Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 325(5): H1133-H1143, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37682237

RESUMO

Children with beta-thalassemia (BT) present with an increase in carotid intima-medial thickness, an early sign suggestive of premature atherosclerosis. However, it is unknown if there is a direct relationship between BT and atherosclerotic disease. To evaluate this, wild-type (WT, littermates) and BT (Hbbth3/+) mice, both male and female, were placed on a 3-mo high-fat diet with low-density lipoprotein receptor suppression via overexpression of proprotein convertase subtilisin/kexin type 9 (PCSK9) gain-of-function mutation (D377Y). Mechanistically, we hypothesize that heme-mediated oxidative stress creates a proatherogenic environment in BT because BT is a hemolytic anemia that has increased free heme and exhausted hemopexin, heme's endogenous scavenger, in the vasculature. We evaluated the effect of hemopexin (HPX) therapy, mediated via an adeno-associated virus, to the progression of atherosclerosis in BT and a phenylhydrazine-induced model of intravascular hemolysis. In addition, we evaluated the effect of deferiprone (DFP)-mediated iron chelation in the progression of atherosclerosis in BT mice. Aortic en face and aortic root lesion area analysis revealed elevated plaque accumulation in both male and female BT mice compared with WT mice. Hemopexin therapy was able to decrease plaque accumulation in both BT mice and mice on our phenylhydrazine (PHZ)-induced model of hemolysis. DFP decreased atherosclerosis in BT mice but did not provide an additive benefit to HPX therapy. Our data demonstrate for the first time that the underlying pathophysiology of BT leads to accelerated atherosclerosis and shows that heme contributes to atherosclerotic plaque development in BT.NEW & NOTEWORTHY This work definitively shows for the first time that beta-thalassemia leads to accelerated atherosclerosis. We demonstrated that intravascular hemolysis is a prominent feature in beta-thalassemia and the resulting increases in free heme are mechanistically relevant. Adeno-associated virus (AAV)-hemopexin therapy led to decreased free heme and atherosclerotic plaque area in both beta-thalassemia and phenylhydrazine-treated mice. Deferiprone-mediated iron chelation led to deceased plaque accumulation in beta-thalassemia mice but provided no additive benefit to hemopexin therapy.


Assuntos
Doenças da Aorta , Aterosclerose , Placa Aterosclerótica , Talassemia beta , Humanos , Criança , Masculino , Feminino , Camundongos , Animais , Pró-Proteína Convertase 9/genética , Talassemia beta/complicações , Talassemia beta/genética , Hemopexina , Deferiprona , Hemólise , Doenças da Aorta/genética , Doenças da Aorta/patologia , Camundongos Knockout , Aterosclerose/genética , Aterosclerose/patologia , Heme , Fenil-Hidrazinas , Quelantes de Ferro , Camundongos Endogâmicos C57BL
2.
PLoS One ; 11(1): e0144561, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26727002

RESUMO

Much attention has been directed to the physiological effects of nitric oxide (NO)-cGMP signaling, but virtually nothing is known about its hematologic effects. We reported for the first time that cGMP signaling induces human γ-globin gene expression. Aiming at developing novel therapeutics for anemia, we examined here the hematologic effects of NO-cGMP signaling in vivo and in vitro. We treated wild-type mice with NO to activate soluble guanylate cyclase (sGC), a key enzyme of cGMP signaling. Compared to untreated mice, NO-treated mice had higher red blood cell counts and total hemoglobin but reduced leukocyte counts, demonstrating that when activated, NO-cGMP signaling exerts hematopoietic effects on multiple types of blood cells in vivo. We next generated mice which overexpressed rat sGC in erythroid and myeloid cells. The forced expression of sGCs activated cGMP signaling in both lineage cells. Compared with non-transgenic littermates, sGC mice exhibited hematologic changes similar to those of NO-treated mice. Consistently, a membrane-permeable cGMP enhanced the differentiation of hematopoietic progenitors toward erythroid-lineage cells but inhibited them toward myeloid-lineage cells by controlling multiple lineage-specific transcription factors. Human γ-globin gene expression was induced at low but appreciable levels in sGC mice carrying the human ß-globin locus. Together, these results demonstrate that NO-cGMP signaling is capable of stimulating erythropoiesis in both in vitro and vivo settings by controlling the expression of multiple lineage-specific transcription factors, suggesting that cGMP signaling upregulates erythropoiesis at the level of gene transcription. The NO-cGMP signaling axis may constitute a novel target to stimulate erythropoiesis in vivo.


