings provide evidence for a mechanism at the level of the male gamete that contributes to the sexual dimorphism in EAE and paternal parent-of-origin effects in female mice, raising the possibility that a similar mechanism may contribute to the sexual dimorphism in multiple sclerosis.


Subject(s)
Autoimmune Diseases/genetics , DNA Copy Number Variations/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Dosage , Genetic Linkage , Sex Characteristics , Y Chromosome/genetics , Animals , Animals, Newborn , CD4-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Female , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Inheritance Patterns/genetics , Linear Models , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Phenotype , Sex Ratio , Spermatogenesis/genetics , Spermatozoa/metabolism , X Chromosome/genetics
6.
J Immunol ; 193(12): 6050-60, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25378596

ABSTRACT

Localized upregulation of type I IFN was previously implicated in development of Borrelia burgdorferi-induced arthritis in C3H mice, and was remarkable due to its absence in the mildly arthritic C57BL/6 (B6) mice. Independently, forward genetics analysis identified a quantitative trait locus on Chr4, termed B. burgdorferi-associated locus 1 (Bbaa1), that regulates Lyme arthritis severity and includes the 15 type I IFN genes. Involvement of Bbaa1 in arthritis development was confirmed in B6 mice congenic for the C3H allele of Bbaa1 (B6.C3-Bbaa1), which developed more severe Lyme arthritis and K/B×N model of rheumatoid arthritis (RA) than did parental B6 mice. Administration of a type I IFN receptor blocking mAb reduced the severity of both Lyme arthritis and RA in B6.C3-Bbaa1 mice, formally linking genetic elements within Bbaa1 to pathological production of type I IFN. Bone marrow-derived macrophages from Bbaa1 congenic mice implicated this locus as a regulator of type I IFN induction and downstream target gene expression. Bbaa1-mediated regulation of IFN-inducible genes was upstream of IFN receptor-dependent amplification; however, the overall magnitude of the response was dependent on autocrine/paracrine responses to IFN-ß. In addition, the Bbaa1 locus modulated the functional phenotype ascribed to bone marrow-derived macrophages: the B6 allele promoted expression of M2 markers, whereas the C3H allele promoted induction of M1 responses. This report identifies a genetic locus physically and functionally linked to type I IFN that contributes to the pathogenesis of both Lyme and RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Borrelia burgdorferi/immunology , Interferon Type I/metabolism , Lyme Disease/genetics , Lyme Disease/metabolism , Quantitative Trait Loci , Alleles , Animals , Arthritis, Rheumatoid/immunology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Interferon Regulatory Factors/genetics , Interferon Type I/pharmacology , Lyme Disease/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/genetics , Phagocytosis/immunology , Phenotype , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/genetics , Transcriptional Activation
7.
PLoS Pathog ; 10(6): e1004212, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24967703

ABSTRACT

MicroRNAs have been shown to be important regulators of inflammatory and immune responses and are implicated in several immune disorders including systemic lupus erythematosus and rheumatoid arthritis, but their role in Lyme borreliosis remains unknown. We performed a microarray screen for expression of miRNAs in joint tissue from three mouse strains infected with Borrelia burgdorferi. This screen identified upregulation of miR-146a, a key negative regulator of NF-κB signaling, in all three strains, suggesting it plays an important role in the in vivo response to B. burgdorferi. Infection of B6 miR-146a-/- mice with B. burgdorferi revealed a critical nonredundant role of miR-146a in modulating Lyme arthritis without compromising host immune response or heart inflammation. The impact of miR-146a was specifically localized to the joint, and did not impact lesion development or inflammation in the heart. Furthermore, B6 miR-146a-/- mice had elevated levels of NF-κB-regulated products in joint tissue and serum late in infection. Flow cytometry analysis of various lineages isolated from infected joint tissue of mice showed that myeloid cell infiltration was significantly greater in B6 miR-146a-/- mice, compared to B6, during B. burgdorferi infection. Using bone marrow-derived macrophages, we found that TRAF6, a known target of miR-146a involved in NF-κB activation, was dysregulated in resting and B. burgdorferi-stimulated B6 miR-146a-/- macrophages, and corresponded to elevated IL-1ß, IL-6 and CXCL1 production. This dysregulated protein production was also observed in macrophages treated with IL-10 prior to B. burgdorferi stimulation. Peritoneal macrophages from B6 miR-146a-/- mice also showed enhanced phagocytosis of B. burgdorferi. Together, these data show that miR-146a-mediated regulation of TRAF6 and NF-κB, and downstream targets such as IL-1ß, IL-6 and CXCL1, are critical for modulation of Lyme arthritis during chronic infection with B. burgdorferi.


Subject(s)
Arthritis, Infectious/genetics , Borrelia burgdorferi/immunology , Lyme Disease/immunology , MicroRNAs/genetics , Myocarditis/genetics , Animals , Arthritis, Infectious/microbiology , Borrelia burgdorferi/pathogenicity , Chemokine CXCL1/immunology , Gene Expression Regulation/genetics , Inflammation/immunology , Inflammation Mediators/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-1beta/immunology , Interleukin-6/immunology , Lyme Disease/genetics , Lyme Disease/pathology , Macrophages/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/microbiology , NF-kappa B/genetics , NF-kappa B/immunology , Phagocytosis/genetics , Phagocytosis/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 6/biosynthesis , TNF Receptor-Associated Factor 6/immunology
8.
J Clin Invest ; 124(1): 311-20, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24334460

ABSTRACT

Lyme disease, caused by the spirochete Borrelia burgdorferi, is the most prevalent arthropod-borne illness in the United States and remains a clinical and social challenge. The spectrum of disease severity among infected patients suggests that host genetics contribute to pathogenic outcomes, particularly in patients who develop arthritis. Using a forward genetics approach, we identified the lysosomal enzyme ß-glucuronidase (GUSB), a member of a large family of coregulated lysosomal enzymes, as a key regulator of Lyme-associated arthritis severity. Severely arthritic C3H mice possessed a naturally occurring hypomorphic allele, Gusbh. C57BL/6 mice congenic for the C3H Gusb allele were prone to increased Lyme-associated arthritis severity. Radiation chimera experiments revealed that resident joint cells drive arthritis susceptibility. C3H mice expressing WT Gusb as a transgene were protected from severe Lyme arthritis. Importantly, the Gusbh allele also exacerbated disease in a serum transfer model of rheumatoid arthritis. A known GUSB function is the prevention of lysosomal accumulation of glycosaminoglycans (GAGs). Development of Lyme and rheumatoid arthritis in Gusbh-expressing mice was associated with heightened accumulation of GAGs in joint tissue. We propose that GUSB modulates arthritis pathogenesis by preventing accumulation of proinflammatory GAGs within inflamed joint tissue, a trait that may be shared by other lysosomal exoglycosidases.


Subject(s)
Arthritis, Experimental/enzymology , Arthritis, Rheumatoid/enzymology , Borrelia burgdorferi , Glucuronidase/metabolism , Lyme Disease/enzymology , Animals , Arthritis, Experimental/microbiology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Cells, Cultured , Glucuronidase/genetics , Glycosaminoglycans/metabolism , Humans , Joints/pathology , Lyme Disease/microbiology , Lyme Disease/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Sequence Deletion , Severity of Illness Index
9.
Genome Res ; 23(9): 1474-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23800453

ABSTRACT

Understanding the DNA elements that constitute and control the regulatory genome is critical for the appropriate therapeutic management of complex diseases. Here, using chromosome Y (ChrY) consomic mouse strains on the C57BL/6J (B6) background, we show that susceptibility to two diverse animal models of autoimmune disease, experimental allergic encephalomyelitis (EAE) and experimental myocarditis, correlates with the natural variation in copy number of Sly and Rbmy multicopy ChrY genes. On the B6 background, ChrY possesses gene regulatory properties that impact genome-wide gene expression in pathogenic CD4(+) T cells. Using a ChrY consomic strain on the SJL background, we discovered a preference for ChrY-mediated gene regulation in macrophages, the immune cell subset underlying the EAE sexual dimorphism in SJL mice, rather than CD4(+) T cells. Importantly, in both genetic backgrounds, an inverse correlation exists between the number of Sly and Rbmy ChrY gene copies and the number of significantly up-regulated genes in immune cells, thereby supporting a link between copy number variation of Sly and Rbmy with the ChrY genetic element exerting regulatory properties. Additionally, we show that ChrY polymorphism can determine the sexual dimorphism in EAE and myocarditis. In humans, an analysis of the CD4(+) T cell transcriptome from male multiple sclerosis patients versus healthy controls provides further evidence for an evolutionarily conserved mechanism of gene regulation by ChrY. Thus, as in Drosophila, these data establish the mammalian ChrY as a member of the regulatory genome due to its ability to epigenetically regulate genome-wide gene expression in immune cells.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Predisposition to Disease , Macrophages/metabolism , Myocarditis/genetics , Transcriptome , Y Chromosome/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport , Animals , DNA Copy Number Variations , Female , Gene Dosage , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sex Characteristics
10.
J Immunol ; 189(5): 2488-501, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22851707

ABSTRACT

Localized elevation in type I IFN has been uniquely linked to the severe Lyme arthritis that develops in C3H mice infected with the spirochete Borrelia burgdorferi. In this study, the dynamic interactions that result in generation of these responses were further examined in C3H mice carrying the type I IFN receptor gene ablation, which effectively blocks all autocrine/paracrine signaling crucial to induction of downstream effectors. Reciprocal radiation chimeras between C3H and IFNAR1⁻/⁻ mice implicated both radiation-sensitive and radiation-resistant cells of the joint tissue in the proarthritic induction of type I IFN. Ex vivo analysis of cells from the naive joint revealed CD45⁺ cells residing in the tissue to be uniquely capable of initiating the type I IFN response to B. burgdorferi. Type I IFN responses were analyzed in real time by lineage sorting of cells from infected joint tissue. This demonstrated that myeloid cells, endothelial cells, and fibroblasts were responsible for propagating the robust IFN response, which peaked at day 7 postinfection and rapidly resolved. Endothelial cells and fibroblasts were the dominant sources of IFN signature transcripts in the joint tissue. Fibroblasts were also the major early source of chemokines associated with polymorphonuclear leukocyte and monocyte/macrophage infiltration, thus providing a focal point for arthritis development. These findings suggest joint-localized interactions among related and unrelated stromal, endothelial, and myeloid cell lineages that may be broadly applicable to understanding the pathogeneses of diseases associated with type I IFN signature, including systemic lupus erythematosus and some rheumatoid arthritides.


Subject(s)
Arthritis, Experimental/immunology , Fibroblasts/immunology , Interferon Type I/biosynthesis , Lyme Disease/immunology , Myeloid Cells/immunology , Up-Regulation/immunology , Animals , Ankle Joint/immunology , Ankle Joint/microbiology , Ankle Joint/pathology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Borrelia burgdorferi/immunology , Borrelia burgdorferi/pathogenicity , Cells, Cultured , Chemokines/biosynthesis , Chemokines/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Interferon Type I/deficiency , Interferon Type I/genetics , Lyme Disease/metabolism , Lyme Disease/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Severity of Illness Index , Signal Transduction/immunology , Transcription, Genetic/immunology , Up-Regulation/genetics
11.
J Immunol ; 188(2): 541-7, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22147765

ABSTRACT

Histamine is a biogenic amine that mediates multiple physiological processes, including immunomodulatory effects in allergic and inflammatory reactions, and also plays a key regulatory role in experimental allergic encephalomyelitis, the autoimmune model of multiple sclerosis. The pleiotropic effects of histamine are mediated by four G protein-coupled receptors, as follows: Hrh1/H(1)R, Hrh2/H(2)R, Hrh3/H(3)R, and Hrh4/H(4)R. H(4)R expression is primarily restricted to hematopoietic cells, and its role in autoimmune inflammatory demyelinating disease of the CNS has not been studied. In this study, we show that, compared with wild-type mice, animals with a disrupted Hrh4 (H(4)RKO) develop more severe myelin oligodendrocyte glycoprotein (MOG)(35\x{2013}55)-induced experimental allergic encephalomyelitis. Mechanistically, we also show that H(4)R plays a role in determining the frequency of T regulatory (T(R)) cells in secondary lymphoid tissues, and regulates T(R) cell chemotaxis and suppressor activity. Moreover, the lack of H(4)R leads to an impairment of an anti-inflammatory response due to fewer T(R) cells in the CNS during the acute phase of the disease and an increase in the proportion of Th17 cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , Blood-Brain Barrier/immunology , CD4 Lymphocyte Count , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Glycoproteins/administration & dosage , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Neurons/immunology , Neurons/pathology , Peptide Fragments/administration & dosage , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine/deficiency , Receptors, Histamine/genetics , Receptors, Histamine H4 , Severity of Illness Index , T-Lymphocytes, Regulatory/metabolism
12.
J Immunol ; 188(3): 1381-93, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22180617

ABSTRACT

IL-10 is a nonredundant inflammatory modulator that suppresses arthritis development in Borrelia burgdorferi-infected mice. Infected C57BL/6 (B6) IL-10(-/-) mice were previously found to have a prolonged IFN-inducible response in joint tissue. Infection of B6 IL-10 reporter mice identified macrophages and CD4(+) T cells as the primary sources of IL-10 in the infected joint tissue, suggesting that early local production of IL-10 dampened the proarthritic IFN response. Treatment of B6 IL-10(-/-) mice with anti-IFN-γ reduced the increase in arthritis severity and suppressed IFN-inducible transcripts to wild-type levels, thereby linking dysregulation of IFN-γ to disease in the B6 IL-10(-/-) mouse. Arthritis in B6 IL-10(-/-) mice was associated with elevated numbers of NK cell, NKT cell, α/ß T cell, and macrophage infiltration of the infected joint. FACS lineage sorting revealed NK cells and CD4(+) T cells as sources of IFN-γ in the joint tissue of B6 IL-10(-/-) mice. These findings suggest the presence of a positive-feedback loop in the joint tissue of infected B6 IL-10(-/-) mice, in which production of inflammatory chemokines, infiltration of IFN-γ-producing cells, and additional production of inflammatory cytokines result in arthritis. This mechanism of arthritis is in contrast to that seen in C3H/He mice, in which arthritis development is linked to transient production of type I IFN and develops independently of IFN-γ. Due to the sustained IFN response driven by NK cells and T cells, we propose the B6 IL-10(-/-) mouse as a potential model to study the persistent arthritis observed in some human Lyme disease patients.


Subject(s)
Cell Movement/immunology , Inflammation/immunology , Interferon-gamma/pharmacology , Interleukin-10/biosynthesis , Lyme Disease/immunology , Animals , Borrelia burgdorferi , Feedback, Physiological , Humans , Interferon-gamma/immunology , Interleukin-10/deficiency , Killer Cells, Natural/immunology , Mice , Mice, Knockout , T-Lymphocytes/immunology
13.
Proc Natl Acad Sci U S A ; 107(44): 18967-72, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956310

ABSTRACT

Disruption of the blood-brain barrier (BBB) underlies the development of experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis. Environmental factors, such as Bordetella pertussis, are thought to sensitize central endothelium to biogenic amines like histamine, thereby leading to increased BBB permeability. B. pertussis-induced histamine sensitization (Bphs) is a monogenic intermediate phenotype of EAE controlled by histamine H(1) receptor (Hrh1/H(1)R). Here, we transgenically overexpressed H(1)R in endothelial cells of Hrh1-KO (H(1)RKO) mice to test the role of endothelial H(1)R directly in Bphs and EAE. Unexpectedly, transgenic H(1)RKO mice expressing endothelial H(1)R under control of the von Willebrand factor promoter (H(1)RKO-vWF(H1R) Tg) were Bphs-resistant. Moreover, H(1)RKO-vWF(H1R) Tg mice exhibited decreased BBB permeability and enhanced protection from EAE compared with H(1)RKO mice. Thus, contrary to prevailing assumptions, our results show that endothelial H(1)R expression reduces BBB permeability, suggesting that endothelial H(1)R signaling may be important in the maintenance of cerebrovascular integrity.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelium, Vascular/metabolism , Receptors, Histamine H1/metabolism , Signal Transduction , Animals , Bordetella pertussis/genetics , Bordetella pertussis/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Predisposition to Disease , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Receptors, Histamine H1/genetics , Whooping Cough/genetics , Whooping Cough/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
14.
Cell Immunol ; 260(2): 119-27, 2010.
Article in English | MEDLINE | ID: mdl-19914609

ABSTRACT

The postnatal maternal environment is known to increase susceptibility to a number of autoimmune diseases. Here we asked whether the postnatal maternal environment could influence autoimmune disease development to day 3 thymectomy (d3tx)-induced autoimmune ovarian disease (AOD) and experimental allergic encephalomyelitis (EAE) in cross-fostered A/J and B6 mice. A/J pups foster-nursed by B6 mothers exhibit an increase in autoimmune disease development while cross-fostering B6 pups on A/J mothers did not alter their susceptibility. The increase in AOD incidence seen in foster-nursed d3tx A/J mice correlated with a decrease in the total number of CD4(+) T cells in the lymph nodes of these animals. Analysis of the cellular composition in the milk revealed that B6 mice shed significantly more maternally derived lymphocytes into their milk compared to A/J mothers. These data suggest that there are maternally derived postnatal factors that influence the development of autoimmune disease in A/J mice.


Subject(s)
Animals, Newborn/immunology , Animals, Suckling/immunology , Autoimmune Diseases/immunology , Disease Susceptibility/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Chemokine CXCL1/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunity, Maternally-Acquired/immunology , Interleukin-13/metabolism , Interleukin-9/metabolism , Lactation/immunology , Lactation/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Ovarian Diseases/immunology , Thymectomy
15.
Infect Immun ; 77(8): 3302-11, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19487472

ABSTRACT

The observation that Borrelia burgdorferi-induced arthritis is severe in C3H mice and milder in C57BL/6 (B6) mice has allowed a forward genetics approach for the identification of genetic elements that regulate the arthritis response. Quantitative trait loci (QTL) on five chromosomes (Chr) were identified previously in segregating crosses between C3H and B6 mice and collectively designated B. burgdorferi arthritis-associated (Bbaa) QTL. Reciprocal interval-specific congenic lines (ISCL) that encompass Bbaa1, Bbaa2-Bbaa3, Bbaa4, Bbaa6, and Bbaa12 on Chr 4, 5, 11, 12, and 1, respectively, have now been generated. Bidirectional transfer of the arthritis severity phenotype in association with Bbaa2-Bbaa3 and Bbaa4 was observed, and unidirectional transfer with the B6 allele of Bbaa6 was noted. These findings confirm the existence of polymorphic loci within Bbaa2-Bbaa3, Bbaa4, and Bbaa6 that regulate the severity of B. burgdorferi-induced arthritis. ISCL were used to assess the regulation of a previously identified interferon transcriptional profile associated with severe disease in C3H mice. The regulation of this transcriptional signature was found to be independent of penetrant Bbaa QTL, both in joint tissues and in isolated macrophages. These results clearly demonstrate the utility of forward genetics for the discovery of novel genes and pathways involved in the regulation of the severity of Lyme arthritis and predict the involvement of regulatory elements not evident from other experimental approaches.


Subject(s)
Borrelia burgdorferi/pathogenicity , Chromosomes , Lyme Disease/genetics , Lyme Disease/pathology , Quantitative Trait Loci , Animals , Ankle/pathology , Crosses, Genetic , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phenotype
16.
Rio de Janeiro; Elsevier; 4 ed; 2009. 1476 p. ilus, tab.
Monography in Portuguese | LILACS, Coleciona SUS, Sec. Munic. Saúde SP, COVISA-Acervo | ID: lil-689190
17.
Rio de Janeiro; Elsevier; 4 ed; 2009. 1476 p. ilus, tab.
Monography in Portuguese | Sec. Munic. Saúde SP, COVISA-Acervo | ID: sms-7126
18.
J Immunol ; 181(12): 8492-503, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050267

ABSTRACT

Gene expression analysis previously revealed a robust IFN-responsive gene induction profile that was selectively up-regulated in Borrelia burgdorferi-infected C3H mice at 1 wk postinfection. This profile was correlated with arthritis development, as it was absent from infected, mildly arthritic C57BL/6 mice. In this report we now demonstrate that profile induction in infected C3H scid mice occurs independently of B or T lymphocyte infiltration in the joint tissue. Additionally, type I IFN receptor-blocking Abs, but not anti-IFN-gamma Abs, dramatically reduced arthritis, revealing a critical but previously unappreciated role for type I IFN in Lyme arthritis development. Certain examined IFN-inducible transcripts were also significantly diminished within joint tissue of mice treated with anti-IFNAR1, whereas expression of other IFN-responsive genes was more markedly altered by anti-IFN-gamma treatment. These data indicate that induction of the entire IFN profile is not necessary for arthritis development. These findings further tie early type I IFN induction to Lyme arthritis development, a connection not previously made. Bone marrow-derived macrophages readily induced IFN-responsive genes following B. burgdorferi stimulation, and this expression required a functional type I IFN receptor. Strikingly, induction of these genes was independent of TLRs 2,4, and 9 and of the adapter molecule MyD88. These data demonstrate that the extracellular pathogen B. burgdorferi uses a previously unidentified receptor and a pathway traditionally associated with viruses and intracellular bacteria to initiate transcription of type I IFN and IFN-responsive genes and to initiate arthritis development.


Subject(s)
Borrelia burgdorferi/immunology , Interferon Type I/physiology , Lyme Disease/immunology , Lyme Disease/microbiology , Animals , Antibodies, Blocking/administration & dosage , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression Profiling , Immunity, Innate/genetics , Interferon Type I/biosynthesis , Interferon Type I/deficiency , Lyme Disease/metabolism , Lyme Disease/therapy , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/immunology , Signal Transduction/genetics , Signal Transduction/immunology
19.
J Acoust Soc Am ; 124(4): 2340-52, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19062872

ABSTRACT

In a previous study, it was hypothesized that ultrasound-induced lung damage was related to the transfer of ultrasonic energy into the lungs (W. D. O'Brien et al. 2002, "Ultrasound-induced lung hemorrhage: Role of acoustic boundary conditions at the pleural surface," J. Acoust. Soc. Am. 111, 1102-1109). From this study a technique was developed to: 1) estimate the impedance (Mrayl) of fresh, excised, ex vivo rat lung versus its level of inflation (cm H(2)O) and 2) predict the fraction of ultrasonic energy transmitted into the lung (M. Oelze et al. 2003, "Impedance measurements of ex vivo rat lung at different volumes of inflation." J. Acoust. Soc. Am. 114, 3384-3393). In the current study, the same technique was used to estimate the frequency-dependent impedance of lungs from rats, rabbits, and pigs of various ages. Impedance values were estimated from lungs under deflation (atmospheric pressure, 0 cm H(2)O) and three volumes of inflation pressure [7 cm H(2)O (5 cm H(2)O for pigs), 10 cm H(2)O, and 15 cm H(2)O]. Lungs were scanned in a tank of degassed 37 degrees C water. The frequency-dependent acoustic pressure reflection coefficient was determined over a frequency range of 3.5-10 MHz. From the reflection coefficient, the frequency-dependent lung impedance was calculated with values ranging from an average of 1.4 Mrayl in deflated lungs (atmospheric pressure) to 0.1 Mrayl for fully inflated lungs (15 cm H(2)O). Across all species, deflated lung (i.e., approximately 7% of the total lung capacity) had impedance values closer to tissue values, suggesting that more acoustic energy was transmitted into the lung under deflated conditions. Finally, the impedance values of deflated lungs from different species at different ages were compared with the thresholds for ultrasound-induced lung damage. The comparison revealed that increases in ultrasonic energy transmission corresponded to lower injury threshold values.


Subject(s)
Acoustics , Lung Injury/etiology , Lung/diagnostic imaging , Age Factors , Animals , Lung/physiopathology , Lung Compliance , Lung Injury/physiopathology , Pressure , Rabbits , Rats , Rats, Sprague-Dawley , Species Specificity , Swine , Total Lung Capacity , Ultrasonography/adverse effects
20.
Am J Pathol ; 173(3): 892-900, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688020

ABSTRACT

Weibel-Palade bodies within endothelial cells are secretory granules known to release von Willebrand Factor (VWF), P-selectin, chemokines, and other stored molecules following histamine exposure. Mice with a disrupted VWF gene (VWFKO) have endothelial cells that are deficient in Weibel-Palade bodies. These mice were used to evaluate the role of VWF and/or Weibel-Palade bodies in Bordetella pertussis toxin-induced hypersensitivity to histamine, a subphenotype of experimental allergic encephalomyelitis, the principal autoimmune model of multiple sclerosis. No significant differences in susceptibility to histamine between wild-type and VWFKO mice were detected after 3 days; however, histamine sensitivity persisted significantly longer in VWFKO mice. Correspondingly, encephalomyelitis onset was earlier, disease was more severe, and blood brain barrier (BBB) permeability was significantly increased in VWFKO mice, as compared with wild-type mice. Moreover, inflammation was selectively increased in the brains, but not spinal cords, of VWFKO mice as compared with wild-type mice. Early increases in BBB permeability in VWFKO mice were not due to increased encephalitogenic T-cell activity since BBB permeability did not differ in adjuvant-treated VWFKO mice as compared with littermates immunized with encephalitogenic peptide plus adjuvant. Taken together, these data indicate that VWF and/or Weibel-Palade bodies negatively regulate BBB permeability changes and autoimmune inflammatory lesion formation within the brain elicited by peripheral inflammatory stimuli.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/pathology , Capillary Permeability/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , von Willebrand Factor/metabolism , Adjuvants, Immunologic/metabolism , Animals , Blood-Brain Barrier/pathology , Brain/immunology , Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Histamine/immunology , Hypersensitivity/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Weibel-Palade Bodies/metabolism , von Willebrand Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...