Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Front Physiol ; 14: 1106662, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36846332

RESUMO

A physiological increase in cardiac workload results in adaptive cardiac remodeling, characterized by increased oxidative metabolism and improvements in cardiac performance. Insulin-like growth factor-1 (IGF-1) has been identified as a critical regulator of physiological cardiac growth, but its precise role in cardiometabolic adaptations to physiological stress remains unresolved. Mitochondrial calcium (Ca2+) handling has been proposed to be required for sustaining key mitochondrial dehydrogenase activity and energy production during increased workload conditions, thus ensuring the adaptive cardiac response. We hypothesized that IGF-1 enhances mitochondrial energy production through a Ca2+-dependent mechanism to ensure adaptive cardiomyocyte growth. We found that stimulation with IGF-1 resulted in increased mitochondrial Ca2+ uptake in neonatal rat ventricular myocytes and human embryonic stem cell-derived cardiomyocytes, estimated by fluorescence microscopy and indirectly by a reduction in the pyruvate dehydrogenase phosphorylation. We showed that IGF-1 modulated the expression of mitochondrial Ca2+ uniporter (MCU) complex subunits and increased the mitochondrial membrane potential; consistent with higher MCU-mediated Ca2+ transport. Finally, we showed that IGF-1 improved mitochondrial respiration through a mechanism dependent on MCU-mediated Ca2+ transport. In conclusion, IGF-1-induced mitochondrial Ca2+ uptake is required to boost oxidative metabolism during cardiomyocyte adaptive growth.

2.
Int J Cardiol ; 356: 88-96, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35395283

RESUMO

Cardiac fibrosis is a key feature of hypertensive cardiac remodeling. In response to microenvironmental stimuli, phenotypic and functional changes in macrophages are considered important determinants of cardiac fibrosis attenuation. VO-OHpic, a phosphatase and tension homolog of chromosome 10 (PTEN) inhibitor, has been demonstrated to be cardioprotective in cardiac remodeling. However, whether VO-OHpic can improve cardiac fibrosis and macrophage polarization remains elusive. The interaction between VO-OHpic and the macrophage phenotype to attenuate cardiac fibrosis was studied in both spontaneously hypertensive rats in vivo and an Ang II-induced hypertension model in vitro. In vitro experiments showed that VO-OHpic promoted M2 macrophage polarization and markedly inhibited proinflammatory M1 macrophages, while VO-OHpic treatment of protein kinase B (AKT)-knockdown/LY294002 (a PI3K inhibitor) macrophages exerted a reduced effect. In a coculture system, culturing cardiac fibroblasts with VO-OHpic-treated macrophages led to significant suppression of proliferation, fibrotic marker expression, and transforming growth factor (TGF)-ß and Smad 2/3 protein expression. Taken together, VO-OHpic mediated a fibro-protective effect and increased M2 macrophage polarization via the phosphatidylinositol 3-kinase (PI3K)/AKT/TGF-ß/Smad2/3 pathway.


Assuntos
Fosfatidilinositol 3-Quinase , Proteínas Proto-Oncogênicas c-akt , Animais , Fibrose , Humanos , Macrófagos/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinase/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Remodelação Ventricular
3.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166290, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34662704

RESUMO

Hepatic fibrosis is characterized by excessive extracellular matrix deposition and ductular reactions, manifested as the expansion of hepatic progenitor cells (HPCs). We previously reported that the Y-box binding protein 1 (YB-1) in HPCs is involved in chronic liver injury. In this study, we constructed YB-1f/f Foxl1-Cre mice and investigated the role of YB-1 in HPC expansion in murine choline-deficient, ethionine-supplemented (CDE), and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) models. Liver injury and fibrosis were measured using hematoxylin and eosin (HE), Masson, and Sirius Red staining. HPC proliferation was detected using EdU and immunofluorescence (IF). Autophagic flow was measured by mCherry-GFP-LC3B staining and transmission electron microscopy (TEM). YB-1 expression was measured by immunofluorescence and western blotting. CUT & Tag analysis, chromatin immunoprecipitation, and RT-PCR were performed to explore the regulation of autophagy-related protein 7 (Atg7) transcription by YB-1. Our results indicated that liver injury was accompanied by high expression of YB-1, proliferative HPCs, and activated autophagy in the CDE and DDC models. YB-1f/f Cre+/- mice displayed less liver injury and fibrosis than YB-1f/f Cre-/- mice in the CDE and DDC models. YB-1 promoted proliferation and autophagy of HPCs in vitro and in vivo. Transforming growth factor-ß (TGF-ß) induced YB-1 nuclear translocation and facilitated the proliferation and autophagy of HPCs. YB-1 nuclear translocation promoted the transcription of Atg7, which is essential for TGF-ß/YB-1 mediated HPCs expansion in vitro and in vivo. In summary, YB-1 nuclear translocation induced by TGF-ß in HPCs promotes the proliferation and autophagy of HPCs and Atg7 participates in YB-1-mediated HPC-expansion and liver fibrosis.


Assuntos
Proteína 7 Relacionada à Autofagia/genética , Doença Hepática Induzida por Substâncias e Drogas/genética , Cirrose Hepática/genética , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/genética , Animais , Autofagia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/patologia , Deficiência de Colina/induzido quimicamente , Deficiência de Colina/genética , Deficiência de Colina/patologia , Modelos Animais de Doenças , Etionina/toxicidade , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Piridinas/toxicidade , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(11): 166510, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35926755

RESUMO

Previously, we reported that the nuclear translocation of Y-box binding protein 1 (YB-1) is induced by transforming growth factor-ß (TGF-ß) and promotes hepatic progenitor cells (HPCs) expansion. Here, we explored the mechanisms underlying YB-1 translocation and the impact of YB-1 on the epithelial-mesenchymal transition (EMT) in HPCs. YB-1flox/floxcre+/- (YB-1f/fcre+/-) mice and YB-1f/fcre-/- mice were fed with a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) or a choline-deficient, ethionine-supplemented (CDE) diet. Liver injury and fibrosis were assessed by performing hematoxylin and eosin (HE) and Masson staining. The expression of collagen and EMT-related markers (E-cadherin, N-cadherin, and Snail) was detected by reverse transcription-polymerase chain reaction (RT-PCR), western blotting, and immunofluorescence analyses. Protein kinase B (AKT) expression in HPCs was silenced via RNA interference. Nuclear YB-1 expression in HPCs was detected via western blotting and immunofluorescence analyses. HPC proliferation was detected by immunofluorescence. Our results indicate that YB-1 transcriptionally regulated the biological behavior of HPCs. HPC-specific YB-1 knockout alleviated liver fibrosis in mice fed with DDC or CDE diet. YB-1 nuclear translocation promoted matrix metallopeptidase 9 transcription. YB-1 depletion in HPCs significantly dampened the EMT and inhibited AKT phosphorylation in vitro and in vivo. AKT knockdown compromised TGF-ß-induced YB-1 nuclear translocation, thereby inhibiting the EMT and HPC proliferation. EMT and AKT were highly activated in HPCs in cirrhotic livers. Collectively, our findings indicate that the loss of YB-1 suppressed EMT in HPCs and alleviated liver fibrosis in mice, and that AKT was essential for TGF-ß-induced YB-1 nuclear translocation and HPC proliferation.


Assuntos
Transição Epitelial-Mesenquimal , Proteínas Proto-Oncogênicas c-akt , Animais , Caderinas/metabolismo , Colina/metabolismo , Colágeno/metabolismo , Amarelo de Eosina-(YS)/metabolismo , Etionina/metabolismo , Hematoxilina/metabolismo , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Metaloproteases/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Fatores de Transcrição , Fator de Crescimento Transformador beta/metabolismo , Fatores de Crescimento Transformadores/metabolismo
5.
Aging (Albany NY) ; 12(8): 7380-7396, 2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32327611

RESUMO

Cardiac fibrosis is a primary phenotype of cardiac remodeling that contributes to cardiac dysfunction and heart failure. The expansion and activation of CD4+ T cells in the heart has been identified to facilitate pathological cardiac remodeling and dysfunction; however, the underlying mechanisms remained not well clarified. Herein, we found that exosomes derived from activated CD4+ T cells (CD4-activated Exos) evoked pro-fibrotic effects of cardiac fibroblasts, and their delivery into the heart aggravated cardiac fibrosis and dysfunction post-infarction. Mechanistically, miR-142-3p that was enriched in CD4-activated Exos recapitulated the pro-fibrotic effects of CD4-activated Exos in cardiac fibroblasts, and vice versa. Furthermore, miR-142-3p directly targeted and inhibited the expression of Adenomatous Polyposis Coli (APC), a negative WNT signaling pathway regulator, contributing to the activation of WNT signaling pathway and cardiac fibroblast activation. Thus, CD4-activated Exos promote post-ischemic cardiac fibrosis through exosomal miR-142-3p-WNT signaling cascade-mediated activation of myofibroblasts. Targeting miR-142-3p in CD4-activated Exos may hold promise for treating cardiac remodeling post-MI.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Regulação da Expressão Gênica , MicroRNAs/genética , Infarto do Miocárdio/genética , Miofibroblastos/metabolismo , Linfócitos T/metabolismo , Remodelação Ventricular , Animais , Modelos Animais de Doenças , Exossomos , Fibroblastos/metabolismo , Fibroblastos/patologia , MicroRNAs/biossíntese , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , RNA/genética , Via de Sinalização Wnt
6.
Atherosclerosis ; 284: 1-10, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30856513

RESUMO

BACKGROUND AND AIMS: Cigarette smoking is an established risk factor for atherosclerosis. Nicotine, the major constituent of cigarettes, mediates the phenotype switching of vascular smooth muscle cells (VSMCs) and contributes to atherogenesis. Recent studies show that autophagy regulates atherogenesis via several pathways. The aim of this study is to determine whether nicotine regulates autophagy and subsequently mediates the phenotypic transition of VSMCs. METHODS AND RESULTS: Oil Red O and HE staining of aortic sections of ApoE-/- mice showed that nicotine promoted atherosclerosis, and in situ expression of α-SMA indicated the involvement of VSMCs. Western blotting documented that nicotine induced the aorta autophagy. Cultured VSMCs treated with nicotine resulted in the increase of LC3 II-to-LC3 I ratio and the decrease of P62, along with GFP-LC3 puncta assay and transmission electron microscopy, further reflecting nicotine-induced autophagy. In addition, Western blotting and quantitative real-time PCR showed that VSMCs exposed to nicotine underwent changes in the expression of differentiation markers (α-SMA, SM22α and osteopontin), confirming the role of nicotine in VSMC differentiation. Transwell migration and scratch assays demonstrated that nicotine increased the migratory capacity of VSMCs. Finally, nicotine also increased the levels of reactive oxygen species (ROS), as measured by DCFH-DA staining. After respectively inhibiting autophagy (3-MA), oxidative stress (NAC), NF-κB activity (BAY 11-7082, si-p65) and nicotinic acetylcholine receptors (nAChRs, hexamethonium), nicotine-induced autophagy and VSMC phenotype switching were reversed. CONCLUSIONS: Nicotine-induced autophagy promotes the phenotype switching of VSMCs and accelerates atherosclerosis, which is partly mediated by the nAChRs/ROS/NF-κB signaling pathway.


Assuntos
Aterosclerose/etiologia , Autofagia/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/diagnóstico por imagem , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , NF-kappa B/fisiologia , Nicotina/farmacologia , Espécies Reativas de Oxigênio , Receptores Nicotínicos/efeitos dos fármacos , Receptores Nicotínicos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Células Cultivadas , Camundongos , Fatores de Tempo
7.
Theranostics ; 9(23): 6901-6919, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31660076

RESUMO

Rationale: During the development of atherosclerosis, macrophages secrete exosomes that regulate vascular smooth muscle cells (VSMCs); however, whether nicotine, a major constituent of cigarettes, can modulate this communication in the context of atherogenesis remains to be further studied. In this study, we hypothesized that nicotine induces macrophages to secrete atherogenic exosomes containing microRNAs (miRNAs) to mediate cell-to-cell crosstalk and encourage proatherogenic phenotypes of VSMCs. Methods: In an in vivo study, nicotine was administered subcutaneously to 8-week-old male ApoE-/- mice fed a high-fat diet (HFD) for 12 weeks. Oil red O and hematoxylin and eosin (HE) were used to stain atherosclerotic lesions. Lesion macrophages, VSMCs and exosomes were stained for CD68, α-smooth muscle actin (α-SMA) and CD9, and plaque exosomes were observed by transmission electron microscopy (TEM). Exosomes derived from control macrophages (M-Exos) and from nicotine-treated macrophages (NM-Exos) were isolated by ultracentrifugation, purified by sucrose density gradient centrifugation and characterized based on specific morphology and surface markers. The IVIS® Spectrum in vivo imaging system showed the biodistribution of NM-Exos and M-Exos in circulation. Chitosan hydrogel-incorporated exosomes were applied to simulate exosome secretion in situ. Scratch wound assay, transwell assay and EdU staining were conducted to assess the effects of NM-Exos on the migration and proliferation of mouse VSMCs. RNA-seq was performed to determine the miRNA profiles of M-Exos and NM-Exos. Quantitative real-time PCR (qRT-PCR) analysis was conducted to detect the expression levels of miRNAs and mRNAs. The roles of the candidate miRNA and its target gene were assessed using specific RNA inhibitors, siRNAs and miRNA mimics. Western blotting was used to detect candidate protein expression levels. A dual-luciferase reporting system was utilized to confirm the binding of a specific miRNA to its target gene. Results: Nicotine induced atherosclerotic lesion progression and resulted in plaque exosome retention in vivo. The biodistribution of NM-Exos showed that plaque-resident exosomes might be secreted in situ. VSMCs cocultured in vitro with nicotine-stimulated macrophages presented an increased capacity for migration and proliferation, which was exosome-dependent. In addition, isolated NM-Exos helped promote VSMC migration and proliferation. miRNA profiling showed that miR-21-3p was enriched in NM-Exos, and this miRNA was shown to play a key role in regulating NM-Exos-induced effects by directly targeting phosphatase and tension homologue (PTEN). Conclusion: Exosomal miR-21-3p from nicotine-treated macrophages may accelerate the development of atherosclerosis by increasing VSMC migration and proliferation through its target PTEN.


Assuntos
Aterosclerose/genética , Exossomos/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Nicotina/toxicidade , PTEN Fosfo-Hidrolase/metabolismo , Animais , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Movimento Celular , Proliferação de Células , Progressão da Doença , Exossomos/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Miócitos de Músculo Liso/metabolismo , PTEN Fosfo-Hidrolase/genética
8.
Mol Immunol ; 101: 74-79, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29886399

RESUMO

CD8+ T cells accumulate in the necrotic myocardium of acute myocardial infarction (AMI). It is unclear whether CD8+CD28+ T cells, a specific subset of CD8+ T cells, contribute to myocardial injury. In this study, 92 consecutive patients with AMI and 28 healthy control subjects were enrolled. The frequency of CD8+CD28+ T cells in peripheral blood samples was assayed by flow cytometry. Plasma cardiac troponin I (TNI) and left ventricular ejection fraction (LVEF) were determined. Long-term prognosis of the patients was evaluated by major adverse cardiac and cerebrovascular events (MACCE) over a 12-month follow-up period. Our findings indicated that patients with AMI who presented with high numbers of CD8+CD28+ T cells had an increased infarction size and aggravated ventricular function. We proposed that cytotoxic CD8+CD28+ T cell-mediated myocardial necrosis may act as a novel and alternative pathway of AMI.


Assuntos
Antígenos CD28/metabolismo , Linfócitos T CD8-Positivos/imunologia , Infarto do Miocárdio/imunologia , Miócitos Cardíacos/patologia , Idoso , Feminino , Humanos , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Modelos Imunológicos , Infarto do Miocárdio/fisiopatologia , Prognóstico , Fatores de Risco , Volume Sistólico , Troponina I/metabolismo
9.
J Thorac Dis ; 10(3): 1815-1824, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29707336

RESUMO

BACKGROUND: Type A acute aortic dissection (A-AAD), involving the ascending aorta, is a life-threatening disease. To detect A-AAD early and rapidly in patients with acute chest pain, especially in patients with acute myocardial infarction (AMI) secondary to A-AAD, we investigated values of combined use of the risk score and the ascending aorta diameter >40 mm for the early identification of A-AAD. METHODS: Our study retrospectively encompassed 239 patients with acute chest pain on admission to our hospital between July 2010 and December 2016. The risk score was calculated according to the aortic dissection detection (ADD) risk score system, and the ascending aorta diameter was accurately obtained from the transthoracic echocardiography (TTE). RESULTS: A risk score ≥1 had an excellent sensitivity of 94.9% and a fair negative predictive value (NPV) of 77.8%, with a poor specificity of 8.7% and a positive predictive value (PPV) of 33.5% for the diagnosis of A-AAD. A risk score ≥2 had an excellent specificity of 91.3% and a fair NPV of 73.1%, whereas it had a lower sensitivity of 30.8% and a PPV of 63.2%. A risk score ≥1, combined with an ascending aorta diameter >40 mm, had a sensitivity, a specificity, a PPV, and an NPV of 84.6%, 87.6%, 76.7%, and 92.2% for the diagnosis of A-AAD, respectively. The combined use of a risk score ≥2 and an ascending aorta diameter >40 mm had an excellent specificity of 98.1% and a PPV of 86.4%, a fair NPV of 72.8%, and a poor sensitivity of 24.4% for the detection of A-AAD. Moreover, the omission diagnostic rate for A-AAD was significantly decreased from 33.3% to 7.4% using a risk score ≥1 combined with an ascending aorta diameter >40 mm in patients with AMI secondary to A-AAD. CONCLUSIONS: The combined use of an ADD risk score ≥1 and an ascending aorta diameter >40 mm was highly indicative of A-AAD in patients presenting with acute chest pain, especially in patients with AMI secondary to A-AAD, which urgently needed computed tomography angiography (CTA) or magnetic resonance imaging (MRI) to confirm the diagnosis of A-AAD.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA