Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cardiovasc Toxicol ; 24(6): 550-562, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38696070

RESUMO

Trastuzumab is widely used in HER2 breast cancer. However, it may cause left ventricular (LV) dysfunction. A decrease in LV global longitudinal strain (GLS) has been previously demonstrated to be a good predictor of subsequent cancer therapy related dysfunction (CTRCD). Left atrial morphological remodeling during Trastuzumab therapy has also been shown. The aim of this study is exploring the relationship between early changes in left atrial function and the development of Trastuzumab-induced cardiotoxicity. Consecutive patients with diagnosis of HER2+non-metastatic breast cancer treated with Trastuzumab were prospectively enrolled. A clinical, conventional, and advanced echocardiographic assessment was performed at baseline and every three months, until a one-year follow-up was reached. One-hundred-sixteen patients completed the 12 months follow-up, 10 (9%) cases of CTRCD were observed, all after the sixth month. GLS and LVEF significantly decreased in the CTRCD group at 6 months of follow-up, with an earlier (3 months) significant worsening in left atrial morpho-functional parameters. Systolic blood pressure, early peak atrial longitudinal strain (PALS), peak atrial contraction (PACS) and left atrial volume (LAVI) changes resulted independent predictors of CTRCD at multivariable logistic regression analysis. Moreover, early changes in PALS and PACS resulted good predictors of CTRCD development (AUC 0.85; p = 0.008, p < 0.001 and 0.77; p = 0.008, respectively). This prospective study emphasizes that the decline in PALS and PACS among trastuzumab-treated patients could possibly increase the accuracy in identifying future CTRCD in non-metastatic HER2 breast cancer cases, adding predictive value to conventional echocardiographic assessment.


Assuntos
Antineoplásicos Imunológicos , Função do Átrio Esquerdo , Neoplasias da Mama , Cardiotoxicidade , Receptor ErbB-2 , Trastuzumab , Função Ventricular Esquerda , Humanos , Trastuzumab/efeitos adversos , Feminino , Neoplasias da Mama/tratamento farmacológico , Pessoa de Meia-Idade , Receptor ErbB-2/metabolismo , Estudos Prospectivos , Antineoplásicos Imunológicos/efeitos adversos , Função Ventricular Esquerda/efeitos dos fármacos , Função do Átrio Esquerdo/efeitos dos fármacos , Adulto , Fatores de Tempo , Fatores de Risco , Resultado do Tratamento , Idoso , Valor Preditivo dos Testes , Medição de Risco , Remodelamento Atrial/efeitos dos fármacos , Cardiopatias/induzido quimicamente , Cardiopatias/fisiopatologia , Cardiopatias/diagnóstico por imagem , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/fisiopatologia , Disfunção Ventricular Esquerda/diagnóstico por imagem , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/fisiopatologia , Átrios do Coração/diagnóstico por imagem , Volume Sistólico/efeitos dos fármacos
2.
J Cell Mol Med ; 24(21): 12331-12340, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32940423

RESUMO

The sodium-glucose cotransporter 2 (SGLT2) inhibitor empagliflozin reduces heart failure in diabetes, but underlying mechanisms remain elusive. We hypothesized that empagliflozin could counteract the senescence of cardiac stromal cells (CSC), the action of which limits cardiac damage and cardiac fibrosis in diabetic-like conditions in vitro and in vivo. CSC were isolated from murine heart biopsies (n = 5) through cardiosphere (CSp) formation and incubated for 3 or 48 hours with 5.5 mmol/L normal glucose (NG), high glucose (12-5 and 30.5 mmol/L, HG) or a hyperosmolar control of mannitol (HM) in the presence or absence of empagliflozin 100 nmol/L. The senescent CSC status was verified by ß-gal staining and expression of the pro-survival marker Akt (pAkt) and the pro-inflammatory marker p38 (p-P38). The cardiac effects of empagliflozin were also studied in vivo by echocardiography and by histology in a murine model of streptozotocin (STZ)-induced diabetes. Compared to NG, incubations with HG and HM significantly reduced the number of CSps, increased the ß-gal-positive CSC and P-p38, while decreasing pAkt, all reversed by empagliflozin (P < .01). Empagliflozin also reversed cardiac dysfunction, cardiac fibrosis and cell senescence in mice with (STZ)-induced diabetes (P < .01). Empagliflozin counteracts the pro-senescence effect of HG and of hyperosmolar stress on CSC, and improves cardiac function via decreasing cardiac fibrosis and senescence in diabetic mice, possibly through SGLT2 off-target effects. These effects may explain empagliflozin unexpected benefits on cardiac function in diabetic patients.


Assuntos
Compostos Benzidrílicos/farmacologia , Senescência Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Glucosídeos/farmacologia , Miocárdio/metabolismo , Células Estromais/efeitos dos fármacos , Animais , Biópsia , Sobrevivência Celular , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Insuficiência Cardíaca/fisiopatologia , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transportador 2 de Glucose-Sódio/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos
3.
Vascul Pharmacol ; 130: 106678, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32229255

RESUMO

BACKGROUND: Hyperglycemia plays a role in promoting insulin resistance in adipocytes, hepatocytes and myocytes. Its effects on insulin signaling in endothelial cells remain, however, incompletely understood. AIM: To investigate the proteomic and metabolomic profiles of human aortic endothelial cells (HAECs) exposed to insulin, normal glucose (NG), high glucose (HG) or its hyperosmolar control high mannitol (HM), and to examine whether and how HG or HM may promote insulin resistance. METHODS AND RESULTS: We exposed HAECs to HG and HM in shorter (3 h) and longer-term experiments (24 h), followed by insulin treatment for 45 min. Label-free proteomics and network analysis showed a downregulation of proteins linked to the PI3K-Akt/mTOR/eNOS signaling pathway in HAECs. Metabolomic profiling showed decreased levels of "odd-chain acylcarnitines" such as C3. At immunoblotting, HG or HM blunted insulin ability to activate the PI3K/AKT/eNOS pathway, which was reverted through a silencing of aquaporin 1 (AQP1) and Tonicity enhancer binding protein (TonEBP), while inducing p-P38 and pERK1/2. CONCLUSIONS: HG impairs the PI3K/AKT/eNOS pathway and shifts insulin signaling towards the activation of mitogenic and pro-inflammatory effectors, such as p38 and ERK1/2. These effects may explain the progression of insulin resistance as a result of endothelial glucotoxicity.


Assuntos
Células Endoteliais/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Glucose/toxicidade , Hiperglicemia/metabolismo , Resistência à Insulina , Insulina/farmacologia , Aquaporina 1/genética , Aquaporina 1/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Hiperglicemia/genética , Hiperglicemia/fisiopatologia , Manitol/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Concentração Osmolar , Pressão Osmótica/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Mapas de Interação de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA