bioRxiv | 2019

SERCA2a-activating PLB mutant improves calcium handling in dilated cardiomyopathy model of hiPSC-CMs

 
 
 
 
 
 
 

Abstract


There is increasing momentum toward the development of gene therapy for heart failure, cardiomyopathy, and chronic cardiac diseases attributed to impaired calcium (Ca) transport and reduced contractility. Here, we established a novel FRET assay using fluorescently-tagged SERCA2a (the cardiac Ca pump) and PLB (its endogenous regulator) that directly tests the capacity of loss-of-function PLB mutants (PLBM) to compete with wild-type PLB (PLBWT) to relieve SERCA2a inhibition. The combination of a mutation conferring loss-of-inhibitory-function (L31A-PLBM) and a mutation associated with increased binding affinity (I40A-PLBM) resulted in a PLBM (L31A/I40A) capable of displacing PLBWT. Overexpression of L31A/I40A-PLBM increased Ca-ATPase activity in PLBWT-expressing HEK293-6E cells. In human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) from healthy individuals, recombinant adeno-associated virus 2 (rAAV2) driven expression of L31A/I40A-PLBM significantly enhanced Ca-release amplitude and removal. Furthermore, L31A/I40A-PLBM expression in a hiPSC-CM dilated cardiomyopathy fully rescued irregular Ca transients and decreased Ca transport. Taken together, these results suggest that gene transfer of PLBM is a potential therapy for cardiac diseases associated with impaired Ca transport and muscle contractility.

Volume None
Pages 699975
DOI 10.1101/699975
Language English
Journal bioRxiv

Full Text