Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Circ Res ; 132(5): 601-624, 2023 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-36786216

RESUMO

BACKGROUND: Hypertension can lead to podocyte damage and subsequent apoptosis, eventually resulting in glomerulosclerosis. Although alleviating podocyte apoptosis has clinical significance for the treatment of hypertensive nephropathy, an effective therapeutic target has not yet been identified. The function of septin4, a proapoptotic protein and an important marker of organ damage, is regulated by post-translational modification. However, the exact role of septin4 in regulating podocyte apoptosis and its connection to hypertensive renal damage remains unclear. METHODS: We investigated the function and mechanism of septin4 in hypertensive nephropathy to discover a theoretical basis for targeted treatment. Mouse models including Rosa 26 (Gt(ROSA)26Sor)-SIRT2 (silent mating type information regulation 2 homolog-2)-Flag-TG (transgenic) (SIRT2-TG) mice SIRT2-knockout, and septin4-K174Q mutant mice, combined with proteomic and acetyl proteomics analysis, followed by multiple molecular biological methodologies, were used to demonstrate mechanisms of SIRT2-mediated deacetylation of septin4-K174 in hypertensive nephropathy. RESULTS: Using transgenic septin4-K174Q mutant mice treated with the antioxidant Tempol, we found that hyperacetylation of the K174 site of septin4 exacerbates Ang II (angiotensin II)- induced hypertensive renal injury resulting from oxidative stress. Proteomics and Western blotting assays indicated that septin4-K174Q activates the cleaved-PARP1 (poly [ADP-ribose] polymerase family, member 1)-cleaved-caspase3 pathway. In septin4-knockdown human renal podocytes, septin4-K174R, which mimics deacetylation at K174, rescues podocyte apoptosis induced by Ang II. Immunoprecipitation and mass spectrometry analyses identified SIRT2 as a deacetylase that interacts with the septin4 GTPase domain and deacetylates septin4-K174. In Sirt2-deficient mice and SIRT2-knockdown renal podocytes, septin4-K174 remains hyperacetylated and exacerbates hypertensive renal injury. By contrast, in Rosa26-Sirt2-Flag (SIRT2-TG) mice and SIRT2-knockdown renal podocytes reexpressing wild-type SIRT2, septin4-K174 is hypoacetylated and mitigates hypertensive renal injury. CONCLUSIONS: Septin4, when activated through acetylation of K174 (K174Q), promotes hypertensive renal injury. Septin4-K174R, which mimics deacetylation by SIRT2, inhibits the cleaved-PARP1-cleaved-caspase3 pathway. Septin4-K174R acts as a renal protective factor, mitigating Ang II-induced hypertensive renal injury. These findings indicate that septin4-K174 is a potential therapeutic target for the treatment of hypertensive renal injury.


Assuntos
Hipertensão Renal , Hipertensão , Animais , Humanos , Camundongos , Apoptose , Hipertensão Renal/genética , Rim/metabolismo , Camundongos Transgênicos , Proteômica , Sirtuína 2/genética , Sirtuína 2/metabolismo
2.
Cardiovasc Diabetol ; 23(1): 140, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664681

RESUMO

BACKGROUND: Diabetic vascular remodeling is the most important pathological basis of diabetic cardiovascular complications. The accumulation of advanced glycation end products (AGEs) caused by elevated blood glucose promotes the proliferation and migration of vascular smooth muscle cells (VSMCs), leading to arterial wall thickening and ultimately vascular remodeling. Therefore, the excessive proliferation and migration of VSMCs is considered as an important therapeutic target for vascular remodeling in diabetes mellitus. However, due to the lack of breakthrough in experiments, there is currently no effective treatment for the excessive proliferation and migration of VSMCs in diabetic patients. Bcl-2-associated athanogene 3 (BAG3) protein is a multifunctional protein highly expressed in skeletal muscle and myocardium. Previous research has confirmed that BAG3 can not only regulate cell survival and apoptosis, but also affect cell proliferation and migration. Since the excessive proliferation and migration of VSMCs is an important pathogenesis of vascular remodeling in diabetes, the role of BAG3 in the excessive proliferation and migration of VSMCs and its molecular mechanism deserve further investigation. METHODS: In this study, BAG3 gene was manipulated in smooth muscle to acquire SM22αCre; BAG3FL/FL mice and streptozotocin (STZ) was used to simulate diabetes. Expression of proteins and aortic thickness of mice were detected by immunofluorescence, ultrasound and hematoxylin-eosin (HE) staining. Using human aorta smooth muscle cell line (HASMC), cell viability was measured by CCK-8 and proliferation was measured by colony formation experiment. Migration was detected by transwell, scratch experiments and Phalloidin staining. Western Blot was used to detect protein expression and Co-Immunoprecipitation (Co-IP) was used to detect protein interaction. RESULTS: In diabetic vascular remodeling, AGEs could promote the interaction between BAG3 and signal transducer and activator of transcription 3 (STAT3), leading to the enhanced interaction between STAT3 and Janus kinase 2 (JAK2) and reduced interaction between STAT3 and extracellular signal-regulated kinase 1/2 (ERK1/2), resulting in accumulated p-STAT3(705) and reduced p-STAT3(727). Subsequently, the expression of matrix metallopeptidase 2 (MMP2) is upregulated, thus promoting the migration of VSMCs. CONCLUSIONS: BAG3 upregulates the expression of MMP2 by increasing p-STAT3(705) and decreasing p-STAT3(727) levels, thereby promoting vascular remodeling in diabetes. This provides a new orientation for the prevention and treatment of diabetic vascular remodeling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Reguladoras de Apoptose , Movimento Celular , Proliferação de Células , Músculo Liso Vascular , Miócitos de Músculo Liso , Fator de Transcrição STAT3 , Transdução de Sinais , Remodelação Vascular , Fator de Transcrição STAT3/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Animais , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Reguladoras de Apoptose/genética , Fosforilação , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/patologia , Angiopatias Diabéticas/fisiopatologia , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/genética , Masculino , Células Cultivadas , Camundongos Knockout , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Humanos , Camundongos Endogâmicos C57BL , Produtos Finais de Glicação Avançada/metabolismo
3.
FASEB J ; 37(8): e23110, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37490283

RESUMO

The ubiquitin-proteasome system is a crucial mechanism for regulating protein levels in cells, with substrate-specific E3 ubiquitin ligases serving as an integral component of this system. Among these ligases are SMAD-specific E3 ubiquitin-protein ligase 1 (SMURF1) and SMAD-specific E3 ubiquitin-protein ligase 2 (SMURF2), which belong to the neural precursor cell-expressed developmentally downregulated 4 (NEDD4) subfamily of Homologous to E6-AP COOH terminus (HECT)-type E3 ligases. As E3 ligases, SMURFs have critical functions in regulating the stability of multiple proteins, thereby maintaining physiological processes such as cell migration, proliferation, and apoptosis. The occurrence of many diseases is attributed to abnormal cell physiology and an imbalance in cell homeostasis. It is noteworthy that SMURFs play pivotal roles in disease progression, with the regulatory functions being complex and either facilitative or inhibitory. In this review, we elucidate the mechanisms by which SMURF1 and SMURF2 can regulate disease progression in non-cancerous diseases. These significant findings offer potential novel therapeutic targets for various diseases and new avenues for research on SMURF proteins.


Assuntos
Apoptose , Ubiquitina-Proteína Ligases , Humanos , Movimento Celular , Progressão da Doença , Ubiquitina
4.
Pharmacol Res ; : 107281, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38942341

RESUMO

Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.

5.
Basic Res Cardiol ; 118(1): 48, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37938421

RESUMO

Cardiovascular disease (CVD) is a major threat to human health, accounting for 46% of non-communicable disease deaths. Glycolysis is a conserved and rigorous biological process that breaks down glucose into pyruvate, and its primary function is to provide the body with the energy and intermediate products needed for life activities. The non-glycolytic actions of enzymes associated with the glycolytic pathway have long been found to be associated with the development of CVD, typically exemplified by metabolic remodeling in heart failure, which is a condition in which the heart exhibits a rapid adaptive response to hypoxic and hypoxic conditions, occurring early in the course of heart failure. It is mainly characterized by a decrease in oxidative phosphorylation and a rise in the glycolytic pathway, and the rise in glycolysis is considered a hallmark of metabolic remodeling. In addition to this, the glycolytic metabolic pathway is the main source of energy for cardiomyocytes during ischemia-reperfusion. Not only that, the auxiliary pathways of glycolysis, such as the polyol pathway, hexosamine pathway, and pentose phosphate pathway, are also closely related to CVD. Therefore, targeting glycolysis is very attractive for therapeutic intervention in CVD. However, the relationship between glycolytic pathway and CVD is very complex, and some preclinical studies have confirmed that targeting glycolysis does have a certain degree of efficacy, but its specific role in the development of CVD has yet to be explored. This article aims to summarize the current knowledge regarding the glycolytic pathway and its key enzymes (including hexokinase (HK), phosphoglucose isomerase (PGI), phosphofructokinase-1 (PFK1), aldolase (Aldolase), phosphoglycerate metatase (PGAM), enolase (ENO) pyruvate kinase (PKM) lactate dehydrogenase (LDH)) for their role in cardiovascular diseases (e.g., heart failure, myocardial infarction, atherosclerosis) and possible emerging therapeutic targets.


Assuntos
Doenças Cardiovasculares , Insuficiência Cardíaca , Humanos , Fosforilação Oxidativa , Aldeído Liases , Redes e Vias Metabólicas
6.
Cardiovasc Diabetol ; 22(1): 107, 2023 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-37149668

RESUMO

BACKGROUND: Endothelial injury caused by Type 2 diabetes mellitus (T2DM) is considered as a mainstay in the pathophysiology of diabetic vascular complications (DVCs). However, the molecular mechanism of T2DM-induced endothelial injury remains largely unknown. Here, we found that endothelial WW domain-containing E3 ubiquitin protein ligase 2 (WWP2) act as a novel regulator for T2DM-induced vascular endothelial injury through modulating ubiquitination and degradation of DEAD-box helicase 3 X-linked (DDX3X). METHODS: Single-cell transcriptome analysis was used to evaluate WWP2 expression in vascular endothelial cells of T2DM patients and healthy controls. Endothelial-specific Wwp2 knockout mice were used to investigate the effect of WWP2 on T2DM-induced vascular endothelial injury. In vitro loss- and gain-of-function studies were performed to assess the function of WWP2 on cell proliferation and apoptosis of human umbilical vein endothelial cells. The substrate protein of WWP2 was verified using mass spectrometry, coimmunoprecipitation assays and immunofluorescence assays. The mechanism of WWP2 regulation on substrate protein was investigated by pulse-chase assay and ubiquitination assay. RESULTS: The expression of WWP2 was significantly down-regulated in vascular endothelial cells during T2DM. Endothelial-specific Wwp2 knockout in mice significantly aggravated T2DM-induced vascular endothelial injury and vascular remodeling after endothelial injury. Our in vitro experiments showed that WWP2 protected against endothelial injury by promoting cell proliferation and inhibiting apoptosis in ECs. Mechanically, we found that WWP2 is down-regulated in high glucose and palmitic acid (HG/PA)-induced ECs due to c-Jun N-terminal kinase (JNK) activation, and uncovered that WWP2 suppresses HG/PA-induced endothelial injury by catalyzing K63-linked polyubiquitination of DDX3X and targeting it for proteasomal degradation. CONCLUSION: Our studies revealed the key role of endothelial WWP2 and the fundamental importance of the JNK-WWP2-DDX3X regulatory axis in T2DM-induced vascular endothelial injury, suggesting that WWP2 may serve as a new therapeutic target for DVCs.


Assuntos
Diabetes Mellitus Tipo 2 , Ubiquitina-Proteína Ligases , Humanos , Camundongos , Animais , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Baixo , Células Endoteliais/metabolismo , Diabetes Mellitus Tipo 2/complicações , Ubiquitinação , Camundongos Knockout , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo
7.
BMC Cancer ; 23(1): 526, 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37291499

RESUMO

NEDD4 family represent an important group of E3 ligases, which regulate various cellular pathways of cell proliferation, cell junction and inflammation. Emerging evidence suggested that NEDD4 family members participate in the initiation and development of tumor. In this study, we systematically investigated the molecular alterations as well as the clinical relevance regarding NEDD4 family genes in 33 cancer types. Finally, we found that NEDD4 members showed increased expression in pancreas cancer and decreased expression in thyroid cancer. NEDD4 E3 ligase family genes had an average mutation frequency in the range of 0-32.1%, of which HECW1 and HECW2 demonstrated relatively high mutation rate. Breast cancer harbors large amount of NEDD4 copy number amplification. NEDD4 family members interacted proteins were enriched in various pathways including p53, Akt, apoptosis and autophagy, which were confirmed by further western blot and flow cytometric analysis in A549 and H1299 lung cancer cells. In addition, expression of NEDD4 family genes were associated with survival of cancer patients. Our findings provide novel insight into the effect of NEDD4 E3 ligase genes on cancer progression and treatment in the future.


Assuntos
Neoplasias , Ubiquitina-Proteína Ligases , Humanos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Ubiquitina-Proteína Ligases Nedd4/genética , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Neoplasias/genética , Proteínas do Tecido Nervoso/genética
8.
J Cell Mol Med ; 25(14): 6470-6478, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34028177

RESUMO

At present, cardiovascular disease is one of the important factors of human death, and there are many kinds of proteins involved. Sirtuins family proteins are involved in various physiological and pathological activities of the human body. Among them, there are more and more studies on the relationship between sirtuin2 (SIRT2) protein and cardiovascular diseases. SIRT2 can effectively inhibit pathological cardiac hypertrophy. The effect of SIRT2 on ischaemia-reperfusion injury has different effects under different conditions. SIRT2 can reduce the level of reactive oxygen species (ROS), which may help to reduce the severity of diabetic cardiomyopathy. SIRT2 can affect a variety of cardiovascular diseases, energy metabolism and the ageing of cardiomyocytes, thereby affecting heart failure. SIRT2 also plays an important role in vascular disease. For endothelial cell damage used by oxidative stress, the role of SIRT2 is bidirectional, which is related to the degree of oxidative stress stimulation. When the degree of stimulation is small, SIRT2 plays a protective role, and when the degree of stimulation increases to a certain level, SIRT2 plays a negative role. In addition, SIRT2 is also involved in the remodelling of blood vessels and the repair of skin damage.


Assuntos
Doenças Cardiovasculares/genética , Estresse Oxidativo/genética , Traumatismo por Reperfusão/genética , Sirtuína 2/genética , Envelhecimento/genética , Envelhecimento/patologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Metabolismo Energético/genética , Humanos , Espécies Reativas de Oxigênio , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Pele/lesões , Pele/metabolismo
9.
J Cell Mol Med ; 24(8): 4600-4611, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32167680

RESUMO

Oxidative stress injury is involved in many cardiovascular diseases, like hypertension and myocardial infarction. Ubiquitination is a ubiquitous protein post-translational modification that controls a wide range of biological functions and plays a crucial role in maintaining the homeostasis of cells in physiology and disease. Many studies have shown that oxidative stress damage is inextricably linked to ubiquitination. We demonstrate that Smurf2, an E3 ubiquitinated ligase, is involved in HUVEC apoptosis induced by oxidative stress to alleviate H2 O2 -induced reactive oxygen species (ROS) production and the apoptosis of human umbilical vein endothelial cells (HUVECs). At the same time, we found that Smurf2 can bind the poly(ADP-ribose) polymerase-1(PARP1), and the interaction is enhanced under the stimulation of oxidative stress. We further study and prove that Smurf2 can promote PARP1 ubiquitination and degradation. Collectively, we demonstrate Smurf2 degradation of overactivated PARP1 by ubiquitin-proteasome pathway to protect HUVEC and alleviate oxidative stress injury.


Assuntos
Estresse Oxidativo/genética , Poli(ADP-Ribose) Polimerase-1/genética , Ubiquitina-Proteína Ligases/genética , Apoptose/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Ubiquitinação/genética , Água/química
10.
J Cell Mol Med ; 24(16): 9041-9054, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32627301

RESUMO

WWP2 is a HECT-type E3 ubiquitin ligase that regulates various physiological and pathological activities by binding to different substrates, but its function and regulatory mechanism in vascular smooth muscle cells (VSMCs) are still unknown. Here, we clarified the role of WWP2 in the regulation of SIRT1-STAT3 and the impact of this regulatory process in VSMCs. We demonstrated that WWP2 expression was significantly increased in angiotensin II-induced VSMCs model. Knockdown of WWP2 significantly inhibited angiotensin II-induced VSMCs proliferation, migration and phenotypic transformation, whereas overexpression of WWP2 had opposite effects. In vivo experiments showed that vascular smooth muscle-specific WWP2 knockout mice significantly relieved angiotensin II-induced hypertensive angiopathy. Mechanistically, mass spectrometry and co-immunoprecipitation assays identified that WWP2 is a novel interacting protein of SIRT1 and STAT3. Moreover, WWP2 formed a complex with SIRT1-STAT3, inhibiting the interaction between SIRT1 and STAT3, then reducing the inhibitory effect of SIRT1 on STAT3, ensuing promoting STAT3-K685 acetylation and STAT3-Y705 phosphorylation in angiotensin II-induced VSMCs and mice. In conclusion, WWP2 modulates hypertensive angiopathy by regulating SIRT1-STAT3 and WWP2 suppression in VSMCs can alleviate hypertensive angiopathy vitro and vivo. These findings provide new insights into the treatment of hypertensive vascular diseases.


Assuntos
Hipertensão/metabolismo , Fosforilação/fisiologia , Fator de Transcrição STAT3/metabolismo , Sirtuína 1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Acetilação/efeitos dos fármacos , Angiotensina II/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Hipertensão/induzido quimicamente , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos
11.
Biochem Biophys Res Commun ; 525(2): 272-279, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32085901

RESUMO

Vascular smooth muscle cells (VSMCs) proliferation and migration play a fundamental role during the process of hypertensive angiopathy. Angiotensin-II (Ang-II) is one of the robust phenotype-modulating agents, which changes VSMCs to efficiently proliferate and migrate. The mechanism of the proliferation and migration is not well understood yet. Septin4, as a member of GTP binding protein family, is widely expressed in the eukaryotic cells and considered to be an essential component of the cytoskeleton which is involved in many important physiological processes. We approved that Septin4 expression was upregulated in mouse aorta by continuous infusion of Ang-II and in cultured VSMCs treated with Ang-II. Overexpression of Septin4 led to lower level of autophagy and decreased capacity of proliferation and migration. In order to identify the mechanism by which Septin4 interacts with these processes, we blocked autophagy by chloroquine (CQ). After inhibiting the autophagy, the ability of proliferation and migration was further restrained in the Septin4 overexpression VSMCs. In conclusion, our results indicated that during the process of VSMCs proliferation and migration induced by Ang-II, Septin4 modulated autophagy and thus regulated the activity of proliferation and migration.


Assuntos
Angiotensina II/farmacologia , Aorta/citologia , Músculo Liso Vascular/citologia , Septinas/fisiologia , Animais , Autofagia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Camundongos
12.
Cell Physiol Biochem ; 46(3): 1009-1018, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29669326

RESUMO

BACKGROUND/AIMS: Many clinical and experimental studies have shown that treatment with statins could prevent myocardial hypertrophy and remodeling induced by hypertension and myocardial infarction. But the molecular mechanism was not clear. We aimed to investigate the beneficial effects of atorvastatin on hypertension-induced myocardial hypertrophy and remodeling in spontaneously hypertensive rats (SHR) with the hope of revealing other potential mechanisms or target pathways to interpret the pleiotropic effects of atorvastatin on myocardial hypertrophy. METHODS: The male and age-matched animals were randomly divided into three groups: control group (8 WKY), SHR (8 rats) and intervention group (8 SHR). The SHR in intervention group were administered by oral gavage with atorvastatin (suspension in distilled water, 10 mg/Kg once a day) for 6 weeks, and the other two groups were administered by gavage with equal quantity distilled water. Blood pressure of rats was measured every weeks using a standard tail cuff sphygmomanometer. Left ventricular (LV) dimensions were measured from short-axis views of LV under M-mode tracings using Doppler echocardiograph. Cardiomyocyte apoptosis was assessed by the TUNEL assay. The protein expression of C/EBPß, PGC-1α and UCP3 were detected by immunohistochemistry or Western blot analysis. RESULTS: At the age of 16 weeks, the mean arterial pressure of rats in three groups were 103.6±6.1, 151.8±12.5 and 159.1±6.2 mmHg respectively, and there wasn't statistically significant difference between the SHR and intervention groups. Staining with Masson's trichrome demonstrated that the increased interstitial fibrosis of LV and ventricular remodeling in the SHR group were attenuated by atorvastatin treatment. Echocardiography examination exhibited that SHR with atorvastatin treatment showed an LV wall thickness that was obviously lower than that of water-treated SHR. In hypertrophic myocardium, accompanied by increasing C/EBPß expression and the percentage of TUNEL-positive cells, the expression of Bcl-2/Bax ratio, PGC-1α and UCP3 were reduced, all of which could be abrogated by treatment with atorvastatin for 6 weeks. CONCLUSION: This study further confirmed that atorvastatin could attenuate myocardial hypertrophy and remodeling in SHR by inhibiting apoptosis and reversing changes in mitochondrial metabolism. The C/EBPß/PGC-1α/UCP3 signaling pathway might also be important for elucidating the beneficial pleiotropic effects of atorvastatin on myocardial hypertrophy.


Assuntos
Atorvastatina/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/patologia , Transdução de Sinais/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Atorvastatina/uso terapêutico , Fator Natriurético Atrial/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Ecocardiografia , Hipertensão/complicações , Hipertrofia/diagnóstico por imagem , Hipertrofia/tratamento farmacológico , Hipertrofia/etiologia , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Encefálico/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Proteína Desacopladora 3/metabolismo , Regulação para Cima/efeitos dos fármacos
13.
Biochem Biophys Res Commun ; 496(2): 621-627, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29366480

RESUMO

Oxidative stress induced vascular endothelial cell injure is one of the key and initial event in the development of atherosclerosis. Septin4, as a member of GTP binding protein family, is widely expressed in the eukaryotic cells and considered to be an essential component of the cytoskeleton which is involved in many important physiological processes. However, whether Septin4 is involved in cardiovascular diseases, such as oxidative stress inducted endothelial cell injury still unclear. PARP1 as a DNA repair enzyme can be activated by identifying DNA damaged fragments, which consumes high levels of energy and leads to vascular endothelial cell apoptosis. Here, our results first found that Septin4 is involved in oxidative stress induced endothelial cell ROS production and apoptosis through knock-down and over-expression Septin4 approaches. Furthermore, to explore how Septin4 is involved in oxidative stress induced endothelial cells injure, we first identified that Septin4 is a novel PARP1 interacting protein and the interaction is enhanced under oxidative stress. In conclusions, our founding indicates that Septin4 is a novel essential factor involved in oxidative stress induced vascular endothelial cell injury by interacting with apoptosis-related protein PARP1.


Assuntos
Células Endoteliais/metabolismo , Estresse Oxidativo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Mapas de Interação de Proteínas , Septinas/metabolismo , Apoptose , Células Endoteliais/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo
14.
Cell Physiol Biochem ; 44(1): 215-228, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29131001

RESUMO

BACKGROUND/AIMS: Abnormal proliferation of vascular smooth muscle cells (VSMCs) is a hallmark of vascular lesions, such as atherosclerosis and restenosis. PDGF-ßß, an isoform of PDGF (platelet-derived growth factor), has been demonstrated to induce proliferation and migration of VSMCs. Atorvastatin calcium, a selective inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, has favorable protective effects on VSMCs. This study examined the effects of atorvastatin calcium on the proliferation and migration of PDGF-ßß-treated VSMCs, as well as its underlying mechanisms. METHODS: MTT assays, Edu imaging, cell cycle analysis, wound healing assays, transwell migration assays, and western blot analysis were performed. RESULTS: Atorvastatin calcium significantly inhibited cell proliferation, DNA synthesis and cell migration of PDGF-ßß-treated VSMCs. We demonstrated that atorvastatin calcium induced cell cycle arrest in the G0/G1 phase in response to PDGF-ßß stimulation and decreased the expression of G0/G1-specific regulatory proteins, including proliferating cell nuclear antigen (PCNA), CDK2, cyclin D1, cyclin E and CDK4 in PDGF-ßß-treated VSMCs. Moreover, pretreatment with atorvastatin calcium inhibited the PDGF-ßß-treated phosphorylation of PDGFRß and Akt, whereas atorvastatin calcium did not affect the phosphorylation of PLC-γ1 or (ERK) 1/2. CONCLUSION: Our data suggested that atorvastatin calcium inhibited abnormal proliferation and migration of VSMCs through G0/G1 cell cycle arrest and suppression of the PDGFRß-Akt signaling cascade.


Assuntos
Atorvastatina/toxicidade , Proliferação de Células/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Becaplermina , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ciclina D1/metabolismo , Ciclina E/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
15.
Biochem Biophys Res Commun ; 486(2): 423-430, 2017 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-28315329

RESUMO

Previous evidences suggested that atorvastatin not only reduced blood lipids but also reduced myocardial hypertrophy and remodeling. And it was reported that C/EBPß (CCAAT/enhancer-binding protein ß) played a pivotal role both in the physiologic and pathological cardiac hypertrophy. However, it has not been reported before whether this signaling pathway of C/EBPß participates in protective effect of atorvastatin for hypertrophy cardiomyocytes. In present study, We found that overexpression of C/EBPß significantly abrogated the effect of atorvastatin on increasing Bcl-2/Bax and PGC-1α while the early and late apoptosis rate increased and mitochondrial membrane potential (MMP) was reduced. In conclusion, we further identified the protective effect of atorvastatin on hypertrophic cardiomyocytes induced by Angiotensin II by rescuing the MMP and inhibiting apoptosis, which might be at least partly attributed to down-regulation of C/EBPß. And C/EBPß might be a new target to rescue mitochondrion function and apoptosis in pathological cardiac hypertrophy.


Assuntos
Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina/farmacologia , Anticolesterolemiantes/farmacologia , Atorvastatina/farmacologia , Proteína beta Intensificadora de Ligação a CCAAT/genética , Miócitos Cardíacos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Transdução de Sinais , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
16.
BMC Psychiatry ; 17(1): 80, 2017 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-28241814

RESUMO

BACKGROUND: To investigate the association between CHADS2 score, depressive symptoms, and quality of life in a large general population from China. METHODS: A cross-sectional study of 11,956 permanent residents of Liaoning Province in China ≥ 35 years of age was conducted between January and August 2013 (response rate 85.3%). All participants completed a questionnaire, had a physical examination, and underwent blood examination. Depressive symptoms were assessed with the Patient Health Questionnaire-9 (PHQ-9), while the quality of life (QoL) was measured using the World Health Organization Quality of Life Brief Scale (WHOQOL-BREF). RESULTS: With increasing CHADS2 score, the prevalence of depressive symptoms increased from 4.9 to 27.8% (P < 0.001), and all scores of WHOQOL-BREF decreased significantly (all Ps < 0.001). After adjusting for confounding risk factors, subjects with CHADS2 score ≥ 3 had higher risk of depressive symptoms than those with CHADS2 score = 0 (all Ps < 0.05). Also, CHADS2 score was negatively associated with all scores of WHOQOL-BREF (all Ps < 0.001). Furthermore, subjects with any item in CHADS2 had higher prevalence of depressive symptoms (all Ps < 0.001). Heart failure and stroke remained independently associated with depressive symptoms after adjusting for confounding risk factors and other items (Ps < 0.001), while heart failure, age ≥ 75 years, diabetes mellitus, and stroke were all independently negatively associated with the total score of WHOQOL-BREF (all Ps < 0.05). CONCLUSIONS: The CHADS2 score is significantly associated with depressive symptoms and impaired quality of life in the general population.


Assuntos
Depressão/diagnóstico , Qualidade de Vida/psicologia , Inquéritos e Questionários , Adulto , Idoso , China , Estudos Transversais , Depressão/psicologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco
17.
Psychol Health Med ; 22(3): 303-309, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27687016

RESUMO

Till now, no evidence illustrates the prevalence and predictors of metabolically healthy obesity (MHO) in rural areas of China. The objective of this study was, firstly, to examine the prevalence of MHO in rural areas of China, and identify contributing determinants of MHO, Secondly, to comprehensively investigate to the different characteristics between MHO and metabolically unhealthy obesity (MUO). We conducted a population-based cross-sectional study of 2037 participants with obesity in rural Liaoning Province during 2012-2013. Obesity was defined as BMI ≥28 kg/m2 and metabolically healthy was defined as not having the metabolic syndrome. The prevalence of MHO was 23.1%, and significantly decreased with advancing age in female group. However there was no significant tendency with advancing age in male group. Independent determinant factors for MHO were age <55 years (odds ratio [OR] 1.659; p = .001), non-current smoking (OR 1.397; p = .038), pre-menopause (OR 1.648; p = .030) and non-hyperuricemia (OR 2.317; p < .001), whereas race, gender, diet score, current drinking, marriage, sleep duration, hyperhomocysteinemia, levels of physical activity, annual income and educational status were not significant contributors. In conclusion, we found that age <55 years, non-current smoking, pre-menopause and non-hyperuricemia were identified as independent determinant factors for MHO in this population.


Assuntos
Obesidade Metabolicamente Benigna/epidemiologia , Obesidade , População Rural , Adulto , Idoso , Consumo de Bebidas Alcoólicas , China/epidemiologia , Estudos Transversais , Dieta , Exercício Físico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Prevalência
18.
Int J Mol Sci ; 18(2)2017 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-28218663

RESUMO

We aimed to investigate the effect of advanced glycation end products (AGEs) on the proliferation and migration ability of human umbilical vein endothelial cells (HUVECs). Cell proliferation was detected by methyl thiazolyl tetrazolium (MTT) assay, real-time cell analyzer and 5-Ethynyl-2'-deoxyuridine (EdU) staining. Cell migration was detected by wound-healing and transwell assay. AGEs significantly inhibited the proliferation and migration of HUVECs in a time-and dose-dependent way. Western blotting revealed that AGEs dramatically increased the expression of microtubule-associated protein 1 light chain 3 (LC3) II/I and p62. Immunofluorescence of p62 and acridine orange staining revealed that AGEs significantly increased the expression of p62 and the accumulation of autophagic vacuoles, respectively. Chloroquine (CQ) could further promote the expression of LC3 II/I and p62, increase the accumulation of autophagic vacuoles and promote cell injury induced by AGEs. In addition, AGEs reduced cathepsin D (CTSD) expression in a time-dependent way. Overexpression of wild-type CTSD significantly decreased the ratio of LC 3 II/I as well as p62 accumulation induced by AGEs, but overexpression of catalytically inactive mutant CTSD had no such effects. Only overexpression of wild-type CTSD could restore the proliferation of HUVECs inhibited by AGEs. However, overexpression of both wild-type CTSD and catalytically inactive mutant CTSD could promote the migration of HUVECs inhibited by AGEs. Collectively, our study found that AGEs inhibited the proliferation and migration in HUVECs and promoted autophagic flux, which in turn played a protective role against AGEs-induced cell injury. CTSD, in need of its catalytic activity, may promote proliferation in AGEs-treated HUVECs independent of the autophagy-lysosome pathway. Meanwhile, CTSD could improve the migration of AGEs-treated HUVECs regardless of its enzymatic activity.


Assuntos
Catepsina D/antagonistas & inibidores , Produtos Finais de Glicação Avançada/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Autofagia/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos
19.
Heart Lung Circ ; 26(3): 251-257, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27555052

RESUMO

BACKGROUND: Obesity has been found to be a predictor of left ventricular hypertrophy (LVH). However, studies which divide obesity into metabolically healthy obesity (MHO) and metabolically unhealthy obesity (MUO) to study the effect of obesity on LVH have been rare. The present study aims to make clear the effects of various obese phenotypes and metabolic abnormalities on LVH. METHODS: A total of 10,804 participants were involved in this cross-sectional study. "Obesity" and "metabolically healthy" were defined as BMI ≥ 25kg/m2 and having none of the metabolic factors respectively. RESULTS: It was found that metabolically unhealthy non-obesity (MUNO) (OR, 2.675; 95%CI, 1.603-4.462, P < 0.001) and MUO (OR, 9.067; 95%CI, 5.474-15.020, P<0.001) were significantly associated with LVH, while it went in reverse for MHO (OR, 1.968; 95%CI, 0.560-6.920, P=0.291), after adjustment for age, race, gender, educational status, physical activity, annual income, current smoking status, current drinking status, sleep duration and BMI. And after further adjustment for metabolic abnormalities, MUNO (OR, 0.567; 95%CI, 0.316-1.018, P=0.794) and MUO (OR, 0.632; 95%CI, 0.342-1.166, P=0.142) tended not to be associated with LVH any longer. However, among the five metabolic components of metabolic abnormalities, high blood pressure (OR, 4.358; 95%CI, 3.266-5.815, P<0.001) and high waist circumference (OR, 1.530; 95%CI, 1.139-2.054, P=0.005) were significantly associated with LVH. CONCLUSIONS: Metabolic abnormalities, but not MHO, were significantly associated with LVH. In addition, metabolic abnormalities were probable to mediate the connection between MUNO/MUO and LVH.


Assuntos
Ventrículos do Coração , Hipertrofia Ventricular Esquerda , Obesidade , Adulto , Idoso , Estudos Transversais , Feminino , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia
20.
BMC Cardiovasc Disord ; 16: 15, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26772538

RESUMO

BACKGROUND: Population-based investigations studying the association between snoring and left ventricular hypertrophy (LVH) are lacking. Therefore, our study aims to investigate whether snoring is significantly associated with LVH, and to make clear the effect of varying degrees of snoring intensity on LVH. METHODS: A total of 10,139 participants were involved in this cross-sectional study. Snoring status and snoring intensity were evaluated by a structured questionnaire. LVH was defined as left ventricular mass index ≥ 51 g/m(2.7) for both men and women. RESULTS: The total prevalence of LVH was 10.0%. the prevalence increased significantly in snorers according to snoring intensity, including low (10.3%), normal (13.1%), strong (14.7%) and very strong (16.7%). After adjustment for age, race, gender, educational status, physical activity, annual income, current smoking status, current drinking status, sleep duration, hypertension, body mass index, waist circumference, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglyceride, total cholesterol and fasting plasma glucose, snoring (OR, 1.371; 95% CI, 1.147-1.637, P < 0.001) was significantly associated with LVH. In addition, among the four kinds of intensity of snoring, normal (OR, 1.436; 95% CI, 1.126-1.832, P = 0.004), strong (OR, 1.462; 95% CI, 1.124-1.902, P < 0.001) and very strong (OR, 1.813; 95% CI, 1.273-2.684, P < 0.001), rather than low (OR, 1.094; 95% CI, 0.834-1.434, P = 0.518) were significantly associated with LVH. CONCLUSIONS: Snoring is independently associated with LVH. What's more, with the rise in snoring intensity, snoring will exert an increasing effect on LVH.


Assuntos
Hiperlipidemias/epidemiologia , Hipertrofia Ventricular Esquerda/epidemiologia , Sobrepeso/epidemiologia , Fumar/epidemiologia , Ronco/epidemiologia , Adulto , Índice de Massa Corporal , China/epidemiologia , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Estudos Transversais , Ecocardiografia , Feminino , Humanos , Hipertensão/epidemiologia , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prevalência , Fatores de Risco , Índice de Gravidade de Doença , Fatores Sexuais , Inquéritos e Questionários , Triglicerídeos/sangue , Circunferência da Cintura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA