Your browser doesn't support javascript.
loading
Using cultured canine cardiac slices to model the autophagic flux with doxorubicin.
Boukhalfa, Asma; Robinson, Sally R; Meola, Dawn M; Robinson, Nicholas A; Ling, Lauren A; LaMastro, Joey N; Upshaw, Jenica N; Pulakat, Lakshmi; Jaffe, Iris Z; London, Cheryl A; Chen, Howard H; Yang, Vicky K.
Afiliação
  • Boukhalfa A; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America.
  • Robinson SR; Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America.
  • Meola DM; Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America.
  • Robinson NA; Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America.
  • Ling LA; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America.
  • LaMastro JN; Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America.
  • Upshaw JN; Tufts University School of Medicine, Boston, Massachusetts, United States of America.
  • Pulakat L; Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, United States of America.
  • Jaffe IZ; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America.
  • London CA; Tufts University School of Medicine, Boston, Massachusetts, United States of America.
  • Chen HH; Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America.
  • Yang VK; Tufts University School of Medicine, Boston, Massachusetts, United States of America.
PLoS One ; 18(3): e0282859, 2023.
Article em En | MEDLINE | ID: mdl-36928870
ABSTRACT
Chemotherapy-induced impairment of autophagy is implicated in cardiac toxicity induced by anti-cancer drugs. Imperfect translation from rodent models and lack of in vitro models of toxicity has limited investigation of autophagic flux dysregulation, preventing design of novel cardioprotective strategies based on autophagy control. Development of an adult heart tissue culture technique from a translational model will improve investigation of cardiac toxicity. We aimed to optimize a canine cardiac slice culture system for exploration of cancer therapy impact on intact cardiac tissue, creating a translatable model that maintains autophagy in culture and is amenable to autophagy modulation. Canine cardiac tissue slices (350 µm) were generated from left ventricular free wall collected from euthanized client-owned dogs (n = 7) free of cardiovascular disease at the Foster Hospital for Small Animals at Tufts University. Cell viability and apoptosis were quantified with MTT assay and TUNEL staining. Cardiac slices were challenged with doxorubicin and an autophagy activator (rapamycin) or inhibitor (chloroquine). Autophagic flux components (LC3, p62) were quantified by western blot. Cardiac slices retained high cell viability for >7 days in culture and basal levels of autophagic markers remained unchanged. Doxorubicin treatment resulted in perturbation of the autophagic flux and cell death, while rapamycin co-treatment restored normal autophagic flux and maintained cell survival. We developed an adult canine cardiac slice culture system appropriate for studying the effects of autophagic flux that may be applicable to drug toxicity evaluations.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Miócitos Cardíacos / Cardiotoxicidade Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Miócitos Cardíacos / Cardiotoxicidade Idioma: En Ano de publicação: 2023 Tipo de documento: Article