Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 149(6): 1298-313, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22682250

RESUMO

Wnt signaling plays critical roles in development of various organs and pathogenesis of many diseases, and augmented Wnt signaling has recently been implicated in mammalian aging and aging-related phenotypes. We here report that complement C1q activates canonical Wnt signaling and promotes aging-associated decline in tissue regeneration. Serum C1q concentration is increased with aging, and Wnt signaling activity is augmented during aging in the serum and in multiple tissues of wild-type mice, but not in those of C1qa-deficient mice. C1q activates canonical Wnt signaling by binding to Frizzled receptors and subsequently inducing C1s-dependent cleavage of the ectodomain of Wnt coreceptor low-density lipoprotein receptor-related protein 6. Skeletal muscle regeneration in young mice is inhibited by exogenous C1q treatment, whereas aging-associated impairment of muscle regeneration is restored by C1s inhibition or C1qa gene disruption. Our findings therefore suggest the unexpected role of complement C1q in Wnt signal transduction and modulation of mammalian aging.


Assuntos
Envelhecimento/metabolismo , Complemento C1q/metabolismo , Via de Sinalização Wnt , Animais , Complemento C1s/metabolismo , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Soro/metabolismo
2.
Circ J ; 2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37438143

RESUMO

Evidence indicates a role of cellular senescence and systemic insulin resistance (hyperinsulinemia) in the pathogenesis of age-related cardiovascular-metabolic disorders, including heart failure, atherosclerotic diseases, obesity, and diabetes. "Metabolic remodeling" is one of the keywords for aging research, and studies with brown adipose tissue have shown that maintaining the homeostasis of this organ is crucial to suppressing the progression of pathologies in obesity and heart failure. The mechanisms contributing to the synchronization of aging (sync-aging) are mysterious and interesting. "Senometabolite" or "senoprotein" are defined as circulating molecules that have causal roles in sync-aging, which requires the establishment of new concepts: age-related fibrotic disorders (A-FiDs), and senometabolite-related disorders (SRDs). Globally, researchers are active in comprehensive and conclusive studies targeting age-related circulating molecules. Recently, the senolytic approach opened a new avenue for aging research. Senolysis, mediated through a genetic/pharmacologic/vaccination approach, reversed aging and pathologies in age-related diseases. Suppression of prosenescent molecules (senocules) and senolysis, the specific depletion of senescent cells, will become next-generation therapies for cardiovascular diseases.

3.
Eur Heart J ; 43(20): 1973-1989, 2022 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-35190817

RESUMO

AIMS: Cereblon (CRBN) is a substrate receptor of the E3 ubiquitin ligase complex that was reported to target ion channel proteins. L-type voltage-dependent Ca2+ channel (LTCC) density and dysfunction is a critical player in heart failure with reduced ejection fraction (HFrEF). However, the underlying cellular mechanisms by which CRBN regulates LTCC subtype Cav1.2α during cardiac dysfunction remain unclear. Here, we explored the role of CRBN in HFrEF by investigating the direct regulatory role of CRBN in Cav1.2α activity and examining how it can serve as a target to address myocardial dysfunction. METHODS AND RESULTS: Cardiac tissues from HFrEF patients exhibited increased levels of CRBN compared with controls. In vivo and ex vivo studies demonstrated that whole-body CRBN knockout (CRBN-/-) and cardiac-specific knockout mice (Crbnfl/fl/Myh6Cre+) exhibited enhanced cardiac contractility with increased LTCC current (ICaL) compared with their respective controls, which was modulated by the direct interaction of CRBN with Cav1.2α. Mechanistically, the Lon domain of CRBN directly interacted with the N-terminal of Cav1.2α. Increasing CRBN levels enhanced the ubiquitination and proteasomal degradation of Cav1.2α and decreased ICaL. In contrast, genetic or pharmacological depletion of CRBN via TD-165, a novel PROTAC-based CRBN degrader, increased surface expression of Cav1.2α and enhanced ICaL. Low CRBN levels protected the heart against cardiomyopathy in vivo. CONCLUSION: Cereblon selectively degrades Cav1.2α, which in turn facilitates cardiac dysfunction. A targeted approach or an efficient method of reducing CRBN levels could serve as a promising strategy for HFrEF therapeutics.


Assuntos
Insuficiência Cardíaca , Ubiquitina-Proteína Ligases , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Humanos , Camundongos , Volume Sistólico , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
J Mol Cell Cardiol ; 129: 105-117, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30790589

RESUMO

p53 is a guardian of the genome that protects against carcinogenesis. There is accumulating evidence that p53 is activated with aging. Such activation has been reported to contribute to various age-associated pathologies, but its role in vascular dysfunction is largely unknown. The aim of this study was to investigate whether activation of endothelial p53 has a pathological effect in relation to endothelial function. We established endothelial p53 loss-of-function and gain-of-function models by breeding endothelial-cell specific Cre mice with floxed Trp53 or floxed Mdm2/Mdm4 mice, respectively. Then we induced diabetes by injection of streptozotocin. In the diabetic state, endothelial p53 expression was markedly up-regulated and endothelium-dependent vasodilatation was significantly impaired. Impairment of vasodilatation was significantly ameliorated in endothelial p53 knockout (EC-p53 KO) mice, and deletion of endothelial p53 also significantly enhanced the induction of angiogenesis by ischemia. Conversely, activation of endothelial p53 by deleting Mdm2/Mdm4 reduced both endothelium-dependent vasodilatation and ischemia-induced angiogenesis. Introduction of p53 into human endothelial cells up-regulated the expression of phosphatase and tensin homolog (PTEN), thereby reducing phospho-eNOS levels. Consistent with these results, the beneficial impact of endothelial p53 deletion on endothelial function was attenuated in EC-p53 KO mice with an eNOS-deficient background. These results show that endothelial p53 negatively regulates endothelium-dependent vasodilatation and ischemia-induced angiogenesis, suggesting that inhibition of endothelial p53 could be a novel therapeutic target in patients with metabolic disorders.


Assuntos
Endotélio Vascular/fisiopatologia , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Isquemia/complicações , Isquemia/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Permeabilidade Capilar , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Endotélio Vascular/metabolismo , Deleção de Genes , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ativação Transcricional/genética , Regulação para Cima/genética , Vasodilatação
5.
Int Heart J ; 59(4): 837-844, 2018 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-29794381

RESUMO

Previous studies have suggested that cellular senescence plays a central role in the progression of pathologic changes in the failing heart. It is well known that the sympathetic nervous system is activated in patients with heart failure, and this change is associated with poor clinical outcomes. Sympathetic activation increases the levels of various catecholamines, such as epinephrine and norepinephrine, but the contribution of these catecholamines to cellular senescence associated with heart failure remains to be determined. We found that catecholamine infusion induced senescence of endothelial cells and bone marrow cells, and promoted cardiac dysfunction in mice. In C57BL/6NCr mice, the continuous infusion of isoproterenol-induced cardiac inflammation and cardiac dysfunction. Expression of p53, a master regulator of cellular senescence, was increased in the cardiac tissue and bone marrow cells of these mice. Suppression of cellular senescence by genetic deletion of p53 in endothelial cells or bone marrow cells led to improvement of isoproterenol-induced cardiac dysfunction. In vitro studies showed that adrenergic signaling increased the expression of p53 and adhesion molecules by endothelial cells and macrophages. Our results indicate that catecholamine-induced senescence of endothelial cells and bone marrow cells plays a pivotal role in the progression of heart failure. Suppression of catecholamine-p53 signaling is crucial for inhibition of remodeling in the failing heart.


Assuntos
Células da Medula Óssea , Catecolaminas , Senescência Celular , Células Endoteliais , Insuficiência Cardíaca , Isoproterenol/farmacologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Catecolaminas/metabolismo , Catecolaminas/farmacologia , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Progressão da Doença , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Expressão Gênica/efeitos dos fármacos , Genes p53/fisiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Simpatomiméticos/farmacologia
6.
Int Heart J ; 59(3): 607-613, 2018 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-29681573

RESUMO

Vascular cells have a finite lifespan and eventually enter irreversible growth arrest called cellular senescence. We have previously suggested that vascular cell senescence contributes to the pathogenesis of human atherosclerosis. Amlodipine is a mixture of two enantiomers, one of which (S- enantiomer) has L-type channel blocking activity, while the other (R+ enantiomer) shows ~1000-fold weaker channel blocking activity than S- enantiomer and has other unknown effects. It has been reported that amlodipine inhibits the progression of atherosclerosis in humans, but the molecular mechanism of this beneficial effect remains unknown. Apolipoprotein E-deficient mice on a high-fat diet were treated with amlodipine, its R+ enantiomer or vehicle for eight weeks. Compared with vehicle treatment, both amlodipine and the R+ enantiomer significantly reduced the number of senescent vascular cells and inhibited plaque formation to a similar extent. Expression of the pro-inflammatory molecule interleukin-1ß was markedly upregulated in vehicle-treated mice, but was inhibited to a similar extent by treatment with amlodipine or the R+ enantiomer. Likewise, activation of p53 (a critical inducer of senescence) was markedly suppressed by treatment with amlodipine or the R+ enantiomer. These results suggest that amlodipine inhibits vascular cell senescence and protects against atherogenesis at least partly by a mechanism that is independent of calcium channel blockade.


Assuntos
Anlodipino/farmacologia , Aterosclerose/tratamento farmacológico , Bloqueadores dos Canais de Cálcio/farmacologia , Senescência Celular/efeitos dos fármacos , Animais , Western Blotting , Canais de Cálcio/efeitos dos fármacos , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiopatologia , Proteína Supressora de Tumor p53/metabolismo
7.
J Biol Chem ; 291(6): 2566-75, 2016 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-26631720

RESUMO

Wnt signaling has diverse actions in cardiovascular development and disease processes. Secreted frizzled-related protein 5 (Sfrp5) has been shown to function as an extracellular inhibitor of non-canonical Wnt signaling that is expressed at relatively high levels in white adipose tissue. The aim of this study was to investigate the role of Sfrp5 in the heart under ischemic stress. Sfrp5 KO and WT mice were subjected to ischemia/reperfusion (I/R). Although Sfrp5-KO mice exhibited no detectable phenotype when compared with WT control at baseline, they displayed larger infarct sizes, enhanced cardiac myocyte apoptosis, and diminished cardiac function following I/R. The ischemic lesions of Sfrp5-KO mice had greater infiltration of Wnt5a-positive macrophages and greater inflammatory cytokine and chemokine gene expression when compared with WT mice. In bone marrow-derived macrophages, Wnt5a promoted JNK activation and increased inflammatory gene expression, whereas treatment with Sfrp5 blocked these effects. These results indicate that Sfrp5 functions to antagonize inflammatory responses after I/R in the heart, possibly through a mechanism involving non-canonical Wnt5a/JNK signaling.


Assuntos
Proteínas de Membrana/metabolismo , Isquemia Miocárdica/metabolismo , Miocardite/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Animais , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Isquemia Miocárdica/genética , Isquemia Miocárdica/patologia , Miocardite/genética , Miocardite/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a
8.
Circ J ; 82(1): 10-16, 2017 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-29225300

RESUMO

It is thought that at least 6,500 low-molecular-weight metabolites exist in humans, and these metabolites have various important roles in biological systems in addition to proteins and genes. Comprehensive assessment of endogenous metabolites is called metabolomics, and recent advances in this field have enabled us to understand the critical role of previously unknown metabolites or metabolic pathways in the cardiovascular system. In this review, we will focus on heart failure and how metabolomic analysis has contributed to improving our understanding of the pathogenesis of this critical condition.


Assuntos
Insuficiência Cardíaca/metabolismo , Metabolômica , Aminoácidos , Glicólise , Humanos , Metabolismo dos Lipídeos , Redes e Vias Metabólicas
9.
J Mol Cell Cardiol ; 97: 245-62, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27262674

RESUMO

The heart must continuously pump blood to supply the body with oxygen and nutrients. To maintain the high energy consumption required by this role, the heart is equipped with multiple complex biological systems that allow adaptation to changes of systemic demand. The processes of growth (hypertrophy), angiogenesis, and metabolic plasticity are critically involved in maintenance of cardiac homeostasis. Cardiac hypertrophy is classified as physiological when it is associated with normal cardiac function or as pathological when associated with cardiac dysfunction. Physiological hypertrophy of the heart occurs in response to normal growth of children or during pregnancy, as well as in athletes. In contrast, pathological hypertrophy is induced by factors such as prolonged and abnormal hemodynamic stress, due to hypertension, myocardial infarction etc. Pathological hypertrophy is associated with fibrosis, capillary rarefaction, increased production of pro-inflammatory cytokines, and cellular dysfunction (impairment of signaling, suppression of autophagy, and abnormal cardiomyocyte/non-cardiomyocyte interactions), as well as undesirable epigenetic changes, with these complex responses leading to maladaptive cardiac remodeling and heart failure. This review describes the key molecules and cellular responses involved in physiological/pathological cardiac hypertrophy.


Assuntos
Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Animais , Autofagia , Cardiomegalia/diagnóstico , Regulação da Expressão Gênica , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Humanos , Neovascularização Patológica , Neovascularização Fisiológica , Fatores de Risco , Transdução de Sinais
10.
Nihon Rinsho ; 74(9): 1491-1496, 2016 Sep.
Artigo em Japonês | MEDLINE | ID: mdl-30557482

RESUMO

Protein p53 is described as guardian of the genome contributing for the suppression of tumorigenesis. p53 is also known to be the critical regulator of cellular senescence. It is activated by various genomic stresses including telomere attrition, excessive mitogenic sig- naling and genotoxic agents(eg. reactive oxygen species), and mediates cellular senescence. Senescent cells develop growth arrest, associated with altered gene profiles. These changes induce chronic inflammation and organ dysfunction, promoting pathologies in age related disorders. In this review, we delineate the role of p53 induced cellular senescence in age related disorders, such as obesity/diabetes and heart failure.


Assuntos
Envelhecimento , Dano ao DNA , Espécies Reativas de Oxigênio , Proteína Supressora de Tumor p53 , Envelhecimento/fisiologia , Proliferação de Células , Transformação Celular Neoplásica , Senescência Celular , Humanos , Espécies Reativas de Oxigênio/metabolismo , Telômero , Proteína Supressora de Tumor p53/fisiologia
11.
J Mol Cell Cardiol ; 85: 183-98, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26055447

RESUMO

The rates of death and disability caused by severe heart failure are still unacceptably high. There is evidence that the sterile inflammatory response has a critical role in the progression of cardiac remodeling in the failing heart. The p53 signaling pathway has been implicated in heart failure, but the pathological link between p53 and inflammation in the failing heart is largely unknown. Here we demonstrate a critical role of p53-induced inflammation in heart failure. Expression of p53 was increased in cardiac endothelial cells and bone marrow cells in response to pressure overload, leading to up-regulation of intercellular adhesion molecule-1 (ICAM1) expression by endothelial cells and integrin expression by bone marrow cells. Deletion of p53 from endothelial cells or bone marrow cells significantly reduced ICAM1 or integrin expression, respectively, as well as decreasing cardiac inflammation and ameliorating systolic dysfunction during pressure overload. Conversely, overexpression of p53 in bone marrow cells led to an increase of integrin expression and cardiac inflammation that reduced systolic function. Norepinephrine markedly increased p53 expression in endothelial cells and macrophages. Reducing ß2-adrenergic receptor expression in endothelial cells or bone marrow cells attenuated cardiac inflammation and improved systolic dysfunction during pressure overload. These results suggest that activation of the sympathetic nervous system promotes cardiac inflammation by up-regulating ICAM1 and integrin expression via p53 signaling to exacerbate cardiac dysfunction. Inhibition of p53-induced inflammation may be a novel therapeutic strategy for heart failure.


Assuntos
Proteína Supressora de Tumor p53/fisiologia , Animais , Antígeno CD11a/metabolismo , Expressão Gênica , Células HEK293 , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hipertensão/complicações , Hipertensão/patologia , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/imunologia , Miocárdio/patologia , Norepinefrina/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Sístole , Pressão Ventricular
12.
Int Heart J ; 56(3): 255-9, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25912906

RESUMO

Obesity has dramatically increased throughout the world, and has become one of the chief healthcare problems in many societies. Evidence has emerged that adipose tissue dysfunction associated with obesity is critically involved in the development of cardiovascular and metabolic disorders. In this review, we delineate the link between adipose tissue abnormalities and systemic metabolic dysfunction in patients with cardio-metabolic diseases and discuss the underlying mechanisms.


Assuntos
Tecido Adiposo/fisiopatologia , Doenças Cardiovasculares/etiologia , Doenças Metabólicas/etiologia , Tecido Adiposo Marrom/fisiopatologia , Coração/fisiopatologia , Humanos , Insulina/fisiologia , Resistência à Insulina/fisiologia , Obesidade/fisiopatologia
13.
Nat Aging ; 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816549

RESUMO

It has been reported that accumulation of senescent cells in various tissues contributes to pathological aging and that elimination of senescent cells (senolysis) improves age-associated pathologies. Here, we demonstrate that inhibition of sodium-glucose co-transporter 2 (SGLT2) enhances clearance of senescent cells, thereby ameliorating age-associated phenotypic changes. In a mouse model of dietary obesity, short-term treatment with the SGLT2 inhibitor canagliflozin reduced the senescence load in visceral adipose tissue and improved adipose tissue inflammation and metabolic dysfunction, but normalization of plasma glucose by insulin treatment had no effect on senescent cells. Canagliflozin extended the lifespan of mice with premature aging even when treatment was started in middle age. Metabolomic analyses revealed that short-term treatment with canagliflozin upregulated 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside, enhancing immune-mediated clearance of senescent cells by downregulating expression of programmed cell death-ligand 1. These findings suggest that inhibition of SGLT2 has an indirect senolytic effect by enhancing endogenous immunosurveillance of senescent cells.

14.
Arterioscler Thromb Vasc Biol ; 32(8): 1902-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22556331

RESUMO

OBJECTIVE: The central nervous system is thought to influence the regulation of the cardiovascular system in response to humoral and neural signals from peripheral tissues, but our understanding of the molecular mechanisms involved is still quite limited. METHODS AND RESULTS: Here, we demonstrate a central nervous system-mediated mechanism by which brain-derived neurotrophic factor (BDNF) has a protective effect against cardiac remodeling after myocardial infarction (MI). We generated conditional BDNF knockout mice, in which expression of BDNF was systemically reduced, by using the inducible Cre-loxP system. Two weeks after MI was induced surgically in these mice, systolic function was significantly impaired and cardiac size was markedly increased in conditional BDNF knockout mice compared with controls. Cardiomyocyte death was increased in these mice, along with decreased expression of survival molecules. Deletion of the BDNF receptor (tropomyosin-related kinase B) from the heart also led to the exacerbation of cardiac dysfunction after MI. The plasma levels of BDNF were markedly increased after MI, and this increase was associated with the upregulation of BDNF expression in the brain, but not in the heart. Ablation of afferent nerves from the heart or genetic disruption of neuronal BDNF expression inhibited the increase of plasma BDNF after MI and led to the exacerbation of cardiac dysfunction. Peripheral administration of BDNF significantly restored the cardiac phenotype of neuronal BDNF-deficient mice. CONCLUSIONS: These results suggest that BDNF expression is upregulated by neural signals from the heart after MI and then protects the myocardium against ischemic injury.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Encéfalo/fisiologia , Infarto do Miocárdio/fisiopatologia , Animais , Fator Neurotrófico Derivado do Encéfalo/sangue , Camundongos , Camundongos Knockout , Receptor trkB/fisiologia , Transdução de Sinais , Sístole , Remodelação Ventricular
15.
Nature ; 446(7134): 444-8, 2007 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-17334357

RESUMO

Cardiac hypertrophy occurs as an adaptive response to increased workload to maintain cardiac function. However, prolonged cardiac hypertrophy causes heart failure, and its mechanisms are largely unknown. Here we show that cardiac angiogenesis is crucially involved in the adaptive mechanism of cardiac hypertrophy and that p53 accumulation is essential for the transition from cardiac hypertrophy to heart failure. Pressure overload initially promoted vascular growth in the heart by hypoxia-inducible factor-1 (Hif-1)-dependent induction of angiogenic factors, and inhibition of angiogenesis prevented the development of cardiac hypertrophy and induced systolic dysfunction. Sustained pressure overload induced an accumulation of p53 that inhibited Hif-1 activity and thereby impaired cardiac angiogenesis and systolic function. Conversely, promoting cardiac angiogenesis by introducing angiogenic factors or by inhibiting p53 accumulation developed hypertrophy further and restored cardiac dysfunction under chronic pressure overload. These results indicate that the anti-angiogenic property of p53 may have a crucial function in the transition from cardiac hypertrophy to heart failure.


Assuntos
Baixo Débito Cardíaco/fisiopatologia , Cardiomegalia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Animais , Aorta/patologia , Aorta/fisiopatologia , Pressão Sanguínea , Cardiomegalia/patologia , Circulação Coronária , Progressão da Doença , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Neovascularização Patológica , Proteína Supressora de Tumor p53/genética
16.
J Hepatol ; 57(4): 837-43, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22641095

RESUMO

BACKGROUND & AIMS: The tumor suppressor p53 is a primary sensor of stressful stimuli, controlling a number of biologic processes. The aim of our study was to examine the roles of p53 in non-alcoholic steatohepatitis (NASH). METHODS: Male wild type and p53-deficient mice were fed a methionine- and choline-deficient diet for 8 weeks to induce nutritional steatohepatitis. mRNA expression profiles in normal liver samples and liver samples from patients with non-alcoholic liver disease (NAFLD) were also evaluated. RESULTS: Hepatic p53 and p66Shc signaling was enhanced in the mouse NASH model. p53 deficiency suppressed the enhanced p66Shc signaling, decreased hepatic lipid peroxidation and the number of apoptotic hepatocytes, and ameliorated progression of nutritional steatohepatitis. In primary cultured hepatocytes, transforming growth factor (TGF)-ß treatment increased p53 and p66Shc signaling, leading to exaggerated reactive oxygen species (ROS) accumulation and apoptosis. Deficient p53 signaling inhibited TGF-ß-induced p66Shc signaling, ROS accumulation, and hepatocyte apoptosis. Furthermore, expression levels of p53, p21, and p66Shc were significantly elevated in human NAFLD liver samples, compared with results obtained with normal liver samples. Among NAFLD patients, those with NASH had significantly higher hepatic expression levels of p53, p21, and p66Shc compared with the group with simple steatosis. A significant correlation between expression levels of p53 and p66Shc was observed. CONCLUSIONS: p53 in hepatocytes regulates steatohepatitis progression by controlling p66Shc signaling, ROS levels, and apoptosis, all of which may be regulated by TGF-ß. Moreover, p53/p66Shc signaling in the liver appears to be a promising target for the treatment of NASH.


Assuntos
Fígado Gorduroso/metabolismo , RNA Mensageiro/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Deficiência de Colina/complicações , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Hepatócitos/metabolismo , Humanos , Masculino , Metionina/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Cultura Primária de Células , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteína Supressora de Tumor p53/genética , Regulação para Cima/efeitos dos fármacos
17.
Circ Res ; 106(2): 391-8, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19940264

RESUMO

RATIONALE: The axon-guiding molecules known as semaphorins and their receptors (plexins) regulate the vascular pattern and play an important role in the development of vascular network during embryogenesis. Semaphorin (Sema)3E is one of the class 3 semaphorins, and plexinD1 is known to be its receptor. Although these molecules have a role in embryonic vascular development, it remains unclear whether the Sema3E/plexinD1 axis is involved in postnatal angiogenesis. OBJECTIVE: The objective of this study was to elucidate the role of Sema3E/plexinD1 in postnatal angiogenesis. METHODS AND RESULTS: Sema3E inhibited cell growth and tube formation by suppressing the vascular endothelial growth factor (VEGF) signaling pathway. Expression of Sema3E and plexinD1 was markedly upregulated in ischemic limbs of mice (2.5- and 4.5-fold increase for Sema3E and plexinD1, respectively), and inhibition of this pathway by introduction of the plexinD1-Fc gene or disruption of Sema3E led to a significant increase of blood flow recovery (1.6- and 1.5-fold increase for the plexinD1-Fc gene treatment and Sema3E disruption, respectively). Hypoxia activated the tumor suppressor protein p53, thereby upregulating Sema3E expression. Expression of p53 and Sema3E was enhanced in diabetic mice compared with normal mice (2- and 1.3-fold increase for p53 and Sema3E, respectively). Consequently, neovascularization after VEGF treatment was poor in the ischemic tissues of diabetic mice, whereas treatment with VEGF plus plexinD1-Fc markedly improved neovascularization. CONCLUSIONS: These results indicate that inhibition of Sema3E may be a novel strategy for therapeutic angiogenesis, especially when VEGF is ineffective.


Assuntos
Moléculas de Adesão Celular Neuronais/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Semaforinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Western Blotting , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Isquemia , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Oncogênica v-akt/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Semaforinas/genética , Semaforinas/metabolismo , Estreptozocina , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Inflamm Regen ; 42(1): 2, 2022 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-35012677

RESUMO

Studies analyzing heterochronic parabiosis mice models showed that molecules in the blood of young mice rejuvenate aged mice. Therefore, blood-based therapies have become one of the therapeutic approaches to be considered for age-related diseases. Blood includes numerous biologically active molecules such as proteins, metabolites, hormones, miRNAs, etc. and accumulating evidence indicates some of these change their concentration with chronological aging or age-related disorders. The level of some circulating molecules showed a negative or positive correlation with all-cause mortality, cardiovascular events, or metabolic disorders. Through analyses of clinical/translation/basic research, some molecules were focused on as therapeutic targets. One approach is the supplementation of circulating anti-aging molecules. Favorable results in preclinical studies let some molecules to be tested in humans. These showed beneficial or neutral results, and some were inconsistent. Studies with rodents and humans indicate circulating molecules can be recognized as biomarkers or therapeutic targets mediating their pro-aging or anti-aging effects. Characterization of these molecules with aging, testing their biological effects, and finding mimetics of young systemic milieu continue to be an interesting and important research topic to be explored.

19.
J Atheroscler Thromb ; 29(7): 971-988, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35370224

RESUMO

Prognosis of heart failure remains poor, and it is urgent to find new therapies for this critical condition. Oxygen and metabolites are delivered through capillaries; therefore, they have critical roles in the maintenance of cardiac function. With aging or age-related disorders, capillary density is reduced in the heart, and the mechanisms involved in these processes were reported to suppress capillarization in this organ. Studies with rodents showed capillary rarefaction has causal roles for promoting pathologies in failing hearts. Drugs used as first-line therapies for heart failure were also shown to enhance the capillary network in the heart. Recently, the approach with senolysis is attracting enthusiasm in aging research. Genetic or pharmacological approaches concluded that the specific depletion of senescent cells, senolysis, led to reverse aging phenotype. Reagents mediating senolysis are described to be senolytics, and these compounds were shown to ameliorate cardiac dysfunction together with enhancement of capillarization in heart failure models. Studies indicate maintenance of the capillary network as critical for inhibition of pathologies in heart failure.


Assuntos
Capilares , Insuficiência Cardíaca , Envelhecimento/fisiologia , Capilares/metabolismo , Capilares/patologia , Vasos Coronários/patologia , Coração , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/patologia , Humanos
20.
iScience ; 25(11): 105424, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36388988

RESUMO

Brown adipose tissue (BAT) has critical roles in thermogenesis and systemic metabolism. Capillary rarefaction was reported to develop in BAT with dietary obesity, and previous studies showed that suppression of vascular endothelial growth factor A (VEGF-A) reduced capillary density in BAT, promoting the functional decline of this organ. Capillarization is regulated through the balance between angiogenesis and vasculogenesis on the one hand and apoptosis of endothelial cells (ECs) on the other; however, the role of EC apoptosis in BAT remained to be explored. In studies testing the role of boysenberry polyphenols (BoyP) in BAT, we found that BoyP decreased EC apoptosis, enhanced capillarization in BAT, and ameliorated dietary BAT dysfunction, which was associated with the upregulation of nicotinamide adenine dinucleotide-dependent protein deacetylase sirtuin 1 (SIRT-1) in ECs. Our studies suggest that EC SIRT-1 would be one of the potential targets of BoyP that contributes to BAT capillarization and function.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA