Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
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
2.
Am J Pathol ; 183(2): 441-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23746656

ABSTRACT

Coxsackievirus B3 (CVB3) variants H3 and H310A1 differ by a single nonconserved amino acid in the VP2 capsid region. C57Bl/6 mice infected with the H3 virus develop myocarditis correlating with activation of T cells expressing the Vγ4 T cell receptor chain. Infecting mice with H310A1 activates natural killer T (NKT; mCD1d-tetramer(+) TCRß(+)) cells, but not Vγ4 T cells, and fails to induce myocarditis. H310A1 infection preferentially activates M2 alternatively activated macrophage and CD4(+)FoxP3 (T regulatory) cells, whereas CD4(+)Th1 (IFN-γ(+)) cells are suppressed. By contrast, H3 virus infection activates M1 proinflammatory and CD4(+)Th1 cells, but not T regulatory cells. The M1 macrophage show significantly increased CD1d expression compared to M2 macrophage. The ability of NKT cells to suppress myocarditis was shown by adoptive transfer of purified NKT cells into H3-infected NKT knockout (Jα18 knockout) mice, which inhibited cardiac inflammation and increased T regulatory cell response. Cardiac virus titers were equivalent in all mouse strains indicating that neither Vγ4 nor NKT cells participate in control of virus infection. These data show that NKT and Vγ4 cells cross-regulate T regulatory cell responses during CVB3 infections and are the primary factor determining viral pathogenesis in this mouse model.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Natural Killer T-Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD1/metabolism , CD4-Positive T-Lymphocytes/immunology , Immunity, Innate/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/virology , T-Lymphocytes, Regulatory/virology
3.
Exp Mol Pathol ; 94(1): 58-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22750431

ABSTRACT

Coxsackievirus B3 (CVB3) infection of C57Bl/6 mice shows a sex bias with males developing more severe cardiac inflammation than females because males develop a Th1 inflammatory response, whereas females develop a Th2 response. Since their discovery, Toll-like receptors have been shown to play an important role in the development of the immune response against harmful pathogens. To assess the role of TLRs in coxsackievirus-induced myocarditis wild type and Toll-like receptor 2-/- male and female mice were infected and assessed for viral replication, myocarditis, helper T-cell generation, and regulatory T-cell generation. TLR2-/- mice show reduced Th1 expression compared to controls. Treatment of wild type mice with either Pam3CSK4 (TLR2) or LPS (TLR4) specific TLR agonists resulted in increased Th1 expression in male and female mice and a decrease in FoxP3+ regulatory T-cells in male mice. The suppression of T regulatory cells by TLR signaling in males but not females correlates with the increased myocarditis susceptibility of the males.


Subject(s)
Coxsackievirus Infections/immunology , Myocarditis/immunology , Myocarditis/virology , Sex Characteristics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , DNA-Binding Proteins , Disease Susceptibility/immunology , Enterovirus/immunology , Female , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/metabolism , Nerve Tissue Proteins/biosynthesis , Nuclear Proteins/biosynthesis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics
4.
Am J Pathol ; 182(2): 401-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23195432

ABSTRACT

There are two major haplotypes of signal lymphocytic activation molecule (Slam) in inbred mouse strains, with the Slam haplotype 1 expressed in C57Bl/6 mice and the Slam haplotype 2 expressed in most other commonly used inbred strains, including 129 mice. Because signaling through Slam family receptors can affect innate immunity [natural killer T cell (NKT) and γ-δ T-cell receptor], and innate immunity can determine susceptibility to coxsackievirus B3 (CVB3) infection, the present study evaluated the response of C57Bl/6 and congenic B6.129c1 mice (expressing the 129-derived Slam locus) to CVB3. CVB3-infected C57Bl/6 male mice developed increased myocarditis but reduced hepatic injury compared with infected B6.129c1 mice. C57Bl/6 mice also had increased γδ(+) and CD8(+)interferon-γ(+) cells but decreased numbers of NKT (T-cell receptor ß chain + mCD1d tetramer(+)) and CD4(+)FoxP3(+) cells compared with B6.129c1 mice. C57Bl/6 mice were infected with CVB3 and treated with either α-galactosylceramide, an NKT cell-specific ligand, or vehicle (dimethyl sulfoxide/PBS). Mice treated with α-galactosylceramide showed significantly reduced myocarditis. Liver injuries, as determined by alanine aminotransferase levels in plasma, were increased significantly, confirming that NKT cells are protective for myocarditis but pathogenic in the liver.


Subject(s)
Antigens, CD/metabolism , Coxsackievirus Infections/immunology , Haplotypes/genetics , Liver/pathology , Lymphocyte Activation/immunology , Myocarditis/pathology , Natural Killer T-Cells/immunology , Receptors, Cell Surface/metabolism , Adaptive Immunity/drug effects , Alanine Transaminase/blood , Animals , Antigens, CD/genetics , Coxsackievirus Infections/complications , Coxsackievirus Infections/pathology , Enterovirus B, Human/drug effects , Enterovirus B, Human/immunology , Galactosylceramides/pharmacology , Hepatitis/complications , Hepatitis/immunology , Hepatitis/pathology , Liver/immunology , Liver/virology , Lymphocyte Activation/drug effects , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Myocarditis/blood , Myocarditis/complications , Myocarditis/immunology , Natural Killer T-Cells/drug effects , Polymorphism, Genetic , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Family Member 1 , Troponin I/blood , Viral Load/immunology
5.
Biol Sex Differ ; 3(1): 25, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23241283

ABSTRACT

BACKGROUND: Coxsackievirus B3 (CVB3) induces myocarditis, an inflammatory heart disease, which affects men more than women. Toll-like receptor (TLR) signaling has been shown to determine the severity of CVB3-induced myocarditis. No direct role for signaling through TLR2 had been shown in myocarditis although published studies show that cardiac myosin is an endogenous TLR2 ligand and stimulates pro-inflammatory cytokine expression by dendritic cells in vitro. The goal of this study is to determine which TLRs show differential expression in CVB3 infected mice corresponding to male susceptibility and female resistance in this disease. METHODS: Male and female C57Bl/6 mice were infected with 102 PFU CVB3 and killed on day 3 or 6 post infection. Hearts were evaluated for virus titer, myocardial inflammation, and TLR mRNA expression by PCR array and microarray analysis. Splenic lymphocytes only were evaluated by flow cytometry for the number of TLR+/CD3+, TLR+/CD4+, TLR+F4/80+ and TLR+/CD11c+ subpopulations and the mean fluorescence intensity to assess upregulation of TLR expression on these cells. Mice were additionally treated with PAM3CSK4 (TLR2 agonist) or ultrapure LPS (TLR4 agonist) on the same day as CVB3 infection or 3 days post infection to confirm their role in myocarditis susceptibility. RESULTS: Despite equivalent viral titers, male C57Bl/6 mice develop more severe myocarditis than females by day 6 after infection. Microarray analysis shows a differential expression of TLR2 at day 3 with female mice having higher levels of TLR2 gene expression compared to males. Disease severity correlates to greater TLR4 protein expression on splenic lymphocytes in male mice 3 days after infection while resistance in females correlates to preferential TLR2 expression, especially in spleen lymphocytes. Treating male mice with PAM reduced mortality from 55% in control CVB3 infected animals to 10%. Treating female mice with LPS increased mortality from 0% in control infected animals to 60%. CONCLUSION: CVB3 infection causes an up-regulation of TLR2 in female and of TLR4 in male mice and this differential expression between the sexes contributes to disease resistance of females and susceptibility of males. While previous reports demonstrated a pathogenic role for TLR4 this is the first report that TLR2 is preferentially up-regulated in CVB3 infected female mice or that signaling through this TLR directly causes myocarditis resistance.

6.
Exp Mol Pathol ; 92(3): 327-33, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22465422

ABSTRACT

IL-21 is a multi-functional cytokine which can promote survival, proliferation and activation of T and B lymphocytes including CD8 T cells. Previous studies have shown that autoimmune CD8+ T cells are the primary pathogenic effector cell in coxsackievirus B3 (CVB3) induced myocarditis in C57Bl/6 mice. To evaluate the role of IL-21 in promoting CD8+ T cell mediated cardiac injury in myocarditis, C57Bl/6 and IL-21RKO mice were infected with CVB3. IL-21RKO mice developed significantly less myocarditis than C57Bl/6 animals although cardiac virus titers were equivalent between the mouse strains. Numbers of CD8+IFNγ+ cells were decreased in IL-21RKO mice but numbers of either CD4+IFNγ+ or CD4+IL-4+ cells were not significantly different from C57Bl/6 animals indicating a selective effect of IL-21 signaling on the CD8+ T cell response. To confirm that IL-21 signaling exclusively functions at the level of the CD8+ T cell in CVB3 induced myocarditis, purified CD8+ cells were isolated from either C57Bl/6 or IL-21RKO donors and adoptively transferred into CD8KO recipients prior to CVB3 infection. CD8KO recipients given either C57Bl/6 or IL-21RKO CD8+ cells showed equivalent reconstitution of the CD8+ cells in the spleen but the recipients given C57Bl/6 CD8+ cells showed significantly greater myocarditis than recipients of IL-21RKO CD8+ cells. These data demonstrate that IL-21 signaling directly in the CD8+ cell population is required for CVB3-induced myocarditis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Myocarditis/immunology , Receptors, Interleukin-21/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Coxsackievirus Infections/genetics , Coxsackievirus Infections/virology , Enterovirus B, Human/physiology , Flow Cytometry , Host-Pathogen Interactions/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukins/immunology , Interleukins/metabolism , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/genetics , Myocarditis/virology , Receptors, Interleukin-21/genetics , Receptors, Interleukin-21/metabolism , Spleen/immunology , Spleen/metabolism
7.
G3 (Bethesda) ; 2(1): 115-21, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22384388

ABSTRACT

Coxsackievirus B3 (CVB3) contributes to the development of myocarditis, an inflammatory heart disease that predominates in males, and infection is a cause of unexpected death in young individuals. Although gonadal hormones contribute significantly to sex differences, sex chromosomes may also influence disease. Increasing evidence indicates that Chromosome Y (ChrY) genetic variants can impact biological functions unrelated to sexual differentiation. Using C57BL/6J (B6)-ChrY consomic mice, we show that genetic variation in ChrY has a direct effect on the survival of CVB3-infected animals. This effect is not due to potential Sry-mediated differences in prenatal testosterone exposure or to differences in adult testosterone levels. Furthermore, we show that ChrY polymorphism influences the percentage of natural killer T cells in B6-ChrY consomic strains but does not underlie CVB3-induced mortality. These data underscore the importance of investigating not only the hormonal regulation but also ChrY genetic regulation of cardiovascular disease and other male-dominant, sexually dimorphic diseases and phenotypes.

8.
Cell Immunol ; 272(2): 269-74, 2012.
Article in English | MEDLINE | ID: mdl-22078271

ABSTRACT

Susceptibility to autoimmune myocarditis has been associated with histamine release by mast cells during the innate immune response to coxsackievirus B3 (CVB3) infection. To investigate the contribution of histamine H(1) receptor (H(1)R) signaling to CVB3-induced myocarditis, we assessed susceptibility to the disease in C57BL/6J (B6) H(1)R(-/-) mice. No difference was observed in mortality between CVB3-infected B6 and H(1)R(-/-) mice. However, analysis of their hearts revealed a significant increase in myocarditis in H(1)R(-/-) mice that is not attributed to increased virus replication. Enhanced myocarditis susceptibility correlated with a significant expansion in pathogenic Th1 and Vγ4(+) γδ T cells in the periphery of these animals. Furthermore, an increase in regulatory T cells was observed, yet these cells were incapable of controlling myocarditis in H(1)R(-/-) mice. These data establish a critical role for histamine and H(1)R signaling in regulating T cell responses and susceptibility to CVB3-induced myocarditis in B6 mice.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Myocarditis/immunology , Receptors, Histamine H1/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Coxsackievirus Infections/metabolism , Disease Susceptibility , Histamine/metabolism , Male , Mice , Mice, Inbred C57BL , Myocarditis/metabolism , Receptors, Histamine H1/deficiency , Receptors, Histamine H1/metabolism , Signal Transduction , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Virus Replication/immunology
9.
Biol Sex Differ ; 2: 8, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21806829

ABSTRACT

BACKGROUND: Both coxsackievirus B3 (CVB3) and influenza A virus (IAV; H1N1) produce sexually dimorphic infections in C57BL/6 mice. Gonadal steroids can modulate sex differences in response to both viruses. Here, the effect of sex chromosomal complement in response to viral infection was evaluated using four core genotypes (FCG) mice, where the Sry gene is deleted from the Y chromosome, and in some mice is inserted into an autosomal chromosome. This results in four genotypes: XX or XY gonadal females (XXF and XYF), and XX or XY gonadal males (XXM and XYM). The FCG model permits evaluation of the impact of the sex chromosome complement independent of the gonadal phenotype. METHODS: Wild-type (WT) male and female C57BL/6 mice were assigned to remain intact or be gonadectomized (Gdx) and all FCG mice on a C57BL/6 background were Gdx. Mice were infected with either CVB3 or mouse-adapted IAV, A/Puerto Rico/8/1934 (PR8), and monitored for changes in immunity, virus titers, morbidity, or mortality. RESULTS: In CVB3 infection, mortality was increased in WT males compared to females and males developed more severe cardiac inflammation. Gonadectomy suppressed male, but increased female, susceptibility to CVB3. Infection with IAV resulted in greater morbidity and mortality in WT females compared with males and this sex difference was significantly reduced by gonadectomy of male and female mice. In Gdx FCG mice infected with CVB3, XY mice were less susceptible than XX mice. Protection correlated with increased CD4+ forkhead box P3 (FoxP3)+ T regulatory (Treg) cell activation in these animals. Neither CD4+ interferon (IFN)γ (T helper 1 (Th1)) nor CD4+ interleukin (IL)-4+ (Th2) responses differed among the FCG mice during CVB3 infection. Infection of Gdx FCG mice revealed no effect of sex chromosome complement on morbidity or mortality following IAV infection. CONCLUSIONS: These studies indicate that sex chromosome complement can influence pathogenicity of some, but not all, viruses.

10.
Am J Reprod Immunol ; 51(2): 180-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14983872

ABSTRACT

Decay accelerating factor (DAF) is a widely distributed glycoprotein which aids in the inactivation of complement. DAF is also a cellular receptor for certain group B coxsackieviruses (CVB) and is responsible for the viral hemagglutinating activity for human red blood cells (RBC). Healthy, young female volunteers donated blood on days 11 and 22 of the ovarian cycle. Samples were categorized into luteal and follicular phases based on serum progesterone level (P4 either < 2.0 ng/mL, follicular; P4 > or = 2.0 ng/mL, luteal) and analyzed by flow cytomtery for DAF expression on RBC and CD21 + B lymphocytes. Cycling females showed significant variation in CVB-induced hemagglutination and % RBC or CD19 + cells which were DAF +. There was a strong correlation between serum estradiol levels and % RBC expressing DAF (P < 0.01) in the follicular, but not in the luteal ovarian phase. Infection of white blood cells with green-fluorescent protein CVB (GFP-CVB) showed a correlation between infectivity of CD19+ cells and DAF expression. This indicates that women may show differential susceptibility to CVB infection in the luteal and follicular phases of the ovarian cycle.


Subject(s)
CD55 Antigens/blood , Enterovirus B, Human/immunology , Erythrocytes/immunology , Estradiol/blood , Menstrual Cycle/blood , Antigens, CD19/immunology , CD55 Antigens/immunology , Enterovirus B, Human/genetics , Enterovirus B, Human/pathogenicity , Enterovirus Infections/blood , Enterovirus Infections/immunology , Erythrocytes/virology , Female , Follicular Phase/blood , Green Fluorescent Proteins , Hemagglutination/immunology , Humans , Leukocytes/cytology , Luminescent Proteins , Luteal Phase/blood , Menstrual Cycle/immunology , Progesterone/blood , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL