Signalosome-Regulated SRF Phosphorylation Determining Myocyte Growth in Width versus Length as a Therapeutic Target for Heart Failure. Circulation Li, J. n., Tan, Y. n., Passariello, C. L., Martinez, E. C., Kritzer, M. D., Li, X. n., Li, X. n., Li, Y. n., Yu, Q. n., Ohgi, K. n., Thakur, H. n., MacArthur, J. W., Ivey, J. R., Woo, Y. J., Emter, C. A., Dodge-Kafka, K. n., Rosenfeld, M. G., Kapiloff, M. S. 2020

Abstract

Background: Concentric and eccentric cardiac hypertrophy are associated with pressure and volume overload, respectively, in cardiovascular disease both conferring an increased risk of heart failure. These contrasting forms of hypertrophy are characterized by asymmetric growth of the cardiac myocyte in mainly width or length, respectively. The molecular mechanisms determining myocyte preferential growth in width versus length remain poorly understood. Identification of the mechanisms governing asymmetric myocyte growth could provide new therapeutic targets for the prevention or treatment of heart failure. Methods: Primary adult rat ventricular myocytes, adeno-associated virus (AAV)-mediated gene delivery in mice, and human tissue samples are used to define a regulatory pathway controlling pathological myocyte hypertrophy. Chromatin Immunoprecipitation Assays with Sequencing (ChIP-seq) and Precision Nuclear Run-On Sequencing (PRO-seq) are used to define a transcriptional mechanism. Results: Here we report that asymmetric cardiac myocyte hypertrophy is modulated by serum response factor (SRF) phosphorylation, constituting an epigenomic switch balancing the growth in width versus length of adult ventricular myocytes in vitro and in vivo. SRF Ser103 phosphorylation is bidirectionally regulated by p90 ribosomal S6 kinase type 3 (RSK3) and protein phosphatase 2A (PP2A) at signalosomes organized by the scaffold protein muscle A-kinase anchoring protein ß (mAKAPß), such that increased SRF phosphorylation activates Activator Protein 1 (AP1)-dependent enhancers that direct myocyte growth in width. AAV are used to express in vivo mAKAPß-derived RSK3 and PP2A anchoring disruptor peptides that block the association of the enzymes with the mAKAPß scaffold. Inhibition of RSK3 signaling prevents concentric cardiac remodeling due to pressure overload, while inhibition of PP2A signaling prevents eccentric cardiac remodeling induced by myocardial infarction, in each case improving cardiac function. SRF Ser103 phosphorylation is significantly decreased in dilated human hearts, supporting the notion that modulation of the mAKAPß-SRF signalosome could be a new therapeutic approach for human heart failure. Conclusions: We have identified a new molecular switch, namely mAKAPß signalosome-regulated SRF phosphorylation, that controls a transcriptional program responsible for modulating changes in cardiac myocyte morphology that occur secondary to pathological stressors. Complementary AAV-based gene therapies constitute rationally-designed strategies for a new translational modality for heart failure.

View details for DOI 10.1161/CIRCULATIONAHA.119.044805

View details for PubMedID 32933333