Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 189(10): 4740-7, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23041568

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily. PPARγ, a ligand-activated transcription factor, has important anti-inflammatory and antiproliferative functions, and it has been associated with diseases including diabetes, scarring, and atherosclerosis, among others. PPARγ is expressed in most bone marrow-derived cells and influences their function. PPARγ ligands can stimulate human B cell differentiation and promote Ab production. A knowledge gap is that the role of PPARγ in B cells under physiological conditions is not known. We developed a new B cell-specific PPARγ (B-PPARγ) knockout mouse and explored the role of PPARγ during both the primary and secondary immune response. In this article, we show that PPARγ deficiency in B cells decreases germinal center B cells and plasma cell development, as well as the levels of circulating Ag-specific Abs during a primary challenge. Inability to generate germinal center B cells and plasma cells is correlated to decreased MHC class II expression and decreased Bcl-6 and Blimp-1 levels. Furthermore, B-PPARγ-deficient mice have an impaired memory response, characterized by low titers of Ag-specific Abs and low numbers of Ag-experienced, Ab-secreting cells. However, B-PPARγ-deficient mice have no differences in B cell population distribution within primary or secondary lymphoid organs during development. This is the first report, to our knowledge, to show that, under physiological conditions, PPARγ expression in B cells is required for an efficient B cell-mediated immune response as it regulates B cell differentiation and Ab production.


Assuntos
Anticorpos/imunologia , Formação de Anticorpos/fisiologia , Especificidade de Anticorpos/fisiologia , Diferenciação Celular/imunologia , PPAR gama/imunologia , Plasmócitos/imunologia , Animais , Anticorpos/genética , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Humanos , Camundongos , Camundongos Knockout , Especificidade de Órgãos , PPAR gama/genética , Plasmócitos/citologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Proto-Oncogênicas c-bcl-6 , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
2.
J Immunol ; 183(11): 6903-12, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19915048

RESUMO

Protective humoral immune responses critically depend on the optimal differentiation of B cells into Ab-secreting cells. Because of the important role of Abs in fighting infections and in successful vaccination, it is imperative to identify mediators that control B cell differentiation. Activation of B cells through TLR9 by CpG-DNA induces plasma cell differentiation and Ab production. Herein, we examined the role of the peroxisome proliferator-activated receptor (PPAR)gamma/RXRalpha pathway on human B cell differentiation. We demonstrated that activated B cells up-regulate their expression of PPARgamma. We also show that nanomolar levels of natural (15-deoxy-Delta(12,14)-prostaglandin J(2)) or synthetic (rosiglitazone) PPARgamma ligands enhanced B cell proliferation and significantly stimulated plasma cell differentiation and Ab production. Moreover, the addition of GW9662, a specific PPARgamma antagonist, abolished these effects. Retinoid X receptor (RXR) is the binding partner for PPARgamma and is required to produce an active transcriptional complex. The simultaneous addition of nanomolar concentrations of the RXRalpha ligand (9-cis-retinoic acid) and PPARgamma ligands to CpG-activated B cells resulted in additive effects on B cell proliferation, plasma cell differentiation, and Ab production. Furthermore, PPARgamma ligands alone or combined with 9-cis-retinoic acid enhanced CpG-induced expression of Cox-2 and the plasma cell transcription factor BLIMP-1. Induction of these important regulators of B cell differentiation provides a possible mechanism for the B cell-enhancing effects of PPARgamma ligands. These new findings indicate that low doses of PPARgamma/RXRalpha ligands could be used as a new type of adjuvant to stimulate Ab production.


Assuntos
Formação de Anticorpos/imunologia , Linfócitos B/citologia , Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , PPAR gama/imunologia , Linfócitos B/imunologia , Western Blotting , Proliferação de Células , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/imunologia , Citometria de Fluxo , Expressão Gênica/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Ligantes , PPAR gama/biossíntese , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Repressoras/biossíntese , Proteínas Repressoras/imunologia , Receptor X Retinoide alfa/biossíntese , Receptor X Retinoide alfa/imunologia , Transfecção , Regulação para Cima
3.
Thromb Haemost ; 99(1): 86-95, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18217139

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) and its ligands are important regulators of lipid metabolism, inflammation, and diabetes. We previously demonstrated that anucleate human platelets express the transcription factor PPARgamma and that PPARgamma ligands blunt platelet activation. To further understand the nature of PPARgamma in platelets, we determined the platelet PPARgamma isoform(s) and investigated the fate of PPARgamma following platelet activation. Our studies demonstrated that human platelets contain only the PPARgamma1 isoform and after activation with thrombin, TRAP, ADP or collagen PPARgamma is released from internal stores. PPARgamma release was blocked by a cytoskeleton inhibitor, Latrunculin A. Platelet-released PPARgamma was complexed with the retinoid X receptor (RXR) and retained its ability to bind DNA. Interestingly, the released PPARgamma and RXR were microparticle associated and the released PPARgamma/RXR complex retained DNA-binding ability. Additionally, a monocytic cell line, THP-1, is capable of internalizing PMPs. Further investigation following treatment of these cells with the PPARgamma agonist, rosiglitazone and PMPs revealed a possible transcellular mechanism to attenuate THP-1 activation. These new findings are the first to demonstrate transcription factor release from platelets, revealing the complex spectrum of proteins expressed and expelled from platelets, and suggests that platelet PPARgamma has an undiscovered role in human biology.


Assuntos
Plaquetas/metabolismo , PPAR gama/metabolismo , Ativação Plaquetária , Receptores X de Retinoides/metabolismo , Vesículas Transportadoras/metabolismo , Adulto , Plaquetas/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular Tumoral , Colágeno/metabolismo , DNA/metabolismo , Dimerização , Feminino , Humanos , Masculino , Megacariócitos/metabolismo , Pessoa de Meia-Idade , Monócitos/metabolismo , PPAR gama/agonistas , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptor X Retinoide alfa/metabolismo , Receptor X Retinoide beta/metabolismo , Receptores X de Retinoides/genética , Rosiglitazona , Tiazolidinedionas/farmacologia , Tiazolidinas/farmacologia , Trombina/metabolismo , Fatores de Tempo
4.
Am J Obstet Gynecol ; 196(4): 346.e1-8, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17403416

RESUMO

OBJECTIVE: The objective of the study was to determine whether vestibular fibroblasts from vulvar vestibulitis (VVS) patients produce higher proinflammatory cytokine levels when provoked with Candida albicans (yeast) and alpha-melanocyte-stimulating hormone (alpha-MSH) in vitro. STUDY DESIGN: Twenty anatomically defined fibroblast strains from patients and age-matched controls were stimulated with 5 regimens: no stimulus, alpha-MSH, heat-killed yeast, alpha-MSH plus yeast, and interleukin (IL)-1beta. Supernatant products included the following: granulocyte macrophage colony-stimulating factor, interferon-gamma, IL-10, IL-12, IL-1beta, IL-2, IL-4, IL-6, IL-8, and tumor necrosis factor-alpha were assayed. RESULTS: Coincubation with alpha-MSH plus yeast significantly increased IL-6 (3-fold) and IL-8 (greater than 40-fold) production in patients and controls. Vestibular fibroblast exceeded external vulvar fibroblast production of IL-1beta, IL-6, and IL-8 following yeast alone and alpha-MSH plus yeast stimuli in patients and controls. Substratified by anatomic origin, vestibular fibroblasts from VVS patients produced the highest relative levels of IL-1beta, IL-6, and IL-8 at baseline and following the yeast-alone regimen. CONCLUSION: Localized pain of VVS may results from regionally elevated cytokines produced by vulvar vestibule-specific fibroblasts.


Assuntos
Candida albicans , Citocinas/biossíntese , Fibroblastos/metabolismo , Vulvite/metabolismo , alfa-MSH/farmacologia , Adulto , Biópsia por Agulha , Candidíase Vulvovaginal/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Feminino , Humanos , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Interleucina-8/biossíntese , Pessoa de Meia-Idade , Probabilidade , Valores de Referência , Estudos de Amostragem , Sensibilidade e Especificidade , Estatísticas não Paramétricas , Vulvite/patologia
5.
J Biotechnol ; 150(3): 417-27, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20888877

RESUMO

Widely known for its role in adipogenesis and energy metabolism, PPARγ also plays a role in platelet function. To further understand functions of platelet-derived PPARγ, we produced rabbit polyclonal (PoAbs) and mouse monoclonal (MoAbs) antibodies against PPARγ 14mer/19mer peptide-immunogens. Unexpectedly, our work produced two key findings. First, MoAbs but not PoAbs produced against PPARγ peptide-immunogens displayed antigenic crossreactivity with highly conserved PPARα and PPARß/δ. Similarly, Santa Cruz PoAb sc-7196 was monospecific for PPARγ while MoAb sc-7273 crossreacted with PPARα and PPARß/δ. Second, immunized rabbits and mice exhibited unusual pathology including cachexia, excessive bleeding, and low platelet counts leading to thrombocytopenia. Spleens from immunized mice were fatty, hemorrhagic and friable. Although passive administration of anti-PPARγ PoAbs failed to induce experimental thrombocytopenia, megakaryocytopoiesis was induced 4-8-fold in mouse spleens. Similarly, marrow megakaryocytopoiesis was enhanced 1.8-4-fold in immunized rabbits. These peptide-immunogens are 100% conserved in human, rabbit and mouse; thus, immune-mediated platelet destruction via crossreactivity with platelet-derived PPARγ likely caused bleeding, thrombocytopenia, and compensatory megakaryocytopoiesis. Such overt pathology would cause significant problems for large-scale production of anti-PPARγ PoAbs. Furthermore, a major pitfall associated with MoAb production against closely related molecules is that monoclonicity does not guarantee monospecificity, an issue worth further scientific scrutiny.


Assuntos
Anticorpos/metabolismo , Homeostase/fisiologia , PPAR gama/fisiologia , Peptídeos/imunologia , Trombocitopenia , Trombopoese , Animais , Anticorpos/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Caquexia , Mapeamento de Epitopos , Feminino , Histocitoquímica , Humanos , Intestinos/patologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ovário/patologia , PPAR gama/antagonistas & inibidores , PPAR gama/química , PPAR gama/imunologia , PPAR gama/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Isoformas de Proteínas , Coelhos , Baço/patologia , Trombocitopenia/patologia , Trombocitopenia/fisiopatologia
6.
J Immunol ; 174(5): 2619-26, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15728468

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) are widely used for the treatment of inflammatory diseases and target cyclooxygenases 1 and 2 (Cox-1, Cox-2) that are responsible for PG production. Newer Cox-2-selective drugs have been heavily prescribed to quench inflammation. Little is known about whether or not these drugs influence human B lymphocytes and their ability to produce Ab. We report herein that activated human B cells not only highly express Cox-2 and produce PGs, but that the NSAID indomethacin and Cox-2-selective drugs profoundly inhibit the ability of human B cells to produce IgG and IgM in vitro. Human blood B cells highly express Cox-2 mRNA and protein and produce PGs after activation with CD40L, pansorbin, or CD40L plus BCR engagement. Cox-2 is also highly expressed by human tonsil B cells, as shown by immunohistochemistry. Cox-inhibiting drugs modestly affect purified B cell proliferation but profoundly reduce Ab production. The ability of whole blood to produce IgM and IgG following stimulation is also strongly inhibited. In support that Cox-2 plays a seminal role in B lymphocyte Ab production, Cox-2 knockout mice have 64% less IgM and 35% less IgG than normal littermate controls. These findings support that NSAIDs and the new Cox-2-selective drugs have an unsuspected target, the B cell, and attenuate Ab production in humans. Use of NSAIDs may therefore influence autoantibody production in autoimmune diseases and may dampen humoral immunity in response to antigenic challenge/vaccination.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Subpopulações de Linfócitos B/enzimologia , Subpopulações de Linfócitos B/imunologia , Inibidores de Ciclo-Oxigenase/farmacologia , Imunossupressores/farmacologia , Ativação Linfocitária/imunologia , Prostaglandina-Endoperóxido Sintases/biossíntese , Animais , Subpopulações de Linfócitos B/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Dinoprosta/biossíntese , Dinoprostona/biossíntese , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Imunoglobulina M/sangue , Imunoglobulina M/deficiência , Ativação Linfocitária/efeitos dos fármacos , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prostaglandina-Endoperóxido Sintases/deficiência , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/fisiologia , RNA Mensageiro/biossíntese , Regulação para Cima/genética , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA