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










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 125(3): 1189-202, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25664851

RESUMO

The lymphocyte adaptor protein LNK (also known as SH2B3) is primarily expressed in hematopoietic and endothelial cells, where it functions as a negative regulator of cytokine signaling and cell proliferation. Single-nucleotide polymorphisms in the gene encoding LNK are associated with autoimmune and cardiovascular disorders; however, it is not known how LNK contributes to hypertension. Here, we determined that loss of LNK exacerbates angiotensin II-induced (Ang II-induced) hypertension and the associated renal and vascular dysfunction. At baseline, kidneys from Lnk-/- mice exhibited greater levels of inflammation, oxidative stress, and glomerular injury compared with WT animals, and these parameters were further exacerbated by Ang II infusion. Aortas from Lnk-/- mice exhibited enhanced inflammation, reduced nitric oxide levels, and impaired endothelial-dependent relaxation. Bone marrow transplantation studies demonstrated that loss of LNK in hematopoietic cells is primarily responsible for the observed renal and vascular inflammation and predisposition to hypertension. Ang II infusion increased IFN-γ-producing CD8+ T cells in the spleen and kidneys of Lnk-/- mice compared with WT mice. Moreover, IFN-γ deficiency resulted in blunted hypertension in response to Ang II infusion. Together, these results suggest that LNK is a potential therapeutic target for hypertension and its associated renal and vascular sequela.


Assuntos
Hipertensão/genética , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células Cultivadas , Quimiotaxia de Leucócito , Hipertensão/imunologia , Interferon gama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Rim/imunologia , Rim/patologia , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite/genética , Nefrite/imunologia , Nefrite/patologia , Estresse Oxidativo , Linfócitos T/imunologia , Vasculite/genética , Vasculite/imunologia , Vasculite/patologia
2.
Hypertension ; 65(3): 569-76, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25601932

RESUMO

Ample genetic and physiological evidence establishes that renal salt handling is a critical regulator of blood pressure. Studies also establish a role for the immune system, T-cell infiltration, and immune cytokines in hypertension. This study aimed to connect immune cytokines, specifically interferon-γ (IFN-γ) and interleukin-17A (IL-17A), to sodium transporter regulation in the kidney during angiotensin-II (Ang-II) hypertension. C57BL/6J (wild-type) mice responded to Ang-II infusion (490 ng/kg per minute, 2 weeks) with a rise in blood pressure (170 mm Hg) and a significant decrease in the rate of excretion of a saline challenge. In comparison, mice that lacked the ability to produce either IFN-γ (IFN-γ(-/-)) or IL-17A (IL-17A(-/-)) exhibited a blunted rise in blood pressure (<150 mm Hg), and both the genotypes maintained baseline diuretic and natriuretic responses to a saline challenge. Along the distal nephron, Ang-II infusion increased abundance of the phosphorylated forms of the Na-K-2Cl cotransporter, Na-Cl cotransporter, and Ste20/SPS-1-related proline-alanine-rich kinase, in both the wild-type and the IL-17A(-/-) but not in IFN-γ(-/-) mice; epithelial Na channel abundance increased similarly in all the 3 genotypes. In the proximal nephron, Ang-II infusion significantly decreased abundance of Na/H-exchanger isoform 3 and the motor myosin VI in IL-17A(-/-) and IFN-γ(-/-), but not in wild-type; the Na-phosphate cotransporter decreased in all the 3 genotypes. Our results suggest that during Ang-II hypertension both IFN-γ and IL-17A production interfere with the pressure natriuretic decrease in proximal tubule sodium transport and that IFN-γ production is necessary to activate distal sodium reabsorption.


Assuntos
Angiotensina II/efeitos adversos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Interferon gama/deficiência , Interleucina-17/deficiência , Túbulos Renais Proximais/metabolismo , Sódio/metabolismo , Angiotensina II/farmacologia , Animais , Transporte Biológico/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Modelos Animais de Doenças , Canais Epiteliais de Sódio/metabolismo , Genótipo , Hipertensão/fisiopatologia , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Simportadores de Cloreto de Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
3.
Hypertension ; 64(5): 1108-15, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25259750

RESUMO

Recent studies have emphasized a role of adaptive immunity, and particularly T cells, in the genesis of hypertension. We sought to determine the T-cell subtypes that contribute to hypertension and renal inflammation in angiotensin II-induced hypertension. Using T-cell receptor spectratyping to examine T-cell receptor usage, we demonstrated that CD8(+) cells, but not CD4(+) cells, in the kidney exhibited altered T-cell receptor transcript lengths in Vß3, 8.1, and 17 families in response to angiotensin II-induced hypertension. Clonality was not observed in other organs. The hypertension caused by angiotensin II in CD4(-/-) and MHCII(-/-) mice was similar to that observed in wild-type mice, whereas CD8(-/-) mice and OT1xRAG-1(-/-) mice, which have only 1 T-cell receptor, exhibited a blunted hypertensive response to angiotensin II. Adoptive transfer of pan T cells and CD8(+) T cells but not CD4(+)/CD25(-) cells conferred hypertension to RAG-1(-/-) mice. In contrast, transfer of CD4(+)/CD25(+) cells to wild-type mice receiving angiotensin II decreased blood pressure. Mice treated with angiotensin II exhibited increased numbers of kidney CD4(+) and CD8(+) T cells. In response to a sodium/volume challenge, wild-type and CD4(-/-) mice infused with angiotensin II retained water and sodium, whereas CD8(-/-) mice did not. CD8(-/-) mice were also protected against angiotensin-induced endothelial dysfunction and vascular remodeling in the kidney. These data suggest that in the development of hypertension, an oligoclonal population of CD8(+) cells accumulates in the kidney and likely contributes to hypertension by contributing to sodium and volume retention and vascular rarefaction.


Assuntos
Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/fisiologia , Hipertensão/patologia , Hipertensão/fisiopatologia , Rim/imunologia , Rim/patologia , Bandas Oligoclonais/fisiologia , Imunidade Adaptativa/fisiologia , Angiotensina II/efeitos adversos , Angiotensina II/farmacologia , Animais , Antígenos CD4/genética , Antígenos CD4/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/fisiologia , Antígenos CD8/genética , Antígenos CD8/fisiologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Hipertensão/induzido quimicamente , Rim/irrigação sanguínea , Complexo Principal de Histocompatibilidade/genética , Complexo Principal de Histocompatibilidade/fisiologia , Masculino , Camundongos , Camundongos Knockout , Remodelação Vascular/efeitos dos fármacos
4.
Circ Res ; 114(4): 616-25, 2014 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-24347665

RESUMO

RATIONALE: Aortic stiffening commonly occurs in hypertension and further elevates systolic pressure. Hypertension is also associated with vascular inflammation and increased mechanical stretch. The interplay between inflammation, mechanical stretch, and aortic stiffening in hypertension remains undefined. OBJECTIVE: Our aim was to determine the role of inflammation and mechanical stretch in aortic stiffening. METHODS AND RESULTS: Chronic angiotensin II infusion caused marked aortic adventitial collagen deposition, as quantified by Masson trichrome blue staining and biochemically by hydroxyproline content, in wild-type but not in recombination activating gene-1-deficient mice. Aortic compliance, defined by ex vivo measurements of stress-strain curves, was reduced by chronic angiotensin II infusion in wild-type mice (P<0.01) but not in recombination activating gene-1-deficient mice (P<0.05). Adoptive transfer of T-cells to recombination activating gene-1-deficient mice restored aortic collagen deposition and stiffness to values observed in wild-type mice. Mice lacking the T-cell-derived cytokine interleukin 17a were also protected against aortic stiffening. In additional studies, we found that blood pressure normalization by treatment with hydralazine and hydrochlorothiazide prevented angiotensin II-induced vascular T-cell infiltration, aortic stiffening, and collagen deposition. Finally, we found that mechanical stretch induces the expression of collagen 1α1, 3α1, and 5a1 in cultured aortic fibroblasts in a p38 mitogen-activated protein kinase-dependent fashion, and that inhibition of p38 prevented angiotensin II-induced aortic stiffening in vivo. Interleukin 17a also induced collagen 3a1 expression via the activation of p38 mitogen-activated protein kinase. CONCLUSIONS: Our data define a pathway in which inflammation and mechanical stretch lead to vascular inflammation that promotes collagen deposition. The resultant increase in aortic stiffness likely further worsens systolic hypertension and its attendant end-organ damage.


Assuntos
Doenças da Aorta/metabolismo , Hipertensão/metabolismo , Inflamação/metabolismo , Rigidez Vascular/fisiologia , Vasculite/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Transferência Adotiva , Angiotensina II/farmacologia , Animais , Doenças da Aorta/fisiopatologia , Antígenos CD4/genética , Antígenos CD8/genética , Células Cultivadas , Colágeno/metabolismo , Modelos Animais de Doenças , Elastina/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Homeodomínio/genética , Hipertensão/induzido quimicamente , Hipertensão/fisiopatologia , Interleucina-17/genética , Masculino , Camundongos , Camundongos Knockout , Estresse Mecânico , Linfócitos T/metabolismo , Linfócitos T/patologia , Vasculite/fisiopatologia , Vasoconstritores/farmacologia
6.
Circ Res ; 107(2): 263-70, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20558826

RESUMO

RATIONALE: We have previously found that T lymphocytes are essential for development of angiotensin II-induced hypertension; however, the mechanisms responsible for T-cell activation in hypertension remain undefined. OBJECTIVE: We sought to study the roles of the CNS and pressure elevation in T-cell activation and vascular inflammation caused by angiotensin II. METHODS AND RESULTS: To prevent the central actions of angiotensin II, we created anteroventral third cerebral ventricle (AV3V) lesions in mice. The elevation in blood pressure in response to angiotensin II was virtually eliminated by AV3V lesions, as was activation of circulating T cells and the vascular infiltration of leukocytes. In contrast, AV3V lesioning did not prevent the hypertension and T-cell activation caused by the peripheral acting agonist norepinephrine. To determine whether T-cell activation and vascular inflammation are attributable to central influences or are mediated by blood pressure elevation, we administered hydralazine (250 mg/L) in the drinking water. Hydralazine prevented the hypertension and abrogated the increase in circulating activated T cells and vascular infiltration of leukocytes caused by angiotensin II. CONCLUSIONS: We conclude that the central and pressor effects of angiotensin II are critical for T-cell activation and development of vascular inflammation. These findings also support a feed-forward mechanism in which modest degrees of blood pressure elevation lead to T-cell activation, which in turn promotes inflammation and further raises blood pressure, leading to severe hypertension.


Assuntos
Hipertensão/imunologia , Ativação Linfocitária , Linfócitos T/imunologia , Terceiro Ventrículo/imunologia , Vasculite/imunologia , Administração Oral , Transferência Adotiva , Angiotensina II , Animais , Anti-Hipertensivos/administração & dosagem , Pressão Sanguínea , Modelos Animais de Doenças , Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Hidralazina/administração & dosagem , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hipertensão/prevenção & controle , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Norepinefrina , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Superóxidos/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/transplante , Terceiro Ventrículo/lesões , Terceiro Ventrículo/fisiopatologia , Fatores de Tempo , Vasculite/induzido quimicamente , Vasculite/genética , Vasculite/metabolismo , Vasculite/fisiopatologia , Vasculite/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...