Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Genomics ; 56(3): 265-275, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38145289

RESUMO

Agouti-related peptide (AgRP/Agrp) within the hypothalamic arcuate nucleus (ARC) contributes to the control of energy balance, and dysregulated Agrp may contribute to metabolic adaptation during prolonged obesity. In mice, three isoforms of Agrp are encoded via distinct first exons. Agrp-A (ENSMUST00000005849.11) contributed 95% of total Agrp in mouse ARC, whereas Agrp-B (ENSMUST00000194654.2) dominated in placenta (73%). Conditional deletion of Klf4 from Agrp-expressing cells (Klf4Agrp-KO mice) reduced Agrp mRNA and increased energy expenditure but had no effects on food intake or the relative abundance of Agrp isoforms in the ARC. Chronic high-fat diet feeding masked these effects of Klf4 deletion, highlighting the context-dependent contribution of KLF4 to Agrp control. In the GT1-7 mouse hypothalamic cell culture model, which expresses all three isoforms of Agrp (including Agrp-C, ENSMUST00000194091.6), inhibition of extracellular signal-regulated kinase (ERK) simultaneously increased KLF4 binding to the Agrp promoter and stimulated Agrp expression. In addition, siRNA-mediated knockdown of Klf4 reduced expression of Agrp. We conclude that the expression of individual isoforms of Agrp in the mouse is dependent upon cell type and that KLF4 directly promotes the transcription of Agrp via a mechanism that is superseded during obesity.NEW & NOTEWORTHY In mice, three distinct isoforms of Agouti-related peptide are encoded via distinct first exons. In the arcuate nucleus of the hypothalamus, Krüppel-like factor 4 stimulates transcription of the dominant isoform in lean mice, but this mechanism is altered during diet-induced obesity.


Assuntos
Proteína Relacionada com Agouti , Fator 4 Semelhante a Kruppel , Neurônios , Animais , Camundongos , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Obesidade/genética , Obesidade/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
2.
Int J Mol Sci ; 24(1)2022 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-36613910

RESUMO

Exosomes have attracted attention due to their ability to promote intercellular communication leading to enhanced cell recruitment, lineage-specific differentiation, and tissue regeneration. The object of this study was to determine the effect of exosomes on cell homing and angiogenic differentiation for pulp regeneration. Exosomes (DPSC-Exos) were isolated from rabbit dental pulp stem cells cultured under a growth (Exo-G) or angiogenic differentiation (Exo-A) condition. The characterization of exosomes was confirmed by nanoparticle tracking analysis and an antibody array. DPSC-Exos significantly promoted cell proliferation and migration when treated with 5 × 108/mL exosomes. In gene expression analysis, DPSC-Exos enhanced the expression of angiogenic markers including vascular endothelial growth factor A (VEGFA), Fms-related tyrosine kinase 1 (FLT1), and platelet and endothelial cell adhesion molecule 1 (PECAM1). Moreover, we identified key exosomal microRNAs in Exo-A for cell homing and angiogenesis. In conclusion, the exosome-based cell homing and angiogenic differentiation strategy has significant therapeutic potential for pulp regeneration.


Assuntos
Exossomos , Fator A de Crescimento do Endotélio Vascular , Animais , Coelhos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Polpa Dentária , Exossomos/metabolismo , Neovascularização Fisiológica/genética , Regeneração , Diferenciação Celular/genética , Proliferação de Células/genética
3.
Arterioscler Thromb Vasc Biol ; 36(3): 466-74, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26769049

RESUMO

OBJECTIVE: Hypercholesterolemia and hypertension are associated with aortic valve stenosis (AVS) in humans. We have examined aortic valve function, structure, and gene expression in hypercholesterolemic/hypertensive mice. APPROACH AND RESULTS: Control, hypertensive, hypercholesterolemic (Apoe(-/-)), and hypercholesterolemic/hypertensive mice were studied. Severe aortic stenosis (echocardiography) occurred only in hypercholesterolemic/hypertensive mice. There was minimal calcification of the aortic valve. Several structural changes were identified at the base of the valve. The intercusp raphe (or seam between leaflets) was longer in hypercholesterolemic/hypertensive mice than in other mice, and collagen fibers at the base of the leaflets were reoriented to form a mesh. In hypercholesterolemic/hypertensive mice, the cusps were asymmetrical, which may contribute to changes that produce AVS. RNA sequencing was used to identify molecular targets during the developmental phase of stenosis. Genes related to the structure of the valve were identified, which differentially expressed before fibrotic AVS developed. Both RNA and protein of a profibrotic molecule, plasminogen activator inhibitor 1, were increased greatly in hypercholesterolemic/hypertensive mice. CONCLUSIONS: Hypercholesterolemic/hypertensive mice are the first model of fibrotic AVS. Hypercholesterolemic/hypertensive mice develop severe AVS in the absence of significant calcification, a feature that resembles AVS in children and some adults. Structural changes at the base of the valve leaflets include lengthening of the raphe, remodeling of collagen, and asymmetry of the leaflets. Genes were identified that may contribute to the development of fibrotic AVS.


Assuntos
Estenose da Valva Aórtica/etiologia , Valva Aórtica/patologia , Hipercolesterolemia/complicações , Hipertensão/complicações , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Valva Aórtica/metabolismo , Valva Aórtica/fisiopatologia , Estenose da Valva Aórtica/metabolismo , Estenose da Valva Aórtica/patologia , Estenose da Valva Aórtica/fisiopatologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Modelos Animais de Doenças , Feminino , Fibrose , Regulação da Expressão Gênica , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Hipertensão/genética , Hipertensão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Renina/genética , Renina/metabolismo , Índice de Gravidade de Doença
4.
Arterioscler Thromb Vasc Biol ; 35(4): 838-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25675995

RESUMO

OBJECTIVE: We tested the hypothesis that endothelial peroxisome proliferator-activated receptor-γ protects against vascular thrombosis using a transgenic mouse model expressing a peroxisome proliferator-activated receptor-γ mutant (E-V290M) selectively in endothelium. APPROACH AND RESULTS: The time to occlusive thrombosis of the carotid artery was significantly shortened in E-V290M mice compared with nontransgenic littermates after either chemical injury with ferric chloride (5.1 ± 0.2 versus 10.1 ± 3.3 minutes; P=0.01) or photochemical injury with rose bengal (48 ± 9 versus 74 ± 9 minutes; P=0.04). Gene set enrichment analysis demonstrated the upregulation of NF-κB target genes, including P-selectin, in aortic endothelial cells from E-V290M mice (P<0.001). Plasma P-selectin and carotid artery P-selectin mRNA were elevated in E-V290M mice (P<0.05). P-selectin-dependent leukocyte rolling on mesenteric venules was increased in E-V290M mice compared with nontransgenic mice (53 ± 8 versus 25 ± 7 per minute; P=0.02). The shortened time to arterial occlusion in E-V290M mice was reversed by administration of P-selectin-blocking antibodies or neutrophil-depleting antibodies (P=0.04 and P=0.02, respectively) before photochemical injury. CONCLUSIONS: Endothelial peroxisome proliferator-activated receptor-γ protects against thrombosis through a mechanism that involves downregulation of P-selectin expression and diminished P-selectin-mediated leukocyte-endothelial interactions.


Assuntos
Doenças das Artérias Carótidas/prevenção & controle , Células Endoteliais/metabolismo , Selectina-P/metabolismo , PPAR gama/metabolismo , Trombose/prevenção & controle , Trombose Venosa/prevenção & controle , Animais , Anticorpos/farmacologia , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/imunologia , Doenças das Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/patologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/patologia , Humanos , Migração e Rolagem de Leucócitos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neutrófilos/imunologia , Neutrófilos/metabolismo , Selectina-P/antagonistas & inibidores , Selectina-P/genética , Selectina-P/imunologia , PPAR gama/genética , RNA Mensageiro/metabolismo , Trombose/genética , Trombose/imunologia , Trombose/metabolismo , Trombose/patologia , Fatores de Tempo , Veia Cava Inferior/metabolismo , Veia Cava Inferior/patologia , Trombose Venosa/genética , Trombose Venosa/imunologia , Trombose Venosa/metabolismo , Trombose Venosa/patologia
5.
Circ Res ; 111(11): 1446-58, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22962432

RESUMO

RATIONALE: Activation of peroxisome proliferator-activated receptor-γ (PPARγ) by thiazolidinediones lowers blood pressure, whereas PPARγ mutations cause hypertension. Previous studies suggest these effects may be mediated through the vasculature, but the underlying mechanisms remain unclear. OBJECTIVE: To identify PPARγ mechanisms and transcriptional targets in vascular smooth muscle and their role in regulating resistance artery tone. METHODS AND RESULTS: We studied mesenteric artery (MA) from transgenic mice expressing dominant-negative (DN) mutant PPARγ driven by a smooth muscle cell-specific promoter. MA from transgenic mice exhibited a robust increase in myogenic tone. Patch clamp analysis revealed a reduced large conductance Ca(2+)-activated K(+) (BKCa) current in freshly dissociated smooth muscle cell from transgenic MA. Inhibition of protein kinase C corrected both enhanced myogenic constriction and impaired the large conductance Ca(2+)-activated K(+) channel function. Gene expression profiling revealed a marked loss of the regulator of G protein signaling 5 (RGS5) mRNA in transgenic MA, which was accompanied by a substantial increase in angiotensin II-induced constriction in MA. Small interfering RNA targeting RGS5 caused augmented myogenic tone in intact mesenteric arteries and increased activation of protein kinase C in smooth muscle cell cultures. PPARγ and PPARδ each bind to a PPAR response element close to the RGS5 promoter. RGS5 expression in nontransgenic MA was induced after activation of either PPARγ or PPARδ, an effect that was markedly blunted by DN PPARγ. CONCLUSIONS: We conclude that RGS5 in smooth muscle is a PPARγ and PPARδ target, which when activated blunts angiotensin II-mediated activation of protein kinase C, and preserves the large conductance Ca(2+)-activated K(+) channel activity, thus providing tight control of myogenic tone in the microcirculation.


Assuntos
Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Artérias Mesentéricas/fisiologia , PPAR gama/fisiologia , Proteína Quinase C/metabolismo , Proteínas RGS/metabolismo , Angiotensina II/farmacologia , Animais , Western Blotting , Feminino , Perfilação da Expressão Gênica , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Masculino , Artérias Mesentéricas/citologia , Artérias Mesentéricas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Mutação , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Naftalenos/farmacologia , PPAR gama/genética , PPAR gama/metabolismo , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteínas RGS/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tetraetilamônio/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
6.
Cell Metab ; 7(3): 215-26, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18316027

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma) is a ligand-activated transcription factor that plays a critical role in metabolism. Thiazolidinediones, high-affinity PPARgamma ligands used clinically to treat type II diabetes, have been reported to lower blood pressure and provide other cardiovascular benefits. Some mutations in PPARgamma (PPARG) cause type II diabetes and severe hypertension. Here we tested the hypothesis that PPARgamma in vascular muscle plays a role in the regulation of vascular tone and blood pressure. Transgenic mice expressing dominant-negative mutations in PPARgamma under the control of a smooth-muscle-specific promoter exhibit a loss of responsiveness to nitric oxide and striking alterations in contractility in the aorta, hypertrophy and inward remodeling in the cerebral microcirculation, and systolic hypertension. These results identify PPARgamma as pivotal in vascular muscle as a regulator of vascular structure, vascular function, and blood pressure, potentially explaining some of the cardioprotective effects of thiazolidinediones.


Assuntos
Hipertensão/metabolismo , Músculo Liso Vascular/metabolismo , Mutação , PPAR gama/metabolismo , Vasoconstrição , Vasodilatação , Animais , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatologia , Pressão Sanguínea , Artérias Cerebrais/metabolismo , Artérias Cerebrais/fisiopatologia , Ritmo Circadiano , GMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Feminino , Humanos , Hipertensão/genética , Hipertensão/patologia , Hipertensão/fisiopatologia , Hipertrofia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Cadeias Pesadas de Miosina/genética , Óxido Nítrico/metabolismo , PPAR gama/genética , Regiões Promotoras Genéticas , Ratos , Sístole , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
7.
Am J Physiol Regul Integr Comp Physiol ; 304(9): R690-701, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23447133

RESUMO

Agonists of the nuclear hormone receptor peroxisome proliferator-activated receptor γ (PPARγ) have potent insulin-sensitizing effects and inhibit atherosclerosis progression in patients with Type II diabetes. Conversely, missense mutations in the ligand-binding domain of PPARγ that render the transcription factor dominant negative (DN) cause early-onset hypertension and Type II diabetes. We tested the hypothesis that DN PPARγ-mediated interference of endogenous wild-type PPARγ in the endothelium and vascular smooth muscle exacerbates atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. Endothelium-specific expression of DN PPARγ on the ApoE(-/-) background unmasked significant impairment of endothelium-dependent relaxation in aortic rings, increased systolic blood pressure, altered expression of atherogenic markers (e.g., Cd36, Mcp1, Catalase), and enhanced diet-induced atherosclerotic lesion formation in aorta. Smooth muscle-specific expression of DN PPARγ, which induces aortic dysfunction and increased systolic blood pressure at baseline, also resulted in enhanced diet-induced atherosclerotic lesion formation in aorta on the ApoE(-/-) background that was associated with altered expression of a shared, yet distinct, set of atherogenic markers (e.g., Cd36, Mcp1, Osteopontin, Vcam1). In particular, induction of Osteopontin expression by smooth muscle-specific DN PPARγ correlated with increased plaque calcification. These data demonstrate that inhibition of PPARγ function specifically in the vascular endothelium or smooth muscle may contribute to cardiovascular disease.


Assuntos
Aterosclerose/genética , Aterosclerose/fisiopatologia , Endotélio Vascular/fisiopatologia , Hipertensão/genética , Hipertensão/fisiopatologia , Músculo Liso Vascular/fisiopatologia , PPAR gama/genética , PPAR gama/fisiologia , Doenças Vasculares/genética , Doenças Vasculares/fisiopatologia , Acetilcolina/farmacologia , Animais , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Apolipoproteínas E/genética , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Dieta , Progressão da Doença , Genes Dominantes/genética , Genes Dominantes/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miografia , Nitroprussiato/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Vasodilatadores/farmacologia
8.
Sci Immunol ; 8(83): eabq6352, 2023 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-37146132

RESUMO

Asthma is a chronic disease most commonly associated with allergy and type 2 inflammation. However, the mechanisms that link airway inflammation to the structural changes that define asthma are incompletely understood. Using a human model of allergen-induced asthma exacerbation, we compared the lower airway mucosa in allergic asthmatics and allergic non-asthmatic controls using single-cell RNA sequencing. In response to allergen, the asthmatic airway epithelium was highly dynamic and up-regulated genes involved in matrix degradation, mucus metaplasia, and glycolysis while failing to induce injury-repair and antioxidant pathways observed in controls. IL9-expressing pathogenic TH2 cells were specific to asthmatic airways and were only observed after allergen challenge. Additionally, conventional type 2 dendritic cells (DC2 that express CD1C) and CCR2-expressing monocyte-derived cells (MCs) were uniquely enriched in asthmatics after allergen, with up-regulation of genes that sustain type 2 inflammation and promote pathologic airway remodeling. In contrast, allergic controls were enriched for macrophage-like MCs that up-regulated tissue repair programs after allergen challenge, suggesting that these populations may protect against asthmatic airway remodeling. Cellular interaction analyses revealed a TH2-mononuclear phagocyte-basal cell interactome unique to asthmatics. These pathogenic cellular circuits were characterized by type 2 programming of immune and structural cells and additional pathways that may sustain and amplify type 2 signals, including TNF family signaling, altered cellular metabolism, failure to engage antioxidant responses, and loss of growth factor signaling. Our findings therefore suggest that pathogenic effector circuits and the absence of proresolution programs drive structural airway disease in response to type 2 inflammation.


Assuntos
Asma , Hipersensibilidade , Humanos , Antioxidantes , Asma/genética , Alérgenos , Inflamação
9.
Microbiol Resour Announc ; 11(7): e0015722, 2022 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-35861552

RESUMO

Trichomonas tenax is a flagellated parasite that plays an important role in periodontal disease, with high prevalence worldwide. Its pathogenesis remains largely unknown, and there is very little information on its genome. Here, we present the whole-genome shotgun sequence of T. tenax strain Hs-4:NIH.

10.
Nat Commun ; 13(1): 1477, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35304452

RESUMO

The epigenetic patterns that are established during early thymic development might determine mature T cell physiology and function, but the molecular basis and topography of the genetic elements involved are not fully known. Here we show, using the Cd4 locus as a paradigm for early developmental programming, that DNA demethylation during thymic development licenses a novel stimulus-responsive element that is critical for the maintenance of Cd4 gene expression in effector T cells. We document the importance of maintaining high CD4 expression during parasitic infection and show that by driving transcription, this stimulus-responsive element allows for the maintenance of histone H3K4me3 levels during T cell replication, which is critical for preventing de novo DNA methylation at the Cd4 promoter. A failure to undergo epigenetic programming during development leads to gene silencing during effector T cell replication. Our study thus provides evidence of early developmental events shaping the functional fitness of mature effector T cells.


Assuntos
Desmetilação do DNA , Metilação de DNA , Linfócitos T CD4-Positivos/metabolismo , Regiões Promotoras Genéticas/genética
11.
J Biol Chem ; 285(13): 9311-9316, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20129921

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPAR gamma) is a ligand-activated transcription factor of the nuclear hormone receptor superfamily. Increasing evidence suggests that PPAR gamma is involved in the regulation of vascular function and blood pressure in addition to its well recognized role in metabolism. Thiazolidinediones, PPAR gamma agonists, lower blood pressure and have protective vascular effects through largely unknown mechanisms. In contrast, loss-of-function dominant-negative mutations in human PPAR gamma cause insulin resistance and severe early onset hypertension. Recent studies using genetically manipulated mouse models have begun to specifically address the importance of PPAR gamma in the vasculature. In this minireview, evidence for a protective role of PPAR gamma in the endothelium and vascular smooth muscle, derived largely from studies of genetically manipulated mice, will be discussed.


Assuntos
Endotélio Vascular/metabolismo , Hipertensão/patologia , Músculo Liso Vascular/metabolismo , PPAR gama/metabolismo , Animais , Pressão Sanguínea , Genes Dominantes , Humanos , Hipertensão/metabolismo , Resistência à Insulina , Ligantes , Camundongos , Modelos Biológicos , Mutação , Ratos , Tiazolidinedionas/farmacologia
12.
Am J Physiol Regul Integr Comp Physiol ; 301(4): R1067-77, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21753145

RESUMO

All components of the renin angiotensin system necessary for ANG II generation and action have been reported to be present in renal proximal convoluted tubules. Given the close relationship between renal sodium handling and blood pressure regulation, we hypothesized that modulating the action of ANG II specifically in the renal proximal tubules would alter the chronic level of blood pressure. To test this, we used a proximal tubule-specific, androgen-dependent, promoter construct (KAP2) to generate mice with either overexpression of a constitutively active angiotensin type 1A receptor transgene or depletion of endogenous angiotensin type 1A receptors. Androgen administration to female transgenic mice caused a robust induction of the transgene in the kidney and increased baseline blood pressure. In the receptor-depleted mice, androgen administration to females resulted in a Cre recombinase-mediated deletion of angiotensin type 1A receptors in the proximal tubule and reduced blood pressure. In contrast to the changes observed at baseline, there was no difference in the blood pressure response to a pressor dose of ANG II in either experimental model. These data, from two separate mouse models, provide evidence that ANG II signaling via the type 1A receptor in the renal proximal tubule is a regulator of systemic blood pressure under baseline conditions.


Assuntos
Pressão Sanguínea/fisiologia , Túbulos Renais Proximais/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Androgênios/farmacologia , Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Feminino , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
13.
Arterioscler Thromb Vasc Biol ; 30(3): 518-25, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20018933

RESUMO

OBJECTIVE: Drugs that activate peroxisome proliferator-activated receptor (PPAR) gamma improve glucose sensitivity and lower blood pressure, whereas dominant-negative mutations in PPARgamma cause severe insulin resistance and hypertension. We hypothesize that these PPARgamma mutants regulate target genes opposite to those of ligand-mediated activation, and we tested this hypothesis on a genomewide scale. METHODS AND RESULTS: We integrated gene expression data in aorta specimens from mice treated with the PPARgamma ligand rosiglitazone with data from mice containing a globally expressed knockin of the PPARgamma P465L dominant-negative mutation. We also integrated our data with publicly available data sets containing the following: (1) gene expression profiles in many human tissues, (2) PPARgamma target genes in 3T3-L1 adipocytes, and (3) experimentally validated PPARgamma binding sites throughout the genome. Many classic PPARgamma target genes were induced by rosiglitazone and repressed by dominant-negative PPARgamma. A similar pattern was observed for about 90% of the gene sets regulated by both rosiglitazone and dominant-negative PPARgamma. Genes exhibiting this pattern of contrasting regulation were significantly enriched for nearby PPARgamma binding sites. CONCLUSIONS: These results provide convincing evidence that the PPARgamma P465L mutation causes transcriptional effects that are opposite to those mediated by PPARgamma ligand, thus validating mice carrying the mutation as a model of PPARgamma interference.


Assuntos
Aorta Torácica/metabolismo , Biologia Computacional , Perfilação da Expressão Gênica , PPAR gama/metabolismo , Animais , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Mutação , PPAR gama/genética , Rosiglitazona , Transdução de Sinais/fisiologia , Tiazolidinedionas/farmacologia , Regulação para Cima/efeitos dos fármacos
14.
Circ Res ; 103(6): 654-61, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18676352

RESUMO

The ligand-activated transcription factor peroxisome proliferator activated receptor gamma (PPARgamma) is expressed in vascular endothelium where it exerts anti-inflammatory and antioxidant effects. However, its role in regulating vascular function remains undefined. We examined endothelial function in transgenic mice expressing dominant-negative mutants of PPARgamma under the control of an endothelial-specific promoter to test the hypothesis that endothelial PPARgamma plays a protective role in the vasculature. Under baseline conditions, responses to the endothelium-dependent agonist acetylcholine were not affected in either aorta or the basilar artery in vitro. In response to feeding a high-fat diet for 12 weeks, acetylcholine produced dilation that was markedly impaired in the basilar artery of mice expressing dominant-negative mutants, but not in mice expressing wild-type PPARgamma controlled by the same promoter. Unlike basilar artery, 12 weeks of a high-fat diet was not sufficient to cause endothelial dysfunction in the aorta of mice expressing dominant-negative PPARgamma, although aortic dysfunction became evident after 25 weeks. The responses to acetylcholine in basilar artery were restored to normal after treatment with a scavenger of superoxide. Baseline blood pressure was only slightly elevated in the transgenic mice, but the pressor response to angiotensin II was augmented. Thus, interference with PPARgamma in the endothelium produces endothelial dysfunction in the cerebral circulation through a mechanism involving oxidative stress. Consistent with its role as a fatty acid sensor, these findings provide genetic evidence that endothelial PPARgamma plays a critical role in protecting blood vessels in response to a high-fat diet.


Assuntos
Circulação Cerebrovascular/genética , Transtornos Cerebrovasculares/etiologia , Transtornos Cerebrovasculares/metabolismo , Gorduras na Dieta/administração & dosagem , Endotélio Vascular/fisiopatologia , PPAR gama/fisiologia , Animais , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatologia , Células Cultivadas , Transtornos Cerebrovasculares/genética , Transtornos Cerebrovasculares/fisiopatologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estresse Oxidativo/genética , PPAR gama/genética
15.
JCI Insight ; 52019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31184598

RESUMO

Patients with mutations in Cullin-3 (CUL3) exhibit severe early onset hypertension but the contribution of the smooth muscle remains unclear. Conditional genetic ablation of CUL3 in vascular smooth muscle (S-CUL3KO) causes progressive impairment in responsiveness to nitric oxide (NO), rapid development of severe hypertension, and increased arterial stiffness. Loss of CUL3 in primary aortic smooth muscle cells or aorta resulted in decreased expression of the NO receptor, soluble guanylate cyclase (sGC), causing a marked reduction in cGMP production and impaired vasodilation to cGMP analogues. Vasodilation responses to a selective large conductance Ca2+-activated K+-channel activator were normal suggesting that downstream signals which promote smooth muscle-dependent relaxation remained intact. We conclude that smooth muscle specific CUL3 ablation impairs both cGMP production and cGMP responses and that loss of CUL3 function selectively in smooth muscle is sufficient to cause severe hypertension by interfering with the NO-sGC-cGMP pathway. Our study provides compelling evidence for the sufficiency of vascular smooth muscle CUL3 as a major regulator of BP. CUL3 mutations cause severe vascular dysfunction, arterial stiffness and hypertension due to defects in vascular smooth muscle.


Assuntos
Proteínas Culina/genética , Proteínas Culina/metabolismo , Predisposição Genética para Doença/genética , Hipertensão/genética , Hipertensão/metabolismo , Músculo Liso/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Hipertensão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Mutação , Miócitos de Músculo Liso/metabolismo , Óxido Nítrico , Guanilil Ciclase Solúvel/metabolismo , Transcriptoma , Rigidez Vascular , Vasodilatação
16.
J Clin Invest ; 129(6): 2318-2332, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30896450

RESUMO

Mice selectively expressing PPARγ dominant negative mutation in vascular smooth muscle exhibit RhoBTB1-deficiency and hypertension. Our rationale was to employ genetic complementation to uncover the mechanism of action of RhoBTB1 in vascular smooth muscle. Inducible smooth muscle-specific restoration of RhoBTB1 fully corrected the hypertension and arterial stiffness by improving vasodilator function. Notably, the cardiovascular protection occurred despite preservation of increased agonist-mediated contraction and RhoA/Rho kinase activity, suggesting RhoBTB1 selectively controls vasodilation. RhoBTB1 augmented the cGMP response to nitric oxide by restraining the activity of phosphodiesterase 5 (PDE5) by acting as a substrate adaptor delivering PDE5 to the Cullin-3 E3 Ring ubiquitin ligase complex for ubiquitination inhibiting PDE5. Angiotensin-II infusion also caused RhoBTB1-deficiency and hypertension which was prevented by smooth muscle specific RhoBTB1 restoration. We conclude that RhoBTB1 protected from hypertension, vascular smooth muscle dysfunction, and arterial stiffness in at least two models of hypertension.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Hipertensão/prevenção & controle , Músculo Liso Vascular/metabolismo , Rigidez Vascular , Vasodilatação , Proteínas rho de Ligação ao GTP/metabolismo , Angiotensina II/efeitos adversos , Angiotensina II/farmacologia , Animais , Proteínas Culina/genética , Proteínas Culina/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/genética , Modelos Animais de Doenças , Células HEK293 , Humanos , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/patologia , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Proteínas rho de Ligação ao GTP/genética , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
17.
JCI Insight ; 3(19)2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30282823

RESUMO

Copeptin, a marker of arginine vasopressin (AVP) secretion, is elevated throughout human pregnancies complicated by preeclampsia (PE), and AVP infusion throughout gestation is sufficient to induce the major phenotypes of PE in mice. Thus, we hypothesized a role for AVP in the pathogenesis of PE. AVP infusion into pregnant C57BL/6J mice resulted in hypertension, renal glomerular endotheliosis, intrauterine growth restriction, decreased placental growth factor (PGF), altered placental morphology, placental oxidative stress, and placental gene expression consistent with human PE. Interestingly, these changes occurred despite a lack of placental hypoxia or elevations in placental fms-like tyrosine kinase-1 (FLT1). Coinfusion of AVP receptor antagonists and time-restricted infusion of AVP uncovered a mid-gestational role for the AVPR1A receptor in the observed renal pathologies, versus mid- and late-gestational roles for the AVPR2 receptor in the blood pressure and fetal phenotypes. These findings demonstrate that AVP is sufficient to initiate phenotypes of PE in the absence of placental hypoxia, and indicate that AVP may mechanistically (independently, and possibly synergistically with hypoxia) contribute to the development of clinical signs of PE in specific subtypes of human PE. Additionally, they identify divergent and gestational time-specific signaling mechanisms that mediate the development of PE phenotypes in response to AVP.


Assuntos
Antagonistas dos Receptores de Hormônios Antidiuréticos/administração & dosagem , Neurofisinas/metabolismo , Pré-Eclâmpsia/etiologia , Precursores de Proteínas/metabolismo , Vasopressinas/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Determinação da Pressão Arterial , Hipóxia Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurofisinas/administração & dosagem , Placenta/efeitos dos fármacos , Placenta/patologia , Pletismografia , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/patologia , Gravidez , Precursores de Proteínas/administração & dosagem , Receptores de Vasopressinas/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Vasopressinas/administração & dosagem
18.
Methods Mol Biol ; 1527: 41-52, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28116705

RESUMO

Hypertension is a complex disorder in which multiple genes, pathways, and organ systems simultaneously interact to contribute to the final level of blood pressure. Fully elucidating these interactions is an important area of hypertension research and one in which high-throughput methods such as microarrays can play a key role. With recent advances in microarray technology, reliable and accurate quantification of all known mRNA transcripts in a sample is now routinely performed. In addition, with improved statistical methods and publicly available tools and resources, robust analysis of the large amount of data generated from microarray experiments is now achievable for all research laboratories as will be outlined in this review.


Assuntos
Hipertensão/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Animais , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Biologia Computacional , Humanos , RNA Mensageiro/genética
19.
JCI Insight ; 2(6): e91738, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28352663

RESUMO

Impaired PPARγ activity in endothelial cells causes oxidative stress and endothelial dysfunction which causes a predisposition to hypertension, but the identity of key PPARγ target genes that protect the endothelium remain unclear. Retinol-binding protein 7 (RBP7) is a PPARγ target gene that is essentially endothelium specific. Whereas RBP7-deficient mice exhibit normal endothelial function at baseline, they exhibit severe endothelial dysfunction in response to cardiovascular stressors, including high-fat diet and subpressor angiotensin II. Endothelial dysfunction was not due to differences in weight gain, impaired glucose homeostasis, or hepatosteatosis, but occurred through an oxidative stress-dependent mechanism which can be rescued by scavengers of superoxide. RNA sequencing revealed that RBP7 was required to mediate induction of a subset of PPARγ target genes by rosiglitazone in the endothelium including adiponectin. Adiponectin was selectively induced in the endothelium of control mice by high-fat diet and rosiglitazone, whereas RBP7 deficiency abolished this induction. Adiponectin inhibition caused endothelial dysfunction in control vessels, whereas adiponectin treatment of RBP7-deficient vessels improved endothelium-dependent relaxation and reduced oxidative stress. We conclude that RBP7 is required to mediate the protective effects of PPARγ in the endothelium through adiponectin, and RBP7 is an endothelium-specific PPARγ target and regulator of PPARγ activity.


Assuntos
Adiponectina/metabolismo , Antioxidantes/metabolismo , Endotélio Vascular/metabolismo , PPAR gama/metabolismo , Proteínas Celulares de Ligação ao Retinol/metabolismo , Adiponectina/genética , Animais , Dieta Hiperlipídica , Endotélio Vascular/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , PPAR gama/genética , RNA Mensageiro/genética , Proteínas Celulares de Ligação ao Retinol/genética
20.
Hypertension ; 70(1): 174-182, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28507170

RESUMO

Selective expression of dominant negative (DN) peroxisome proliferator-activated receptor γ (PPARγ) in vascular smooth muscle cells (SMC) results in hypertension, atherosclerosis, and increased nuclear factor-κB (NF-κB) target gene expression. Mesenteric SMC were cultured from mice designed to conditionally express wild-type (WT) or DN-PPARγ in response to Cre recombinase to determine how SMC PPARγ regulates expression of NF-κB target inflammatory genes. SMC-specific overexpression of WT-PPARγ or agonist-induced activation of endogenous PPARγ blunted tumor necrosis factor α (TNF-α)-induced NF-κB target gene expression and activity of an NF-κB-responsive promoter. TNF-α-induced gene expression responses were enhanced by DN-PPARγ in SMC. Although expression of NF-κB p65 was unchanged, nuclear export of p65 was accelerated by WT-PPARγ and prevented by DN-PPARγ in SMC. Leptomycin B, a nuclear export inhibitor, blocked p65 nuclear export and inhibited the anti-inflammatory action of PPARγ. Consistent with a role in facilitating p65 nuclear export, WT-PPARγ coimmunoprecipitated with p65, and WT-PPARγ was also exported from the nucleus after TNF-α treatment. Conversely, DN-PPARγ does not bind to p65 and was retained in the nucleus after TNF-α treatment. Transgenic mice expressing WT-PPARγ or DN-PPARγ specifically in SMC (S-WT or S-DN) were bred with mice expressing luciferase controlled by an NF-κB-responsive promoter to assess effects on NF-κB activity in whole tissue. TNF-α-induced NF-κB activity was decreased in aorta and carotid artery from S-WT but was increased in vessels from S-DN mice. We conclude that SMC PPARγ blunts expression of proinflammatory genes by inhibition of NF-κB activity through a mechanism promoting nuclear export of p65, which is abolished by DN mutation in PPARγ.


Assuntos
Hipertensão , Músculo Liso Vascular , NF-kappa B , PPAR gama/genética , Fator de Transcrição RelA/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Núcleo Celular/metabolismo , Células Cultivadas , Ácidos Graxos Insaturados/farmacologia , Hipertensão/genética , Hipertensão/metabolismo , Inflamação/genética , Inflamação/metabolismo , Camundongos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Mutação , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA