Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(13)2023 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-37446344

RESUMO

Mutations in the LMNA gene (encoding lamin A/C proteins) cause several human cardiac diseases, including dilated cardiomyopathies (LMNA-DCM). The main clinical risks in LMNA-DCM patients are sudden cardiac death and progressive left ventricular ejection fraction deterioration, and therefore most human and animal studies have sought to define the mechanisms through which LMNA mutations provoke cardiac alterations, with a particular focus on cardiomyocytes. To investigate if LMNA mutations also cause vascular alterations that might contribute to the etiopathogenesis of LMNA-DCM, we generated and characterized Lmnaflox/floxSM22αCre mice, which constitutively lack lamin A/C in vascular smooth muscle cells (VSMCs), cardiac fibroblasts, and cardiomyocytes. Like mice with whole body or cardiomyocyte-specific lamin A/C ablation, Lmnaflox/floxSM22αCre mice recapitulated the main hallmarks of human LMNA-DCM, including ventricular systolic dysfunction, cardiac conduction defects, cardiac fibrosis, and premature death. These alterations were associated with elevated expression of total and phosphorylated (active) Smad3 and cleaved (active) caspase 3 in the heart. Lmnaflox/floxSM22αCre mice also exhibited perivascular fibrosis in the coronary arteries and a switch of aortic VSMCs from the 'contractile' to the 'synthetic' phenotype. Ex vivo wire myography in isolated aortic rings revealed impaired maximum contraction capacity and an altered response to vasoconstrictor and vasodilator agents in Lmnaflox/floxSM22αCre mice. To our knowledge, our results provide the first evidence of phenotypic alterations in VSMCs that might contribute significantly to the pathophysiology of some forms of LMNA-DCM. Future work addressing the mechanisms underlying vascular defects in LMNA-DCM may open new therapeutic avenues for these diseases.


Assuntos
Cardiomiopatia Dilatada , Miócitos Cardíacos , Humanos , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Músculo Liso Vascular/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Volume Sistólico , Função Ventricular Esquerda , Cardiomiopatia Dilatada/patologia , Mutação
2.
Front Cell Dev Biol ; 11: 1128594, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37025175

RESUMO

Population aging and age-related cardiovascular disease (CVD) are becoming increasingly prevalent worldwide, generating a huge medical and socioeconomic burden. The complex regulation of aging and CVD and the interaction between these processes are crucially dependent on cellular stress responses. Interferon-stimulated gene-15 (ISG15) encodes a ubiquitin-like protein expressed in many vertebrate cell types that can be found both free and conjugated to lysine residues of target proteins via a post-translational process termed ISGylation. Deconjugation of ISG15 (deISGylation) is catalyzed by the ubiquitin-specific peptidase 18 (USP18). The ISG15 pathway has mostly been studied in the context of viral and bacterial infections and in cancer. This minireview summarizes current knowledge on the role of ISG15 in age-related telomere shortening, genomic instability, and DNA damage accumulation, as well as in hypertension, diabetes, and obesity, major CVD risk factors prevalent in the elderly population.

3.
An. R. Acad. Nac. Farm. (Internet) ; 88(número extraordinario): 149-185, diciembre 2022. ilus
Artigo em Espanhol | IBECS | ID: ibc-225696

