Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
2.
Virology ; 381(2): 155-60, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18829062

ABSTRACT

Ultraviolet (u.v.) inactivated coxsackievirus B3 (CVB3) induces rapid calcium flux in naïve BALB/c CD4+ T cells. CD4+ cells lacking decay accelerating factor (DAF-/-) show little calcium flux indicating that virus cross-linking of this virus receptor protein is necessary for calcium signaling in CVB3 infection. Interaction of CVB3 with CD4+ cells also activates NFAT DNA binding. To show that NFAT activation is crucial to CVB3 induced disease, wild-type mice and transgenic mice expressing dominant-negative NFAT (dnNFAT) mutant in T cells were infected and evaluated for myocarditis and pancreatitis 7 days later. Inhibition of NFAT in T cells prevented myocarditis but had no effect on pancreatitis. Virus titers in pancreas were equivalent in wild-type and dnNFAT animals but cardiac virus titers were increased in dnNFAT mice. Interferon-gamma (IFN gamma) expression was reduced in both CD4+ and V gamma 4+ T cells from dnNFAT mice compared to controls. FasL expression by V gamma 4+ cells was also suppressed. Inhibition of FasL expression by V gamma 4+ cells is consistent with myocarditis protection in dnNFAT mice.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Coxsackievirus Infections/physiopathology , Enterovirus B, Human/physiology , Myocarditis/physiopathology , Myocarditis/virology , NFATC Transcription Factors/metabolism , Animals , Calcium/metabolism , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Disease Susceptibility/metabolism , Enterovirus B, Human/immunology , Gene Expression Regulation , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Myocarditis/immunology , Myocardium/pathology , Pancreas/pathology , Spleen/cytology
3.
Virology ; 378(2): 292-8, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18586295

ABSTRACT

Transgenic female mice expressing the TNFalpha gene under the cardiac myosin promoter (TNF1.6) develop substantially increased myocarditis and increased numbers of CD4+Th1 (interferon gamma+) cells when infected with coxsackievirus B3 (CVB3) during the diestrus and proestrus phases of the estrus cycle compared to females infected during the estrus and metestrus phases. Cardiac virus titers were increased in females infected in estrus compared to females infected during the other phases. T regulatory cells (CD4+CD25+FoxP3+) were increased in both peripheral blood and inflammatory cells in the heart in females infected during estrus. Exogenous administration of 200 ng/mouse 17-beta-estradiol to females protected against CVB3 induced myocarditis and increased CD4+CD25+FoxP3+ cells. These results demonstrate that hormonal fluctuations occurring in normally cycling females can determine T regulatory cell response and control virus-induced pathogenesis.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human/isolation & purification , Estrus/immunology , Myocarditis/immunology , Myocarditis/virology , T-Lymphocytes, Regulatory/immunology , Animals , Body Weight , CD4-Positive T-Lymphocytes/immunology , Coxsackievirus Infections/virology , Estradiol/administration & dosage , Female , Forkhead Transcription Factors/analysis , Heart/virology , Interleukin-2 Receptor alpha Subunit/analysis , Mice , Mice, Transgenic , Myocardium/pathology , Th1 Cells/immunology
4.
J Lipid Res ; 47(11): 2525-37, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16914769

ABSTRACT

Although the immune system, inflammation, and cellular metabolism are linked to diseases associated with dyslipidemias, the mechanism(s) remain unclear. To determine whether there is a mechanistic link between lipid availability and inflammation/immune activation, we evaluated macrophage cell lines incubated under conditions of altered exogenous and endogenous lipid availability. Limiting exogenous lipids results in decreased lysosomal acidity and decreased lysosomal enzymatic activity. Both lysosomal parameters are restored with the addition of oleoyl-CoA, suggesting that fatty acids play a role in the regulation of lysosomal function. Cell surface expression of major histocompatibility complex (MHC)-encoded molecules is also decreased in the absence of exogenous lipids. Additionally, we observe decreased gamma-interferon stimulation of cell surface MHC class II. Using cerulenin to limit the endogenous synthesis of fatty acids results in decreased cell surface expression of MHC class II but does not appear to alter lysosomal acidity, suggesting that lysosomal acidity is dependent on exogenous, but not endogenous, fatty acid availability. Testing these conclusions in an in vivo mouse model, we observed statistically significant, diet-dependent differences in lysosomal acidity and MHC class II cell surface expression. Collectively, these data demonstrate a mechanistic link between lipid availability and early events in the immune response.


Subject(s)
Fatty Acids/metabolism , Genes, MHC Class II , Histocompatibility Antigens Class II/metabolism , Lysosomes/metabolism , Animals , Cell Membrane/metabolism , Female , Glucosylceramidase/chemistry , Humans , Immune System/metabolism , Inflammation , Lipids/chemistry , Lysosomes/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
5.
Virology ; 352(1): 226-36, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16730774

ABSTRACT

Coxsackievirus B3 induced murine myocarditis depends upon CD1d expression and upon a population of CD1d-restricted Vgamma4+ T cells. Infection upregulates CD1d expression in CD4+ T cells. Bone marrow chimeras were made between BALB/c and BALB/c CD1d-/- mice and showed that CD1d expression in either hemopoietic and non-hemopoietic cells induces myocarditis, although CD1d expression on hemopoietic cells was more effective in increasing Vgamma4+ cell numbers and activation, and CD4+ IFNgamma+ cell response than CD1d expression on non-hemopoietic cells. Co-culture of enriched CD4+ cells from infected CD1d-/- and BALB/c mice with Vgamma4+ T cells demonstrated that the Vgamma4+ cells bias the CD4+ cell response to the Th1 phenotype through CD1d. Anti-CD1d antibody effectively blocked promotion of IFNgamma expression by the CD4+ cell population. These results show that Vgamma4+ cells modulate developing adaptive immunity through recognition of CD1d on CD4+ T cells, and that this interaction, more than Vgamma4+ cell interaction with infected cardiocytes, determines pathogenicity.


Subject(s)
Antigens, CD1/metabolism , CD4-Positive T-Lymphocytes/immunology , Enterovirus B, Human/pathogenicity , Hematopoietic System/cytology , Myocarditis/immunology , Th1 Cells/immunology , Animals , Antigens, CD1d , Hematopoietic System/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Myocarditis/virology , Receptors, Antigen, T-Cell, gamma-delta/metabolism
6.
Microbes Infect ; 7(3): 537-43, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15777711

ABSTRACT

Coxsackieviruses are a cause of clinical myocarditis. Both virus replication and host defense mechanisms, including virus-induced autoimmunity, mediate heart injury and cardiac dysfunction. Vgamma4+ cells kill infected cardiocytes and virus-specific CD4+ Th2 cells through Fas-dependent apoptosis and CD1d. The CD4+ Th1 response is necessary for activation of the autoimmune CD8+ T cells, which kill uninfected cardiocytes through perforin-dependent mechanisms.


Subject(s)
Autoimmunity/immunology , Coxsackievirus Infections/immunology , Enterovirus B, Human , Myocarditis/immunology , Myocarditis/virology , T-Lymphocyte Subsets/immunology , Animals , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/virology , Mice , Signal Transduction
7.
J Virol ; 79(5): 2659-65, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15708985

ABSTRACT

Giving C57BL/6 mice 10(4) PFU of coxsackievirus B3 (H3 variant) fails to induce myocarditis, but increasing the initial virus inoculum to 10(5) or 10(6) PFU causes significant cardiac disease. Virus titers in the heart were equivalent at days 3 and 7 in mice given all three virus doses, but day 3 titers in the pancreases of mice inoculated with 10(4) PFU were reduced. Tumor necrosis factor alpha (TNF-alpha) concentrations in the heart were increased in all infected mice, but cytokine levels were highest in mice given the larger virus inocula. TNF-alpha(-/-) and p55 TNF receptor-negative (TNFR(-/-)) mice developed minimal myocarditis compared to B6;129 or C57BL/6 control mice. p75 TNFR(-/-) mice were as disease susceptible as C57BL/6 animals. No significant differences in virus titers in heart or pancreas were observed between the groups, but C57BL/6 and p75 TNFR(-/-) animals showed 10-fold more inflammatory cells in the heart than p55 TNFR(-/-) mice, and the cell population was comprised of high concentrations of CD4(+) gamma interferon-positive and Vgamma4(+) cells. Cardiac endothelial cells isolated from C57BL/6 and p75 TNFR(-/-) mice upregulate CD1d, the molecule recognized by Vgamma4(+) cells, but infection of TNF(-/-) or p55 TNFR(-/-) endothelial cells failed to upregulate CD1d. Infection of C57BL/6 endothelial cells with a nonmyocarditic coxsackievirus B3 variant, H310A1, which is a poor inducer of TNF-alpha, failed to elicit CD1d expression, but TNF-alpha treatment of H310A1-infected endothelial cells increased CD1d levels to those seen in H3-infected cells. TNF-alpha treatment of uninfected endothelial cells had only a modest effect on CD1d expression, suggesting that optimal CD1d upregulation requires both infection and TNF-alpha signaling.


Subject(s)
Antigens, CD1/biosynthesis , Enterovirus B, Human/pathogenicity , Enterovirus Infections/etiology , Myocarditis/etiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Antigens, CD1d , Cell Death , Enterovirus B, Human/isolation & purification , Enterovirus Infections/immunology , Enterovirus Infections/pathology , Enterovirus Infections/virology , Heart/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/virology , Myocardium/immunology , Myocardium/pathology , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
8.
J Virol ; 76(21): 10785-90, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12368321

ABSTRACT

T cells expressing the Vgamma4 T-cell receptor (TCR) promote myocarditis in coxsackievirus B3 (CVB3)-infected BALB/c mice. CD1, a major histocompatibility complex (MHC) class I-like molecule, is required for activation of Vgamma4(+) cells. Once activated, Vgamma4(+) cells initiate myocarditis through gamma interferon (IFN-gamma)-mediated induction of CD4(+) T helper type 1 (Th1) cells in the infected animal. These CD4(+) Th1 cells are required for activation of an autoimmune CD8(+) alphabeta TCR(+) effector, which is the predominant pathogenic agent in this model of CVB3-induced myocarditis. Activated Vgamma4(+) cells can adoptively transfer myocarditis into BALB/c mice infected with a nonmyocarditic variant of CVB3 (H310A1) but cannot transfer myocarditis into either uninfected or CD1(-/-) recipients, demonstrating the need for both infection and CD1 expression for Vgamma4(+) cell function. In contrast, CD8(+) alphabeta TCR(+) cells transfer myocarditis into either infected CD1(-/-) or uninfected recipients, showing that once activated, the CD8(+) alphabeta TCR(+) effectors function independently of both virus and CD1. Vgamma4(+) cells given to mice lacking CD4(+) T cells minimally activate the CD8(+) alphabeta TCR(+) cells. These studies show that Vgamma4(+) cells determine CVB3 pathogenicity by their ability to influence both the CD4(+) and CD8(+) adaptive immune response. Vgamma4(+) cells enhance CD4(+) Th1 (IFN-gamma(+)) cell activation through IFN-gamma- and CD1-dependent mechanisms. CD4(+) Th1 cells promote activation of the autoimmune CD8(+) alphabeta TCR(+) effectors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Enterovirus B, Human/immunology , Lymphocyte Activation/immunology , Myocarditis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Adoptive Transfer , Animals , Antigens, CD1 , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Myocarditis/virology
10.
J Virol ; 75(13): 5860-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390587

ABSTRACT

Two coxsackievirus B3 (CVB3) variants (H3 and H310A1) differ by a single amino acid mutation in the VP2 capsid protein. H3 induces severe myocarditis in BALB/c mice, but H310A1 is amyocarditic. Infection with H3, but not H310A1, preferentially activates Vgamma4 Vdelta4 cells, which are strongly positive for gamma interferon (IFN-gamma), whereas Vgamma1 Vdelta4 cells are increased in both H3 and H310A1 virus-infected animals. Depletion of Vgamma1(+) cells using monoclonal anti-Vgamma1 antibody enhanced myocarditis and CD4(+)-, IFN-gamma(+)-cell responses in both H3- and H310A1-infected mice yet decreased the CD4(+)-, IL-4(+)-cell response. Depleting Vgamma4(+) cells suppressed myocarditis and reduced CD4(+) IFN-gamma(+) cells but increased CD4(+) IL-4(+) T cells. The role of cytokine production by Vgamma1(+) and Vgamma4(+) T cells was investigated by adoptively transferring these cells isolated from H3-infected BALB/c Stat4 knockout (Stat4ko) (defective in IFN-gamma expression) or BALB/c Stat6ko (defective in IL-4 expression) mice into H3 virus-infected wild-type BALB/c recipients. Vgamma4 and Vgamma1(+) T cells from Stat4ko mice expressed IL-4 but no or minimal IFN-gamma, whereas these cell populations derived from Stat6ko mice expressed IFN-gamma but no IL-4. Stat4ko Vgamma1(+) cells (IL-4(+)) suppress myocarditis. Stat6ko Vgamma1(+) cells (IFN-gamma(+)) were not inhibitory. Stat6ko Vgamma4(+) cells (IFN-gamma(+)) significantly enhanced myocarditis. Stat4ko Vgamma4(+) cells (IL-4(+)) neither inhibited nor enhanced disease. These results show that distinct gammadelta-T-cell subsets control myocarditis susceptibility and bias the CD4(+)-Th-cell response. The cytokines produced by the Vgamma subpopulation have a significant influence on the CD4(+)-Th-cell phenotype.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Coxsackievirus Infections/etiology , Cytokines/biosynthesis , Enterovirus B, Human , Myocarditis/etiology , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/physiology , Adoptive Transfer , Animals , DNA-Binding Proteins/physiology , Immunophenotyping , Lymphocyte Depletion , Male , Mice , Mice, Inbred BALB C , STAT4 Transcription Factor , STAT6 Transcription Factor , Trans-Activators/physiology
11.
Circulation ; 103(21): 2610-6, 2001 May 29.
Article in English | MEDLINE | ID: mdl-11382732

ABSTRACT

BACKGROUND: T cells are implicated in atherosclerosis, but little is known about the genetic control or molecular pathways, especially under conditions of mild hypercholesterolemia. METHODS AND RESULTS: BALB/c mice, making a CD4+ Th2 (IL-4+) cell response, express both MHC class II antigens (IA(d), IE(d)) and are atherosclerosis-resistant. C57Bl/6 mice produce a CD4+ Th1 (interferon [IFN]gamma+) response, express IA(b) but no IE, and are atherosclerosis-prone. To evaluate T helper-cell phenotype in fatty streak formation, wild-type C57Bl/6 mice (IA(b)+IE-) and transgenic mice, either AB(o), IA(b)-IE-; ABEalpha, IA-IE(k)+; or BL:TG:Ealpha, IA(b)+IE(k)+, were fed a high-cholesterol diet for 16 weeks and evaluated histomorphometrically for aortic lesions. Lesion size in AB(o), ABEalpha, and BL:TG:Ealpha strains was decreased by 54%, 79%, and 82%, respectively, compared with wild-type, correlating with decreased Th1 and increased Th2 expression and suggesting that T helper-cell phenotype is important in fatty lesion development. Decreasing Th1 cells by antibodies (alpha-CD4) or cytokines (IL-4) also caused >/=80% reductions in lesion size. Immunohistology revealed IFN-gamma, but not IL-4, colocalized with activated macrophages. Confirming these findings in a different mouse strain, BALB/c Stat 6 knockout mice (Th2 cell-deficient) developed aortic lesions comparable to C57Bl/6 mice on the same diet. CONCLUSIONS: In mildly hypercholesterolemic C57Bl/6 mice, presence of IA(b) and absence of IE regulated CD4+ T helper-cell phenotype; fatty lesions were proportional to IFNgamma+ Th1 cells in both C57Bl/6 and BALB/c strains. IFN-gamma may participate through macrophage activation, whereas IL-4 may act to limit Th1-cell response.


Subject(s)
Arteriosclerosis/pathology , Hypercholesterolemia/physiopathology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , CD4 Antigens/genetics , CD4 Antigens/metabolism , Cholesterol/blood , Cytokines/metabolism , Dietary Fats/administration & dosage , Female , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , STAT6 Transcription Factor , Trans-Activators/genetics , Trans-Activators/metabolism
12.
J Immunol ; 165(11): 6472-9, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11086087

ABSTRACT

Gammadelta T lymphocytes have been shown to regulate immune responses in diverse experimental systems. Because distinct gammadelta T cell subsets, as defined by the usage of certain TCR V genes, preferentially respond in various diseases and disease models, we have hypothesized that the various gammadelta T cell subsets carry out different functions. To test this, we compared one particular gammadelta T cell subset, the Vgamma1(+) subset, which represents a major gammadelta T cell type in the lymphoid organs and blood of mice, to other subsets and to gammadelta T cells as a whole. Using Listeria monocytogenes infection as an infectious disease model, we found that bacterial containment improves in mice depleted of Vgamma1(+) gammadelta T cells, albeit mice lacking all gammadelta T cells are instead impaired in their ability to control Listeria expansion. Our findings indicate that Vgamma1(+) gammadelta T cells reduce the ability of the innate immune system to destroy Listeria, even though other gammadelta T cells as a whole promote clearance of this pathogen.


Subject(s)
Listeriosis/genetics , Listeriosis/immunology , Lymphocyte Depletion , Receptors, Antigen, T-Cell, gamma-delta/deficiency , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Female , Gene Targeting , Immunity, Cellular/genetics , Immunity, Innate/genetics , Injections, Intravenous , Listeria monocytogenes/immunology , Listeriosis/microbiology , Liver/immunology , Liver/microbiology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Spleen/immunology , Spleen/microbiology , T-Lymphocyte Subsets/microbiology
13.
J Immunol ; 165(8): 4174-81, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035049

ABSTRACT

Coxsackievirus B3 infections of C57BL/6 mice, which express the MHC class II IA but not IE Ag, results in virus replication in the heart but minimal myocarditis. In contrast, Bl.Tg.Ealpha mice, which are C57BL/6 mice transgenically induced to express IE Ag, develop significant myocarditis upon Coxsackievirus B3 infection. Despite this difference in inflammatory damage, cardiac virus titers are similar between C57BL/6 and Bl.Tg.Ealpha mice. Removing gammadelta T cells from either strain by genetic manipulation (gammadelta knockout(ko)) changes the disease phenotype. C57BL/6 gammadelta ko mice show increased myocarditis. In contrast, Bl.Tg.Ealpha gammadelta ko mice show decreased cardiac inflammation. Flow cytometry revealed a difference in the gammadelta cell subsets in the two strains, with Vgamma1 dominating in C57BL/6 mice, and Vgamma4 predominating Bl.Tg.Ealpha mice. This suggests that these two Vgamma-defined subsets might have different functions. To test this possibility, we used mAb injection to deplete each subset. Mice depleted of Vgamma1 cells showed enhanced myocarditis, whereas those depleted of Vgamma4 cells suppressed myocarditis. Adoptively transfusing enriched Vgamma4(+) cells to the C57BL/6 and Bl.Tg. Ealpha gammadelta ko strains confirmed that the Vgamma4 subset promoted myocarditis. Th subset analysis suggests that Vgamma1(+) cells biased the CD4(+) T cells to a dominant Th2 cell response, whereas Vgamma4(+) cells biased CD4(+) T cells toward a dominant Th1 cell response.


Subject(s)
Enterovirus B, Human/immunology , Genetic Predisposition to Disease , Myocarditis/immunology , Receptors, Antigen, T-Cell, gamma-delta/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/prevention & control , Female , Immune Sera/administration & dosage , Injections, Intravenous , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocarditis/genetics , Myocarditis/pathology , Myocarditis/prevention & control , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/transplantation
14.
J Anim Sci ; 78(3): 726-34, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10764081

ABSTRACT

Two digestion studies were conducted to evaluate the use of pretanned leather shavings as a component of a protein supplement. In Exp. 1, the in situ and in vitro disappearance of pretanned leather shavings and soybean meal was evaluated. Results revealed that less than 18.4% of the pretanned leather shavings was solubilized and disappeared when exposed to McDougall's buffer for 48 h, but there was 90.0% disappearance with 48-h exposure to a .1 N HCl/pepsin treatment and 97.0% disappearance with exposure to a two-stage digestion. In situ disappearance following 72 h in the rumen allowed 6.8% disappearance. Thus, leather shavings seem to be relatively indigestible in the rumen, but postruminal digestion may be possible. In Exp. 2, six Angus x Holstein steers, fitted with ruminal and duodenal cannulas, were used in a replicated 3 x 3 Latin square to evaluate ruminal and digestion effects of the following supplements combined with fescue hay at 1.7% of BW (DM basis): no supplementation (control); supplementation intraruminally with soybean meal at .07% of BW (as-fed basis); and supplementation intraruminally with a combination of soybean meal and pretanned leather shavings (17:8 ratio) at .05% of BW (isonitrogenous to soybean meal; as-fed basis). Ruminal fluid passage rate was greater and fluid turnover time was shorter in steers fed leather shavings than in those fed soybean meal (P = .10). Ruminal pH was lower (P = .04) for supplemented steers than for control steers and ruminal NH3 N concentration was greater (P = .01) in steers fed soybean meal than in those fed leather shavings. Total VFA concentration was increased (P = .02) by supplementation. Supplementation with soybean meal increased (P < .05) ruminal molar proportions of butyrate, valerate, and isovalerate compared with leather shavings. Duodenal OM flow and OM disappearing in the intestines were increased by supplementation (P < .10), but not by the type of supplement fed (P > .10). Ruminal digestion of OM and total tract OM digestion were unaffected (P > .10) by supplementation and the type of supplement fed. Flow and digestion of NDF were unaffected (P > .10) by the treatments. Flow of N and the quantity of N disappearing in the intestines were increased (P < .05) by supplementation but did not differ (P > .10) between supplementation groups. Microbial N flow, N utilization for net microbial protein synthesis, and ruminal N disappearance were unaffected (P > .10) by supplementation and the type of supplement provided. Combining pretanned leather shavings with soybean meal seemed to have no deleterious effects on digestion or fermentation and to allow for escape of some N to the lower tract.


Subject(s)
Animal Feed , Cattle/metabolism , Dietary Fiber/metabolism , Nitrogen/metabolism , Rumen/metabolism , Tanning , Animals , Digestion , Fermentation , Poaceae , Glycine max
15.
Cardiovasc Res ; 45(3): 579-87, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10728379

ABSTRACT

OBJECTIVE: Enterovirus infections are major etiological factors in myocarditis and dilated cardiomyopathy. Using an experimental murine model of this disease, previous studies have shown that myocarditis susceptibility depends upon activation of T lymphocytes expressing the gamma delta T cell receptor (TcR), and that only mouse strains which accumulate gamma delta T cells in the myocardium show apoptosis of myocytes or evidence of dilated cardiomyopathy-like disease. The objective of the present studies is to demonstrate that gamma delta T cells directly induce greater Fas-dependent apoptosis of cultured myocytes than T cells expressing the alpha beta TcR. METHODS: Bl.Tg.E alpha mice were infected for 7 days with coxsackievirus B3 (CVB3). Hearts were removed and were either formalin-fixed, sectioned and stained with hematoxylin and eosin for inflammation, and using TdT-TUNEL for apoptosis, or were minced and collagenase digested for isolation of gamma delta+ and alpha beta+ T cells using immunomagnetic bead separation. Neonatal cultures of cardiac myocytes were isolated from mice less than 2 days old by collagenase and pancreatin digestion, and were either untreated or infected with virus. Levels of Fas (CD95) were measured using FITC-conjugated hamster anti-mouse Fas monoclonal antibody and flow cytometry. Susceptibility of myocytes to Fas-dependent killing was measured by 51Cr-release by labeled myocytes incubated for 4 h on either 3T3-mock or 3T3-FasL transfected cell monolayers. Killing by T cells was also measured in a 4 h 51Cr-release assay. Fas-dependent and perforin-dependent cytotoxicity was determined by specific blocking using either Fas-Fc or concanamycin A. RESULTS: Virally infected myocyte cultures showed significantly enhanced Fas expression compared to uninfected cells, with maximal upregulation of Fas occurring 18-24 h after virus infection. Both infected and uninfected myocytes were selectively killed by FasL-transfected 3T3 cells but not by mock control cells. Approximately 38% of CD3+ lymphocytes isolated from the heart express the gamma delta TcR with the remainder expressing the alpha beta TcR. Both gamma delta+ and alpha beta+ T cells lysed myocyte targets. Blocking studies indicate that gamma delta+ T cells induced predominantly Fas-mediated killing, while alpha beta+ cell produced more perforin-mediated death, although these effectors were capable of Fas-dependent killing as well. CONCLUSIONS: These studies demonstrate that T cells expressing the gamma delta TcR are more effective mediators of myocyte apoptosis than alpha beta+ T cells in vitro and suggests that these effectors may be primarily responsible for myocardial injury associated with dilated cardiomyopathy-like signs during coxsackievirus B3-induced myocarditis.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/immunology , Myocarditis/immunology , Myocardium/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Animals , Cell Communication , Cells, Cultured , Coculture Techniques , Cytotoxicity Tests, Immunologic , Flow Cytometry , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/physiopathology , Receptors, Antigen, T-Cell, gamma-delta/genetics , fas Receptor/metabolism
16.
Arterioscler Thromb Vasc Biol ; 19(10): 2364-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10521365

ABSTRACT

Acute-phase proteins, which respond to systemic proinflammatory cytokines such as interleuken-6, are elevated in cardiovascular disease and are predictive markers of future ischemic events, even over decades. This suggests a role for proinflammatory cytokines and/or acute phase proteins in early lesion development. To explore this issue, we fed C57Bl/6 and nonobese diabetic male mice high-fat (20% total fat, 1.5% cholesterol) diets and ApoE-deficient male mice both high-fat and normal chow diets for 6 to 21 weeks, injecting them weekly with either 5000 U recombinant interleukin-6 (rIL-6) or saline buffer. Blood was collected when animals were euthanized and assayed for cytokines, acute-phase proteins, and cholesterol. Across all mice, IL-6 injection resulted in significant increases in proinflammatory cytokines (IL-6, 4.6-fold; IL-1beta, 1.6-fold; and tissue necrosis factor-alpha, 1.7-fold) and fibrinogen (1.2-fold) and with decreased concentrations of albumin (0.9-fold) in plasma. Total cholesterol levels were unchanged between rIL-6-treated and nontreated groups. Serial sections through the aortic sinus were stained with oil red O to detect fatty streaks, and area of the lesions was determined by image analysis. Although no fatty streaks were detected in the nonobese diabetic mice with or without rIL-6 treatment, rIL-6 treatment increased lesion size in C57Bl/6 and ApoE-deficient mice 1.9- to 5.1-fold over lesions in saline-treated animals. These results suggest that under the appropriate circumstances changes in circulating proinflammatory cytokines and acute-phase proteins may be more than just markers of atherosclerosis but actual participants in early lesion development.


Subject(s)
Arteriosclerosis/chemically induced , Arteriosclerosis/immunology , Diabetes Mellitus, Type 1/immunology , Interleukin-6/pharmacology , Animals , Apolipoproteins E/genetics , Cholesterol/blood , Interleukin-6/blood , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Recombinant Proteins/pharmacology , Vasculitis/chemically induced , Vasculitis/immunology
17.
Lupus ; 8(5): 384-7, 1999.
Article in English | MEDLINE | ID: mdl-10455518

ABSTRACT

Coxsackievirus B3 (CVB3) induces myocarditis in male BALB/c mice. Female mice are resistant to viral myocarditis, except in the third trimester of pregnancy and postpartum. Cardiac damage is mediated by T lymphocytes activated during virus infection. Th1 (interferon-gamma+) cell responses promote cardiac injury, while disease resistance correlates to preferential activation of Th2 (interleukin-4+) cell responses. CVB3-specific Th1 and Th2 cell clones were established, treated with between 0 and 100 ng/ml 17beta estradiol and 4-androsten-17beta-ol-one (testosterone) for two days, 51Cr-labeled and cultured on FasL-transfected 3T3 cells to determine susceptibility to Fas-dependent apoptosis. Testosterone treatment enhanced Th2 cell lysis while estradiol treatment was protective. Staining of Th2 cells for Bcl 2, an anti-apoptotic factor, indicates that Bcl 2 expression increased in these cells with estradiol but decreased with testosterone exposure. Hormone-induced changes in Bcl 2 expression likely explain the selective survival of Th2 cells in females and prevention of viral myocarditis.


Subject(s)
Apoptosis/drug effects , Estradiol/pharmacology , Proto-Oncogene Proteins c-bcl-2/analysis , Testosterone/pharmacology , Th2 Cells/drug effects , fas Receptor/physiology , 3T3 Cells , Animals , Dose-Response Relationship, Drug , Female , Male , Mice , Rabbits , Th1 Cells/drug effects , Th1 Cells/physiology , Th2 Cells/physiology
18.
J Virol ; 73(7): 5630-6, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364312

ABSTRACT

Coxsackievirus B3 (CVB3) infection induces myocardial inflammation and myocyte necrosis in some, but not all, strains of mice. C57BL/6 mice, which inherently lack major histocompatibility complex (MHC) class II IE antigen, develop minimal cardiac lesions despite high levels of virus in the heart. The present experiments evaluate the relative roles of class II IA and IE expression on myocarditis susceptibility in four transgenic C57BL/6 mouse strains differing in MHC class II antigen expression. Animals lacking MHC class II IE antigen (C57BL/6 [IA+ IE-] and ABo [IA- IE-]) developed minimal cardiac lesions subsequent to infection despite high concentrations of virus in the heart. In contrast, strains expressing IE (ABo Ealpha [IA- IE+] and Bl.Tg.Ealpha [IA+ IE+]) had substantial cardiac injury. Myocarditis susceptibility correlated to a Th1 (gamma interferon-positive) cell response in the spleen, while disease resistance correlated to a preferential Th2 (interleukin-4-positive) phenotype. Vgamma/Vdelta analysis indicates that distinct subpopulations of gamma delta+ T cells are activated after CVB3 infection of C57BL/6 and Bl.Tg.Ealpha mice. Depletion of gamma delta+ T cells abrogated myocarditis susceptibility in IE+ animals and resulted in a Th1-->Th2 phenotype shift. These studies indicate that the MHC class II antigen haplotype controls myocarditis susceptibility, that this control is most likely mediated through the type of gamma delta T cells activated during CVB3 infection, and finally that different subpopulations of gamma delta+ T cells may either promote or inhibit Th1 cell responses.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human , Histocompatibility Antigens Class II/immunology , Myocarditis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/virology , Coxsackievirus Infections/pathology , Disease Models, Animal , Disease Susceptibility/immunology , Female , Histocompatibility Antigens Class II/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/pathology , Myocarditis/virology , Th1 Cells/immunology
19.
J Virol ; 73(6): 4689-95, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10233928

ABSTRACT

Coxsackievirus B3 infection causes significant cardiac inflammation in male, but not female, B1.Tg.Ealpha mice. This gender difference in disease susceptibility correlates with selective induction of CD4(+) Th1 (gamma interferon-positive) cell responses in animals with testosterone, whereas estradiol promotes preferential CD4(+) Th2 (interleukin-4 positive [IL-4(+)]) cell responses. Differences in immune deviation of CD4(+) T cells cannot be explained by variation in B7-1 or B7-2 expression. Infection significantly upregulated both molecules, but no differences were detected between estradiol- and testosterone-treated groups. Significantly increased numbers of activated (CD69(+)) T cells expressing the gammadelta T-cell receptor were found in male and testosterone-treated male and female mice. In vivo depletion of gammadelta+ cells by using monoclonal antibodies inhibited myocarditis and resulted in a shift from a Th1 to Th2 response phenotype. Taken together, our results indicate that testosterone promotes a CD4(+) Th1 cell response and myocarditis by promoting increased gammadelta+ cell activation.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Coxsackievirus Infections/immunology , Enterovirus B, Human , Myocarditis/immunology , Testosterone/pharmacology , Animals , CD4-Positive T-Lymphocytes/immunology , Estradiol/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/analysis
20.
Ann N Y Acad Sci ; 887: 77-82, 1999.
Article in English | MEDLINE | ID: mdl-10668465

ABSTRACT

Here we discuss the possibility that the way cells utilize fuel(s) for energy confers the properties that can be recognized by the immune system and, reciprocally, that recognition by the immune system can alter the balance of the cell's energy metabolism. We propose that immune recognition, of somatic cells via MHC can alter the their energy metabolism and induce a metabolic shift. We demonstrate the reciprocal relationship that inducing a shift in metabolism toward glycolysis by supplying glucose and insulin results in the upregulation of immunologically recognizable molecules such as cell surface Fas. Thus, immune recognition can induce metabolic deviation. Metabolic deviation can result in altered immune recognition and ultimately in cell proliferation, cell differentiation, or cell death.


Subject(s)
Cell Death/physiology , Cell Division/physiology , Energy Metabolism , Glycolysis , Animals , Apoptosis , Cell Death/immunology , Cell Differentiation , Cell Division/immunology , Glucose/metabolism , Humans , Insulin/physiology , Major Histocompatibility Complex , Models, Biological , fas Receptor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...