RT Journal Article SR Electronic T1 EPAC1 Inhibition Protects the Heart from Doxorubicin-Induced Toxicity JF bioRxiv FD Cold Spring Harbor Laboratory SP 2021.06.16.448655 DO 10.1101/2021.06.16.448655 A1 Marianne Mazevet A1 Maxance Ribeiro A1 Anissa Belhadef A1 Delphine Dayde A1 Anna Llach A1 Marion Laudette A1 Tiphaine Belleville A1 Philippe Mateo A1 Mélanie Gressette A1 Florence Lefebvre A1 Ju Chen A1 Christilla Bachelot-Loza A1 Catherine Rucker-Martin A1 Frank Lezoualc’h A1 Bertrand Crozatier A1 Jean-Pierre Benitah A1 Marie-Catherine Vozenin A1 Rodolphe Fischmeister A1 Ana-Maria Gomez A1 Christophe Lemaire A1 Eric Morel YR 2021 UL http://biorxiv.org/content/early/2021/06/17/2021.06.16.448655.abstract AB Rationale The widely used chemotherapeutic agent Doxorubicin (Dox) induces cardiotoxicity leading to dilated cardiomyopathy and heart failure. This cardiotoxicity has been related to ROS generation, DNA intercalation, bioenergetic distress and cell death. However, alternative mechanisms are emerging, focusing on signaling pathways.Objective We investigated the role of Exchange Protein directly Activated by cAMP (EPAC), key factor in cAMP signaling, in Dox-induced cardiotoxicity.Methods and Results Dox was administrated in vivo (10 ± 2 mg/kg, i.v.; with analysis at 2, 6 and 15 weeks post injection) in WT and EPAC1 KO C57BL6 mice. Cardiac function was analyzed by echocardiography and intracellular Ca2+ homeostasis by confocal microscopy in isolated ventricular cardiomyocytes. 15 weeks post-injections, Dox-treated WT mice, developed a dilated cardiomyopathy with decreased ejection fraction, increased telediastolic volume and impaired Ca2+ homeostasis, which were totally prevented in the EPAC1 KO mice.The underlying mechanisms were investigated in neonatal and adult rat cardiac myocytes under Dox treatment (1-10 μM). Flow cytometry, Western blot, BRET sensor assay, and RT-qPCR analysis showed that Dox induced DNA damage and cardiomyocyte cell death with apoptotic features rather than necrosis, including Ca2+-CaMKKβ-dependent opening of the Mitochondrial Permeability Transition Pore, dissipation of the Mitochondrial membrane potential (Δψm), caspase activation, cell size reduction, and DNA fragmentation. Dox also led to an increase in both cAMP concentration and EPAC1 protein level and activity. The pharmacological inhibition of EPAC1 (CE3F4) but not EPAC2 alleviated the whole Dox-induced pattern of alterations including DNA damage, Δψm, apoptosis, mitochondrial biogenesis, dynamic, and fission/fusion balance, and respiratory chain activity, suggesting a crucial role of EPAC1 in these processes. Importantly, while preserving cardiomyocyte integrity, EPAC1 inhibition potentiated Dox-induced cell death in several human cancer cell lines.Conclusion Thus, EPAC1 inhibition could be a valuable therapeutic strategy to limit Dox-induced cardiomyopathy without interfering with its antitumoral activity.Competing Interest StatementThe authors have declared no competing interest.8-CPT8-(4-chloro-phenylthio)-2’-O-methyladenosine-3’-5’cyclic monophosphateβ-ARβ-adrenergic receptorARVMAdult Rat Ventricular MyocyteBAABiotin-conjugated ADTM analogCa2+CalciumCaMKKβCa2+/calmodulin-dependent protein kinase kinase βDCMDilated cardiomyopathyDoxDoxorubicinEFEjection fractionEPACExchange Protein directly Activated by cAMPFDAFluorescein di-acetateHFHeart failureKOKnock-OutLVEDVLeft ventricle end-diastolic volumeMCUMitochondrial Ca2+ UniporterMPTPMitochondrial Permeability Transition PoreNRVMNeonatal Rat Ventricular MyocytesPIPropidium IodideRuthRed Ruthenium RedTopIIβTopoisomerase IIβWTWild Type