Abstract

Introduction: Cardiac fibrosis can independently contribute to the impairment of cardiac function and development of heart failure. Early activation of pro-fibrotic genetic pathways has been suggested to occur well before adverse ventricular remodeling occurs in fibrosis as it progresses to heart failure. Delta-like homolog 1 (Dlk1), a paternally imprinted gene that encodes a transmembrane protein that contains multiple epidermal growth factor repeats, has been shown to play a critical role as a regulator of cell differentiation, neuronal differentiation, bone differentiation, and hematopoiesis. We hypothesize that Dlk-1 could regulate myocardial fibrosis via inhibition of fibroblast-myofibroblast conversion and therefore restrain cardiac fibrosis progression. Methods: Wildtype (WT) and Dlk1 knockout (Dlk1-/-) mice were subjected to myocardial infarction (MI) by permanent ligation of the left anterior descending artery to induce heart failure. A recombinant adeno-associated virus 9 (AAV9) was used to deliver Dlk1 into the myocardium in vivo. Cardiac function and hemodynamic changes were measured by echocardiography and left ventricular catheterization. Immunohistochemistry and confocal microscopy were used to determine the expression of pro-fibrotic markers. Total RNA was prepared from left ventricular (LV) tissue or cardiomyocytes and fibroblasts from Dlk1-/- and WT hearts. Masson trichrome staining was used to determine abnormal collagen deposition in the heart. Result: Myocardial fibrosis was demonstrated by increased expression of alpha-smooth muscle actin (αSMA), collagen, fibronectin, connective tissue growth factor, along with a substantial increase in the deposition of collagen as evident from Masson's trichrome staining. We observed downregulated Dlk-1 mRNA and protein expression in ischemic tissues from human patients and infarcted pigs’ hearts. The reduced levels of Dlk1 were associated with increased mRNA expression of pro-fibrotic molecules, collagen1a, LOX and αSMA in the scar area. Echocardiographic analysis revealed substantial LV chamber dilatation and reduced fractional shortening (FS %) in Dlk1 null mice as compared to WT, whereas the mice treated with AAV9-Dlk1 for 8-10 weeks showed a significant improvement in cardiac function. Consistently, Dlk1-deficient mice exhibited impairment in heart function, cardiac fibrosis and myocyte hypertrophy and increased expression of atrial natriuretic peptide, and B-type natriuretic peptide, while AAV9-Dlk1-treated mice significantly reduced cardiac remodeling and myocardial fibrosis. Conclusion: Restoration of Dlk1 in the heart following MI-induced heart failure improved cardiac function and attenuated myocardial fibrosis. The findings from the current study suggest a critical role for Dlk1 in myocardial fibrosis-induced cardiac remodeling, indicating that Dlk1 gene therapy may be a viable therapeutic approach for heart failure. This is the full abstract presented at the American Physiology Summit 2023 meeting and is only available in HTML format. There are no additional versions or additional content available for this abstract. Physiology was not involved in the peer review process.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call