RESUMO

La hipertensión y la obesidad son importantes problemas de salud en todo el mundo con notables consecuencias sobre la morbilidad y la mortalidad. De hecho, tanto la hipertensión como la obesidad son importantes factores de riesgo para el desarrollo de enfermedades cardiovasculares. La disfunción endotelial, el remodelado vascular y las alteraciones en la mecánica vascular son aspectos comunes del daño vascular en la hipertensión, la obesidad y los aneurismas. Durante las últimas décadas, se ha demostrado la importancia de la inflamación de bajo grado en el daño vascular asociado a las enfermedades cardiovasculares. Dicha inflamación se caracteriza por la acumulación de células inflamatorias en la vasculatura, así como por el aumento de citoquinas proinflamatorias locales y circulantes. Por tanto, la identificación de nuevos mediadores inflamatorios implicados en dicho daño se ha convertido en un área de investigación muy importante.El interferón-γ (IFNγ) o el factor de necrosis tumoral-α (TNFα) son importantes citoquinas implicadas en el daño vascular asociado a la hipertensión. Además, se acepta que el TNFα es un mediador clave implicado en la resistencia a la insulina y el daño vascular observado en la obesidad. Ambas citoquinas inducen la expresión del gen 15 estimulado por interferón (ISG15), una proteína similar a la ubiquitina que induce una modificación postraducional reversible (ISGilación) y que también puede secretarse como forma libre. Las funciones de ISG15 están principalmente relacionadas con la respuesta inmune frente a infecciones, sin embargo, podría ser también una interesante nueva diana del daño cardiovascular. (AU)


Hypertension and obesity are major health problems worldwide with significant consequences on morbidity and mortality. In fact, both hypertension and obesity are important risk factors for the development of cardiovascular diseases. Endothelial dysfunction, vascular remodeling, and alterations in vascular mechanics are common aspects of vascular damage in hypertension, obesity, and aneurysms. During the last decades, the importance of low-grade inflammation in vascular damage associated with cardiovascular diseases has been demonstrated. This inflammation is characterized by the accumulation of inflammatory cells in the vasculature, as well as by the increase of local and circulating pro-inflammatory cytokines. Therefore, the identification of new inflammatory mediators involved in this damage has become a very important area of research.Interferón-γ (IFNγ) or tumor necrosis tumoral-α (TNFα) are important cytokines involved in the vascular damage associated with hypertension. Furthermore, it is accepted that TNFα is a key mediator involved in insulin resistance and vascular damage observed in obesity. Both cytokines induce the expression of interferon-stimulated gene 15 (ISG15), a protein similar to ubiquitin that induces a reversible post-translational modification (ISGylation) and that can also be secreted as a free form. The functions of ISG15 are mainly related to the immune response against infections, however, it could also be an interesting new target for cardiovascular damage. (AU)


Assuntos
Humanos , Hipertensão , Obesidade , Estresse Oxidativo , Inflamação , Doenças Cardiovasculares , Mortalidade
4.
An. R. Acad. Nac. Farm. (Internet) ; 88(2): 149-185, abr-jun 2022. ilus
Artigo em Espanhol | IBECS | ID: ibc-206554

RESUMO

La hipertensión y la obesidad son importantes problemas de salud en todo el mundo con notables consecuencias sobre la morbilidad y la mortalidad. De hecho, tanto la hipertensión como la obesidad son importantes factores de riesgo para el desarrollo de enfermedades cardiovasculares. La disfunción endotelial, el remodelado vascular y las alteraciones en la mecánica vascular son aspectos comunes del daño vascular en la hipertensión, la obesidad y los aneurismas. Durante las últimas décadas, se ha demostrado la importancia de la inflamación de bajo grado en el daño vascular asociado a las enfermedades cardiovasculares. Dicha inflamación se caracteriza por la acumulación de células inflamatorias en la vasculatura, así como por el aumento de citoquinas proinflamatorias locales y circulantes. Por tanto, la identificación de nuevos mediadores inflamatorios implicados en dicho daño se ha convertido en un área de investigación muy importante. El interferón-γ (IFNγ) o el factor de necrosis tumoral-α (TNFα) son importantes citoquinas implicadas en el daño vascular asociado a la hipertensión. Además, se acepta que el TNFα es un mediador clave implicado en la resistencia a la insulina y el daño vascular observado en la obesidad. Ambas citoquinas inducen la expresión del gen 15 estimulado por interferón (ISG15), una proteína similar a la ubiquitina que induce una modificación postraducional reversible (ISGilación) y que también puede secretarse como forma libre. Las funciones de ISG15 están principalmente relacionadas con la respuesta inmune frente a infecciones, sin embargo, podría ser también una interesante nueva diana del daño cardiovascular.(AU)


Hypertension and obesity are major health problems worldwide with significant consequences on morbidity and mortality. In fact, both hypertension and obesity are important risk factors for the development of cardiovascular diseases. Endothelial dysfunction, vascular remodeling, and alterations in vascular mechanics are common aspects of vascular damage in hypertension, obesity, and aneurysms. During the last decades, the importance of low-grade inflammation in vascular damage associated with cardiovascular diseases has been demonstrated. This inflammation is characterized by the accumulation of inflammatory cells in the vasculature, as well as by the increase of local and circulating pro-inflammatory cytokines. Therefore, the identification of new inflammatory mediators involved in this damage has become a very important area of research. Interferón-γ (IFNγ) or tumor necrosis tumoral-α (TNFα) are important cytokines involved in the vascular damage associated with hypertension. Furthermore, it is accepted that TNFα is a key mediator involved in insulin resistance and vascular damage observed in obesity. Both cytokines induce the expression of interferon-stimulated gene 15 (ISG15), a protein similar to ubiquitin that induces a reversible post-translational modification (ISGylation) and that can also be secreted as a free form. The functions of ISG15 are mainly related to the immune response against infections, however, it could also be an interesting new target for cardiovascular damage.(AU)


Assuntos
Humanos , Hipertensão , Obesidade , Interferon gama , Fator de Necrose Tumoral alfa , Aneurisma da Aorta Abdominal , Resistência à Insulina , Estresse Oxidativo
5.
Cardiovasc Res ; 118(16): 3250-3268, 2022 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-34672341

RESUMO

AIMS: Interferon-stimulated gene 15 (ISG15) encodes a ubiquitin-like protein that induces a reversible post-translational modification (ISGylation) and can also be secreted as a free form. ISG15 plays an essential role as host-defence response to microbial infection; however, its contribution to vascular damage associated with hypertension is unknown. METHODS AND RESULTS: Bioinformatics identified ISG15 as a mediator of hypertension-associated vascular damage. ISG15 expression positively correlated with systolic and diastolic blood pressure and carotid intima-media thickness in human peripheral blood mononuclear cells. Consistently, Isg15 expression was enhanced in aorta from hypertension models and in angiotensin II (AngII)-treated vascular cells and macrophages. Proteomics revealed differential expression of proteins implicated in cardiovascular function, extracellular matrix and remodelling, and vascular redox state in aorta from AngII-infused ISG15-/- mice. Moreover, ISG15-/- mice were protected against AngII-induced hypertension, vascular stiffness, elastin remodelling, endothelial dysfunction, and expression of inflammatory and oxidative stress markers. Conversely, mice with excessive ISGylation (USP18C61A) show enhanced AngII-induced hypertension, vascular fibrosis, inflammation and reactive oxygen species (ROS) generation along with elastin breaks, aortic dilation, and rupture. Accordingly, human and murine abdominal aortic aneurysms showed augmented ISG15 expression. Mechanistically, ISG15 induces vascular ROS production, while antioxidant treatment prevented ISG15-induced endothelial dysfunction and vascular remodelling. CONCLUSION: ISG15 is a novel mediator of vascular damage in hypertension through oxidative stress and inflammation.


Assuntos
Aneurisma da Aorta Abdominal , Hipertensão , Camundongos , Humanos , Animais , Elastina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Angiotensina II/metabolismo , Interferons/metabolismo , Leucócitos Mononucleares/metabolismo , Espessura Intima-Media Carotídea , Estresse Oxidativo , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/metabolismo , Oxirredução , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/prevenção & controle , Inflamação , Camundongos Endogâmicos C57BL
6.
Br J Pharmacol ; 179(11): 2733-2753, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34877656

RESUMO

BACKGROUND AND PURPOSE: Microsomal prostaglandin E synthase-1 (mPGES-1) is an inducible isomerase responsible for prostaglandin E2 production in inflammatory conditions. We evaluated the role of mPGES-1 in the development and the metabolic and cardiovascular alterations of obesity. EXPERIMENTAL APPROACH: mPGES-1+/+ and mPGES-1-/- mice were fed with normal or high fat diet (HFD, 60% fat). The glycaemic and lipid profile was evaluated by glucose and insulin tolerance tests and colorimetric assays. Vascular function, structure and mechanics were assessed by myography. Histological studies, q-RT-PCR, and western blot analyses were performed in adipose tissue depots and cardiovascular tissues. Gene expression in abdominal fat and perivascular adipose tissue (PVAT) from patients was correlated with vascular damage. KEY RESULTS: Male mPGES-1-/- mice fed with HFD were protected against body weight gain and showed reduced adiposity, better glucose tolerance and insulin sensitivity, lipid levels and less white adipose tissue and PVAT inflammation and fibrosis, compared with mPGES-1+/+ mice. mPGES-1 knockdown prevented cardiomyocyte hypertrophy, cardiac fibrosis, endothelial dysfunction, aortic insulin resistance, and vascular inflammation and remodelling, induced by HFD. Obesity-induced weight gain and endothelial dysfunction of resistance arteries were ameliorated in female mPGES-1-/- mice. In humans, we found a positive correlation between mPGES-1 expression in abdominal fat and vascular remodelling, vessel stiffness, and systolic blood pressure. In human PVAT, there was a positive correlation between mPGES-1 expression and inflammatory markers. CONCLUSIONS AND IMPLICATIONS: mPGES-1 inhibition might be a novel therapeutic approach to the management of obesity and the associated cardiovascular and metabolic alterations.


Assuntos
Resistência à Insulina , Obesidade , Prostaglandina-E Sintases , Tecido Adiposo/metabolismo , Animais , Dieta Hiperlipídica , Feminino , Fibrose , Glucose/metabolismo , Humanos , Inflamação/metabolismo , Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Prostaglandina-E Sintases/genética , Prostaglandina-E Sintases/metabolismo
7.
Antioxidants (Basel) ; 9(10)2020 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-33020373

RESUMO

Perivascular adipose tissue (PVAT) is increasingly being regarded as an important endocrine organ that directly impacts vessel function, structure, and contractility in obesity-associated diseases. We uncover here a role for myeloid G protein-coupled receptor kinase 2 (GRK2) in the modulation of PVAT-dependent vasodilation responses. GRK2 expression positively correlates with myeloid- (CD68) and lymphoid-specific (CD3, CD4, and CD8) markers and with leptin in PVAT from patients with abdominal aortic aneurysms. Using mice hemizygous for GRK2 in the myeloid lineage (LysM-GRK2+/-), we found that GRK2 deficiency in myeloid cells allows animals to preserve the endothelium-dependent acetylcholine or insulin-induced relaxation, which is otherwise impaired by PVAT, in arteries of animals fed a high fat diet (HFD). Downregulation of GRK2 in myeloid cells attenuates HFD-dependent infiltration of macrophages and T lymphocytes in PVAT, as well as the induction of tumor necrosis factor-α (TNFα) and NADPH oxidase (Nox)1 expression, whereas blocking TNFα or Nox pathways by pharmacological means can rescue the impaired vasodilator responses to insulin in arteries with PVAT from HFD-fed animals. Our results suggest that myeloid GRK2 could be a potential therapeutic target in the development of endothelial dysfunction induced by PVAT in the context of obesity.

8.
Arterioscler Thromb Vasc Biol ; 40(10): 2408-2424, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32847388

RESUMO

OBJECTIVE: microRNAs are master regulators of gene expression with essential roles in virtually all biological processes. miR-217 has been associated with aging and cellular senescence, but its role in vascular disease is not understood. Approach and Results: We have used an inducible endothelium-specific knock-in mouse model to address the role of miR-217 in vascular function and atherosclerosis. miR-217 reduced NO production and promoted endothelial dysfunction, increased blood pressure, and exacerbated atherosclerosis in proatherogenic apoE-/- mice. Moreover, increased endothelial miR-217 expression led to the development of coronary artery disease and altered left ventricular heart function, inducing diastolic and systolic dysfunction. Conversely, inhibition of endogenous vascular miR-217 in apoE-/- mice improved vascular contractility and diminished atherosclerosis. Transcriptome analysis revealed that miR-217 regulates an endothelial signaling hub and downregulates a network of eNOS (endothelial NO synthase) activators, including VEGF (vascular endothelial growth factor) and apelin receptor pathways, resulting in diminished eNOS expression. Further analysis revealed that human plasma miR-217 is a biomarker of vascular aging and cardiovascular risk. CONCLUSIONS: Our results highlight the therapeutic potential of miR-217 inhibitors in aging-related cardiovascular disease.


Assuntos
Envelhecimento/metabolismo , Aterosclerose/metabolismo , Células Endoteliais/metabolismo , MicroRNAs/metabolismo , Placa Aterosclerótica , Fatores Etários , Idoso de 80 Anos ou mais , Envelhecimento/genética , Animais , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/metabolismo , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Hemodinâmica , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , MicroRNAs/sangue , MicroRNAs/genética , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda
9.
Antioxidants (Basel) ; 9(9)2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32854368

RESUMO

Antioxidant compounds, including polyphenols, have therapeutic effects because of their anti-inflammatory, antihypertensive, antithrombotic and antiproliferative properties. They play important roles in protecting the cardiovascular and neurological systems, by having preventive or protective effects against free radicals produced by either normal or pathological metabolism in such systems. For instance, resveratrol, a well-known potent antioxidant, has a counteracting effect on the excess of reactive oxygen species (ROS) and has a number of therapeutic benefits, like anti-inflammatory, anti-cancer and cardioprotective activities. Based on previous work from our group, and on the most frequent OH substitutions of natural polyphenols, we designed two series of synthetically accessible bis-polyhydroxyphenyl derivatives, separated by amide or urea linkers. These compounds exhibit high antioxidant ability (oxygen radical absorbance capacity (ORAC) assay) and interesting radical scavenging activity (RSA) values (2,2'-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and α,α-diphenyl-ß-picrylhydrazyl (DPPH) tests). Some of the best polyphenols were evaluated in two biological systems, endothelial cells (in vitro) and whole aorta (ex vivo), highly susceptible for the deleterious effects of prooxidants under different inflammatory conditions, showing protection against oxidative stress induced by inflammatory stimuli relevant in cardiovascular diseases, i.e., Angiotensin II and IL-1ß. Selected compounds also showed strong in vivo antioxidant properties when evaluated in the model organism Saccharomyces cerevisiae.

10.
J Cell Mol Med ; 22(10): 4948-4962, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30063118

RESUMO

Branched-chain amino acids (BCAA: leucine, isoleucine and valine) are essential amino acids implicated in glucose metabolism and maintenance of correct brain function. Elevated BCAA levels can promote an inflammatory response in peripheral blood mononuclear cells. However, there are no studies analysing the direct effects of BCAA on endothelial cells (ECs) and its possible modulation of vascular function. In vitro and ex vivo studies were performed in human ECs and aorta from male C57BL/6J mice, respectively. In ECs, BCAA (6 mmol/L) increased eNOS expression, reactive oxygen species production by mitochondria and NADPH oxidases, peroxynitrite formation and nitrotyrosine expression. Moreover, BCAA induced pro-inflammatory responses through the transcription factor NF-κB that resulted in the release of intracellular adhesion molecule-1 and E-selectin conferring endothelial activation and adhesion capacity to inflammatory cells. Pharmacological inhibition of mTORC1 intracellular signalling pathway decreased BCAA-induced pro-oxidant and pro-inflammatory effects in ECs. In isolated murine aorta, BCAA elicited vasoconstrictor responses, particularly in pre-contracted vessels and after NO synthase blockade, and triggered endothelial dysfunction, effects that were inhibited by different antioxidants, further demonstrating the potential of BCAA to induce oxidative stress with functional impact. In summary, we demonstrate that elevated BCAA levels generate inflammation and oxidative stress in ECs, thereby facilitating inflammatory cells adhesion and endothelial dysfunction. This might contribute to the increased cardiovascular risk observed in patients with elevated BCAA blood levels.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Aorta/metabolismo , Células Endoteliais/efeitos dos fármacos , Inflamação/metabolismo , Animais , Antioxidantes/administração & dosagem , Aorta/efeitos dos fármacos , Selectina E/genética , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Glucose/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Molécula 1 de Adesão Intercelular/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/genética , Mitocôndrias/metabolismo , NF-kappa B/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Ácido Peroxinitroso/biossíntese , Ácido Peroxinitroso/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirosina/análogos & derivados , Tirosina/biossíntese , Tirosina/metabolismo , Vasoconstritores/administração & dosagem
11.
Hypertension ; 72(2): 492-502, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29891646

RESUMO

mPGES-1 (microsomal prostaglandin E synthase-1), the downstream enzyme responsible for PGE2 (prostaglandin E2) synthesis in inflammatory conditions and oxidative stress are increased in vessels from hypertensive animals. We evaluated the role of mPGES-1-derived PGE2 in the vascular dysfunction and remodeling in hypertension and the possible contribution of oxidative stress. We used human peripheral blood mononuclear cells from asymptomatic patients, arteries from untreated and Ang II (angiotensin II)-infused mPGES-1-/- and mPGES-1+/+ mice, and vascular smooth muscle cells exposed to PGE2 In human cells, we found a positive correlation between mPGES-1 mRNA and carotid intima-media thickness (r=0.637; P<0.001) and with NADPH oxidase-dependent superoxide production (r=0.417; P<0.001). In Ang II-infused mice, mPGES-1 deletion prevented all of the following: (1) the augmented wall:lumen ratio, vascular stiffness, and altered elastin structure; (2) the increased gene expression of profibrotic and proinflammatory markers; (3) the increased vasoconstrictor responses and endothelial dysfunction; (4) the increased NADPH oxidase activity and the diminished mitochondrial membrane potential; and (5) the increased reactive oxygen species generation and reduced NO bioavailability. In vascular smooth muscle cells or aortic segments, PGE2 increased NADPH oxidase expression and activity and reduced mitochondrial membrane potential, effects that were abolished by antagonists of the PGE2 receptors (EP), EP1 and EP3, and by JNK (c-Jun N-terminal kinase) and ERK1/2 (extracellular-signal-regulated kinases 1/2) inhibition. Deletion of mPGES-1 augmented vascular production of PGI2 suggesting rediversion of the accumulated PGH2 substrate. In conclusion, mPGES-1-derived PGE2 is involved in vascular remodeling, stiffness, and endothelial dysfunction in hypertension likely through an increase of oxidative stress produced by NADPH oxidase and mitochondria.


Assuntos
Artérias Carótidas/fisiopatologia , Regulação da Expressão Gênica , Hipertensão/genética , Músculo Liso Vascular/metabolismo , Estresse Oxidativo , Prostaglandina-E Sintases/genética , Rigidez Vascular , Animais , Artérias Carótidas/metabolismo , Modelos Animais de Doenças , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/fisiologia , Prostaglandina-E Sintases/biossíntese , RNA/genética
12.
Pharmacol Res ; 133: 236-249, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29309904

RESUMO

Cyclooxygenase-2 (COX-2) derived-prostanoids participate in the altered vascular function and mechanical properties in cardiovascular diseases. We investigated whether regulator of calcineurin 1 (Rcan1) participates in vascular contractility and stiffness through the regulation of COX-2. For this, wild type (Rcan1+/+) and Rcan1-deficient (Rcan1-/-) mice untreated or treated with the COX-2 inhibitor rofecoxib were used. Vascular function and structure were analysed by myography. COX-2 and phospo-p65 expression were studied by western blotting and immunohistochemistry and TXA2 production by ELISA. We found that Rcan1 deficiency increases COX-2 and IL-6 expression and NF-κB activation in arteries and vascular smooth muscle cells (VSMC). Adenoviral-mediated re-expression of Rcan1.4 in Rcan1-/- VSMC normalized COX-2 expression. Phenylephrine-induced vasoconstrictor responses were greater in aorta from Rcan1-/- compared to Rcan1+/+ mice. This increased response were diminished by etoricoxib, furegrelate, SQ 29548, cyclosporine A and parthenolide, inhibitors of COX-2, TXA2 synthase, TP receptors, calcineurin and NF-κB, respectively. Endothelial removal and NOS inhibition increased phenylephrine responses only in Rcan1+/+ mice. TXA2 levels were greater in Rcan1-/- mice. In small mesenteric arteries, vascular function and structure were similar in both groups of mice; however, vessels from Rcan1-/- mice displayed an increase in vascular stiffness that was diminished by rofecoxib. In conclusion, our results suggest that Rcan1 might act as endogenous negative modulator of COX-2 expression and activity by inhibiting calcineurin and NF-kB pathways to maintain normal contractility and vascular stiffness in aorta and small mesenteric arteries, respectively. Our results uncover a new role for Rcan1 in vascular contractility and mechanical properties.


Assuntos
Aorta Torácica/fisiologia , Ciclo-Oxigenase 2/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Artérias Mesentéricas/fisiologia , Proteínas Musculares/fisiologia , Músculo Liso Vascular/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Células Cultivadas , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia
13.
Sci Rep ; 7(1): 16802, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29196758

RESUMO

We have investigated whether mineralocorticoid receptor activation can participate in the profibrotic effects of leptin in cardiac myofibroblasts, as well as the potential mechanisms involved. The presence of eplerenone reduced the leptin-induced increase in protein levels of collagen I, transforming growth factor ß, connective tissue growth factor and galectin-3 and the levels of both total and mitochondrial of superoxide anion (O2.-) in cardiac myofibroblasts. Likewise, the MEK/ERK inhibitor, PD98059, and the PI3/Akt inhibitor, LY294002, showed a similar pattern. Mitochondrial reactive oxygen species (ROS) scavenger (MitoTempo) attenuated the increase in body weight observed in rats fed a high fat diet (HFD). No differences were found in cardiac function or blood pressure among any group. However, the cardiac fibrosis and enhanced O2.-levels observed in HFD rats were attenuated by MitoTempo, which also prevented the increased circulating leptin and aldosterone levels in HFD fed animals. This study supports a role of mineralocorticoid receptor in the cardiac fibrosis induced by leptin in the context of obesity and highlights the role of the mitochondrial ROS in this process.


Assuntos
Fibrose Endomiocárdica/metabolismo , Leptina/metabolismo , Miocárdio/citologia , Obesidade/complicações , Espécies Reativas de Oxigênio/metabolismo , Receptores de Mineralocorticoides/metabolismo , Animais , Colágeno Tipo I/metabolismo , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Fibrose Endomiocárdica/etiologia , Eplerenona/farmacologia , Fibroblastos/citologia , Galectina 3/metabolismo , Masculino , Mitocôndrias/metabolismo , Obesidade/induzido quimicamente , Obesidade/metabolismo , Estresse Oxidativo , Ratos , Ratos Wistar , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...