Your browser doesn't support javascript.
loading
Deletion of delta-like 1 homologue accelerates fibroblast-myofibroblast differentiation and induces myocardial fibrosis.
Rodriguez, Patricia; Sassi, Yassine; Troncone, Luca; Benard, Ludovic; Ishikawa, Kiyotake; Gordon, Ronald E; Lamas, Santiago; Laborda, Jorge; Hajjar, Roger J; Lebeche, Djamel.
Afiliación
  • Rodriguez P; Department of Medicine, Cardiovascular Research Institute.
  • Sassi Y; Department of Medicine, Cardiovascular Research Institute.
  • Troncone L; Department of Medicine, Cardiovascular Research Institute.
  • Benard L; Department of Medicine, Cardiovascular Research Institute.
  • Ishikawa K; Department of Medicine, Cardiovascular Research Institute.
  • Gordon RE; Department of Pathology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
  • Lamas S; Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Nicolás Cabrera 1, Campus UAM, Madrid, Spain.
  • Laborda J; Department of Inorganic and Organic Chemistry and Biochemistry, Pharmacy School/Biomedical Unit/CRIB, University of Castilla-La Mancha/CSIC, Dr. José María Sánchez Ibáñez Street, s/n Albacete, Spain.
  • Hajjar RJ; Department of Medicine, Cardiovascular Research Institute.
  • Lebeche D; Department of Medicine, Cardiovascular Research Institute.
Eur Heart J ; 40(12): 967-978, 2019 03 21.
Article en En | MEDLINE | ID: mdl-29668883
ABSTRACT

AIMS:

Myocardial fibrosis is associated with profound changes in ventricular architecture and geometry, resulting in diminished cardiac function. There is currently no information on the role of the delta-like homologue 1 (Dlk1) in the regulation of the fibrotic response. Here, we investigated whether Dlk1 is involved in cardiac fibroblast-to-myofibroblast differentiation and regulates myocardial fibrosis and explored the molecular mechanism underpinning its effects in this process. METHODS AND

RESULTS:

Using Dlk1-knockout mice and adenoviral gene delivery, we demonstrate that overexpression of Dlk1 in cardio-fibroblasts resulted in inhibition of fibroblast proliferation and differentiation into myofibroblasts. This process is mediated by TGF-ß1 signalling, since isolated fibroblasts lacking Dlk1 exhibited a higher activation of the TGF-ß1/Smad-3 pathway at baseline, leading to an earlier acquisition of a myofibroblast phenotype. Likewise, Dlk1-null mice displayed increased TGF-ß1/Smad3 cardiac activity, resulting in infiltration/accumulation of myofibroblasts, induction and deposition of extra-domain A-fibronectin isoform and collagen, and activation of pro-fibrotic markers. Furthermore, these profibrotic events were associated with disrupted myofibril integrity, myocyte hypertrophy, and cardiac dysfunction. Interestingly, Dlk1 expression was down-regulated in ischaemic human and porcine heart tissues. Mechanistically, miR-370 mediated Dlk1's regulation of cardiac fibroblast-myofibroblast differentiation by directly targeting TGFß-R2/Smad-3 signalling, while the Dlk1 canonical target, Notch pathway, does not seem to play a role in this process.

CONCLUSION:

These findings are the first to demonstrate an inhibitory role of Dlk1 of cardiac fibroblast-to-myofibroblast differentiation by interfering with TGFß/Smad-3 signalling in the myocardium. Given the deleterious effects of continuous activation of this pathway, we propose Dlk1 as a new potential candidate for therapy in cases where aberrant TGFß signalling leads to chronic fibrosis.
Asunto(s)
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Fibrosis / Proteínas de Unión al Calcio / Miofibroblastos / Fibroblastos / Miocardio Límite: Animals / Humans / Male Idioma: En Revista: Eur Heart J Año: 2019 Tipo del documento: Article

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Fibrosis / Proteínas de Unión al Calcio / Miofibroblastos / Fibroblastos / Miocardio Límite: Animals / Humans / Male Idioma: En Revista: Eur Heart J Año: 2019 Tipo del documento: Article