Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Transplant ; 16(1): 334-41, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26372909

RESUMO

The strongly immunogenic environment in autoimmune diseases such as lupus may pose a stringent barrier to transplantation. Despite available murine models of lupus, transplant tolerance in this setting has yet to be fully investigated in highly penetrant genetic models of disease. Such studies are of clear clinical importance because lupus is a transplant indication in which transplanted kidneys have a substantially increased risk of rejection including a role for recurrent nephritis. In the fully penetrant B6.SLE123 mouse, we determined that CD4 T follicular helper and germinal center B cells were significantly expanded compared with healthy controls. We traced this expansion to resistance of effector CD4 T and B cells in B6.SLE123 mice to regulation by either CD4 T regulatory cells (CD4Tregs) or CD8 T regulatory cells (CD8Tregs), despite demonstrating normal function by Tregs in this strain. Finally, we determined that B6.SLE123 mice resist anti-CD45RB-mediated tolerance induction to foreign islet allografts, even in the absence of islet autoimmunity. Overall, B6.SLE123 lupus-prone mice are highly resistant to transplant tolerance induction, which provides a new model of failed tolerance in autoimmunity that may elucidate barriers to clinical transplantation in lupus through further cellular and genetic dissection.


Assuntos
Autoanticorpos/imunologia , Autoimunidade/imunologia , Rejeição de Enxerto/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Nefrite Lúpica/etiologia , Linfócitos T Reguladores/imunologia , Tolerância ao Transplante/imunologia , Animais , Autoanticorpos/sangue , Células Cultivadas , Células Dendríticas/imunologia , Sobrevivência de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Lúpus Eritematoso Sistêmico/complicações , Nefrite Lúpica/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Endogâmicos NOD
2.
Thromb Haemost ; 106(5): 849-57, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21979131

RESUMO

Rheumatic autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus (SLE), are associated with antibodies to "self" antigens. Persons with autoimmune diseases, most notably SLE, are at increased risk for developing accelerated cardiovascular disease. The link between immune and inflammatory responses in the pathogenesis of cardiovascular disease has been firmly established; yet, despite our increasing knowledge, accelerated atherosclerosis continues to be a significant co-morbidity and cause of mortality in SLE. Recent animal models have been generated in order to identify mechanism(s) behind SLE-accelerated atherosclerosis. In addition, clinical studies have been designed to examine potential treatments options. This review will highlight data from recent studies of immunity in SLE and atherosclerosis and discuss the potential implications of these investigations.


Assuntos
Artérias/imunologia , Aterosclerose/imunologia , Autoimunidade , Inflamação/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Aterosclerose/epidemiologia , Aterosclerose/terapia , Comorbidade , Modelos Animais de Doenças , Progressão da Doença , Humanos , Inflamação/epidemiologia , Inflamação/terapia , Lúpus Eritematoso Sistêmico/epidemiologia , Lúpus Eritematoso Sistêmico/terapia , Medição de Risco , Fatores de Risco , Fatores de Tempo
3.
Lupus ; 19(1): 34-42, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19850656

RESUMO

Cardiovascular disease risk is increased in individuals suffering from systemic lupus erythematosus. Understanding the mechanism(s) of systemic lupus erythematosus-accelerated atherosclerosis is critical for the development of effective therapies. Our laboratory previously demonstrated that radiation chimeras of systemic lupus erythematosus-susceptible B6.Sle1.2.3 and low density lipoprotein receptor (LDLr)(-/-) mice have augmented atherosclerosis, which is associated with increased T-cell burden and activation in the lesion. The goals of this study were to further define specific immune mechanisms that mediate accelerated atherosclerosis and to determine whether the gene interval Sle3, which is linked to lupus-associated T-cell dysregulation, was sufficient to modulate atherogenesis. We transferred B6.Sle3 or C57Bl/6-derived bone marrow cells into lethally irradiated LDLr( -/-) mice (hereafter referred to as LDLr.Sle3 and LDLr.B6, respectively). Sixteen weeks after transplantation, the mice were placed on a western-type diet for 8 weeks. Our analyses revealed that LDLr.Sle3 mice had increased auto-antibody production against double-stranded DNA and cardiolipin compared with LDLr.B6 controls. We also found an increase in atherosclerosis-associated oxLDL antibodies. Antibody isotypes and serum cytokine analysis suggested that the humoral immune response in LDLr.Sle3 mice was skewed toward a Th2 phenotype. This finding is consistent with lupus-associated immune dysregulation. Additionally, LDLr.Sle3 mice had decreased serum cholesterol and triglyceride levels. However, there was no difference in lesion area or cellular composition of lesions between the two groups. These data demonstrate that, despite no change in lesion area, transfer of Sle3-associated T-cell dysregulation alone to LDLr-deficient mice is sufficient to decrease serum cholesterol and to exacerbate humoral immune responses that are frequently associated with atherosclerosis.


Assuntos
Aterosclerose/etiologia , Predisposição Genética para Doença , Lúpus Eritematoso Sistêmico/genética , Receptores de LDL/fisiologia , Animais , Formação de Anticorpos , Mapeamento Cromossômico , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/deficiência , Linfócitos T/imunologia
4.
Lupus ; 17(12): 1070-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19029274

RESUMO

Individuals suffering from systemic lupus erythematosus (SLE) are predisposed to accelerate cardiovascular disease. Our laboratory has recently developed an animal model of SLE-accelerated atherosclerosis. We have shown that, following 8 weeks feeding high fat Western diet, radiation chimeras consisting of SLE-derived haematopoietic cells transferred to low-density lipoprotein (LDL)r(-/-) mice (LDLr.Sle) have increased atherosclerosis compared with C57Bl/6 bone marrow recipients (LDLr.B6). However, this feeding regimen resulted in significant mortality in SLE-susceptible mice compared with controls with surviving animals having extremely elevated serum cholesterol (>500 mg/dL) and increased serum markers of kidney pathology. To test the hypothesis that SLE-associated autoimmune dysregulation can exacerbate atherosclerosis under more mild serum cholesterol conditions (approximately 200 mg/dL), we examined SLE and lesion development in radiation chimeras fed either a normal chow or high fat Western diet for 8 weeks. High fat fed LDLr.Sle mice exhibited increased mortality and were significantly more hypertensive. LDLr.Sle mice had greater titres of antibodies against dsDNA, oxLDL and phospholipid compared with controls. Lupus-susceptibility increased the atherosclerotic lesions and the percentage of CD4(+) T cells in the lesions of proximal aortas, independent of diet. These data show that increased dyslipidemia resulting from high-fat feeding can exacerbate autoimmunity and associated vascular complications. Conversely, they also show that autoimmune dysregulation can accelerate atherosclerosis in LDLr-deficient animals independent of feeding high fat diet. Collectively this study provides additional evidence that the accelerated atherosclerosis observed in SLE is autoimmune associated.


Assuntos
Aterosclerose , Gorduras na Dieta/farmacologia , Lúpus Eritematoso Sistêmico , Receptores de LDL/genética , Animais , Anticorpos Antifosfolipídeos/metabolismo , Aterosclerose/complicações , Aterosclerose/genética , Aterosclerose/imunologia , Autoimunidade/imunologia , LDL-Colesterol/metabolismo , VLDL-Colesterol/metabolismo , DNA/imunologia , Dislipidemias/complicações , Dislipidemias/genética , Dislipidemias/imunologia , Lipoproteínas LDL/metabolismo , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptores de LDL/metabolismo
5.
Circulation ; 104(23): 2820-5, 2001 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-11733401

RESUMO

BACKGROUND: We recently used a bone marrow-based gene therapy approach to show that small amounts of retrovirus-derived human apolipoprotein E3 (apoE3) produced by macrophages are protective against early atherosclerosis in apoE-deficient mice. METHODS AND RESULTS: In the present study, we evaluated whether the effect produced by macrophage-derived apoE3 is related to its ability to bind cellular membranes. To this end, we used apoE2 and apoEcys142, dysfunctional human variants with reduced binding to the LDL receptor or to heparan sulfate proteoglycans, respectively. ApoE-deficient mice, 5 weeks of age, received transplants of apoE(-/-) bone marrow cells transduced with either parental retrovirus or apoE3, apoE2, or apoEcys142 retroviral vectors. Human apoE was detected by ELISA in the serum of apoE3, apoE2, and apoEcys142 mice as early as 4 weeks after bone marrow transplantation, and at 8 weeks, plasma apoE levels were 55.5+/-20.3, 50.5+/-8.7, and 15.3+/-7.3 microgram/dL, respectively. In all groups, cholesterol levels increased with age but were not affected by apoE expression. As previously demonstrated, the lesion area in male apoE3 mice (3808+/-2224 micrometer(2)/section) was 40% smaller than that in control mice (6503+/-3475 micrometer(2)/section). In apoE2 mice, however, the lesion area was similar to that of controls (5991+/-2771 micrometer(2)/section), and apoEcys142 mice showed an unexpected and significant increase in lesion size (10 320+/-6128 micrometer(2)/section). Thus, transplantation with marrow transfected with receptor binding-defective apoE variants did not replicate the antiatherogenic effect of apoE3. CONCLUSIONS: These data provide in vivo evidence suggesting that macrophage-derived apoE delays development of atherosclerosis through a receptor-dependent pathway.


Assuntos
Apolipoproteínas E/fisiologia , Arteriosclerose/patologia , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/sangue , Apolipoproteínas E/genética , Arteriosclerose/genética , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Clonagem Molecular , Feminino , Técnicas de Transferência de Genes , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Masculino , Camundongos , Camundongos Knockout , Isoformas de Proteínas/sangue , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Retroviridae/genética , Fatores de Tempo
6.
Arterioscler Thromb Vasc Biol ; 21(11): 1790-5, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11701467

RESUMO

The concentration of apolipoprotein (apo) AI in the artery wall is thought to enhance cellular cholesterol efflux and protect against atherosclerosis. It has been shown that although macrophages do not make apoAI, they respond to it by increased cholesterol efflux. We hypothesized that macrophage production of apoAI would increase cholesterol efflux and reduce atherogenesis. In this study, we produced mice expressing human apoAI under the control of the macrophage-specific scavenger receptor-A promoter (mphi-AI). Human apoAI was detectable in the serum HDL fraction of mphi-AI transgenic mice at concentrations too low to affect serum cholesterol or HDL levels. Immunoblotting showed the presence of human apoAI in transgenic macrophage culture supernatants, mostly as lipoprotein-free protein, with a small component associated with HDL-like particles. Atherosclerosis studies using apoAI((-/-)) mice transplanted with mphi-AI bone marrow showed that in the absence of macrophage-derived apoE, local expression of apoAI reduced diet-induced lesions in the proximal aorta. Additionally, mphi-AI macrophages showed a 40% increase in cholesterol efflux compared with control macrophages. These data support the hypothesis that apoAI production by macrophages in the artery wall is protective against atherosclerosis. This protection is likely mediated by increased cholesterol efflux and decreased foam cell formation in vivo.


Assuntos
Apolipoproteína A-I/biossíntese , Apolipoproteína A-I/genética , Arteriosclerose/metabolismo , Colesterol/metabolismo , Macrófagos/metabolismo , Animais , Arteriosclerose/etiologia , Transporte Biológico , Células Cultivadas , Dieta Aterogênica , Células Espumosas/fisiologia , Humanos , Lipoproteínas HDL/química , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
7.
J Biol Chem ; 276(39): 36742-8, 2001 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-11477092

RESUMO

We have previously reported that the lack of apolipoprotein (apo) E expression by macrophages promotes foam cell formation in vivo. Because transgenic mice overexpressing human apoA-I from the liver (h-apoA-I TgN) are protected from the atherogenesis induced by apoE deficiency, we hypothesized that the presence of apoA-I in the vessel wall could reduce the negative effect of apoE deficiency on lesion growth. To address this issue, we used both retroviral transduction and transgenic approaches to produce in vivo systems where apoA-I is expressed from macrophages. In the retroviral transduction study, apoA-I-deficient (apoA-I(-/-)) mice reconstituted with apoE-deficient (apoE(-/-)) bone marrow cells that were infected with a retroviral vector expressing human apoA-I (MFG-HAI) had 95% lower atherosclerotic lesion area than that of recipients of apoE(-/-) bone marrow cells infected with the parental virus (MFG). To determine whether the protective effect of locally produced apoA-I was due to the lack of systemic apoA-I, we conducted a different experiment using h-apoA-I TgN mice as recipients of apoE(-/-) bone marrow with or without human apoA-I (driven by a macrophage-specific transgene defined as mphi-AI). Aortic lesion area in apoE(-/-)/mphi-AI --> h-apoA-I TgN mice was decreased by 85% compared with apoE(-/-) --> h-apoA-I TgN mice. These data demonstrate that expression of apoA-I from macrophages protects against atherogenesis without affecting plasma apoA-I and high density lipoprotein cholesterol levels.


Assuntos
Apolipoproteína A-I/biossíntese , Arteriosclerose/metabolismo , Macrófagos/metabolismo , Retroviridae/genética , Animais , Apolipoproteína A-I/química , Apolipoproteínas/metabolismo , Western Blotting , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Células Cultivadas , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Imuno-Histoquímica , Metabolismo dos Lipídeos , Lipoproteínas HDL/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Retroviridae/metabolismo , Fatores de Tempo , Transdução Genética , Transgenes
8.
J Biol Chem ; 276(25): 22965-70, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11304532

RESUMO

We have investigated the recycling of apoE in livers of apoE(-)/- mice transplanted with wild type bone marrow (apoE(+/+) --> apoE(-)/-), a model in which circulating apoE is derived exclusively from macrophages. Nascent Golgi lipoproteins were recovered from livers of apoE(+/+) --> apoE(-)/- mice 8 weeks after transplantation. ApoE was identified with nascent d < 1.006 and with d 1.006-1.210 g/ml lipoproteins at a level approximately 6% that of nascent lipoproteins from C57BL/6 mice. Hepatocytes from apoE(+/+) --> apoE(-)/- mice were isolated and cultured in media free of exogenous apoE. ApoE was found in the media primarily on the d < 1.006 g/ml fraction, indicating a resecretion of internalized apoprotein. Secretion of apoE from C57BL/6 hepatocytes was consistent with constitutive production, whereas the majority of apoE secreted from apoE(+/+) --> apoE(-)/- hepatocytes was recovered in the last 24 h of culture. This suggests that release may be triggered by accumulation of an acceptor, such as very low density lipoproteins, in the media. In agreement with the in vivo data, total recovery of apoE from apoE(+/+) --> apoE(-)/- hepatocytes was approximately 6% that of the apoE recovered from C57BL/6 hepatocytes. Since plasma apoE levels in the transplanted mice are approximately 10% of control levels, the findings indicate that up to 60% of the internalized apoE may be reutilized under physiologic conditions. These studies provide definitive evidence for the sparing of apoE and its routing through the secretory pathway and demonstrate that internalized apoE can be resecreted in a quantitatively significant fashion.


Assuntos
Apolipoproteínas E/metabolismo , Endocitose , Hepatócitos/metabolismo , Animais , Apolipoproteínas E/genética , Células Cultivadas , Hepatócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL
9.
J Clin Invest ; 107(2): 163-71, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11160132

RESUMO

During atherogenesis, circulating macrophages migrate into the subendothelial space, internalize cholesterol-rich lipoproteins, and become foam cells by progressively accumulating cholesterol esters. The inhibition of macrophage acyl coenzyme A:cholesterol acyltransferase (ACAT), which catalyzes the formation of cholesterol esters, has been proposed as a strategy to reduce foam cell formation and to treat atherosclerosis. We show here, however, that hypercholesterolemic LDL receptor-deficient (LDLR(-/-)) mice reconstituted with ACAT1-deficient macrophages unexpectedly develop larger atherosclerotic lesions than control LDLR(-/-) mice. The ACAT1-deficient lesions have reduced macrophage immunostaining and more free cholesterol than control lesions. Our findings suggest that selective inhibition of ACAT1 in lesion macrophages in the setting of hyperlipidemia can lead to the accumulation of free cholesterol in the artery wall, and that this promotes, rather than inhibits, lesion development.


Assuntos
Arteriosclerose/genética , Macrófagos/enzimologia , Receptores de LDL/deficiência , Esterol O-Aciltransferase/deficiência , Animais , Aorta/metabolismo , Aorta/patologia , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Transplante de Medula Óssea , Transplante de Células , Colesterol/metabolismo , Corantes , Feminino , Imuno-Histoquímica , Fígado/citologia , Fígado/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esterol O-Aciltransferase/antagonistas & inibidores , Regulação para Cima
11.
J Virol ; 71(8): 5782-9, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9223466

RESUMO

Enteric infection of mice with respiratory enteric orphan virus (reovirus) type 1, strain Lang elicits both humoral and cellular immune responses. To investigate the role of CD8+, alpha/beta T-cell receptor (TCR)+ T cells in mucosal immunity to an enteric pathogen, we examined immune responses and viral clearance following enteric reovirus infection in C57BL/6, B6129F2, and beta2-microglobulin-deficient (beta2m-/-) mice. Analysis of Peyer's patch and lamina propria culture supernatants revealed a two- to threefold increase in levels of reovirus-specific immunoglobulin A in beta2m-/- mice compared to normal controls. These data corresponded to a similar increase in the frequency of virus-specific immunoglobulin A-producing cells in Peyer's patches and lamina propria and an increase in immunoglobulin G-producing cells in spleens from beta2m-/- mice compared to controls. These increased humoral immune responses were not due to a difference in B-cell populations because cell counts and flow cytometric analyses showed that beta2m-/- and control mice had similar numbers and percentages of B cells in mucosal and systemic tissues. Analysis of cytokine message by reverse transcriptase-PCR 5 and 10 days after infection revealed no difference in message level for transforming growth factor beta, gamma interferon, interleukin-4, interleukin-5, or interleukin-6 for all mouse strains. Virus tissue titers determined by plaque assay at 5 and 10 days after infection demonstrated that beta2m-/- mice cleared reovirus from the small intestines with the same efficiency as control mice. Collectively, these data suggest that CD8+, alpha/beta TCR+ T cells may regulate mucosal and systemic humoral immune responses to oral infection with reovirus.


Assuntos
Enteropatias/imunologia , Infecções por Reoviridae/imunologia , Microglobulina beta-2/deficiência , Animais , Citocinas/genética , Imunidade nas Mucosas , Imunoglobulina A Secretora/biossíntese , Imunoglobulina G/biossíntese , Imunoglobulina G/classificação , Tecido Linfoide/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Linfócitos T Citotóxicos/imunologia
12.
J Virol ; 70(9): 5968-74, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8709219

RESUMO

Reovirus serotype 1, strain Lang (T1/L), a well characterized enteric virus, elicits humoral and cellular immune responses in mice. Although orally and intradermally induced infections generate comparable reovirus-specific serum antibody titers, little is known about the effects of the route of infection on the systemic immunoglobulin G (IgG) response. To assess whether the route of exposure affects virus-specific humoral immunity, we infected various strains of mice with reovirus T1/L by the oral or intradermal routes. At day 10 following infection, virus-specific serum antibody titers and IgG subclasses were determined by enzyme-linked immunosorbent assay. Serum IgG2a and IgG2b antibodies were detected in all mouse strains independent of the route of infection. Mice of the H-2d haplotype that received an intradermal infection also had high levels of reovirus-specific serum IgG1. This dichotomy of responses was not associated with differences in the types of cytokine produced by draining peripheral lymph nodes. However, peripheral lymph node lymphocytes from C3H mice produced significantly higher levels of gamma interferon than did BALB/c, C57BL/6, and B10.D2 mice. Additionally, peripheral lymph node lymphocytes from all strains of mice produced only low levels of interleukin-5, with no detectable level of interleukin-4 or interleukin-6. Analysis of specific antibody at inductive sites of the immune response showed that orally infected Peyer's patches produced predominantly IgA and intradermally infected peripheral lymph nodes produced predominantly IgG2a. Western blot (immunoblot) analysis showed that virus-specific IgA, IgG1, and IgG2a reacted with reovirus structural proteins. These data suggest that the route of infection affects the isotype and IgG subclasses, but not the antigen specificity, of the local antibody response. In addition, virus-specific IgG1 generated following an intradermally induced infection is linked to the H-2d major histocompatibility complex haplotype.


Assuntos
Imunoglobulina G/biossíntese , Orthoreovirus , Infecções por Reoviridae/imunologia , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Anticorpos Antivirais/classificação , Formação de Anticorpos , Especificidade de Anticorpos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Imunidade Celular , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Linfonodos/imunologia , Linfócitos/imunologia , Complexo Principal de Histocompatibilidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Orthoreovirus/imunologia , Baço/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...