Assuntos
GMP Cíclico/fisiologia , Eritropoese/fisiologia , Óxido Nítrico/farmacologia , Sistemas do Segundo Mensageiro/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica , Animais , Células da Medula Óssea/metabolismo , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Linhagem da Célula , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Contagem de Eritrócitos , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Eritropoese/genética , Feminino , Guanilato Ciclase/genética , Guanilato Ciclase/fisiologia , Hemoglobinas/análise , Humanos , Contagem de Leucócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Óxido Nítrico/administração & dosagem , Óxido Nítrico/fisiologia , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Globinas beta/biossíntese , Globinas beta/genética , gama-Globinas/biossíntese , gama-Globinas/genética
3.
J Lipid Res ; 56(3): 599-611, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25598081

RESUMO

Retinal hyperpermeability and subsequent macular edema is a cardinal feature of early diabetic retinopathy (DR). Here, we investigated the role of bioactive lipid metabolites, in particular 12/15-lipoxygenase (LOX)-derived metabolites, in this process. LC/MS lipidomic screen of human retinal endothelial cells (HRECs) demonstrated that 15-HETE was the only significantly increased metabolite (2.4 ± 0.4-fold, P = 0.0004) by high glucose (30 mM) treatment. In the presence of arachidonic acid, additional eicosanoids generated by 12/15-LOX, including 12- and 11-HETEs, were significantly increased. Fluorescein angiography and retinal albumin leakage showed a significant decrease in retinal hyperpermeability in streptozotocin-induced diabetic mice lacking 12/15-LOX compared with diabetic WT mice. Our previous studies demonstrated the potential role of NADPH oxidase in mediating the permeability effect of 12- and 15-HETEs, therefore we tested the impact of intraocular injection of 12-HETE in mice lacking the catalytic subunit of NADPH oxidase (NOX2). The permeability effect of 12-HETE was significantly reduced in NOX2(-/-) mice compared with the WT mice. In vitro experiments also showed that 15-HETE induced HREC migration and tube formation in a NOX-dependent manner. Taken together our data suggest that 12/15-LOX is implicated in DR via a NOX-dependent mechanism.


Assuntos
Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo , Retinopatia Diabética/tratamento farmacológico , Ácidos Hidroxieicosatetraenoicos/farmacologia , Hiperglicemia/tratamento farmacológico , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Animais , Araquidonato 12-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/genética , Retinopatia Diabética/enzimologia , Retinopatia Diabética/genética , Retinopatia Diabética/patologia , Humanos , Hiperglicemia/enzimologia , Hiperglicemia/genética , Hiperglicemia/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/genética
4.
Am J Physiol Cell Physiol ; 307(7): C648-56, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25099734

RESUMO

The low-voltage-activated T-type Ca(2+) channels play an important role in mediating the cellular responses to altered oxygen tension. Among three T-type channel isoforms, α1G, α1H, and α1I, only α1H was found to be upregulated under hypoxia. However, mechanisms underlying such hypoxia-dependent isoform-specific gene regulation remain incompletely understood. We, therefore, studied the hypoxia-dependent transcriptional regulation of α1G and α1H gene promoters with the aim to identify the functional hypoxia-response elements (HREs). In rat pulmonary artery smooth muscle cells (PASMCs) and pheochromocytoma (PC12) cells after hypoxia (3% O2) exposure, we observed a prominent increase in α1H mRNA at 12 h along with a significant rise in α1H-mediated T-type current at 24 and 48 h. We then cloned two promoter fragments from the 5'-flanking regions of rat α1G and α1H gene, 2,000 and 3,076 bp, respectively, and inserted these fragments into a luciferase reporter vector. Transient transfection of PASMCs and PC12 cells with these recombinant constructs and subsequent luciferase assay revealed a significant increase in luciferase activity from the reporter containing the α1H, but not α1G, promoter fragment under hypoxia. Using serial deletion and point mutation analysis strategies, we identified a functional HRE at site -1,173cacgc-1,169 within the α1H promoter region. Furthermore, an electrophoretic mobility shift assay using this site as a DNA probe demonstrated an increased binding activity to nuclear protein extracts from the cells after hypoxia exposure. Taken together, these findings indicate that hypoxia-induced α1H upregulation involves binding of hypoxia-inducible factor to an HRE within the α1H promoter region.


Assuntos
Canais de Cálcio Tipo T/genética , Transcrição Gênica , Animais , Sítios de Ligação , Canais de Cálcio Tipo T/metabolismo , Hipóxia Celular , Potenciais da Membrana , Músculo Liso Vascular/metabolismo , Mutação , Miócitos de Músculo Liso/metabolismo , Células PC12 , Artéria Pulmonar/metabolismo , Veias Pulmonares/metabolismo , RNA Mensageiro/metabolismo , Ratos , Elementos de Resposta , Fatores de Tempo , Transfecção , Regulação para Cima
5.
Biochim Biophys Acta ; 1823(10): 1666-75, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22766303

RESUMO

The transcriptional activator ß-catenin is a key mediator of the canonical Wnt signaling pathway. ß-catenin itself does not bind DNA but functions via interaction with T-cell factor (TCF)/lymphoid-enhancing factor (LEF) transcription factors. Thus, in the case of active Wnt signaling, ß-catenin, in cooperation with TCF/LEF proteins family, activates the expression of a wide variety of genes. To date, the list of established ß-catenin interacting targets is far from complete. In this study, we aimed to establish the interaction between ß-catenin and transcription factors that might affect TCF activity. We took advantage of EMSA, using TCF as a probe, to screen oligonucleotides known to bind specific transcription factors that might dislodge or antagonize ß-catenin/TCF binding. We found that Sox9 and KLF4 antagonize ß-catenin/TCF binding in HEK293, A549, SW480, and T47D cells. This inhibition of TCF binding was concentration-dependent and correlated to the in vitro TCF-luciferase functional assays. Overexpression of Sox9 and KLF4 transcription factors in cancer cells shows a concentration-dependent reduction of TCF-luciferase as well as the TCF-binding activities. In addition, we demonstrated that both Sox9 and KLF4 interact with ß-catenin in an immunoprecipitation assay and reduce its binding to TCF4. Together, these results demonstrate that Sox9 and KLF4 transcription factors antagonize ß-catenin/TCF in cancer cells.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição/metabolismo , beta Catenina/antagonistas & inibidores , Ligação Competitiva/efeitos dos fármacos , Linhagem Celular Tumoral , Células HEK293 , Humanos , Fator 4 Semelhante a Kruppel , Luciferases/metabolismo , Oligonucleotídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Fator de Transcrição 4 , beta Catenina/metabolismo
6.
Front Biosci ; 11: 356-67, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16146737

RESUMO

This basic science review examines the role of cGMP and cGMP-dependent protein kinase (PKG) in the regulation of vascular smooth muscle cell (VSMC) phenotype. The first such studies suggested a role for nitric oxide (NO) and atrial natriuretic peptides (ANP), and the downstream second messenger cGMP, in the inhibition of VSMC proliferation. Subsequently, many laboratories confirmed the anti-proliferative effects of the cGMP pathway in cultured cells and the anti-atherosclerotic effects of the pathway in in vivo animal models. Other studies suggested that the cGMP target, PKG, mediated the anti-proliferative effects of cGMP although other laboratories have not consistently observed these effects. On the other hand, PKG mediates cGMP-dependent increases in smooth muscle-specific gene expression, and in vivo studies suggest that PKG expression itself reduces vascular lesions. The mechanisms by which PKG regulates gene expression are addressed, but it still unknown how the cGMP-PKG pathway is involved in smooth muscle-specific gene expression and phenotype.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/química , GMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação da Expressão Gênica , Miócitos de Músculo Liso/metabolismo , Actinas/química , Animais , Aorta/metabolismo , Fator Natriurético Atrial/química , Western Blotting , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação a Calmodulina/química , Proliferação de Células , Colágeno/química , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Endotélio Vascular/citologia , Humanos , Integrinas/metabolismo , Proteínas dos Microfilamentos/química , Modelos Biológicos , Músculo Liso/metabolismo , Músculo Liso Vascular/citologia , Miosinas/metabolismo , Óxido Nítrico/química , Fenótipo , Plasminogênio/química , Conformação Proteica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores de Angiotensina/metabolismo , Transdução de Sinais , Calponinas
7.
Am J Physiol Cell Physiol ; 287(1): C88-96, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-14985234

RESUMO

NO and cGMP have antigrowth and anti-inflammatory effects on the vessel wall in response to injury. It is well established that after vascular injury proinflammatory cytokines are involved in vascular wall remodeling. The purpose of this study was to ascertain the signaling mechanisms involved in cGMP-dependent protein kinase (PKG) suppression by inflammatory cytokines in primary bovine aortic vascular smooth muscle cells (VSMC). Interleukin (IL)-Ibeta, tumor necrosis factor (TNF)-alpha, and LPS decreased the mRNA and protein levels of PKG in VSMC. IL-Ibeta, TNF-alpha, and LPS increased inducible nitric oxide synthase (iNOS) expression and cGMP production. Treatment of cells with selective inhibitors of iNOS or soluble guanylate cyclase (sGC) reversed the downregulation of PKG expression induced by cytokines and LPS. The NO donor (Z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (DETA NONOate) and 3-(5-hydroxymethyl-2-furyl)-1-benzylindazole (YC-1), a NO-independent sGC activator, decreased PKG mRNA and protein expression in bovine aortic VSMC. Cyclic nucleotide analogs [8-(4-chlorophenylthio)guanosine 3',5'-cyclic monophosphate (CPT-cGMP) and 8-(4-chlorophenylthio)adenosine 3,5'-cyclic monophosphate (CPT-cAMP)] also suppressed PKG mRNA and protein expression. However, CPT-cAMP was more effective than CPT-cGMP in decreasing PKG mRNA levels. Selective inhibition of PKA with the Rp isomer of 8-(4-chlorophenylthio)adenosine 3',5'-cyclic monophosphorothioate (Rp-8p-CPT cAMPS) prevented the downregulation of PKG by LPS. In contrast, the Rp isomer of 8-(4-chlorophenylthio)guanosine 3,5'-cyclic monophosphorothioate (Rp-8p-CPT cGMPS; inhibitor of PKG) had no effect on LPS-induced inhibition of PKG mRNA and protein expression. These studies suggest that cross-activation of PKA in response to iNOS expression by inflammatory mediators downregulates PKG expression in bovine aortic VSMC.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Interleucina-1/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Aorta , Bovinos , Células Cultivadas , Regulação para Baixo , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Indazóis/farmacologia , Interleucina-1/farmacologia , Lipopolissacarídeos/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Nucleotídeos Cíclicos/farmacologia , Oxidiazóis/farmacologia , Quinoxalinas/farmacologia , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
8.
Circ Res ; 90(4): 405-12, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11884369

RESUMO

cGMP-dependent protein kinase (PKG) expression is highly variable and decreases in cultured vascular smooth muscle cells (VSMCs), exposure of cells to nitric oxide (NO), or in response to balloon catheter injury in vivo. In this study, the mechanisms of human type I PKG-alpha (PKG-Ialpha) gene expression were examined. Three structurally unrelated NO donors decreased PKG-Ialpha promoter activity after transfection of a promoter/luciferase construct in VSMCs. Promoter deletion analysis demonstrated that (1) a 120-bp promoter containing tandem Sp1 sites was sufficient to drive basal PKG-Ialpha promoter activity, and (2) NO was inhibitory at this site. Cyclic nucleotide analogues also suppressed PKG-Ialpha promoter activity with cAMP being more potent than cGMP. The effects of cyclic nucleotides to suppress PKG-Ialpha promoter activity were attenuated by a specific cAMP-dependent protein kinase (PKA) inhibitor. Single or double mutation of Sp1 binding sites abolished PKG-Ialpha expression. Moreover, Sp1 binding activity on the PKG-Ialpha promoter was detected in A7r5 cells, and this binding was inhibited by NO and cyclic nucleotides. These results indicate that PKG-Ialpha gene expression is driven by an Sp1 transcription mechanism, and that NO and cAMP inhibit Sp1-mediated PKG-Ialpha gene expression through separate mechanisms.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Músculo Liso Vascular/metabolismo , Óxido Nítrico/farmacologia , Nucleotídeos Cíclicos/farmacologia , Fator de Transcrição Sp1/metabolismo , Animais , Sítios de Ligação/fisiologia , Bovinos , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I , Proteínas Quinases Dependentes de GMP Cíclico/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Genes Reporter , Guanilato Ciclase , Humanos , Lipopolissacarídeos/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Óxido Nítrico/biossíntese , Doadores de Óxido Nítrico/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Guanilil Ciclase Solúvel , Fator de Transcrição Sp1/antagonistas & inibidores , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA