DNA damage response/TP53 pathway is activated and contributes to the pathogenesis of dilated cardiomyopathy associated with LMNA (Lamin A/C) mutations

SN Chen, R Lombardi, J Karmouch, JY Tsai… - Circulation …, 2019 - Am Heart Assoc
SN Chen, R Lombardi, J Karmouch, JY Tsai, G Czernuszewicz, MRG Taylor, L Mestroni
Circulation research, 2019Am Heart Assoc
Rationale: Mutations in the LMNA gene, encoding LMNA (lamin A/C), are responsible for
laminopathies. Dilated cardiomyopathy (DCM) is a major cause of mortality and morbidity in
laminopathies. Objective: To gain insights into the molecular pathogenesis of DCM in
laminopathies. Methods and Results: We generated a tet-off bigenic mice expressing either
a WT (wild type) or a mutant LMNA (D300N) protein in cardiac myocytes. LMNAD300N
mutation is associated with DCM in progeroid syndromes. Expression of LMNAD300N led to …
Rationale:
Mutations in the LMNA gene, encoding LMNA (lamin A/C), are responsible for laminopathies. Dilated cardiomyopathy (DCM) is a major cause of mortality and morbidity in laminopathies.
Objective:
To gain insights into the molecular pathogenesis of DCM in laminopathies.
Methods and Results:
We generated a tet-off bigenic mice expressing either a WT (wild type) or a mutant LMNA (D300N) protein in cardiac myocytes. LMNAD300N mutation is associated with DCM in progeroid syndromes. Expression of LMNAD300N led to severe myocardial fibrosis, apoptosis, cardiac dysfunction, and premature death. Administration of doxycycline suppressed LMNAD300N expression and prevented the phenotype. Whole-heart RNA sequencing in 2-week-old WT and LMNAD300N mice led to identification of ≈6000 differentially expressed genes. Gene Set Enrichment and Hallmark Pathway analyses predicted activation of E2F (E2F transcription factor), DNA damage response, TP53 (tumor protein 53), NFκB (nuclear factor κB), and TGFβ (transforming growth factor-β) pathways, which were validated by Western blotting, quantitative polymerase chain reaction of selected targets, and immunofluorescence staining. Differentially expressed genes involved cell death, cell cycle regulation, inflammation, and epithelial-mesenchymal differentiation. RNA sequencing of human hearts with DCM associated with defined LMNA pathogenic variants corroborated activation of the DNA damage response/TP53 pathway in the heart. Increased expression of CDKN2A (cyclin-dependent kinase inhibitor 2A)—a downstream target of E2F pathway and an activator of TP53—provided a plausible mechanism for activation of the TP53 pathway. To determine pathogenic role of TP53 pathway in DCM, Tp53 gene was conditionally deleted in cardiac myocytes in mice expressing the LMNAD300N protein. Deletion of Tp53 partially rescued myocardial fibrosis, apoptosis, proliferation of nonmyocyte cells, left ventricular dilatation and dysfunction, and slightly improved survival.
Conclusions:
Cardiac myocyte-specific expression of LMNAD300N, associated with DCM, led to pathogenic activation of the E2F/DNA damage response/TP53 pathway in the heart and induction of myocardial fibrosis, apoptosis, cardiac dysfunction, and premature death. The findings denote the E2F/DNA damage response/TP53 axis as a responsible mechanism for DCM in laminopathies and as a potential intervention target.
Am Heart Assoc