Abstract

Cardiovascular diseases (CVDs) arise from a complex interplay among genomic, proteomic, and metabolomic abnormalities. Emerging evidence has recently consolidated the presence of robust DNA damage in a variety of cardiovascular disorders. DNA damage triggers a series of cellular responses termed DNA damage response (DDR) including detection of DNA lesions, cell cycle arrest, DNA repair, cellular senescence, and apoptosis, in all organ systems including hearts and vasculature. Although transient DDR in response to temporary DNA damage can be beneficial for cardiovascular function, persistent activation of DDR promotes the onset and development of CVDs. Moreover, therapeutic interventions that target DNA damage and DDR have the potential to attenuate cardiovascular dysfunction and improve disease outcome. In this review, we will discuss molecular mechanisms of DNA damage and repair in the onset and development of CVDs, and explore how DDR in specific cardiac cell types contributes to CVDs. Moreover, we will highlight the latest advances regarding the potential therapeutic strategies targeting DNA damage signalling in CVDs.

1. Introduction

Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality globally.1 To date, multi-omics approaches have identified a multi-facet aetiology for CVDs, including genomic, proteomic, and metabolomic derrangements.2,3 However, much focus has been geared towards impairment of proteostasis and metabolic harmony.3,4 Possible genomic instability and DNA damage have been indicated in the pathophysiology of CVDs albeit remains largely unexplored.5,6 Clinical and pre-clinical evidence has indicated the presence of DNA damage in CVDs.7–9 Besides, DNA damage triggers unfavourable sequelae in mammalian cells, including cell senescence, apoptosis, and inflammation,6,10 which are well perceived for the pathophysiology of CVDs. In this review, we outline the emerging role for DNA damage and repair mechanisms in CVDs, the DNA damage response (DDR) in various cardiac cell types, and potential therapies targeting DNA damage and DDR for the management of CVDs.

2. DNA damage and repair mechanism in the cardiovascular system

DNA damage can be provoked by intrinsic or extrinsic insults, including oxidative stress, metabolic stress, chemotherapy drugs, and radiotherapy.11 Among these genomic insults, reactive oxygen species (ROS) represent the main endogenous culprit which leads to oxidative DNA damage, including a range of DNA lesions such as base oxidations, single-strand breaks (SSBs), double-strand breaks (DSBs), and telomere shortening.12,13 DNA repair system is evolved to overcome various types of DNA damage, including homologous recombination (HR), non-homologous end joining (NHEJ), base excision repair (BER), nucleotide excision repair (NER), and mismatch repair (MMR). In general, DSBs are repaired through HR or NHEJ, while SSBs and individual base damage are revamped through BER, and large adducts through NER.12Figure 1 denotes perceived causes of DNA damage, DNA repair system, and cellular consequences in CVDs. Although mitochondrial DNA damage also contributes to the pathogenesis of CVDs,14–16 such as heart failure (HF) and coronary heart disease, we will only focus on the nuclear DNA damage signalling in CVDs.

DNA damage and DDR activation drive the development of CVDs. Extrinsic and intrinsic molecules stimulate various DNA lesions in cardiovascular cells, which are repaired by several mechanisms. Accumulation of DNA damage due to incomplete or incorrect repair evokes persistent DDR and genomic instability, thus driving unfavourable cellular outcomes, including senescence, apoptosis, and inflammation. These unfavourable sequelae and DNA damage lead to the onset and development of CVDs. Thus, suppression of DNA damage and inhibition of DDR activation may have therapeutic promises in the management of CVDs. ROS, reactive oxygen species; AGEs, advanced glycation end products; UV, ultraviolet; DSB, double-strand break; SSB, single-strand break; HR, homologous recombination; NHEJ, non-homologous end joining; BER, base excision repair; NER, nucleotide excision repair; MMR, mismatch repair; DDR, DNA damage response.
Figure 1

DNA damage and DDR activation drive the development of CVDs. Extrinsic and intrinsic molecules stimulate various DNA lesions in cardiovascular cells, which are repaired by several mechanisms. Accumulation of DNA damage due to incomplete or incorrect repair evokes persistent DDR and genomic instability, thus driving unfavourable cellular outcomes, including senescence, apoptosis, and inflammation. These unfavourable sequelae and DNA damage lead to the onset and development of CVDs. Thus, suppression of DNA damage and inhibition of DDR activation may have therapeutic promises in the management of CVDs. ROS, reactive oxygen species; AGEs, advanced glycation end products; UV, ultraviolet; DSB, double-strand break; SSB, single-strand break; HR, homologous recombination; NHEJ, non-homologous end joining; BER, base excision repair; NER, nucleotide excision repair; MMR, mismatch repair; DDR, DNA damage response.

2.1 DSB signalling pathway in the cardiovascular system

In the HR modality, DSBs are detected by the MRE11-RAD50-NBS1 (MRN) complex which activates ataxia–telangiectasia mutated (ATM) kinase to transduce DDR signalling.17 HR repair is generally restricted to the S and G2 phases of the cell cycle, since this repair system uses sister-chromatid sequences as the template to execute the faithful repair.12 Normally, HR is initiated by detection of the MRN complex, the 5′–3′ strand resection of exonuclease 1 (EXO1) and bloom syndrome heterodimer, leading to the generation of a 3′ single-stranded DNA (ssDNA) tail. The ssDNA is then rapidly coated by replication protein A (RPA), and the ssDNA–RPA complex recruits ATRIP, leading to activation of another primary kinase namely ataxia–telangiectasia and Rad3-related (ATR). RPA is subsequently displaced by Rad51 to form a nucleoprotein filament. In cellular processes catalysed by RAD51 and the breast-cancer susceptibility proteins BRCA1 and BRCA2, ssDNA invades undamaged template, helicases, and other components with the assistance of polymerases and nucleases.12 Besides, BRCA1 interacts and co-localizes with RAD51 at the sites of DSBs to turn on RAD51-mediated HR repair. Notably, BRCA1-deficient cells exhibit deficiency in RAD51-mediated HR of DSBs along with hypersensitivity to genetic insults.18

In the NHEJ modality, DSBs are detected by the Ku70 and Ku80 proteins which bind and activate the protein kinase DNA-dependent protein kinase (DNA-PK) catalytic subunit (DNA-PKcs).12 The DNA-PK complex recruits and activates DNA ligase IV and associated scaffolding factors XRCC4 (X-ray repair cross-complementing protein 4) and XLF (XRCC4-like factor) to repair DSBs.12 In DSB signalling, the ATM, ATR, and DNA-PK complexes serve as major DDR mediators for phosphorylation of downstream substrates, including H2AX, checkpoint kinase 1 (CHK1)/CHK2, and p53 to transduce DDR signals (i.e. cell cycle arrest, senescence, apoptosis, and inflammation).19 Notably, phosphorylation of H2AX at serine 139 (termed γH2AX) is perceived as a benchmark for DSBs.20 The pathogenic and therapeutic value of ATM, ATR, and DNA-PK in the cardiovascular system will be discussed in Section 7.

DSBs are identified under genomic stress in nearly all cardiovascular cell types.6,9,21,22 However, only limited studies directly assessed the role of DSB repair in cardiovascular cells. In theory, HR repair is unlikely to occur in post-mitotic cardiomyocytes due to the restriction of the cell cycle. However, up-regulated BRCA1 was observed in post-ischaemic human myocardium.9 BRCA1 depletion in cardiomyocytes led to impaired DSB repair (indicated by RAD51-foci formation) and cardiac function in myocardial ischaemia.9 However, no direct evidence was offered for possible HR in cardiomyocytes.9 Further study is warranted to discern the presence of HR in cardiac diseases. In another independent study, vascular smooth muscle cells (VSMCs) from human atherosclerotic plaques displayed DNA damage and up-regulated MRN complex, favouring the presence of HR in VSMCs.23 Distinct from HR, a role for NHEJ in the cardiovascular system was only reported in endothelial cells.24 The results revealed reduced lncRNA SNHG12 in atherosclerotic plaque endothelial cells, leading to reduced interaction between DNA-PKcs and Ku70/Ku80, compromised DNA repair, DDR activation, and vascular senescence.24 These findings demonstrate that deficiency in DSB repair may lead to genomic instability, activation of DDR, and progression of CVDs.

2.2 SSB and excision repair in the cardiovascular system

The most common trigger for SSBs is oxidative attack evoked by ROS. SSBs can occur either by direct disintegration of oxidized sugar or indirectly during DNA BER of modified bases, such as oxidized bases, abasic sites (aka, apurinic, or apyrimidinic sites), or other base modifications.25 Besides, SSBs can also be evoked due to the abortive or erroneous function of enzymes including DNA topoisomerase 1.25 These base or nucleotide modifications and SSBs are repaired by excision repair mechanisms, including SSB repair, BER, and NER. The most likely consequence of SSBs in dividing cells is the blockage of DNA replication forks during the S phase, leading to DSB formation.25 In non-proliferating cells such as post-mitotic cardiomyocytes, cell death induced by SSBs might involve RNA polymerase implemented during transcription, and overactivation of the SSB sensor protein poly(ADP-ribose) polymerase 1 (PARP1).25

Compared to DSB repair, PARP enzymes play critical roles in various steps of SSB repair.26,27 In brief, SSBs are rapidly recognized and bound with PARP1, followed by attachment of PAR onto PARP1, leading to PARP1 activation. During BER, PARP1 activates SSB repair through recruiting and stabilizing a series of repair components. PARP1 recruits a scaffold protein XRCC1 and assemblies with DNA polymerase β and DNA ligase III to foster the repair process.28

NER pathway is conserved to repair bulky DNA lesions due to multiple sources of mutagenic agents, such as ultraviolet (UV) irradiation. This repair system includes the following processes namely (i) DNA damage detection by xeroderma pigmentosum group C-complementing protein (XPC) assisted by the UV-DDB1/2 (XPE) complex, (ii) local unwinding of DNA, (iii) damage verification by XPA, (iv) excision of damaged DNA by endonucleases ERCC1/XPF and XPG, and (v) replacement of excised DNA using intact strand as a template.25 Notably, PARP1 was shown to play a vital role in NER by mediating the PARylation of NER proteins including XPA and XPC.26

Although SSB is established in CVDs including HF, there is limited evidence for the precise role of SSB repair in given cardiovascular cells and CVDs.29,30 It was previously shown that cardiomyocyte-specific depletion of XRCC1 led to SSB accumulation, DDR activation, cardiac inflammatory response, and exacerbated HF.29 In addition, an essential role for NER in vascular disease was shown by two NER-defective mouse models, Ercc1d/ and XpdTTD, where increased vascular senescence, stiffness, hypertension, and impaired vasodilator function were evident with ageing.30 These findings further underline the importance of SSB repair machinery for the maintenance of genomic stability of the cardiovascular system and protection against CVDs.

2.3 Oxidative bases and OGG1-mediated BER in the cardiovascular system

DNA bases are especially vulnerable to oxidation damage evoked by ROS. 8-oxoguanine (8-oxoG) is the most abundant DNA lesion marker formed upon oxidative exposure. Accumulation of 8-oxoG is a cellular biomarker for oxidative stress.5 BER is the primary 8-oxoG repair machinery. Among these, 8-oxoguanine glycosylase (OGG1), the predominant form of DNA glycosylase, is deemed responsible for the removal of 8-oxoG.5

To date, ample studies have linked a deficiency in OGG1-mediated BER with the aetiology of CVDs.5,7,31 Hearts from patients with end-stage cardiomyopathies exhibited a loss in OGG1 content.7 Besides, nutrient deprivation also contributed to impaired BER (8-oxoG accumulation) in cardiomyocytes in vitro, accompanied by loss of OGG1, while BER activity was rescued by recombinant OGG1. OGG1 was also verified in vascular cells in atherosclerosis progression. In particular, 8-oxoG accumulation in endothelial cells, VSMCs, and macrophages were closely linked with the onset of atherosclerosis.31 In this context, OGG1 deficiency in macrophages was shown to promote oxidative DNA damage and accelerated atherogenesis through induction of inflammation.31 Further investigations revealed reduced protein levels and activity of OGG1 in VSMCs from human plaques, leading to BER deficiency and 8-oxoG accumulation, en route to the facilitation of atherosclerosis.5 Thus, detrimental outcomes of oxidative stress on CVDs involve a vicious cycle among oxidative DNA damage, down-regulation of OGG1, and BER deficiency.

3. DDR in specific cardiovascular cell types

DDR encompasses a complex intertwined signalling network to prevent genomic instability. In particular, proteins undergo post-translational modifications to reconcile DNA damage, while DDR proteins are categorized into sensors, transducers, and effectors12 (Figure 2). Normally, if DNA lesions are minor and properly repaired, cells will undergo a transient cell cycle arrest, and DDR inactivation ensues to restore normal function. Cells undergo excessive or permanent DDR activation triggering cellular senescence and apoptosis with severe unrepairable DNA damage.12 Moreover, proteomics analysis has identified a variety of uncharacterized ATM/ATR-mediated phosphorylation sites,32 denoting more cellular processes as possible DDR regulatory targets.

DDR pathways and key components being targeted in CVDs. Specific types of DNA damage, such as DSBs, SSBs, and base mismatch, lead to activation of DDR and repair cascades. DDR components can be categorized into sensors, transductors and effectors. The PARP enzymes are crucial to activate a host of downstream signalling and are primary proteins involved in BER. DSBs trigger the activation of a number of kinases including ATM, ATR, and DNA-PK, as well as their downstream targets including CHK1 and CHK2 to regulate cell cycle control checkpoints. The p53 activation directly triggers cellular senescence, apoptosis, and induction of inflammation. A number of drugs targeting key DDR components are deemed effective in the treatment of CVDs in animal models. ATM, ataxia–telangiectasia mutated; ATR, ataxia–telangiectasia and Rad3-related; ATRIP, ATR-interacting protein; BER, base excision repair; CHK, checkpoint kinase; DDR, DNA damage response; DNA-PK, DNA-dependent protein kinase; DSB, double-strand break; DPQ, 3,4-dihydro-5-[4-(1-piperidinyl)butoxyl]-1 (2H)-isoquinolinone; HR, homologous recombination; MMR, mismatch repair; MRN, MRE11, RAD50 and NBS1 complex; NHEJ, non-homologous end joining; PARP, poly(ADP-ribose) polymerase; POLB, DNA polymerase-β; RPA, replication protein A; SSB, single-strand break; TIQ-A, thieno[2,3-c]isoquinolin-5-one; 3-AB, 3-aminobenzamide.
Figure 2

DDR pathways and key components being targeted in CVDs. Specific types of DNA damage, such as DSBs, SSBs, and base mismatch, lead to activation of DDR and repair cascades. DDR components can be categorized into sensors, transductors and effectors. The PARP enzymes are crucial to activate a host of downstream signalling and are primary proteins involved in BER. DSBs trigger the activation of a number of kinases including ATM, ATR, and DNA-PK, as well as their downstream targets including CHK1 and CHK2 to regulate cell cycle control checkpoints. The p53 activation directly triggers cellular senescence, apoptosis, and induction of inflammation. A number of drugs targeting key DDR components are deemed effective in the treatment of CVDs in animal models. ATM, ataxia–telangiectasia mutated; ATR, ataxia–telangiectasia and Rad3-related; ATRIP, ATR-interacting protein; BER, base excision repair; CHK, checkpoint kinase; DDR, DNA damage response; DNA-PK, DNA-dependent protein kinase; DSB, double-strand break; DPQ, 3,4-dihydro-5-[4-(1-piperidinyl)butoxyl]-1 (2H)-isoquinolinone; HR, homologous recombination; MMR, mismatch repair; MRN, MRE11, RAD50 and NBS1 complex; NHEJ, non-homologous end joining; PARP, poly(ADP-ribose) polymerase; POLB, DNA polymerase-β; RPA, replication protein A; SSB, single-strand break; TIQ-A, thieno[2,3-c]isoquinolin-5-one; 3-AB, 3-aminobenzamide.

DNA damage differentially affects cells depending on cell types. The cardiovascular system involves a wide range of cell types, including cardiomyocytes, VSMCs, fibroblasts, endothelial, immune, progenitor, and epithelial cells. In vivo and in vitro studies have shown that nearly all cell types undergo DNA damage and DDR activation with ageing or development of CVDs.21,23,24,33 Therefore, possible contributions of DNA damage to CVD pathology can be originated from a variety of cell types. Although activation of DDR was initially thought to be a cell cycle arrest response unique to dividing cells, it became evident that DDR activation is also generalized in post-mitotic cells such as cardiomyocytes.33

3.1 DDR in cardiomyocytes

The role of DDR activation in cardiomyocytes has been evaluated in only limited studies.21,29,33–35 Mammalian hearts possess a remarkable regenerative capacity for a brief period following birth, after which the majority of cardiomyocytes permanently exit the cell cycle. Notably, DDR serves as a mediator of physiological post-natal cell cycle arrest in cardiomyocytes. Post-natal exposure to environmental oxygen was shown to trigger mitochondrial oxidative metabolism, ROS production, oxidative DNA damage (8-oxoG level), and ATM-mediated DDR activation in the heart during the first post-natal week, triggering cell cycle arrest.34 Intriguingly, post-natal exposure to mild hypoxia, ROS scavenging, and DDR inhibition promote cardiomyocyte proliferation and regenerative capacity.34 Moreover, gradual exposure to systemic hypoxaemia after myocardial infarction (MI), where inspired oxygen was slowly dropped by 1% and was ultimately maintained at 7% for 2 weeks, contributed to a robust regenerative response in mice, along with reduced myocardial fibrosis and improved cardiac function.35 Similarly, moderate hypoxia (blood oxygen saturation: 75–85%) was associated with increased cell cycle markers possibly through reduced oxidative DNA damage in post-natal human cardiomyocytes.36

Under pathological stress, persistent DNA damage and DDR activation are evident in cardiomyocytes, prompting a shift from a non-dividing physiological to a non-dividing pathological state (e.g. hypertrophic growth). DNA damage in cardiomyocytes leads to up-regulation of several DDR markers such as phosphorylated ATM, ATR, PARP1, and p53, with increased cellular senescence and apoptosis.21,37–39 DDR is shown to regulate pathological cardiomyocyte hypertrophy.21,29 Pressure overload induces DSBs, DDR activation, ATM-dependent calcineurin activation, and protein synthesis, leading to cardiac hypertrophy.21 During telomeric DNA damage and DDR activation, cardiomyocytes acquire senescence-like phenotypes, characterized by p53, p21, and p16 activation, and a non-canonical SASP secretome including inflammatory cytokines TNF-α, IL-1, and IL-6, growth differentiation factor 15 (GDF15), TGFβ2, and endothelin 3 (EDN3), contributing to cardiac hypertrophy and interstitial fibrosis.33

Cardiomyocytes usually display impaired contractile and electrical patterns in response to DDR activation, resulting in cardiomyopathies and arrhythmias.40 For example, lamin A (LMNA) and TMEM43 (transmembrane protein 43) mutations led to dilated cardiomyopathy (DCM) and arrhythmogenic cardiomyopathy, associated with genomic instability, DDR and p53 activation in cardiomyocytes.39,41 Besides, anthracyclines, a class of chemotherapeutic agents, trigger a DCM phenotype associated with cardiomyocyte DNA damage.42 These findings suggest an essential role of DNA damage and DDR activation, in particular through ATM signalling, in physiological cell cycle arrest and pathological hypertrophy in cardiomyocytes.

3.2 DDR in dividing cells in the cardiovascular system

In dividing cardiovascular cells, such as endothelial cells, VSMCs, cardiac fibroblasts (CFs), immune, and epithelial cells, DDR is characterized by ATM and p53 activation, reduced proliferation, cellular senescence, and apoptosis.6 Senescence is the major DDR consequence in dividing cardiovascular cells, characterized by cell cycle arrest, DNA damage, and up-regulated senescence markers (p16, p21, and p53), as well as elevated canonical SASP components, such as IL-6, CXC-chemokine ligand 1 (CXCL1) and CXCL2, TNF-α, vascular endothelial growth factor (VEGF), matrix metalloproteinase 1 (MMP1), MMP3, and plasminogen activator inhibitor 1 (PAI1).43

In VSMCs, DNA damage due to defective 8-oxoG BER is involved in cell cycle arrest, lost proliferative capacity, and senescence, through an ATM/ATR-p53-p21-dependent pathway.5,23 Abnormal processing of pre-lamin A compromised formation of the nuclear lamina, making VSMC more prone to DNA damage. Accumulation of pre-lamin A resulted from LMNA mutation was shown to promote VSMC senescence via DDR activation.44 Activation of DDR such as in SIRT6 deficiency was shown to promote VSMC senescence and atherosclerosis.45 Importantly, DDR activation in VSMC led to the progression and destabilization of atherosclerotic plaque.23 In brief, DNA damage leads to loss of proliferative potential of VSMCs, while senescent VSMC triggers plaque instability directly through shrinkage of VSMC content in fibrous cap (reduced VSMC proliferation and collagen synthesis).46 Further evidence also revealed a more proactive role for DNA damage from VSMC in plaque instability by fostering inflammation, secretion of MMPs, and inflammatory cytokines to favour matrix degradation.45–47

Increased senescence and apoptosis have been shown in endothelial cells in response to DNA damage, in an ATM/Akt/p53/p21-dependent manner.24,48 DDR activation in the endothelial cell leads to a pro-atherosclerotic, pro-thrombotic, and anti-angiogenic phenotype.49 Under DNA damage stress, endothelial cells undergo premature senescence, and secret pro-inflammatory mediators (IL-6, IL-8, and TNF-α), endothelin-1 (ET-1), chemokines, MMPs, and growth factors, contributing to endothelial dysfunction, ultimately atherosclerosis, pulmonary hypertension, and cardiac diseases.49

Several studies have reported a role for DDR in CFs.50–52 Normally, activation of DDR and cellular senescence in CFs would suppress CF proliferation and limit cardiac fibrosis.50 CCN1 (cellular communication network factor 1) belongs to SASP capable of inhibiting cardiac fibrosis through induction of CF senescence following MI, indicating a beneficial role from DDR and senescence of fibroblasts.51 Up-regulation of p53 following MI insult induces senescence of CFs. ATM activation was also noted in CFs in doxorubicin cardiotoxicity. ATM activation turns on Fas ligand in CF to interact with Fas and regulates cardiomyocyte apoptosis.52

3.3 Crosstalk among various cells in the cardiovascular system during DDR and senescence

Recent studies have noted a rather complex interplay among various cardiovascular cells during DDR and senescence, through the paracrine SASP components, leading to unfavourable consequences such as fibroblast activation and fibrosis, prothrombotic phenotype in endothelial cells, and cardiomyocyte hypertrophy and death33,53 (Figure 3). For example, telomeric DNA damage triggers cardiomyocyte secretion of EDN3, TGF-β, and GDF15, leading to myofibroblast activation and collagen accumulation.33 Besides, inflammatory cytokines (IL-1, IL-6, and TNF-α) released by senescent cardiomyocytes induce local cardiac inflammation.51 CFs under DDR activation also communicate through paracrine signalling to regulate cardiomyocyte senescence and hypertrophic growth.40 In heart injury, IGF-1 (insulin-like growth factor-1) is produced by CFs, leading to collagen synthesis and cardiomyocyte hypertrophy.54 Senescent endothelial cells show decreased nitric oxide production and elevated ET-1 release, leading to crosstalk between surrounding cardiac cell populations.40 This interaction contributes to vascular inflammation and impaired vasodilation. Taken together, DNA damage triggers cellular senescence in various cardiac cell types, while their crosstalk via SASP secretome leads to unfavourable consequences, including fibrosis, myofibroblast activation, collagen synthesis, hypertrophy, and inflammation, thereby drives the development of CVDs.

DNA damage-induced senescence in different cardiac cell types leads to cardiovascular pathologies. DNA damage activates a signalling cascade involving DDR via ATR and ATM kinases, CHK1 and CHK2 kinases, p53 up-regulation, and increased p21, leading to cellular senescence. Senescent cells in the cardiovascular system can adversely influence the neighbouring cells via SASP products. Fibroblasts, endothelial cells, and cardiomyocytes secrete a number of SASP molecules, such as IL-6, CXCL1, CXCL2, MMPs, and TGFβ2. Detrimental consequences of SASP can be mediated by autocrine or paracrine effects, including fibroblast activation and cardiac fibrosis; decreased eNOS activity and a switch of prothrombotic phenotype in endothelial cells; as well as cardiomyocyte death and hypertrophy. ATM, ataxia–telangiectasia mutated; ATR, ataxia–telangiectasia and Rad3-related; CHK, checkpoint kinase; CXCL, CXC-chemokine ligand; EDN3, endothelin 3; eNOS, endothelial nitric oxide synthase; GDF15, growth differentiation factor 15; MMP, matrix metalloproteinase; PAI1 plasminogen activator inhibitor 1; SASP, senescence-associated secretory phenotype; TGFβ2, transforming growth factor β2; TNF, tumour necrosis factor; VEGF, vascular endothelial growth factor; IL-6, interleukin 6.
Figure 3

DNA damage-induced senescence in different cardiac cell types leads to cardiovascular pathologies. DNA damage activates a signalling cascade involving DDR via ATR and ATM kinases, CHK1 and CHK2 kinases, p53 up-regulation, and increased p21, leading to cellular senescence. Senescent cells in the cardiovascular system can adversely influence the neighbouring cells via SASP products. Fibroblasts, endothelial cells, and cardiomyocytes secrete a number of SASP molecules, such as IL-6, CXCL1, CXCL2, MMPs, and TGFβ2. Detrimental consequences of SASP can be mediated by autocrine or paracrine effects, including fibroblast activation and cardiac fibrosis; decreased eNOS activity and a switch of prothrombotic phenotype in endothelial cells; as well as cardiomyocyte death and hypertrophy. ATM, ataxia–telangiectasia mutated; ATR, ataxia–telangiectasia and Rad3-related; CHK, checkpoint kinase; CXCL, CXC-chemokine ligand; EDN3, endothelin 3; eNOS, endothelial nitric oxide synthase; GDF15, growth differentiation factor 15; MMP, matrix metalloproteinase; PAI1 plasminogen activator inhibitor 1; SASP, senescence-associated secretory phenotype; TGFβ2, transforming growth factor β2; TNF, tumour necrosis factor; VEGF, vascular endothelial growth factor; IL-6, interleukin 6.

4. Evidence of DNA damage and DDR in cardiovascular pathology

Emerging evidence has depicted a vital role of DNA damage and DDR in the aetiology of CVDs. As demonstrated in Table 1, DNA damage and DDR occur in multiple cardiovascular pathologies, including HF, arrhythmia, ischaemic heart disease (IHD), metabolic cardiomyopathy, atherosclerosis, and hypertension. Besides, genetically engineered mouse models for genes involved in DDR display pathological cardiovascular phenotypes (Table 2), supporting a role for DNA damage and DDR in the pathogenesis of CVDs.

Table 1

Emerging evidence for DNA damage and DDR in patients with CVDs

CVDSample sourceDNA damage and DDR marker alterationsMajor clinical outcomeRef
HFSerum8-OHdG↑Higher serum 8-OHdG levels in HF patients, elevated with disease severity55
Myocardiump-ATM↑, 8-oxoG↑, γH2AX↑, NBS1↑Increased oxidative DNA damage and DDR activation in human failing hearts7,56
DCMEMBsPAR↑, γH2AX↑Decreased PAR and γH2AX levels in patients with LVRR57
Serum and EMBs8-OHdG↑Increased 8-OHdG levels in serum and myocardium of HF patients58
AFUrine8-OHdG↑Elevated oxidative DNA damage in AF patients, reversed with the restoration of sinus rhythm59
Serum8-OHdG↑Gradually and significantly increased 8-OHdG levels with AF progression60,61
I/RLVγH2AX↑, p-ATM↑, p53↑Extensive DNA damage and ATM activation in myocardial ischaemia62
Myocardial ischaemiaAtrium and LVBRCA1↑, γH2AX↑DSBs and up-regulated BRCA1 to repair DNA damage in myocardial ischaemia9
CADPBMCDifferentially expressedDifference in DDR genes in PBMCs between SA and NSTEMI63
AtherosclerosisCarotid plaques8-oxoG↑, PARP1↑, p53↑, DNA-PK↑Increased oxidative DNA damage and DDR in human atherosclerotic plaques64
Plaque VSMCsMRE11↑, RAD50↑, NBS1↑, γH2AX↑Increased DSBs and DDR activation in human atherosclerotic plaque VSMCs, altered plaque phenotype and suppressed fibrous cap areas in advanced lesions upon VSMC DNA damage23
HypertensionUrine8-OHdG↑Urine 8-OHdG used as a marker for oxidative DNA damage in patients with hypertension65
LymphocyteNot assessedIncreased oxidative DNA damage in hypertensive patients66
PAHPulmonary arteries53BP1↑, γH2AX↑, PARP1↑DNA damage/PARP1 signalling is essential for PAH development67
PAECs and PBMCsγH2AX↑Elevated DNA damage in PAH68
CVDSample sourceDNA damage and DDR marker alterationsMajor clinical outcomeRef
HFSerum8-OHdG↑Higher serum 8-OHdG levels in HF patients, elevated with disease severity55
Myocardiump-ATM↑, 8-oxoG↑, γH2AX↑, NBS1↑Increased oxidative DNA damage and DDR activation in human failing hearts7,56
DCMEMBsPAR↑, γH2AX↑Decreased PAR and γH2AX levels in patients with LVRR57
Serum and EMBs8-OHdG↑Increased 8-OHdG levels in serum and myocardium of HF patients58
AFUrine8-OHdG↑Elevated oxidative DNA damage in AF patients, reversed with the restoration of sinus rhythm59
Serum8-OHdG↑Gradually and significantly increased 8-OHdG levels with AF progression60,61
I/RLVγH2AX↑, p-ATM↑, p53↑Extensive DNA damage and ATM activation in myocardial ischaemia62
Myocardial ischaemiaAtrium and LVBRCA1↑, γH2AX↑DSBs and up-regulated BRCA1 to repair DNA damage in myocardial ischaemia9
CADPBMCDifferentially expressedDifference in DDR genes in PBMCs between SA and NSTEMI63
AtherosclerosisCarotid plaques8-oxoG↑, PARP1↑, p53↑, DNA-PK↑Increased oxidative DNA damage and DDR in human atherosclerotic plaques64
Plaque VSMCsMRE11↑, RAD50↑, NBS1↑, γH2AX↑Increased DSBs and DDR activation in human atherosclerotic plaque VSMCs, altered plaque phenotype and suppressed fibrous cap areas in advanced lesions upon VSMC DNA damage23
HypertensionUrine8-OHdG↑Urine 8-OHdG used as a marker for oxidative DNA damage in patients with hypertension65
LymphocyteNot assessedIncreased oxidative DNA damage in hypertensive patients66
PAHPulmonary arteries53BP1↑, γH2AX↑, PARP1↑DNA damage/PARP1 signalling is essential for PAH development67
PAECs and PBMCsγH2AX↑Elevated DNA damage in PAH68

LVRR, left ventricular reverse remodelling; PAR, poly (ADP-ribose); PBMC, peripheral blood mononuclear cell.

Table 1

Emerging evidence for DNA damage and DDR in patients with CVDs

CVDSample sourceDNA damage and DDR marker alterationsMajor clinical outcomeRef
HFSerum8-OHdG↑Higher serum 8-OHdG levels in HF patients, elevated with disease severity55
Myocardiump-ATM↑, 8-oxoG↑, γH2AX↑, NBS1↑Increased oxidative DNA damage and DDR activation in human failing hearts7,56
DCMEMBsPAR↑, γH2AX↑Decreased PAR and γH2AX levels in patients with LVRR57
Serum and EMBs8-OHdG↑Increased 8-OHdG levels in serum and myocardium of HF patients58
AFUrine8-OHdG↑Elevated oxidative DNA damage in AF patients, reversed with the restoration of sinus rhythm59
Serum8-OHdG↑Gradually and significantly increased 8-OHdG levels with AF progression60,61
I/RLVγH2AX↑, p-ATM↑, p53↑Extensive DNA damage and ATM activation in myocardial ischaemia62
Myocardial ischaemiaAtrium and LVBRCA1↑, γH2AX↑DSBs and up-regulated BRCA1 to repair DNA damage in myocardial ischaemia9
CADPBMCDifferentially expressedDifference in DDR genes in PBMCs between SA and NSTEMI63
AtherosclerosisCarotid plaques8-oxoG↑, PARP1↑, p53↑, DNA-PK↑Increased oxidative DNA damage and DDR in human atherosclerotic plaques64
Plaque VSMCsMRE11↑, RAD50↑, NBS1↑, γH2AX↑Increased DSBs and DDR activation in human atherosclerotic plaque VSMCs, altered plaque phenotype and suppressed fibrous cap areas in advanced lesions upon VSMC DNA damage23
HypertensionUrine8-OHdG↑Urine 8-OHdG used as a marker for oxidative DNA damage in patients with hypertension65
LymphocyteNot assessedIncreased oxidative DNA damage in hypertensive patients66
PAHPulmonary arteries53BP1↑, γH2AX↑, PARP1↑DNA damage/PARP1 signalling is essential for PAH development67
PAECs and PBMCsγH2AX↑Elevated DNA damage in PAH68
CVDSample sourceDNA damage and DDR marker alterationsMajor clinical outcomeRef
HFSerum8-OHdG↑Higher serum 8-OHdG levels in HF patients, elevated with disease severity55
Myocardiump-ATM↑, 8-oxoG↑, γH2AX↑, NBS1↑Increased oxidative DNA damage and DDR activation in human failing hearts7,56
DCMEMBsPAR↑, γH2AX↑Decreased PAR and γH2AX levels in patients with LVRR57
Serum and EMBs8-OHdG↑Increased 8-OHdG levels in serum and myocardium of HF patients58
AFUrine8-OHdG↑Elevated oxidative DNA damage in AF patients, reversed with the restoration of sinus rhythm59
Serum8-OHdG↑Gradually and significantly increased 8-OHdG levels with AF progression60,61
I/RLVγH2AX↑, p-ATM↑, p53↑Extensive DNA damage and ATM activation in myocardial ischaemia62
Myocardial ischaemiaAtrium and LVBRCA1↑, γH2AX↑DSBs and up-regulated BRCA1 to repair DNA damage in myocardial ischaemia9
CADPBMCDifferentially expressedDifference in DDR genes in PBMCs between SA and NSTEMI63
AtherosclerosisCarotid plaques8-oxoG↑, PARP1↑, p53↑, DNA-PK↑Increased oxidative DNA damage and DDR in human atherosclerotic plaques64
Plaque VSMCsMRE11↑, RAD50↑, NBS1↑, γH2AX↑Increased DSBs and DDR activation in human atherosclerotic plaque VSMCs, altered plaque phenotype and suppressed fibrous cap areas in advanced lesions upon VSMC DNA damage23
HypertensionUrine8-OHdG↑Urine 8-OHdG used as a marker for oxidative DNA damage in patients with hypertension65
LymphocyteNot assessedIncreased oxidative DNA damage in hypertensive patients66
PAHPulmonary arteries53BP1↑, γH2AX↑, PARP1↑DNA damage/PARP1 signalling is essential for PAH development67
PAECs and PBMCsγH2AX↑Elevated DNA damage in PAH68

LVRR, left ventricular reverse remodelling; PAR, poly (ADP-ribose); PBMC, peripheral blood mononuclear cell.

Table 2

Cardiovascular phenotypes of DDR and DNA repair alterations in mouse models

ModelOrgan specificityCardiovascular stressCardiovascular phenotypeRef
PARP1 deficiencySystemicHFD and ApoE−/−Reduced atherosclerotic plaque formation and size69–72
I/RAttenuated myocardial injury, disrupted myocardial structure, mitochondrial dysfunction, MMP, and neutrophil infiltration73,74
HyperglycemiaReduced diabetic arteriosclerotic calcification, regulated synthetic phenotype switching of VSMC75
Ang II-induced cardiac hypertrophyAttenuated cardiac hypertrophy and interstitial fibrosis76
Ang II-induced AAADecreased AAA incidence, abdominal aortic diameter, and macrophage infiltration77
PARP2 deficiencySystemicDoxorubicinDecreased vascular damage and mitochondrial dysfunction78
ATM deficiencySystemicTACAttenuated LV dysfunction, improved mortality, and alleviated NF-κB-mediated inflammation29
MIAttenuated LV dysfunction, dilation and inflammation in early post-MI (1–7 days), exacerbated LV dysfunction, fibrosis, apoptosis, and cardiac hypertrophy in late post-MI (14–28 days)79–83
ApoE−/−Accelerated atherosclerosis and multiple features of metabolic syndrome84,85
HyperglycemiaAttenuated endothelial dysfunction and senescence induced by injection of STZ48
Cardiomyocyte-specificTACAttenuated cardiac hypertrophy21
Cardiac fibroblast-specificDoxorubicinAttenuated LV dysfunction, cardiac apoptosis and mortality52
DNA-PKcs deficiencyCardiacI/RReserved cardiac function, reduced myocardial injury, attenuated apoptosis and inflammation86
p53 deficiencyCardiacTACAttenuated cardiac hypertrophy, injury, and dysfunction87
SystemicMIReduced incidence of LV rupture post-MI88
BRCA1 deficiencyCardiomyocyte-specificMIIncreased mortality, exaggerated LV dilation, increased apoptosis, and impaired DSB repair9
NBS1 overexpressionVSMC-specificHFD and ApoE−/−Enhanced DSB repair, decreased apoptosis and growth arrest, increased fibrous cap area and plaque stability23
OGG1 deficiencySystemicWestern diet and Ldlr−/−Increased plaque size and lipid content, DNA damage, apoptosis, and inflammation31
ModelOrgan specificityCardiovascular stressCardiovascular phenotypeRef
PARP1 deficiencySystemicHFD and ApoE−/−Reduced atherosclerotic plaque formation and size69–72
I/RAttenuated myocardial injury, disrupted myocardial structure, mitochondrial dysfunction, MMP, and neutrophil infiltration73,74
HyperglycemiaReduced diabetic arteriosclerotic calcification, regulated synthetic phenotype switching of VSMC75
Ang II-induced cardiac hypertrophyAttenuated cardiac hypertrophy and interstitial fibrosis76
Ang II-induced AAADecreased AAA incidence, abdominal aortic diameter, and macrophage infiltration77
PARP2 deficiencySystemicDoxorubicinDecreased vascular damage and mitochondrial dysfunction78
ATM deficiencySystemicTACAttenuated LV dysfunction, improved mortality, and alleviated NF-κB-mediated inflammation29
MIAttenuated LV dysfunction, dilation and inflammation in early post-MI (1–7 days), exacerbated LV dysfunction, fibrosis, apoptosis, and cardiac hypertrophy in late post-MI (14–28 days)79–83
ApoE−/−Accelerated atherosclerosis and multiple features of metabolic syndrome84,85
HyperglycemiaAttenuated endothelial dysfunction and senescence induced by injection of STZ48
Cardiomyocyte-specificTACAttenuated cardiac hypertrophy21
Cardiac fibroblast-specificDoxorubicinAttenuated LV dysfunction, cardiac apoptosis and mortality52
DNA-PKcs deficiencyCardiacI/RReserved cardiac function, reduced myocardial injury, attenuated apoptosis and inflammation86
p53 deficiencyCardiacTACAttenuated cardiac hypertrophy, injury, and dysfunction87
SystemicMIReduced incidence of LV rupture post-MI88
BRCA1 deficiencyCardiomyocyte-specificMIIncreased mortality, exaggerated LV dilation, increased apoptosis, and impaired DSB repair9
NBS1 overexpressionVSMC-specificHFD and ApoE−/−Enhanced DSB repair, decreased apoptosis and growth arrest, increased fibrous cap area and plaque stability23
OGG1 deficiencySystemicWestern diet and Ldlr−/−Increased plaque size and lipid content, DNA damage, apoptosis, and inflammation31

AAA, abdominal aortic aneurysm; HFD, high-fat diet; STZ, streptozotocin; TAC, transverse aortic constriction.

Table 2

Cardiovascular phenotypes of DDR and DNA repair alterations in mouse models

ModelOrgan specificityCardiovascular stressCardiovascular phenotypeRef
PARP1 deficiencySystemicHFD and ApoE−/−Reduced atherosclerotic plaque formation and size69–72
I/RAttenuated myocardial injury, disrupted myocardial structure, mitochondrial dysfunction, MMP, and neutrophil infiltration73,74
HyperglycemiaReduced diabetic arteriosclerotic calcification, regulated synthetic phenotype switching of VSMC75
Ang II-induced cardiac hypertrophyAttenuated cardiac hypertrophy and interstitial fibrosis76
Ang II-induced AAADecreased AAA incidence, abdominal aortic diameter, and macrophage infiltration77
PARP2 deficiencySystemicDoxorubicinDecreased vascular damage and mitochondrial dysfunction78
ATM deficiencySystemicTACAttenuated LV dysfunction, improved mortality, and alleviated NF-κB-mediated inflammation29
MIAttenuated LV dysfunction, dilation and inflammation in early post-MI (1–7 days), exacerbated LV dysfunction, fibrosis, apoptosis, and cardiac hypertrophy in late post-MI (14–28 days)79–83
ApoE−/−Accelerated atherosclerosis and multiple features of metabolic syndrome84,85
HyperglycemiaAttenuated endothelial dysfunction and senescence induced by injection of STZ48
Cardiomyocyte-specificTACAttenuated cardiac hypertrophy21
Cardiac fibroblast-specificDoxorubicinAttenuated LV dysfunction, cardiac apoptosis and mortality52
DNA-PKcs deficiencyCardiacI/RReserved cardiac function, reduced myocardial injury, attenuated apoptosis and inflammation86
p53 deficiencyCardiacTACAttenuated cardiac hypertrophy, injury, and dysfunction87
SystemicMIReduced incidence of LV rupture post-MI88
BRCA1 deficiencyCardiomyocyte-specificMIIncreased mortality, exaggerated LV dilation, increased apoptosis, and impaired DSB repair9
NBS1 overexpressionVSMC-specificHFD and ApoE−/−Enhanced DSB repair, decreased apoptosis and growth arrest, increased fibrous cap area and plaque stability23
OGG1 deficiencySystemicWestern diet and Ldlr−/−Increased plaque size and lipid content, DNA damage, apoptosis, and inflammation31
ModelOrgan specificityCardiovascular stressCardiovascular phenotypeRef
PARP1 deficiencySystemicHFD and ApoE−/−Reduced atherosclerotic plaque formation and size69–72
I/RAttenuated myocardial injury, disrupted myocardial structure, mitochondrial dysfunction, MMP, and neutrophil infiltration73,74
HyperglycemiaReduced diabetic arteriosclerotic calcification, regulated synthetic phenotype switching of VSMC75
Ang II-induced cardiac hypertrophyAttenuated cardiac hypertrophy and interstitial fibrosis76
Ang II-induced AAADecreased AAA incidence, abdominal aortic diameter, and macrophage infiltration77
PARP2 deficiencySystemicDoxorubicinDecreased vascular damage and mitochondrial dysfunction78
ATM deficiencySystemicTACAttenuated LV dysfunction, improved mortality, and alleviated NF-κB-mediated inflammation29
MIAttenuated LV dysfunction, dilation and inflammation in early post-MI (1–7 days), exacerbated LV dysfunction, fibrosis, apoptosis, and cardiac hypertrophy in late post-MI (14–28 days)79–83
ApoE−/−Accelerated atherosclerosis and multiple features of metabolic syndrome84,85
HyperglycemiaAttenuated endothelial dysfunction and senescence induced by injection of STZ48
Cardiomyocyte-specificTACAttenuated cardiac hypertrophy21
Cardiac fibroblast-specificDoxorubicinAttenuated LV dysfunction, cardiac apoptosis and mortality52
DNA-PKcs deficiencyCardiacI/RReserved cardiac function, reduced myocardial injury, attenuated apoptosis and inflammation86
p53 deficiencyCardiacTACAttenuated cardiac hypertrophy, injury, and dysfunction87
SystemicMIReduced incidence of LV rupture post-MI88
BRCA1 deficiencyCardiomyocyte-specificMIIncreased mortality, exaggerated LV dilation, increased apoptosis, and impaired DSB repair9
NBS1 overexpressionVSMC-specificHFD and ApoE−/−Enhanced DSB repair, decreased apoptosis and growth arrest, increased fibrous cap area and plaque stability23
OGG1 deficiencySystemicWestern diet and Ldlr−/−Increased plaque size and lipid content, DNA damage, apoptosis, and inflammation31

AAA, abdominal aortic aneurysm; HFD, high-fat diet; STZ, streptozotocin; TAC, transverse aortic constriction.

4.1 HF and cardiac remodelling

The involvement of DNA damage in the aetiology of HF has drawn some recent attention. Overt DNA damage (e.g. oxidative DNA damage, SSB, and DSB) or DDR activation is observed in patients with end-stage HF55–57,89 and pressure overload-induced murine model of HF.21,29,90 The end-stage cardiomyopathic human hearts presented marked DNA damage and DDR activation, as evidenced by increased 8-oxoG, γH2AX, ATM phosphorylation, and NBS1.7 Besides, these DNA damage markers are found in not only heart tissues but also peripheral blood and urine. In a study including 56 HF patients and 20 healthy controls, elevated levels of 8-hydroxy-2′-deoxyguanosine (8-OHdG) were noted in both serum and myocardium of patients.58 In another study, serum 8-OHdG levels were higher in HF patients, in particular advanced New York Heart Association classes.55 Furthermore, the incidence of cardiac events was significantly higher in patients with high 8-OHdG levels during the follow-up. In a meta-analysis including six independent studies, 8-OHdG levels are drastically higher in urine and blood samples from HF patients.91 These findings suggest the presence of extensive oxidative stress and DNA damage in HF, and the extent of DNA damage correlates to the disease severity in human samples.

Mechanistically, current available evidence has revealed that DNA damage in HF is caused by deficiency of both OGG1-mediated BER and SSB repair.7,29 ATM activation has recently garnered much attention as a critical DDR regulator closely related to myocardial inflammation, cardiac remodelling, and eventually HF development. ATM deletion was shown to suppress the activation of the nuclear factor-κB (NF-κB) pathway, cardiac inflammation, and HF progression.29 In another parallel study, pressure overload led to DNA DSBs (specifically indicated by DNA-PKcs phosphorylation) in cardiomyocytes, triggering subsequent ATM-mediated DDR activation.21 Mechanistically, activated ATM phosphorylated and degraded Cain, leading to calcineurin activation. Besides, ATM also phosphorylated 4E-BP1 (eukaryotic translation initiation factor 4E-binding protein 1) to initiate protein synthesis, triggering cardiac hypertrophy. Both calcineurin and 4E-BP1 served as mediators of cardiomyocyte hypertrophy in response to DNA DSBs and ATM activation. Accordingly, genetic or pharmacological inhibition of ATM attenuated pressure overload-induced cardiomyocyte hypertrophy.21 These studies suggested that overwhelmed DNA damage triggers ATM activation in failing hearts. ATM-mediated DDR activation may represent a crucial pathway to initiate cardiac inflammation and remodelling, promoting HF development, by phosphorylation of key substrates related to cardiac pathologies.

4.2 Cardiac arrhythmia

A limited number of studies have shown the presence of DNA damage (mostly oxidative DNA damage) in arrhythmias, in particular atrial fibrillation (AF).37,59–61,92,93 DNA damage is noted in both tachypaced atrial cardiomyocytes and atrial tissue samples from persistent AF patients.37 Importantly, 8-OHdG, a classical oxidative DNA damage marker, can be detectable in blood and urine samples, depicting their potential as biomarkers for AF.59 Besides, 8-OHdG level was greatly reduced following rhythm restoration. In another recent study, serum 8-OHdG levels were gradually elevated in AF progression.60 Notably, a significant rise in 8-OHdG levels was noted in patients with AF recurrence, as compared to those without AF recurrence. These clinical findings favour the utility of 8-OHdG as a diagnostic, severity, and recurrent biomarker for AF.

A recent study explored the potential mechanisms linking DDR to the pathogenesis of AF. PARP1 activation might foster cardiomyocyte dysfunction in an experimental AF model. Oxidative DNA damage was precipitated in atrial cardiomyocytes, leading to excess PARP1 activation, nicotinamide adenine dinucleotide (NAD+) depletion, and subsequent electrical and contractile impairment.37 NAD+ loss was shown to cause a progressive decline in ATP levels, energy stress, and cell death during excessive PARP1 activation. Accordingly, PARP1 depletion or NAD+ replenishment was demonstrated to retard AF progression. Targeting PARP1 should represent a novel therapeutic approach to preserve cardiac function in clinical AF.

4.3 IHD

Infarcted hearts exhibit various types of DNA damage including oxidative DNA damage, SSB, and DSB. DDR activation plays an essential role in post-infarction remodelling through induction of cardiomyocyte apoptosis.9,94–96 Corbucci et al.62 first noted DNA damage and ATM activation following acute myocardial ischaemia. In their study, myocardial ischaemia up-regulated p53 and γH2AX levels as well as ATM activation in human left ventricular samples, indicating the presence of DSBs in ischaemic hearts. In a clinical study involving patients with stable angina and non-ST-segment elevation MI, peripheral blood mononuclear cells exhibited evident differences in DNA ligase activity and DDR genes.63

Current evidence has suggested that BRCA1 and ATM represent the two most important DDR proteins regulating the development of MI. BRCA1 depletion in cardiomyocytes led to compromised HR repair and p53 activation, resulting in ventricular dysfunction, adverse cardiac remodelling, and increased mortality in response to ischaemic stress.9 In addition, post-MI ATM activation was shown to regulate cardiac remodelling, inflammation, and systolic function79,80,97,98 (discussed in Section 7). Taken together, DDR activation is involved in the pathogenesis of IHD.

4.4 Metabolic cardiomyopathy

Ample evidence has noted the accumulation of DNA damage in metabolic disorders, including obesity99 and diabetes mellitus.100 Recently, a close interplay was noted between metabolic abnormalities and cardiovascular dysfunction through DNA damage.101–103ob/ob and db/db mice demonstrated increased apoptosis in cardiomyocytes, associated with increased DNA damage and premature mortality.101 HF was reported to trigger DNA damage in both myocardial tissues and visceral fat, thereby promoting inflammation of adipose tissues and systemic insulin resistance.102 HF-related overactivation of the sympathetic nervous system led to oxidative stress and subsequent DNA damage via p53-dependent signalling. Interestingly, p53 deletion inhibited adipose tissue inflammation and improved metabolic and cardiac dysfunction, indicating a vicious circle between adipose tissues and hearts.102 In another study, high-caloric diet feeding led to up-regulated p53 in endothelial cells, due to DNA damage associated with metabolic stress.103 Inhibition of endothelial p53 activation improved insulin resistance and obesity, suggesting a potential role of DNA damage linking diabetes to vascular function. These findings demonstrate the promises of suppressing DDR activation in the interruption of the vicious cycle between metabolic disorders and CVDs.

4.5 Cancer therapy-related cardiotoxicity

The field of cardio-oncology is evolved given the ever-rising prevalence of CVDs in cancer patients. Chemotherapeutic drugs and radiation therapy are tied with cardiotoxicity and risks of HF, coronary artery disease, and arrhythmia.104 Besides, the recent advance has demonstrated a more complex relationship between cancer and CVDs, leading to a new concept of CVD-induced cancer incidence termed ‘reverse cardio-oncology’.105,106 A number of mechanisms have been postulated to explain overtly increased cancer risk in CVD patients, including enrolment of inflammatory mediators, hypoxia, and secretion of circulating factors (e.g. extracellular vesicles, cardiokines, and microRNAs) from cardiovascular system.107 Herein we only focus on the role of DNA damage in chemotherapy-related cardiotoxicity. The precise mechanisms of cardiotoxicity are complex and may involve (but not restrict to) oxidative stress, DNA damage, mitochondrial dysfunction, apoptosis, and other regulated cell deaths.104 To date, compelling evidence suggests a cardinal role for DDR activation in radiation and chemotherapy-induced cardiomyopathy.40,42,52,108 For example, heart samples from patients receiving doxorubicin exhibited DNA damage, cell senescence, cardiac progenitor cell (CPC) migration impairment, and increased ROS production in the majority of CPCs.109

Several animal models have been explored to unveil the mechanisms underlying cardiac DNA damage and DDR in doxorubicin-challenged animals.42,52,110,111 Doxorubicin was shown to induce DSBs in cardiomyocytes via topoisomerase 2β, while cardiomyocyte-specific deletion of topoisomerase 2β protected cardiomyocytes from doxorubicin-induced DSBs, defective mitochondrial biogenesis, and ROS accumulation.42 Chronic doxorubicin-induced cardiotoxicity is mediated by the oxidative DNA damage-ATM-p53-apoptosis axis in cardiomyocytes.110 Further evidence revealed ATM activation in CFs, associated with elevated Fas ligand and cardiomyocyte apoptosis.52 In addition, doxorubicin increased the cardiac level of p53 which binds to the promoter of Ppargc1a (encoding PGC1α to regulate mitochondrial function), contributing to dampened Ppargc1a expression,112 suggesting a role of DDR in regulating mitochondrial dysfunction after doxorubicin therapy.112 These findings depict a crucial role for DDR activation in the complex interplay with oxidative stress and mitochondrial dysfunction, en route to the development of doxorubicin cardiotoxicity.

Given the essential role of DDR activation in doxorubicin cardiotoxicity, inhibition of DDR components may be a novel therapeutic approach for chemotherapy-related cardiotoxicity. At present, only one study showed that systemic administration of ATM inhibitor (KU55933) was able to prevent doxorubicin cardiotoxicity in animal models.52 Notably, genomic instability is a promising hallmark of cancer owing to defects in DDR and replication stress.13 A number of DDR inhibitors have been proven to be effective in various types of cancers from pre-clinical findings.13 Moreover, these drugs exhibited synergy with classical chemotherapy, radiotherapy, and immunotherapy.13 Therefore, the translational perspective may reside in that DDR inhibitors offer therapeutic advantage by simultaneously diminishing doxorubicin cardiotoxicity while boosting its anticancer effect. To the best of our knowledge, few studies have examined the role of DDR inhibition in animal models with concurrent cancer and cardiotoxicity.

4.6 Atherosclerosis

Ample evidence has revealed DNA damage and DDR in both plaques and circulating monocytes in atherosclerosis.23,113 VSMCs and inflammatory cells within plaques exhibit various types of DNA damage, including oxidation of bases, strand breaks, and telomere shortening.64,114,115 DNA damage and DDR activation increase in proportion to the disease severity. Besides, these cells within atherosclerotic plaques exhibit a series of consequences of DNA damage including premature senescence, apoptosis, and inflammation.116–118 Human atherosclerotic VSMCs exhibit evident DNA DSBs and DDR activation as evidenced by increased MRN complex.23 VSMC DNA damage was shown to alter plaque phenotype through inhibiting fibrous cap in advanced lesions. Thus, inhibition of DNA damage in atherosclerotic plaque may represent a novel approach to promote plaque stability.23

Mechanistically, defective BER due to OGG1 deficiency is considered the major cause for DNA damage in atherosclerosis.5,31 Overexpression of OGG1 in VSMCs alleviated DNA damage, cellular senescence, apoptosis, and NLRP3 inflammasome activation, thereby leading to reduced atherosclerotic plaque formation.5 These findings favour the presence of a vicious cycle among oxidative DNA damage, decreased OGG1 activity, and defective BER in the development of atherosclerosis. Moreover, PARP1 also serves as a critical DDR protein in atherogenesis.69 Genetic and pharmacological inhibition of PARP1 was shown to interfere with plaque development and prevent plaque instability, through inhibition of NF-κB and inflammation in plaque dynamics. Collectively, DNA damage and DDR activation are involved in the onset and progression of atherosclerosis.

4.7 Hypertension and pulmonary artery hypertension

Only a handful of studies suggested a role for DNA damage in the aetiology of hypertension. Oxidative DNA damage is prominent in patients with hypertension.66 Interestingly, DNA damage and oxidative stress were significantly attenuated following anti-hypertensive therapy. Urine 8-OHdG was elevated in hypertensive patients.65 Thus, urine 8-OHdG levels may have promises as a marker for oxidative DNA damage in hypertension.

Although limited evidence favours a link between DNA damage and hypertension, possible roles of DNA damage and DDR activation in the pathogenesis of pulmonary artery hypertension (PAH) and associated right ventricular dysfunction have been unveiled.67,68,119,120 Genetic mutations were involved in PAH pathogenesis, as evidenced by a monoclonal origin of pulmonary artery endothelial cells (PAECs) in the majority of plexiform lesions in idiopathic PAH.121,122 Subsequent studies showed that DNA damage and abnormal DNA repair provoked pulmonary vascular cell proliferation and vascular resistance in PAH progression.67,119,120 DNA damage in PAECs, VSMCs, and peripheral blood mononuclear cells precedes the clinical manifestation of PAH, as shown by the elevated susceptibility to DNA damage prior to disease onset.68 DNA damage may be attributable to ROS, inflammation, and hypoxia stress in PAH, and such damage persists for years even after the removal of the triggering exposure.123

Among DDR machineries, PARP1 is the most extensively studied in PAH, as overactivation of PARP1 was noted in PAH and induced activation of inflammatory response and cell dysfunction. PARP1 inhibition was shown to reverse miR-204-mediated up-regulation of transcription factors nuclear factor of activated T cells (NFAT) and hypoxia-inducible factor 1-α (HIF-1α), contributing to the proliferation of pulmonary VSMCs. Genetic and pharmacological inhibition of PARP1 improved pulmonary vascular remodelling, endothelial function, and right ventricular hypertrophy and remodelling.67,124

5. Relationship between DNA damage and CVDs in laminopathies

LMNA (lamin A/C) is a nuclear lamina protein that interacts with the genome to regulate gene expression. Mutations in LMNA cause a group of divergent tissue-specific disorders, including DCM, termed as laminopathies.125 These disorders can also be caused by defects in the lamin interacting partners including TMEM43, and nesprins.126 The linkers of the nucleoskeleton and cytoskeleton (LINC) complex are composed of nesprins 1–4 in the outer nuclear membrane, and SUN1/2 in the inner nuclear membrane. Disruption of these proteins is shown to evoke cardiomyopathies. Previous studies suggested an important role of DDR activation in pre-lamin A (Lamin A precursor protein) accumulation in VSMC senescence and vascular calcification.44,127 Recent findings indicated a role for nuclear lamina in the DDR. Disruption of the nuclear lamina is closely tied with DCM.39 Mice with cardiac-specific LMNA missense mutation p.Asp300Asn (LMNAD300N), which is associated with DCM and progeroid syndrome in cardiomyocytes,39 presented DCM phenotype including severe cardiac dysfunction, myocardial fibrosis, apoptosis, and premature death. Further cardiac transcriptome analysis revealed that these phenotypes were related to aberrant activation of the E2F transcription factor-DDR–p53 pathway. Targeted p53 deletion in cardiomyocytes rescued these phenotypes. This study suggested that DDR/p53 pathway is activated to drive the pathogenesis of DCM associated with Lamin A/C mutations.

Similar myopathies were noted in mice deficient in Lamin A/C interacting nuclear envelope protein TMEM43.41 Activation of DDR, p53, and SASP was observed in cardiomyocytes with TMEM43 haploinsufficiency, in association with increased mortality, apoptosis, and a pro-fibrotic cardiomyopathy phenotype in mice. In addition, nesprin-2, initially identified as a nuclear envelope Lamin A-binding protein, was shown to regulate DDR activation in pre-lamin accumulation and VSMC senescence.128 A recent study noted a critical role of the cytoskeleton to maintain nuclear lamina structure in cardiomyocytes.129 Acute loss of desmin or its LINC-binding partner nesprin-3, led to abnormal nuclear morphology in cardiomyocytes, accompanied with functional defects in Ca2+ cycling and contractility. Desmin depletion led to DNA damage and compromised key lamina-associated chromatin structures. These findings have demonstrated the key regulatory role of lamina-interacting LINC and cytoskeletal components in DNA damage, DDR, and nuclear homeostasis. Taken together, DNA damage and DDR may represent potential therapeutic targets in cardiomyopathies caused by the LMNA and associated gene mutations.

6. Therapeutic approach to combat DNA damage in CVDs

Given that accumulation of DNA damage greatly hampers cellular function and increases susceptibility for CVDs, therapeutic interventions should be targeted towards genomic instability and DNA damage. Alleviation of exogenous DNA damage, such as tobacco smoke and UV exposure, lowers morbidity and mortality of CVDs. Therapies including antioxidants and dietary intervention have been established to combat endogenous DNA damage. Emerging evidence has also unveiled efficacy towards DNA damage for multiple cardiovascular drugs including statins, angiotensin-converting enzyme inhibitors (ACEIs)/angiotensin II receptor blockers (ARBs), and β-blockers (Table 3). These findings further demonstrate the promises of targeting DNA damage in the management of CVDs.

Table 3

Emerging evidence for cardiovascular drugs to reduce DNA damage

CV drugsCV stressMechanism of action and cardiovascular protective propertiesRef
Statins
AtorvastatinHypercholesterolemiaReduced plasma TC, LDL-C, and TG, attenuated oxidative protein and DNA damage (plasma 8-OHdG level)130
AtorvastatinAtherosclerosisAccelerated NBS-1-mediated DNA repair, reduced VSMC senescence and telomere attrition, attenuated ATM/ATR activation, reduced apoptosis115
SimvastatinAtherosclerosisReduced chromosomal damage (sister-chromatid exchanges and high-frequency cells)131
SimvastatinMyocardial I/RReduced cardiac oxidative damage (8-OHdG level) and apoptosis, protection against I/R injury132
PitavastatinDoxorubicin-induced cardiotoxicityAttenuated p53 accumulation, ATM activation, and oxidative stress, reduced cardiomyocyte apoptosis110
ACEIs/ARBs
CaptoprilClozapine-induced myocarditisAttenuated oxidative stress, NO, and cardiac and serum 8-OHdG levels, reduced biochemical markers (CK-MB and LDH) of myocarditis133
Ramipril ValsartanMIReduced oxidative stress and 8-oxoG levels134
CandesartanHypertensionDecreased blood pressure, oxidative stress, and DNA damage markers (γH2AX, PAR, and 8-oxoG) in kidneys and heart135
β-blockers
CarvedilolDCMDecreased serum 8-OHdG levels (oxidative DNA damage) and ameliorates HF58
CarvedilolDoxorubicin-induced cardiotoxicityAttenuated doxorubicin-induced DNA damage (γH2AX), myocardial apoptosis, fibrosis, hypertrophy, and cardiac function136
CarvedilolHypertensionReduced plasma 8-OHdG level137
CV drugsCV stressMechanism of action and cardiovascular protective propertiesRef
Statins
AtorvastatinHypercholesterolemiaReduced plasma TC, LDL-C, and TG, attenuated oxidative protein and DNA damage (plasma 8-OHdG level)130
AtorvastatinAtherosclerosisAccelerated NBS-1-mediated DNA repair, reduced VSMC senescence and telomere attrition, attenuated ATM/ATR activation, reduced apoptosis115
SimvastatinAtherosclerosisReduced chromosomal damage (sister-chromatid exchanges and high-frequency cells)131
SimvastatinMyocardial I/RReduced cardiac oxidative damage (8-OHdG level) and apoptosis, protection against I/R injury132
PitavastatinDoxorubicin-induced cardiotoxicityAttenuated p53 accumulation, ATM activation, and oxidative stress, reduced cardiomyocyte apoptosis110
ACEIs/ARBs
CaptoprilClozapine-induced myocarditisAttenuated oxidative stress, NO, and cardiac and serum 8-OHdG levels, reduced biochemical markers (CK-MB and LDH) of myocarditis133
Ramipril ValsartanMIReduced oxidative stress and 8-oxoG levels134
CandesartanHypertensionDecreased blood pressure, oxidative stress, and DNA damage markers (γH2AX, PAR, and 8-oxoG) in kidneys and heart135
β-blockers
CarvedilolDCMDecreased serum 8-OHdG levels (oxidative DNA damage) and ameliorates HF58
CarvedilolDoxorubicin-induced cardiotoxicityAttenuated doxorubicin-induced DNA damage (γH2AX), myocardial apoptosis, fibrosis, hypertrophy, and cardiac function136
CarvedilolHypertensionReduced plasma 8-OHdG level137
Table 3

Emerging evidence for cardiovascular drugs to reduce DNA damage

CV drugsCV stressMechanism of action and cardiovascular protective propertiesRef
Statins
AtorvastatinHypercholesterolemiaReduced plasma TC, LDL-C, and TG, attenuated oxidative protein and DNA damage (plasma 8-OHdG level)130
AtorvastatinAtherosclerosisAccelerated NBS-1-mediated DNA repair, reduced VSMC senescence and telomere attrition, attenuated ATM/ATR activation, reduced apoptosis115
SimvastatinAtherosclerosisReduced chromosomal damage (sister-chromatid exchanges and high-frequency cells)131
SimvastatinMyocardial I/RReduced cardiac oxidative damage (8-OHdG level) and apoptosis, protection against I/R injury132
PitavastatinDoxorubicin-induced cardiotoxicityAttenuated p53 accumulation, ATM activation, and oxidative stress, reduced cardiomyocyte apoptosis110
ACEIs/ARBs
CaptoprilClozapine-induced myocarditisAttenuated oxidative stress, NO, and cardiac and serum 8-OHdG levels, reduced biochemical markers (CK-MB and LDH) of myocarditis133
Ramipril ValsartanMIReduced oxidative stress and 8-oxoG levels134
CandesartanHypertensionDecreased blood pressure, oxidative stress, and DNA damage markers (γH2AX, PAR, and 8-oxoG) in kidneys and heart135
β-blockers
CarvedilolDCMDecreased serum 8-OHdG levels (oxidative DNA damage) and ameliorates HF58
CarvedilolDoxorubicin-induced cardiotoxicityAttenuated doxorubicin-induced DNA damage (γH2AX), myocardial apoptosis, fibrosis, hypertrophy, and cardiac function136
CarvedilolHypertensionReduced plasma 8-OHdG level137
CV drugsCV stressMechanism of action and cardiovascular protective propertiesRef
Statins
AtorvastatinHypercholesterolemiaReduced plasma TC, LDL-C, and TG, attenuated oxidative protein and DNA damage (plasma 8-OHdG level)130
AtorvastatinAtherosclerosisAccelerated NBS-1-mediated DNA repair, reduced VSMC senescence and telomere attrition, attenuated ATM/ATR activation, reduced apoptosis115
SimvastatinAtherosclerosisReduced chromosomal damage (sister-chromatid exchanges and high-frequency cells)131
SimvastatinMyocardial I/RReduced cardiac oxidative damage (8-OHdG level) and apoptosis, protection against I/R injury132
PitavastatinDoxorubicin-induced cardiotoxicityAttenuated p53 accumulation, ATM activation, and oxidative stress, reduced cardiomyocyte apoptosis110
ACEIs/ARBs
CaptoprilClozapine-induced myocarditisAttenuated oxidative stress, NO, and cardiac and serum 8-OHdG levels, reduced biochemical markers (CK-MB and LDH) of myocarditis133
Ramipril ValsartanMIReduced oxidative stress and 8-oxoG levels134
CandesartanHypertensionDecreased blood pressure, oxidative stress, and DNA damage markers (γH2AX, PAR, and 8-oxoG) in kidneys and heart135
β-blockers
CarvedilolDCMDecreased serum 8-OHdG levels (oxidative DNA damage) and ameliorates HF58
CarvedilolDoxorubicin-induced cardiotoxicityAttenuated doxorubicin-induced DNA damage (γH2AX), myocardial apoptosis, fibrosis, hypertrophy, and cardiac function136
CarvedilolHypertensionReduced plasma 8-OHdG level137

6.1 Natural compounds and antioxidants

Several natural compounds capable of reducing DNA damage have displayed promises in the treatment of CVDs.138–140 Cardiac benefits from fruits and vegetables have been attributed to polyphenols, classical natural antioxidants.141 Pre-clinical and clinical studies have shown that polyphenols mitigate endogenous DNA damage in various pathological conditions including CVDs,138 in association with activation of antioxidant enzymes and scavenging ROS.141 In particular, the beneficial effects of polyphenols such as resveratrol in CVDs have been confirmed in clinical trials.142

Excessive ROS-induced DNA damage is established in all types of CVDs, supporting the role of antioxidants in the therapeutics for CVDs. Antioxidants including vitamin C, vitamin E, and combinations are found therapeutically effective in pre-clinical studies of CVDs.143,144 However, large clinical trials for dietary supplementation with antioxidants were disappointing. Randomized controlled trials (RCTs) have demonstrated the little additive cardiovascular benefit of antioxidants (such as vitamin C and E) in humans.145,146 The discrepancy between experimental studies and RCTs might suggest the usage of antioxidants is intrinsically flawed. It is possible that RCTs were inadequately designed for methodological reasons. The cardiovascular benefit of antioxidants may depend on baseline antioxidant status, and reliable biomarkers of oxidative stress are still unavailable for large RCTs.

6.2 Statins

Statins exert proven benefits in patients with CVDs, mainly through cholesterol-lowering, anti-inflammatory, antioxidant, and anti-thrombotic properties. Many pharmacological effects of statins are attributed to abrogation of DNA damage in multiple CVDs including dyslipidemia,130,147 atherosclerosis,115,131 MI,132 and chemotherapy-induced cardiotoxicity.110 Statins can reduce oxidant stress-induced DNA damage, and inhibit DDR downstream signalling. For example, statins lead to phosphorylation of the ubiquitin ligase mdm2, promoting its nuclear localization and interaction with p300 and inhibiting its interaction with p19ARF, favouring p53 degradation. In addition, statins were shown to improve telomere dysfunction by up-regulating telomere repeat-binding factor TRF2, thus enhancing the migratory capacity of endothelial progenitor cells.148 In atherosclerosis, atorvastatin therapy reduced VSMC senescence, telomere shortening, and apoptosis, as well as inhibited ATM/ATR-mediated DDR activation.115 Mechanistically, statin treatment markedly accelerated NBS-1-induced DNA repair. Statin-induced accelerated DNA repair was mediated by NBS-1 and the human double minute protein Hdm2. In the presence of Statin, Hdm2 is phosphorylated, triggering dissociation of Hdm2 from NBS-1, thus inhibiting NBS-1 degradation. Moreover, statin therapy up-regulates a subset of DNA repair genes,149 and inhibits ionizing radiation-induced DDR including activation of p53 and CHK1 in primary human endothelial cells.150 In a clinical study involving 19 795 participants, statin users display a 4.3–6.0% lower urinary 8-oxoG level which represents oxidative DNA damage.151 Based on these findings, statin therapy for CVDs may be mediated by reduction of DNA damage and facilitation of DNA repair.

6.3 ACEIs/ARBs

Overactivation of the renin-angiotensin system with high levels of angiotensin II (Ang II) causes oxidative stress in CVDs including endothelial dysfunction and atherosclerosis.152 Previous findings showed that Ang II promotes ROS production and oxidative DNA damage via AT1 receptor in human VSMCs to accelerate vascular senescence.153 The ARB losartan was shown to reduce oxidative stress-induced DNA damage and cell senescence through inhibition of telomere attribution-induced replicative senescence, or acute stress-induced premature senescence independent of the telomere.153 Preceding studies also revealed that ACEI/ARB exerted cardiovascular protective effects by attenuation of oxidative stress, reduction of DNA damage, and improvement of endothelial function.133,154 In an experimental MI model, administration of ramipril and valsartan decreased MI severity and attenuated DNA damage by reducing oxidative stress.134 Similarly, oxidative stress and DNA damage (DSBs and mutagenic DNA base modification) were present in the hearts and kidneys of Ang II-induced hypertensive mice.155 Furthermore, oxidative DNA damage was prevented by ARB in hypertensive mice.135 These findings favour a role for the attention of oxidative stress and DNA damage in ACEI/ARB-offered cardiovascular benefits.

6.4 β-blockers

β-adrenergic receptor blockade prevents DNA damage, thereby protecting against CVDs.58,136,137 In an experimental model of doxorubicin cardiomyopathy, early carvedilol therapy was shown to alleviate doxorubicin-induced DNA damage and cardiotoxicity.136 Carvedilol therapy has been shown to reduce oxidative DNA damage indicated by 8-OHdG level in DCM patients, together with amelioration of HF.58 Similarly, oxidative DNA damage was reduced by carvedilol in hypertensive patients.137 These findings suggest that β-blockers may alleviate DNA damage by attenuating oxidative stress, although little is known with regards to the mechanistic insights behind β-blocker-induced response on DNA damage. These studies indicate that β-blockers may be a therapeutic medication to prevent DNA damage in CVDs.

7. DDR inhibition: novel therapeutic targets for CVDs

Given the cardinal role of DDR activation in CVDs, emerging strategies targeting DDR signalling components may be considered. Genetic and pharmacological inhibition of key DDR components has been confirmed beneficial in CVDs (Tables 2 and 4). In this section, we will focus on the inhibition of various DDR proteins, including PARP, ATM, ATR, DNA-PK, CHK1/2, and p53, in the management of CVDs.

Table 4

The application of DDR inhibitors in the treatment of CVDs

DDR targetDrug nameAnimal model/patientCardioprotective effectsRef
PARP inhibitor3-ABAtherosclerosisImproved endothelial function156
I/RReduced infarct size, myocardial injury, and preserved the myocardial ATP levels94
ABT-888PAHReversed PAH, decreased pulmonary pressure, and right ventricular hypertrophy67
AFAttenuated NAD+ depletion, oxidative stress, atrial contractile dysfunction37
TIQ-AAtherosclerosisInduced regression of plaques, strengthened plaque stability69,157
PJ-34Endothelial dysfunctionAttenuated endothelial dysfunction158,159
DPQI/RReduced infarct size, cardiomyocyte apoptosis, and improved cardiac function160,161
INO-1001STEMIDecreased CRP and IL-6 levels162
AtherosclerosisReduced atherosclerotic lesion development and inflammatory response163,164
ATM inhibitorKU55933Vascular senescenceAttenuated endothelial cell dysfunction and senescence48
Doxorubicin-induced cardiotoxicityAttenuated systolic dysfunction and cardiac apoptosis, improved mortality52
KU60019Pressure overloadAttenuated cardiac hypertrophy and dysfunction21,22
MIAttenuated systolic dysfunction and reduced the infarct size98
PAHIncreased pulmonary arterial VSMC proliferation and inhibited cell apoptosis165
CaffeineVascular senescenceAttenuated endothelial cell dysfunction and senescence48
ATR inhibitorAZD6738Sarcomeric cardiomyopathyReduced pathological ventricular remodelling38
DNA-PK inhibitorNU7026
DMNB
Arterial injuryAttenuated neointimal formation166
CHK1/2 inhibitorMK-8776PAHImproved PAH, reduced pulmonary VSMC proliferation and resistance to apoptosis167
LY2606368PAHAttenuated fibrosis and pulmonary vascular remodelling, improved respiratory mechanics and hemodynamic parameters168
p53 inhibitorPifithrin-αDoxorubicin-induced cardiotoxicityReduced apoptosis, attenuated cardiotoxicity169–171
MIImproved cardiac function, reduced apoptosis172,173
DCMReduced cardiac apoptosis174
Diabetic cardiomyopathyAttenuated cardiac apoptosis and senescence, reduced glycolysis, impaired angiogenesis175
DDR targetDrug nameAnimal model/patientCardioprotective effectsRef
PARP inhibitor3-ABAtherosclerosisImproved endothelial function156
I/RReduced infarct size, myocardial injury, and preserved the myocardial ATP levels94
ABT-888PAHReversed PAH, decreased pulmonary pressure, and right ventricular hypertrophy67
AFAttenuated NAD+ depletion, oxidative stress, atrial contractile dysfunction37
TIQ-AAtherosclerosisInduced regression of plaques, strengthened plaque stability69,157
PJ-34Endothelial dysfunctionAttenuated endothelial dysfunction158,159
DPQI/RReduced infarct size, cardiomyocyte apoptosis, and improved cardiac function160,161
INO-1001STEMIDecreased CRP and IL-6 levels162
AtherosclerosisReduced atherosclerotic lesion development and inflammatory response163,164
ATM inhibitorKU55933Vascular senescenceAttenuated endothelial cell dysfunction and senescence48
Doxorubicin-induced cardiotoxicityAttenuated systolic dysfunction and cardiac apoptosis, improved mortality52
KU60019Pressure overloadAttenuated cardiac hypertrophy and dysfunction21,22
MIAttenuated systolic dysfunction and reduced the infarct size98
PAHIncreased pulmonary arterial VSMC proliferation and inhibited cell apoptosis165
CaffeineVascular senescenceAttenuated endothelial cell dysfunction and senescence48
ATR inhibitorAZD6738Sarcomeric cardiomyopathyReduced pathological ventricular remodelling38
DNA-PK inhibitorNU7026
DMNB
Arterial injuryAttenuated neointimal formation166
CHK1/2 inhibitorMK-8776PAHImproved PAH, reduced pulmonary VSMC proliferation and resistance to apoptosis167
LY2606368PAHAttenuated fibrosis and pulmonary vascular remodelling, improved respiratory mechanics and hemodynamic parameters168
p53 inhibitorPifithrin-αDoxorubicin-induced cardiotoxicityReduced apoptosis, attenuated cardiotoxicity169–171
MIImproved cardiac function, reduced apoptosis172,173
DCMReduced cardiac apoptosis174
Diabetic cardiomyopathyAttenuated cardiac apoptosis and senescence, reduced glycolysis, impaired angiogenesis175

3-AB, 3-aminobenzamide; DPQ, 3,4-dihydro-5-[4-(1-piperidinyl)butoxyl]-1 (2H)-isoquinolinone; TIQ-A, thieno[2,3-c]isoquinolin-5-one.

Table 4

The application of DDR inhibitors in the treatment of CVDs

DDR targetDrug nameAnimal model/patientCardioprotective effectsRef
PARP inhibitor3-ABAtherosclerosisImproved endothelial function156
I/RReduced infarct size, myocardial injury, and preserved the myocardial ATP levels94
ABT-888PAHReversed PAH, decreased pulmonary pressure, and right ventricular hypertrophy67
AFAttenuated NAD+ depletion, oxidative stress, atrial contractile dysfunction37
TIQ-AAtherosclerosisInduced regression of plaques, strengthened plaque stability69,157
PJ-34Endothelial dysfunctionAttenuated endothelial dysfunction158,159
DPQI/RReduced infarct size, cardiomyocyte apoptosis, and improved cardiac function160,161
INO-1001STEMIDecreased CRP and IL-6 levels162
AtherosclerosisReduced atherosclerotic lesion development and inflammatory response163,164
ATM inhibitorKU55933Vascular senescenceAttenuated endothelial cell dysfunction and senescence48
Doxorubicin-induced cardiotoxicityAttenuated systolic dysfunction and cardiac apoptosis, improved mortality52
KU60019Pressure overloadAttenuated cardiac hypertrophy and dysfunction21,22
MIAttenuated systolic dysfunction and reduced the infarct size98
PAHIncreased pulmonary arterial VSMC proliferation and inhibited cell apoptosis165
CaffeineVascular senescenceAttenuated endothelial cell dysfunction and senescence48
ATR inhibitorAZD6738Sarcomeric cardiomyopathyReduced pathological ventricular remodelling38
DNA-PK inhibitorNU7026
DMNB
Arterial injuryAttenuated neointimal formation166
CHK1/2 inhibitorMK-8776PAHImproved PAH, reduced pulmonary VSMC proliferation and resistance to apoptosis167
LY2606368PAHAttenuated fibrosis and pulmonary vascular remodelling, improved respiratory mechanics and hemodynamic parameters168
p53 inhibitorPifithrin-αDoxorubicin-induced cardiotoxicityReduced apoptosis, attenuated cardiotoxicity169–171
MIImproved cardiac function, reduced apoptosis172,173
DCMReduced cardiac apoptosis174
Diabetic cardiomyopathyAttenuated cardiac apoptosis and senescence, reduced glycolysis, impaired angiogenesis175
DDR targetDrug nameAnimal model/patientCardioprotective effectsRef
PARP inhibitor3-ABAtherosclerosisImproved endothelial function156
I/RReduced infarct size, myocardial injury, and preserved the myocardial ATP levels94
ABT-888PAHReversed PAH, decreased pulmonary pressure, and right ventricular hypertrophy67
AFAttenuated NAD+ depletion, oxidative stress, atrial contractile dysfunction37
TIQ-AAtherosclerosisInduced regression of plaques, strengthened plaque stability69,157
PJ-34Endothelial dysfunctionAttenuated endothelial dysfunction158,159
DPQI/RReduced infarct size, cardiomyocyte apoptosis, and improved cardiac function160,161
INO-1001STEMIDecreased CRP and IL-6 levels162
AtherosclerosisReduced atherosclerotic lesion development and inflammatory response163,164
ATM inhibitorKU55933Vascular senescenceAttenuated endothelial cell dysfunction and senescence48
Doxorubicin-induced cardiotoxicityAttenuated systolic dysfunction and cardiac apoptosis, improved mortality52
KU60019Pressure overloadAttenuated cardiac hypertrophy and dysfunction21,22
MIAttenuated systolic dysfunction and reduced the infarct size98
PAHIncreased pulmonary arterial VSMC proliferation and inhibited cell apoptosis165
CaffeineVascular senescenceAttenuated endothelial cell dysfunction and senescence48
ATR inhibitorAZD6738Sarcomeric cardiomyopathyReduced pathological ventricular remodelling38
DNA-PK inhibitorNU7026
DMNB
Arterial injuryAttenuated neointimal formation166
CHK1/2 inhibitorMK-8776PAHImproved PAH, reduced pulmonary VSMC proliferation and resistance to apoptosis167
LY2606368PAHAttenuated fibrosis and pulmonary vascular remodelling, improved respiratory mechanics and hemodynamic parameters168
p53 inhibitorPifithrin-αDoxorubicin-induced cardiotoxicityReduced apoptosis, attenuated cardiotoxicity169–171
MIImproved cardiac function, reduced apoptosis172,173
DCMReduced cardiac apoptosis174
Diabetic cardiomyopathyAttenuated cardiac apoptosis and senescence, reduced glycolysis, impaired angiogenesis175

3-AB, 3-aminobenzamide; DPQ, 3,4-dihydro-5-[4-(1-piperidinyl)butoxyl]-1 (2H)-isoquinolinone; TIQ-A, thieno[2,3-c]isoquinolin-5-one.

7.1 PARP inhibition

PARP family denotes a major enzymatic group to catalyse poly(ADP-ribosyl)ation.28 PARP activation in response to DNA damage mediates the assembly of a poly(ADP-ribose) chain through catalysing transfer of ADP-ribose residues from NAD+ onto target substrates. Recent work noted an essential role for PARP in cellular processes including DDR, gene transcription, and cell death.27,176 In this context, PARP inhibition displays therapeutic promise for a variety of diseases, in particular cancers where potent PARP inhibitors have entered clinical trials and received approval for therapeutics.13 Emerging evidence has revealed aberrant activation of PARP in CVDs,70,71,77,157–160,162–164,177–179 and validated PARP inhibitors in pre-clinical studies and clinical trials for the treatment of CVDs (Table 5).

Table 5

Clinical trials of PARP inhibitors in the treatment of CVDs

CVDPARP inhibitor treatmentsNCT identifierClinical statusPrimary outcome
AMIINO-1001 (200, 400, and 800 mg)NCT00271765Phase IISafety evaluated by symptoms, vital signs, physical examination, laboratory data, ECGs
PAHOlaparib up to 300 mg BID for 24 weeksNCT03782818Phase I/IIOccurrence of treatment-emergent AEs
PAHOlaparib up to 400 mg BID (100–400 mg) for 16 weeksNCT03251872Phase IChange in pulmonary vascular resistance
StrokeJPI-289 through I.V for 30 minNCT01983358Phase IDifferences of ECG, vital sign, and diagnostic test
Heart diseasesINO-1001 with 4 dosesNCT00271167Phase IIReduction in serious post-operative complications
CVDPARP inhibitor treatmentsNCT identifierClinical statusPrimary outcome
AMIINO-1001 (200, 400, and 800 mg)NCT00271765Phase IISafety evaluated by symptoms, vital signs, physical examination, laboratory data, ECGs
PAHOlaparib up to 300 mg BID for 24 weeksNCT03782818Phase I/IIOccurrence of treatment-emergent AEs
PAHOlaparib up to 400 mg BID (100–400 mg) for 16 weeksNCT03251872Phase IChange in pulmonary vascular resistance
StrokeJPI-289 through I.V for 30 minNCT01983358Phase IDifferences of ECG, vital sign, and diagnostic test
Heart diseasesINO-1001 with 4 dosesNCT00271167Phase IIReduction in serious post-operative complications

AE, adverse event; ECG, electrocardiogram.

Table 5

Clinical trials of PARP inhibitors in the treatment of CVDs

CVDPARP inhibitor treatmentsNCT identifierClinical statusPrimary outcome
AMIINO-1001 (200, 400, and 800 mg)NCT00271765Phase IISafety evaluated by symptoms, vital signs, physical examination, laboratory data, ECGs
PAHOlaparib up to 300 mg BID for 24 weeksNCT03782818Phase I/IIOccurrence of treatment-emergent AEs
PAHOlaparib up to 400 mg BID (100–400 mg) for 16 weeksNCT03251872Phase IChange in pulmonary vascular resistance
StrokeJPI-289 through I.V for 30 minNCT01983358Phase IDifferences of ECG, vital sign, and diagnostic test
Heart diseasesINO-1001 with 4 dosesNCT00271167Phase IIReduction in serious post-operative complications
CVDPARP inhibitor treatmentsNCT identifierClinical statusPrimary outcome
AMIINO-1001 (200, 400, and 800 mg)NCT00271765Phase IISafety evaluated by symptoms, vital signs, physical examination, laboratory data, ECGs
PAHOlaparib up to 300 mg BID for 24 weeksNCT03782818Phase I/IIOccurrence of treatment-emergent AEs
PAHOlaparib up to 400 mg BID (100–400 mg) for 16 weeksNCT03251872Phase IChange in pulmonary vascular resistance
StrokeJPI-289 through I.V for 30 minNCT01983358Phase IDifferences of ECG, vital sign, and diagnostic test
Heart diseasesINO-1001 with 4 dosesNCT00271167Phase IIReduction in serious post-operative complications

AE, adverse event; ECG, electrocardiogram.

PARP enzymes play a critical role in the pathogenesis of CVDs including HF, MI and ischaemia and reperfusion (I/R) injury, atherosclerosis, and PAH. Inhibition of PARP was shown to exert cardioprotective effects in animal models of MI, chronic HF, and cardiopulmonary bypass.73,94 Subsequent studies in patient specimens and pre-clinical samples demonstrated PARP activation in the heart and circulating leucocytes after MI.180,181 The third-generation PARP inhibitor INO-1001 was shown to improve cardiac function and reduce infarct size in myocardial I/R.182–184 Based on these studies, INO-1001 became the first PARP inhibitor to enter clinical trials.162 This trial displayed that PARP inhibition may inhibit the inflammatory response in MI patients, indicated by plasma C-reactive protein and IL-6 levels.

Ample pharmacological mechanisms have been postulated for the cardioprotective effects of PARP inhibitors.185 DNA damage-triggered excessive PARP1 activation leads to unfavourable consequences including (i) NF-κB and activator protein 1-mediated inflammatory response; (ii) NAD+ and ATP depletion, down-regulation of cardioprotective SIRT1, as well as necrosis; (iii) acetyl-coenzyme A acetyltransferase 1-mediated foam cell death and oxidized LDL accumulation; (iv) decreased endothelial nitric oxide synthase (eNOS) activity; and (v) apoptosis-inducing factor translocation to the nucleus. PARP inhibitors suppress these detrimental consequences to protect against cardiovascular damage.185

7.2 ATM inhibition

ATM is an apical activator of DDR in the face of DSBs.186 ATM-mediated phosphorylation plays cardinal roles in response to genomic stress to preserve cellular homeostasis. Recently, ATM activation has garnered much attention as a DDR regulator in CVDs, in particular, HF (see Section 4).21,22,29,98 Accordingly, ATM depletion or inhibitors preserves cardiac function. Genetic deletion of ATM and pharmacological inhibition using KU60019 exhibited protective effects in cardiac function in response to pressure overload.21 In another study, long non-coding RNA Caren was shown to maintain cardiac function under pressure overload through inactivation of ATM-mediated DDR and activation of mitochondrial bioenergetics.22 ATM inhibitor KU60019 partially rescues cardiac function in Caren loss-of-function mice. Taken together, these findings suggest inactivating DDR through genetic or pharmacological loss of ATM kinase function may become a new therapeutic strategy against HF.

Although promising therapeutic effects of ATM inhibition in preserving cardiac function in HF, discrepancies in the role of ATM in MI have been noted. Several reports also demonstrated aberrant activation of ATM kinase in MI.79,80,97,98 ATM deficiency has shown to attenuate cardiac dysfunction and dilation post-MI with increased fibrosis and apoptosis.79 Moreover, ATM promotes cardiac inflammation during myocardial ischaemia.80 Post-MI cardiac remodelling was attenuated in ATM heterozygous knockout mice. A recent study also showed that KU60019 attenuated cardiac systolic dysfunction and reduced infarct size in MI.98 These studies suggest the promises of ATM inhibition in post-MI remodelling. However, several studies showed that ATM deficiency exacerbated post-MI cardiac dysfunction and remodelling through impaired angiogenesis, and disturbed autophagy.81–83 These studies have shown that ATM inhibition attenuated cardiac dysfunction, remodelling, and inflammation during the early post-MI period (1–7 days) while exacerbating cardiomyocyte dysfunction, fibrosis, apoptosis, and cardiac hypertrophy during late post-MI (14–28 days).

ATM inhibitors have been developed for the treatment of solid tumours. Besides, combination treatment with chemotherapy and ATM inhibitors are being tested in clinical trials.187 At present, there is no clinical trial on ATM inhibitors in CVDs. Overall, modulating ATM represents a new therapeutic option in the management of CVDs.

7.3 ATR inhibition

ATR represents another logical therapeutic DDR target for CVDs, given its association with ATM and its central role in regulating DNA damage. To date, only a recent study demonstrated the essential role of ATR in sarcomeric cardiomyopathy.38 In this study, increased DNA damage and selective ATR-p53 activation were found in Mybpc3−/− cardiomyopathy, secondary to replication stress. Highly selective ATR inhibitor AZD6738 was shown to reduce pathological remodelling in Mybpc3−/− cardiomyopathy. Besides, depletion of cardiomyocyte p53 mimicked the protective function of AZD6738. These findings suggest that replication stress-induced ATR activation regulates the remodelling of sarcomeric cardiomyopathy.

7.4 DNA-PK inhibition

DNA-PK, a heterotrimeric protein complex with a heterodimer of Ku proteins Ku70/Ku80, and a catalytic subunit DNA-PKcs, is a member of the phosphoinositide-3-kinase-related kinase family involved in NHEJ for DSB repair.188–190 DNA-PK activation is noted in CVDs, including atherosclerosis,24,64,166 DCM,191 cardiac hypertrophy,21 and myocardial I/R.86

Genetic and pharmacological inhibition of DNA-PK demonstrated beneficial effects in atherosclerosis. DNA-PK and neuron-derived orphan receptor 1 activation were found within neointima of human atherosclerotic tissue specimens.166 The specific DNA-PK inhibitors NU7024 and DMNB were shown to inhibit NOR-1-dependent proliferation of human aortic VSMCs.166 Furthermore, DNA-PK inhibition attenuates neointimal lesion size following wire injury. Similarly, recent evidence from our laboratory suggested a role for DNA-PK inhibition in myocardial I/R.86 DNA-PK is turned on in response to I/R stress to promote myocardial damage. Intriguingly, cardiomyocyte-specific DNA-PK depletion inhibited inflammatory response and apoptosis to preserve cardiac function under I/R injury. Therefore, targeting DNA-PK-mediated DDR may provide therapeutic promise in CVDs.

7.5 CHK1/2 inhibition

CHK1 and CHK2 are functionally overlapped serine/threonine kinases and immediate targets for ATR and ATM, respectively.192 CHK1 and CHK2 trigger cell cycle arrest and cellular senescence through phosphorylation of p53 and CDC25A, both with a vital role in the pathogenesis of CVDs. Recent studies identified a role for CHK1/2 kinases in CVDs including HF193 and PAH.194 In human HF, cardiomyocyte apoptosis is specifically related to down-regulated telomere repeat-binding factor TRF2, telomere shortening, and CHK2 activation.193 In another study, patients with PAH exhibited up-regulated CHK1 expression in pulmonary VSMCs and distal pulmonary arteries.167,194 CHK1 triggers VSMC proliferation and resistance to apoptosis. The CHK1 inhibitor MK8776, with a proven efficacy in clinical trials in cancer patients, attenuates vascular remodelling, and improves hemodynamic parameters in rodent models of PAH.194 These findings argue the potential of CHK1 inhibitors in the management of PAH.

7.6 p53 inhibition

DDR-mediated p53 activation leads to cell cycle arrest, cellular senescence, and apoptosis.195 A number of upstream kinases may mediate p53 phosphorylation and activation including ATM, ATR, DNA-PK, CHK1, and CHK2.196 Mounting evidence has demonstrated DDR and p53 activation in CVDs, including HF, cardiac remodelling, myocardial I/R injury, diabetic cardiomyopathy, and chemotherapy-induced cardiotoxicity. Besides, p53 activation participates in the pathogenesis of CVDs, including anti-angiogenesis, apoptosis, autophagy, necrosis, metabolic alterations, and senescence. Genetic and pharmacological loss-of-function of p53 exerts cardioprotective effects. Currently, pifithrin, a small molecule inhibitor of p53, is used in animal models of CVDs, while further studies are required to test its clinical efficacy.

8. Future directions and conclusions

Many questions regarding DNA damage and DDR remain to be addressed in CVDs. While extensive attention was given for the involvement of DDR in HF, atherosclerosis or chemotherapy-evoked cardiovascular pathologies,6,21,69,111 a role for DDR remains elusive in other crucial areas (e.g. arrhythmias and hypertension). In addition, susceptibilities and consequences of DNA damage in various cardiovascular cell types, including cardiomyocytes, CFs, endothelial cells, and immune cells, are poorly understood. A heterogeneity for DNA damage likely exists at the cellular level. Single-cell transcriptomics and spatial transcriptomics should be applied to better appreciate cell complexity in response to DNA damage, and to determine the impact of DDR in proliferative vs. non-proliferative cells.

DDR exerts both beneficial and detrimental effects on certain cardiovascular pathologies. For example, ATM inhibition may attenuate cardiac dysfunction and inflammation in the early post-MI period, while such inhibition exacerbates cardiac dysfunction and remodelling during late post-MI.82,98 DDR components such as PARP and ATM may participate in DNA repair to maintain genome stability and promote cell survival in response to mild DNA damage.12 Nonetheless, more studies have revealed rather detrimental roles of these molecules to promote DDR activation and cell death in the aetiology of CVDs.21,22,29 Further clarification is needed with regards to the role of these proteins in either DNA repair or persistent DDR activation.

Although a number of compounds targeting DDR components have displayed therapeutic potential for CVDs, clinical trials evaluating these DDR inhibitors in CVDs are still lacking. Much of our knowledge on DNA damage is derived from animal studies which limit the translational value for DNA damage and DDR in human CVDs. However, it remains to be tested with regards to the efficacy and safety of DDR inhibitors in humans. Also, the safety and pharmacokinetics of PARP1 inhibitors were assessed in humans up to 2 weeks,162 lacking information for the long-term safety of PARP1 inhibitors. Thus, adverse consequences associated with the long-term application of DDR inhibitors would take longer observational periods, which cannot be simply evaluated using current models.

In conclusion, DNA damage and DDR have crucial roles in the pathogenesis of CVDs, while many classic cardiovascular drugs can modulate DNA damage. Understanding how DDR of individual cardiovascular cell types contributes to disease phenotypes will help to develop targeted therapies to prevent or delay DNA damage-associated CVDs. Such an approach should have a high impact in patients at risk of long-term DNA damage (e.g. chemotherapy). Overall, DDR-associated protein therapy is being recognized as an important therapeutic strategy for CVDs.

Authors’ contributions

L.W. and J.R.S. drafted the manuscript. All other authors edited the manuscript. All authors have approved the final version.

Acknowledgements

We wish to sincerely apologize to those authors whose important work cannot be included due to space limitations. Some of the icons and elements of diagrams were created with BioRender.com.

Funding

This work was supported in part by the National Key R&D Program of China (2017YFA0506000), the National Natural Science Foundation of China (82130011), and the Program of Shanghai Academic/Technology Research Leader (20XD1420900).

References

1

GBD 2015 Disease and Injury Incidence and Prevalence Collaborators
.
Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015
.
Lancet
2016
;
388
:
1545
1602
.

2

Doran
S
,
Arif
M
,
Lam
S
,
Bayraktar
A
,
Turkez
H
,
Uhlen
M
,
Boren
J
,
Mardinoglu
A
.
Multi-omics approaches for revealing the complexity of cardiovascular disease
.
Brief Bioinform
2021
;
22
:
bbab061
.

3

Ren
J
,
Bi
Y
,
Sowers
JR
,
Hetz
C
,
Zhang
Y
.
Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases
.
Nat Rev Cardiol
2021
;
18
:
499
521
.

4

Zhang
Y
,
Sowers
JR
,
Ren
J
.
Targeting autophagy in obesity: from pathophysiology to management
.
Nat Rev Endocrinol
2018
;
14
:
356
376
.

5

Shah
A
,
Gray
K
,
Figg
N
,
Finigan
A
,
Starks
L
,
Bennett
M
.
Defective base excision repair of oxidative DNA damage in vascular smooth muscle cells promotes atherosclerosis
.
Circulation
2018
;
138
:
1446
1462
.

6

Uryga
A
,
Gray
K
,
Bennett
M
.
DNA damage and repair in vascular disease
.
Annu Rev Physiol
2016
;
78
:
45
66
.

7

Siggens
L
,
Figg
N
,
Bennett
M
,
Foo
R
.
Nutrient deprivation regulates DNA damage repair in cardiomyocytes via loss of the base-excision repair enzyme OGG1
.
FASEB J
2012
;
26
:
2117
2124
.

8

Sano
M
,
Minamino
T
,
Toko
H
,
Miyauchi
H
,
Orimo
M
,
Qin
Y
,
Akazawa
H
,
Tateno
K
,
Kayama
Y
,
Harada
M
,
Shimizu
I
,
Asahara
T
,
Hamada
H
,
Tomita
S
,
Molkentin
JD
,
Zou
Y
,
Komuro
I
.
p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload
.
Nature
2007
;
446
:
444
448
.

9

Shukla
PC
,
Singh
KK
,
Quan
A
,
Al-Omran
M
,
Teoh
H
,
Lovren
F
,
Cao
L
,
Rovira
II
,
Pan
Y
,
Brezden-Masley
C
,
Yanagawa
B
,
Gupta
A
,
Deng
CX
,
Coles
JG
,
Leong-Poi
H
,
Stanford
WL
,
Parker
TG
,
Schneider
MD
,
Finkel
T
,
Verma
S
.
BRCA1 is an essential regulator of heart function and survival following myocardial infarction
.
Nat Commun
2011
;
2
:
593
.

10

Shah
A
,
Bennett
M
.
Controlling inflammation through DNA damage and repair
.
Circ Res
2016
;
119
:
698
700
.

11

Schumacher
B
,
Pothof
J
,
Vijg
J
,
Hoeijmakers
JHJ
.
The central role of DNA damage in the ageing process
.
Nature
2021
;
592
:
695
703
.

12

Jackson
SP
,
Bartek
J
.
The DNA-damage response in human biology and disease
.
Nature
2009
;
461
:
1071
1078
.

13

Pilié
PG
,
Tang
C
,
Mills
GB
,
Yap
TA
.
State-of-the-art strategies for targeting the DNA damage response in cancer
.
Nat Rev Clin Oncol
2019
;
16
:
81
104
.

14

Yu
EP
,
Bennett
MR
.
Mitochondrial DNA damage and atherosclerosis
.
Trends Endocrinol Metab
2014
;
25
:
481
487
.

15

Ait-Aissa
K
,
Blaszak
SC
,
Beutner
G
,
Tsaih
SW
,
Morgan
G
,
Santos
JH
,
Flister
MJ
,
Joyce
DL
,
Camara
AKS
,
Gutterman
DD
,
Donato
AJ
,
Porter
GA
Jr
,
Beyer
AM
.
Mitochondrial oxidative phosphorylation defect in the heart of subjects with coronary artery disease
.
Sci Rep
2019
;
9
:
7623
.

16

Guarini
G
,
Kiyooka
T
,
Ohanyan
V
,
Pung
YF
,
Marzilli
M
,
Chen
YR
,
Chen
CL
,
Kang
PT
,
Hardwick
JP
,
Kolz
CL
,
Yin
L
,
Wilson
GL
,
Shokolenko
I
,
Dobson
JG
Jr
,
Fenton
R
,
Chilian
WM
.
Impaired coronary metabolic dilation in the metabolic syndrome is linked to mitochondrial dysfunction and mitochondrial DNA damage
.
Basic Res Cardiol
2016
;
111
:
29
.

17

Uziel
T
,
Lerenthal
Y
,
Moyal
L
,
Andegeko
Y
,
Mittelman
L
,
Shiloh
Y
.
Requirement of the MRN complex for ATM activation by DNA damage
.
EMBO J
2003
;
22
:
5612
5621
.

18

Jasin
M
.
Homologous repair of DNA damage and tumorigenesis: the BRCA connection
.
Oncogene
2002
;
21
:
8981
8993
.

19

Blackford
AN
,
Jackson
SP
.
ATM, ATR, and DNA-PK: the trinity at the heart of the DNA damage response
.
Mol Cell
2017
;
66
:
801
817
.

20

Kuo
LJ
,
Yang
LX
.
Gamma-H2AX - a novel biomarker for DNA double-strand breaks
.
In Vivo
2008
;
22
:
305
309
.

21

Nakada
Y
,
Nhi Nguyen
NU
,
Xiao
F
,
Savla
JJ
,
Lam
NT
,
Abdisalaam
S
,
Bhattacharya
S
,
Mukherjee
S
,
Asaithamby
A
,
Gillette
TG
,
Hill
JA
,
Sadek
HA
.
DNA damage response mediates pressure overload-induced cardiomyocyte hypertrophy
.
Circulation
2019
;
139
:
1237
1239
.

22

Sato
M
,
Kadomatsu
T
,
Miyata
K
,
Warren
JS
,
Tian
Z
,
Zhu
S
,
Horiguchi
H
,
Makaju
A
,
Bakhtina
A
,
Morinaga
J
,
Sugizaki
T
,
Hirashima
K
,
Yoshinobu
K
,
Imasaka
M
,
Araki
M
,
Komohara
Y
,
Wakayama
T
,
Nakagawa
S
,
Franklin
S
,
Node
K
,
Araki
K
,
Oike
Y
.
The lncRNA Caren antagonizes heart failure by inactivating DNA damage response and activating mitochondrial biogenesis
.
Nat Commun
2021
;
12
:
2529
.

23

Gray
K
,
Kumar
S
,
Figg
N
,
Harrison
J
,
Baker
L
,
Mercer
J
,
Littlewood
T
,
Bennett
M
.
Effects of DNA damage in smooth muscle cells in atherosclerosis
.
Circ Res
2015
;
116
:
816
826
.

24

Haemmig
S
,
Yang
D
,
Sun
X
,
Das
D
,
Ghaffari
S
,
Molinaro
R
,
Chen
L
,
Deng
Y
,
Freeman
D
,
Moullan
N
,
Tesmenitsky
Y
,
Wara
A
,
Simion
V
,
Shvartz
E
,
Lee
JF
,
Yang
T
,
Sukova
G
,
Marto
JA
,
Stone
PH
,
Lee
WL
,
Auwerx
J
,
Libby
P
,
Feinberg
MW
.
Long noncoding RNA SNHG12 integrates a DNA-PK-mediated DNA damage response and vascular senescence
.
Sci Transl Med
2020
;
12
:
eaaw1868
.

25

Caldecott
KW
.
Single-strand break repair and genetic disease
.
Nat Rev Genet
2008
;
9
:
619
631
.

26

Ray Chaudhuri
A
,
Nussenzweig
A
.
The multifaceted roles of PARP1 in DNA repair and chromatin remodelling
.
Nat Rev Mol Cell Biol
2017
;
18
:
610
621
.

27

Wang
Y
,
Luo
W
,
Wang
Y
.
PARP-1 and its associated nucleases in DNA damage response
.
DNA Repair (Amst)
2019
;
81
:
102651
.

28

Curtin
NJ
,
Szabo
C
.
Poly(ADP-ribose) polymerase inhibition: past, present and future
.
Nat Rev Drug Discov
2020
;
19
:
711
736
.

29

Higo
T
,
Naito
AT
,
Sumida
T
,
Shibamoto
M
,
Okada
K
,
Nomura
S
,
Nakagawa
A
,
Yamaguchi
T
,
Sakai
T
,
Hashimoto
A
,
Kuramoto
Y
,
Ito
M
,
Hikoso
S
,
Akazawa
H
,
Lee
JK
,
Shiojima
I
,
McKinnon
PJ
,
Sakata
Y
,
Komuro
I
.
DNA single-strand break-induced DNA damage response causes heart failure
.
Nat Commun
2017
;
8
:
15104
.

30

Durik
M
,
Kavousi
M
,
van der Pluijm
I
,
Isaacs
A
,
Cheng
C
,
Verdonk
K
,
Loot
AE
,
Oeseburg
H
,
Bhaggoe
UM
,
Leijten
F
,
van Veghel
R
,
de Vries
R
,
Rudez
G
,
Brandt
R
,
Ridwan
YR
,
van Deel
ED
,
de Boer
M
,
Tempel
D
,
Fleming
I
,
Mitchell
GF
,
Verwoert
GC
,
Tarasov
KV
,
Uitterlinden
AG
,
Hofman
A
,
Duckers
HJ
,
van Duijn
CM
,
Oostra
BA
,
Witteman
JC
,
Duncker
DJ
,
Danser
AH
,
Hoeijmakers
JH
,
Roks
AJ
.
Nucleotide excision DNA repair is associated with age-related vascular dysfunction
.
Circulation
2012
;
126
:
468
478
.

31

Tumurkhuu
G
,
Shimada
K
,
Dagvadorj
J
,
Crother
TR
,
Zhang
W
,
Luthringer
D
,
Gottlieb
RA
,
Chen
S
,
Arditi
M
.
Ogg1-dependent DNA repair regulates NLRP3 inflammasome and prevents atherosclerosis
.
Circ Res
2016
;
119
:
e76
e90
.

32

Matsuoka
S
,
Ballif
BA
,
Smogorzewska
A
,
McDonald
ER
III
,
Hurov
KE
,
Luo
J
,
Bakalarski
CE
,
Zhao
Z
,
Solimini
N
,
Lerenthal
Y
,
Shiloh
Y
,
Gygi
SP
,
Elledge
SJ
.
ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage
.
Science
2007
;
316
:
1160
1166
.

33

Anderson
R
,
Lagnado
A
,
Maggiorani
D
,
Walaszczyk
A
,
Dookun
E
,
Chapman
J
,
Birch
J
,
Salmonowicz
H
,
Ogrodnik
M
,
Jurk
D
,
Proctor
C
,
Correia-Melo
C
,
Victorelli
S
,
Fielder
E
,
Berlinguer-Palmini
R
,
Owens
A
,
Greaves
LC
,
Kolsky
KL
,
Parini
A
,
Douin-Echinard
V
,
LeBrasseur
NK
,
Arthur
HM
,
Tual-Chalot
S
,
Schafer
MJ
,
Roos
CM
,
Miller
JD
,
Robertson
N
,
Mann
J
,
Adams
PD
,
Tchkonia
T
,
Kirkland
JL
,
Mialet-Perez
J
,
Richardson
GD
,
Passos
JF
.
Length-independent telomere damage drives post-mitotic cardiomyocyte senescence
.
EMBO J
2019
;
38
:
e100492
.

34

Puente
BN
,
Kimura
W
,
Muralidhar
SA
,
Moon
J
,
Amatruda
JF
,
Phelps
KL
,
Grinsfelder
D
,
Rothermel
BA
,
Chen
R
,
Garcia
JA
,
Santos
CX
,
Thet
S
,
Mori
E
,
Kinter
MT
,
Rindler
PM
,
Zacchigna
S
,
Mukherjee
S
,
Chen
DJ
,
Mahmoud
AI
,
Giacca
M
,
Rabinovitch
PS
,
Asaithamby
A
,
Shah
AM
,
Szweda
LI
,
Sadek
HA
.
The oxygen-rich postnatal environment induces cardiomyocyte cell-cycle arrest through DNA damage response
.
Cell
2014
;
157
:
565
579
.

35

Nakada
Y
,
Canseco
DC
,
Thet
S
,
Abdisalaam
S
,
Asaithamby
A
,
Santos
CX
,
Shah
AM
,
Zhang
H
,
Faber
JE
,
Kinter
MT
,
Szweda
LI
,
Xing
C
,
Hu
Z
,
Deberardinis
RJ
,
Schiattarella
G
,
Hill
JA
,
Oz
O
,
Lu
Z
,
Zhang
CC
,
Kimura
W
,
Sadek
HA
.
Hypoxia induces heart regeneration in adult mice
.
Nature
2017
;
541
:
222
227
.

36

Ye
L
,
Qiu
L
,
Feng
B
,
Jiang
C
,
Huang
Y
,
Zhang
H
,
Zhang
H
,
Hong
H
,
Liu
J
.
Role of blood oxygen saturation during post-natal human cardiomyocyte cell cycle activities
.
JACC Basic Transl Sci
2020
;
5
:
447
460
.

37

Zhang
D
,
Hu
X
,
Li
J
,
Liu
J
,
Baks-Te Bulte
L
,
Wiersma
M
,
Malik
NU
,
van Marion
DMS
,
Tolouee
M
,
Hoogstra-Berends
F
,
Lanters
EAH
,
van Roon
AM
,
de Vries
AAF
,
Pijnappels
DA
,
de Groot
NMS
,
Henning
RH
,
Brundel
B
.
DNA damage-induced PARP1 activation confers cardiomyocyte dysfunction through NAD+ depletion in experimental atrial fibrillation
.
Nat Commun
2019
;
10
:
1307
.

38

Pal
S
,
Nixon
BR
,
Glennon
MS
,
Shridhar
P
,
Satterfield
SL
,
Su
YR
,
Becker
JR
.
Replication stress response modifies sarcomeric cardiomyopathy remodeling
.
J Am Heart Assoc
2021
;
10
:
e021768
.

39

Chen
SN
,
Lombardi
R
,
Karmouch
J
,
Tsai
JY
,
Czernuszewicz
G
,
Taylor
MRG
,
Mestroni
L
,
Coarfa
C
,
Gurha
P
,
Marian
AJ
.
DNA damage response/TP53 pathway is activated and contributes to the pathogenesis of dilated cardiomyopathy associated with LMNA (Lamin A/C) mutations
.
Circ Res
2019
;
124
:
856
873
.

40

Chen
MS
,
Lee
RT
,
Garbern
JC
.
Senescence mechanisms and targets in the heart
.
Cardiovasc Res
2022
;
118
:
1173
1187
.

41

Rouhi
L
,
Cheedipudi
SM
,
Chen
SN
,
Fan
S
,
Lombardi
R
,
Chen
X
,
Coarfa
C
,
Robertson
MJ
,
Gurha
P
,
Marian
AJ
.
Haploinsufficiency of Tmem43 in cardiac myocytes activates the DNA damage response pathway leading to a late-onset senescence-associated pro-fibrotic cardiomyopathy
.
Cardiovasc Res
2021
;
117
:
2377
2394
.

42

Zhang
S
,
Liu
X
,
Bawa-Khalfe
T
,
Lu
LS
,
Lyu
YL
,
Liu
LF
,
Yeh
ET
.
Identification of the molecular basis of doxorubicin-induced cardiotoxicity
.
Nat Med
2012
;
18
:
1639
1642
.

43

Shimizu
I
,
Minamino
T
.
Cellular senescence in cardiac diseases
.
J Cardiol
2019
;
74
:
313
319
.

44

Ragnauth
CD
,
Warren
DT
,
Liu
Y
,
McNair
R
,
Tajsic
T
,
Figg
N
,
Shroff
R
,
Skepper
J
,
Shanahan
CM
.
Prelamin A acts to accelerate smooth muscle cell senescence and is a novel biomarker of human vascular aging
.
Circulation
2010
;
121
:
2200
2210
.

45

Grootaert
MOJ
,
Finigan
A
,
Figg
NL
,
Uryga
AK
,
Bennett
MR
.
SIRT6 protects smooth muscle cells from senescence and reduces atherosclerosis
.
Circ Res
2021
;
128
:
474
491
.

46

Grootaert
MOJ
,
Moulis
M
,
Roth
L
,
Martinet
W
,
Vindis
C
,
Bennett
MR
,
De Meyer
GRY
.
Vascular smooth muscle cell death, autophagy and senescence in atherosclerosis
.
Cardiovasc Res
2018
;
114
:
622
634
.

47

Regnault
V
,
Challande
P
,
Pinet
F
,
Li
Z
,
Lacolley
P
.
Cell senescence: basic mechanisms and the need for computational networks in vascular ageing
.
Cardiovasc Res
2021
;
117
:
1841
1858
.

48

Zhan
H
,
Suzuki
T
,
Aizawa
K
,
Miyagawa
K
,
Nagai
R
.
Ataxia telangiectasia mutated (ATM)-mediated DNA damage response in oxidative stress-induced vascular endothelial cell senescence
.
J Biol Chem
2010
;
285
:
29662
29670
.

49

Xu
S
,
Ilyas
I
,
Little
PJ
,
Li
H
,
Kamato
D
,
Zheng
X
,
Luo
S
,
Li
Z
,
Liu
P
,
Han
J
,
Harding
IC
,
Ebong
EE
,
Cameron
SJ
,
Stewart
AG
,
Weng
J
,
Ma
Q
.
Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies
.
Pharmacol Rev
2021
;
73
:
924
967
.

50

Shibamoto
M
,
Higo
T
,
Naito
AT
,
Nakagawa
A
,
Sumida
T
,
Okada
K
,
Sakai
T
,
Kuramoto
Y
,
Yamaguchi
T
,
Ito
M
,
Masumura
Y
,
Higo
S
,
Lee
JK
,
Hikoso
S
,
Komuro
I
,
Sakata
Y
.
Activation of DNA damage response and cellular senescence in cardiac fibroblasts limit cardiac fibrosis after myocardial infarction
.
Int Heart J
2019
;
60
:
944
957
.

51

Cui
S
,
Xue
L
,
Yang
F
,
Dai
S
,
Han
Z
,
Liu
K
,
Liu
B
,
Yuan
Q
,
Cui
Z
,
Zhang
Y
,
Xu
F
,
Chen
Y
.
Postinfarction hearts are protected by premature senescent cardiomyocytes via GATA 4-dependent CCN 1 secretion
.
J Am Heart Assoc
2018
;
7
:
e009111
.

52

Zhan
H
,
Aizawa
K
,
Sun
J
,
Tomida
S
,
Otsu
K
,
Conway
SJ
,
McKinnon
PJ
,
Manabe
I
,
Komuro
I
,
Miyagawa
K
,
Nagai
R
,
Suzuki
T
.
Ataxia telangiectasia mutated in cardiac fibroblasts regulates doxorubicin-induced cardiotoxicity
.
Cardiovasc Res
2016
;
110
:
85
95
.

53

Bochenek
ML
,
Schutz
E
,
Schafer
K
.
Endothelial cell senescence and thrombosis: ageing clots
.
Thromb Res
2016
;
147
:
36
45
.

54

Ren
J
,
Samson
WK
,
Sowers
JR
.
Insulin-like growth factor I as a cardiac hormone: physiological and pathophysiological implications in heart disease
.
J Mol Cell Cardiol
1999
;
31
:
2049
2061
.

55

Suzuki
S
,
Shishido
T
,
Ishino
M
,
Katoh
S
,
Sasaki
T
,
Nishiyama
S
,
Miyashita
T
,
Miyamoto
T
,
Nitobe
J
,
Watanabe
T
,
Takeishi
Y
,
Kubota
I
.
8-Hydroxy-2′-deoxyguanosine is a prognostic mediator for cardiac event
.
Eur J Clin Invest
2011
;
41
:
759
766
.

56

Takahashi
T
,
Shishido
T
,
Kinoshita
D
,
Watanabe
K
,
Toshima
T
,
Sugai
T
,
Narumi
T
,
Otaki
Y
,
Tamura
H
,
Nishiyama
S
,
Arimoto
T
,
Takahashi
H
,
Miyamoto
T
,
Watanabe
T
,
Woo
CH
,
Abe
JI
,
Takeishi
Y
,
Kubota
I
,
Watanabe
M
.
Cardiac nuclear high-mobility group box 1 ameliorates pathological cardiac hypertrophy by inhibiting DNA damage response
.
JACC Basic Transl Sci
2019
;
4
:
234
247
.

57

Ko
T
,
Fujita
K
,
Nomura
S
,
Uemura
Y
,
Yamada
S
,
Tobita
T
,
Katoh
M
,
Satoh
M
,
Ito
M
,
Domoto
Y
,
Hosoya
Y
,
Amiya
E
,
Hatano
M
,
Morita
H
,
Fukayama
M
,
Aburatani
H
,
Komuro
I
.
Quantification of DNA damage in heart tissue as a novel prediction tool for therapeutic prognosis of patients with dilated cardiomyopathy
.
JACC Basic Transl Sci
2019
;
4
:
670
680
.

58

Kono
Y
,
Nakamura
K
,
Kimura
H
,
Nishii
N
,
Watanabe
A
,
Banba
K
,
Miura
A
,
Nagase
S
,
Sakuragi
S
,
Kusano
KF
,
Matsubara
H
,
Ohe
T
.
Elevated levels of oxidative DNA damage in serum and myocardium of patients with heart failure
.
Circ J
2006
;
70
:
1001
1005
.

59

Toyama
K
,
Yamabe
H
,
Uemura
T
,
Nagayoshi
Y
,
Morihisa
K
,
Koyama
J
,
Kanazawa
H
,
Hoshiyama
T
,
Ogawa
H
.
Analysis of oxidative stress expressed by urinary level of 8-hydroxy-2′-deoxyguanosine and biopyrrin in atrial fibrillation: effect of sinus rhythm restoration
.
Int J Cardiol
2013
;
168
:
80
85
.

60

Li
J
,
Zhang
D
,
Ramos
KS
,
Baks
L
,
Wiersma
M
,
Lanters
EAH
,
Bogers
A
,
de Groot
NMS
,
Brundel
B
.
Blood-based 8-hydroxy-2′-deoxyguanosine level: a potential diagnostic biomarker for atrial fibrillation
.
Heart Rhythm
2021
;
18
:
271
277
.

61

Tascanov
MB
,
Tanriverdi
Z
,
Gungoren
F
,
Besli
F
,
Erkus
ME
,
Altiparmak
IH
,
Gonel
A
,
Koyuncu
I
,
Demirbag
R
.
Relationships between paroxysmal atrial fibrillation, total oxidant status, and DNA damage
.
Rev Port Cardiol (Engl Ed)
2021
;
40
:
5
10
.

62

Corbucci
GG
,
Perrino
C
,
Donato
G
,
Ricchi
A
,
Lettieri
B
,
Troncone
G
,
Indolfi
C
,
Chiariello
M
,
Avvedimento
EV
.
Transient and reversible deoxyribonucleic acid damage in human left ventricle under controlled ischemia and reperfusion
.
J Am Coll Cardiol
2004
;
43
:
1992
1999
.

63

Shah
N
,
Meira
LB
,
Elliott
RM
,
Hoole
SP
,
West
NE
,
Brown
AJ
,
Bennett
MR
,
Garcia-Garcia
HM
,
Kuku
KO
,
Dan
K
,
Kolm
P
,
Mariathas
M
,
Curzen
N
,
Mahmoudi
M
.
DNA damage and repair in patients with coronary artery disease: correlation with plaque morphology using optical coherence tomography (DECODE study)
.
Cardiovasc Revasc Med
2019
;
20
:
812
818
.

64

Martinet
W
,
Knaapen
MW
,
De Meyer
GR
,
Herman
AG
,
Kockx
MM
.
Elevated levels of oxidative DNA damage and DNA repair enzymes in human atherosclerotic plaques
.
Circulation
2002
;
106
:
927
932
.

65

Yavuzer
S
,
Yavuzer
H
,
Cengiz
M
,
Erman
H
,
Demirdag
F
,
Doventas
A
,
Balci
H
,
Erdincler
DS
,
Uzun
H
.
The role of protein oxidation and DNA damage in elderly hypertension
.
Aging Clin Exp Res
2016
;
28
:
625
632
.

66

Subash
P
.
Assessment of oxidative DNA damage by alkaline comet assay in human essential hypertension
.
Indian J Clin Biochem
2016
;
31
:
185
193
.

67

Meloche
J
,
Pflieger
A
,
Vaillancourt
M
,
Paulin
R
,
Potus
F
,
Zervopoulos
S
,
Graydon
C
,
Courboulin
A
,
Breuils-Bonnet
S
,
Tremblay
E
,
Couture
C
,
Michelakis
ED
,
Provencher
S
,
Bonnet
S
.
Role for DNA damage signaling in pulmonary arterial hypertension
.
Circulation
2014
;
129
:
786
797
.

68

Federici
C
,
Drake
KM
,
Rigelsky
CM
,
McNelly
LN
,
Meade
SL
,
Comhair
SA
,
Erzurum
SC
,
Aldred
MA
.
Increased mutagen sensitivity and DNA damage in pulmonary arterial hypertension
.
Am J Respir Crit Care Med
2015
;
192
:
219
228
.

69

Oumouna-Benachour
K
,
Hans
CP
,
Suzuki
Y
,
Naura
A
,
Datta
R
,
Belmadani
S
,
Fallon
K
,
Woods
C
,
Boulares
AH
.
Poly(ADP-ribose) polymerase inhibition reduces atherosclerotic plaque size and promotes factors of plaque stability in apolipoprotein E-deficient mice: effects on macrophage recruitment, nuclear factor-κB nuclear translocation, and foam cell death
.
Circulation
2007
;
115
:
2442
2450
.

70

Hans
CP
,
Zerfaoui
M
,
Naura
AS
,
Catling
A
,
Boulares
AH
.
Differential effects of PARP inhibition on vascular cell survival and ACAT-1 expression favouring atherosclerotic plaque stability
.
Cardiovasc Res
2008
;
78
:
429
439
.

71

Hans
CP
,
Feng
Y
,
Naura
AS
,
Zerfaoui
M
,
Rezk
BM
,
Xia
H
,
Kaye
AD
,
Matrougui
K
,
Lazartigues
E
,
Boulares
AH
,
Bonini
M
.
Protective effects of PARP-1 knockout on dyslipidemia-induced autonomic and vascular dysfunction in ApoE−/− mice: effects on eNOS and oxidative stress
.
PLoS One
2009
;
4
:
e7430
.

72

Hans
CP
,
Feng
Y
,
Naura
AS
,
Troxclair
D
,
Zerfaoui
M
,
Siddiqui
D
,
Ju
J
,
Kim
H
,
Kaye
AD
,
Matrougui
K
,
Lazartigues
E
,
Boulares
AH
.
Opposing roles of PARP-1 in MMP-9 and TIMP-2 expression and mast cell degranulation in dyslipidemic dilated cardiomyopathy
.
Cardiovasc Pathol
2011
;
20
:
e57
e68
.

73

Zingarelli
B
,
Salzman
AL
,
Szabó
C
.
Genetic disruption of poly (ADP-ribose) synthetase inhibits the expression of P-selectin and intercellular adhesion molecule-1 in myocardial ischemia/reperfusion injury
.
Circ Res
1998
;
83
:
85
94
.

74

Zhou
HZ
,
Swanson
RA
,
Simonis
U
,
Ma
X
,
Cecchini
G
,
Gray
MO
.
Poly(ADP-ribose) polymerase-1 hyperactivation and impairment of mitochondrial respiratory chain complex I function in reperfused mouse hearts
.
Am J Physiol Heart Circ Physiol
2006
;
291
:
H714
H723
.

75

Li
P
,
Wang
Y
,
Liu
X
,
Liu
B
,
Wang
ZY
,
Xie
F
,
Qiao
W
,
Liang
ES
,
Lu
QH
,
Zhang
MX
.
Loss of PARP-1 attenuates diabetic arteriosclerotic calcification via Stat1/Runx2 axis
.
Cell Death Dis
2020
;
11
:
22
.

76

Pillai
JB
,
Gupta
M
,
Rajamohan
SB
,
Lang
R
,
Raman
J
,
Gupta
MP
.
Poly(ADP-ribose) polymerase-1-deficient mice are protected from angiotensin II-induced cardiac hypertrophy
.
Am J Physiol Heart Circ Physiol
2006
;
291
:
H1545
H1553
.

77

Liang
ES
,
Bai
WW
,
Wang
H
,
Zhang
JN
,
Zhang
F
,
Ma
Y
,
Jiang
F
,
Yin
M
,
Zhang
MX
,
Chen
XM
,
Qin
WD
.
PARP-1 (Poly[ADP-Ribose] Polymerase 1) inhibition protects from Ang II (Angiotensin II)-induced abdominal aortic aneurysm in mice
.
Hypertension
2018
;
72
:
1189
1199
.

78

Szanto
M
,
Rutkai
I
,
Hegedus
C
,
Czikora
A
,
Rozsahegyi
M
,
Kiss
B
,
Virag
L
,
Gergely
P
,
Toth
A
,
Bai
P
.
Poly(ADP-ribose) polymerase-2 depletion reduces doxorubicin-induced damage through SIRT1 induction
.
Cardiovasc Res
2011
;
92
:
430
438
.

79

Foster
CR
,
Daniel
LL
,
Daniels
CR
,
Dalal
S
,
Singh
M
,
Singh
K
,
Gadeau
A-P
.
Deficiency of ataxia telangiectasia mutated kinase modulates cardiac remodeling following myocardial infarction: involvement in fibrosis and apoptosis
.
PLoS One
2013
;
8
:
e83513
.

80

Daniel
LL
,
Daniels
CR
,
Harirforoosh
S
,
Foster
CR
,
Singh
M
,
Singh
K
.
Deficiency of ataxia telangiectasia mutated kinase delays inflammatory response in the heart following myocardial infarction
.
J Am Heart Assoc
2014
;
3
:
e001286
.

81

Daniel
LL
,
Scofield
SL
,
Thrasher
P
,
Dalal
S
,
Daniels
CR
,
Foster
CR
,
Singh
M
,
Singh
K
.
Ataxia telangiectasia-mutated kinase deficiency exacerbates left ventricular dysfunction and remodeling late after myocardial infarction
.
Am J Physiol Heart Circ Physiol
2016
;
311
:
H445
H452
.

82

Jia
L
,
Zhang
W
,
Ma
Y
,
Chen
B
,
Liu
Y
,
Piao
C
,
Wang
Y
,
Yang
M
,
Liu
T
,
Zhang
J
,
Li
T
,
Nie
S
,
Du
J
.
Haplodeficiency of ataxia telangiectasia mutated accelerates heart failure after myocardial infarction
.
J Am Heart Assoc
2017
;
6
:
e006349
.

83

Thrasher
PR
,
Scofield
SLC
,
Dalal
S
,
Crawford
CC
,
Singh
M
,
Singh
K
.
Ataxia telangiectasia mutated kinase deficiency impairs the autophagic response early during myocardial infarction
.
Am J Physiol Heart Circ Physiol
2018
;
315
:
H48
H57
.

84

Wu
D
,
Yang
H
,
Xiang
W
,
Zhou
L
,
Shi
M
,
Julies
G
,
Laplante
JM
,
Ballard
BR
,
Guo
Z
.
Heterozygous mutation of ataxia-telangiectasia mutated gene aggravates hypercholesterolemia in apoE-deficient mice
.
J Lipid Res
2005
;
46
:
1380
1387
.

85

Mercer
JR
,
Cheng
KK
,
Figg
N
,
Gorenne
I
,
Mahmoudi
M
,
Griffin
J
,
Vidal-Puig
A
,
Logan
A
,
Murphy
MP
,
Bennett
M
.
DNA damage links mitochondrial dysfunction to atherosclerosis and the metabolic syndrome
.
Circ Res
2010
;
107
:
1021
1031
.

86

Zhou
H
,
Toan
S
,
Zhu
P
,
Wang
J
,
Ren
J
,
Zhang
Y
.
DNA-PKcs promotes cardiac ischemia reperfusion injury through mitigating BI-1-governed mitochondrial homeostasis
.
Basic Res Cardiol
2020
;
115
:
11
.

87

Mak
TW
,
Hauck
L
,
Grothe
D
,
Billia
F
.
p53 regulates the cardiac transcriptome
.
Proc Natl Acad Sci U S A
2017
;
114
:
2331
2336
.

88

Matsusaka
H
,
Ide
T
,
Matsushima
S
,
Ikeuchi
M
,
Kubota
T
,
Sunagawa
K
,
Kinugawa
S
,
Tsutsui
H
.
Targeted deletion of p53 prevents cardiac rupture after myocardial infarction in mice
.
Cardiovasc Res
2006
;
70
:
457
465
.

89

Canseco
DC
,
Kimura
W
,
Garg
S
,
Mukherjee
S
,
Bhattacharya
S
,
Abdisalaam
S
,
Das
S
,
Asaithamby
A
,
Mammen
PP
,
Sadek
HA
.
Human ventricular unloading induces cardiomyocyte proliferation
.
J Am Coll Cardiol
2015
;
65
:
892
900
.

90

Funayama
A
,
Shishido
T
,
Netsu
S
,
Narumi
T
,
Kadowaki
S
,
Takahashi
H
,
Miyamoto
T
,
Watanabe
T
,
Woo
CH
,
Abe
J
,
Kuwahara
K
,
Nakao
K
,
Takeishi
Y
,
Kubota
I
.
Cardiac nuclear high mobility group box 1 prevents the development of cardiac hypertrophy and heart failure
.
Cardiovasc Res
2013
;
99
:
657
664
.

91

Di Minno
A
,
Turnu
L
,
Porro
B
,
Squellerio
I
,
Cavalca
V
,
Tremoli
E
,
Di Minno
MN
.
8-Hydroxy-2-deoxyguanosine levels and heart failure: a systematic review and meta-analysis of the literature
.
Nutr Metab Cardiovasc Dis
2017
;
27
:
201
208
.

92

Lin
PH
,
Lee
SH
,
Su
CP
,
Wei
YH
.
Oxidative damage to mitochondrial DNA in atrial muscle of patients with atrial fibrillation
.
Free Radic Biol Med
2003
;
35
:
1310
1318
.

93

Carlquist
JF
,
Knight
S
,
Cawthon
RM
,
Le
VT
,
Jared Bunch
T
,
Horne
BD
,
Rollo
JS
,
Huntinghouse
JA
,
Brent Muhlestein
J
,
Anderson
JL
.
Shortened telomere length is associated with paroxysmal atrial fibrillation among cardiovascular patients enrolled in the Intermountain Heart Collaborative Study
.
Heart Rhythm
2016
;
13
:
21
27
.

94

Zingarelli
B
,
Cuzzocrea
S
,
Zsengeller
Z
,
Salzman
AL
,
Szabo
C
.
Protection against myocardial ischemia and reperfusion injury by 3-aminobenzamide, an inhibitor of poly (ADP-ribose) synthetase
.
Cardiovasc Res
1997
;
36
:
205
215
.

95

Cordis
GA
,
Maulik
G
,
Bagchi
D
,
Riedel
W
,
Das
DK
.
Detection of oxidative DNA damage to ischemic reperfused rat hearts by 8-hydroxydeoxyguanosine formation
.
J Mol Cell Cardiol
1998
;
30
:
1939
1944
.

96

Stefani
GP
,
Nunes
RB
,
Rossato
DD
,
Hentschke
VS
,
Domenico
MD
,
Lago
PD
,
Rhoden
CR
.
Quantification of DNA damage in different tissues in rats with heart failure
.
Arq Bras Cardiol
2020
;
114
:
234
242
.

97

Chiang
MH
,
Liang
CJ
,
Lin
LC
,
Yang
YF
,
Huang
CC
,
Chen
YH
,
Kao
HL
,
Chen
YC
,
Ke
SR
,
Lee
CW
,
Lin
MS
,
Chen
YL
.
miR-26a attenuates cardiac apoptosis and fibrosis by targeting ataxia–telangiectasia mutated in myocardial infarction
.
J Cell Physiol
2020
;
235
:
6085
6102
.

98

Yu
H
,
Zhang
F
,
Yan
P
,
Zhang
S
,
Lou
Y
,
Geng
Z
,
Li
Z
,
Zhang
Y
,
Xu
Y
,
Lu
Y
,
Chen
C
,
Wang
D
,
Zhu
W
,
Hu
X
,
Wang
J
,
Zhuang
T
,
Zhang
Y
,
Wu
G
,
Liu
J
,
Zeng
C
,
Pu
WT
,
Sun
K
,
Zhang
B
.
LARP7 protects against heart failure by enhancing mitochondrial biogenesis
.
Circulation
2021
;
143
:
2007
2022
.

99

Włodarczyk
M
,
Nowicka
G
.
Obesity, DNA damage, and development of obesity-related diseases
.
Int J Mol Sci
2019
;
20
:
1146
.

100

Kumar
V
,
Agrawal
R
,
Pandey
A
,
Kopf
S
,
Hoeffgen
M
,
Kaymak
S
,
Bandapalli
OR
,
Gorbunova
V
,
Seluanov
A
,
Mall
MA
,
Herzig
S
,
Nawroth
PP
.
Compromised Dna repair is responsible for diabetes-associated fibrosis
.
EMBO J
2020
;
39
:
e103477
.

101

Barouch
LA
,
Gao
D
,
Chen
L
,
Miller
KL
,
Xu
W
,
Phan
AC
,
Kittleson
MM
,
Minhas
KM
,
Berkowitz
DE
,
Wei
C
,
Hare
JM
.
Cardiac myocyte apoptosis is associated with increased DNA damage and decreased survival in murine models of obesity
.
Circ Res
2006
;
98
:
119
124
.

102

Shimizu
I
,
Yoshida
Y
,
Katsuno
T
,
Tateno
K
,
Okada
S
,
Moriya
J
,
Yokoyama
M
,
Nojima
A
,
Ito
T
,
Zechner
R
,
Komuro
I
,
Kobayashi
Y
,
Minamino
T
.
p53-induced adipose tissue inflammation is critically involved in the development of insulin resistance in heart failure
.
Cell Metab
2012
;
15
:
51
64
.

103

Yokoyama
M
,
Okada
S
,
Nakagomi
A
,
Moriya
J
,
Shimizu
I
,
Nojima
A
,
Yoshida
Y
,
Ichimiya
H
,
Kamimura
N
,
Kobayashi
Y
,
Ohta
S
,
Fruttiger
M
,
Lozano
G
,
Minamino
T
.
Inhibition of endothelial p53 improves metabolic abnormalities related to dietary obesity
.
Cell Rep
2014
;
7
:
1691
1703
.

104

Herrmann
J
.
Adverse cardiac effects of cancer therapies: cardiotoxicity and arrhythmia
.
Nat Rev Cardiol
2020
;
17
:
474
502
.

105

Koelwyn
GJ
,
Aboumsallem
JP
,
Moore
KJ
,
de Boer
RA
.
Reverse cardio-oncology: exploring the effects of cardiovascular disease on cancer pathogenesis
.
J Mol Cell Cardiol
2022
;
163
:
1
8
.

106

Koelwyn
GJ
,
Newman
AAC
,
Afonso
MS
,
van Solingen
C
,
Corr
EM
,
Brown
EJ
,
Albers
KB
,
Yamaguchi
N
,
Narke
D
,
Schlegel
M
,
Sharma
M
,
Shanley
LC
,
Barrett
TJ
,
Rahman
K
,
Mezzano
V
,
Fisher
EA
,
Park
DS
,
Newman
JD
,
Quail
DF
,
Nelson
ER
,
Caan
BJ
,
Jones
LW
,
Moore
KJ
.
Myocardial infarction accelerates breast cancer via innate immune reprogramming
.
Nat Med
2020
;
26
:
1452
1458
.

107

Aboumsallem
JP
,
Moslehi
J
,
de Boer
RA
.
Reverse cardio-oncology: cancer development in patients with cardiovascular disease
.
J Am Heart Assoc
2020
;
9
:
e013754
.

108

Mehdizadeh
M
,
Aguilar
M
,
Thorin
E
,
Ferbeyre
G
,
Nattel
S
.
The role of cellular senescence in cardiac disease: basic biology and clinical relevance
.
Nat Rev Cardiol
2022
;
19
:
250
264
.

109

Piegari
E
,
De Angelis
A
,
Cappetta
D
,
Russo
R
,
Esposito
G
,
Costantino
S
,
Graiani
G
,
Frati
C
,
Prezioso
L
,
Berrino
L
,
Urbanek
K
,
Quaini
F
,
Rossi
F
.
Doxorubicin induces senescence and impairs function of human cardiac progenitor cells
.
Basic Res Cardiol
2013
;
108
:
334
.

110

Yoshida
M
,
Shiojima
I
,
Ikeda
H
,
Komuro
I
.
Chronic doxorubicin cardiotoxicity is mediated by oxidative DNA damage-ATM-p53-apoptosis pathway and attenuated by pitavastatin through the inhibition of Rac1 activity
.
J Mol Cell Cardiol
2009
;
47
:
698
705
.

111

Kuno
A
,
Hosoda
R
,
Tsukamoto
M
,
Sato
T
,
Sakuragi
H
,
Ajima
N
,
Saga
Y
,
Tada
K
,
Taniguchi
Y
,
Iwahara
N
,
Horio
Y
.
SIRT1 in the cardiomyocyte counteracts doxorubicin-induced cardiotoxicity via regulating histone H2AX
.
Cardiovasc Res
2022
;
118
:
3360
3373
.

112

Sahin
E
,
Colla
S
,
Liesa
M
,
Moslehi
J
,
Müller
FL
,
Guo
M
,
Cooper
M
,
Kotton
D
,
Fabian
AJ
,
Walkey
C
,
Maser
RS
,
Tonon
G
,
Foerster
F
,
Xiong
R
,
Wang
YA
,
Shukla
SA
,
Jaskelioff
M
,
Martin
ES
,
Heffernan
TP
,
Protopopov
A
,
Ivanova
E
,
Mahoney
JE
,
Kost-Alimova
M
,
Perry
SR
,
Bronson
R
,
Liao
R
,
Mulligan
R
,
Shirihai
OS
,
Chin
L
,
DePinho
RA
.
Telomere dysfunction induces metabolic and mitochondrial compromise
.
Nature
2011
;
470
:
359
365
.

113

Jacinto
TA
,
Meireles
GS
,
Dias
AT
,
Aires
R
,
Porto
ML
,
Gava
AL
,
Vasquez
EC
,
Pereira
TMC
,
Campagnaro
BP
,
Meyrelles
SS
.
Increased ROS production and DNA damage in monocytes are biomarkers of aging and atherosclerosis
.
Biol Res
2018
;
51
:
33
.

114

Matthews
C
,
Gorenne
I
,
Scott
S
,
Figg
N
,
Kirkpatrick
P
,
Ritchie
A
,
Goddard
M
,
Bennett
M
.
Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress
.
Circ Res
2006
;
99
:
156
164
.

115

Mahmoudi
M
,
Gorenne
I
,
Mercer
J
,
Figg
N
,
Littlewood
T
,
Bennett
M
.
Statins use a novel Nijmegen breakage syndrome-1-dependent pathway to accelerate DNA repair in vascular smooth muscle cells
.
Circ Res
2008
;
103
:
717
725
.

116

Bennett
MR
,
Evan
GI
,
Schwartz
SM
.
Apoptosis of human vascular smooth muscle cells derived from normal vessels and coronary atherosclerotic plaques
.
J Clin Invest
1995
;
95
:
2266
2274
.

117

Tabas
I
.
Consequences and therapeutic implications of macrophage apoptosis in atherosclerosis: the importance of lesion stage and phagocytic efficiency
.
Arterioscler Thromb Vasc Biol
2005
;
25
:
2255
2264
.

118

Gray
K
,
Bennett
M
.
Role of DNA damage in atherosclerosis—Bystander or participant?
Biochem Pharmacol
2011
;
82
:
693
700
.

119

Li
M
,
Vattulainen
S
,
Aho
J
,
Orcholski
M
,
Rojas
V
,
Yuan
K
,
Helenius
M
,
Taimen
P
,
Myllykangas
S
,
De Jesus Perez
V
,
Koskenvuo
JW
,
Alastalo
TP
.
Loss of bone morphogenetic protein receptor 2 is associated with abnormal DNA repair in pulmonary arterial hypertension
.
Am J Respir Cell Mol Biol
2014
;
50
:
1118
1128
.

120

Ranchoux
B
,
Meloche
J
,
Paulin
R
,
Boucherat
O
,
Provencher
S
,
Bonnet
S
.
DNA damage and pulmonary hypertension
.
Int J Mol Sci
2016
;
17
:
990
.

121

Lee
SD
,
Shroyer
KR
,
Markham
NE
,
Cool
CD
,
Voelkel
NF
,
Tuder
RM
.
Monoclonal endothelial cell proliferation is present in primary but not secondary pulmonary hypertension
.
J Clin Invest
1998
;
101
:
927
934
.

122

Tuder
RM
,
Radisavljevic
Z
,
Shroyer
KR
,
Polak
JM
,
Voelkel
NF
.
Monoclonal endothelial cells in appetite suppressant-associated pulmonary hypertension
.
Am J Respir Crit Care Med
1998
;
158
:
1999
2001
.

123

Chen
PI
,
Cao
A
,
Miyagawa
K
,
Tojais
NF
,
Hennigs
JK
,
Li
CG
,
Sweeney
NM
,
Inglis
AS
,
Wang
L
,
Li
D
,
Ye
M
,
Feldman
BJ
,
Rabinovitch
M
.
Amphetamines promote mitochondrial dysfunction and DNA damage in pulmonary hypertension
.
JCI Insight
2017
;
2
:
e90427
.

124

Kaur
G
,
Singh
N
,
Lingeshwar
P
,
Siddiqui
HH
,
Hanif
K
.
Poly (ADP-ribose) polymerase-1: an emerging target in right ventricle dysfunction associated with pulmonary hypertension
.
Pulm Pharmacol Ther
2015
;
30
:
66
79
.

125

Schreiber
KH
,
Kennedy
BK
.
When lamins go bad: nuclear structure and disease
.
Cell
2013
;
152
:
1365
1375
.

126

Banerjee
I
,
Zhang
J
,
Moore-Morris
T
,
Pfeiffer
E
,
Buchholz
KS
,
Liu
A
,
Ouyang
K
,
Stroud
MJ
,
Gerace
L
,
Evans
SM
,
McCulloch
A
,
Chen
J
,
Wang
D-Z
.
Targeted ablation of nesprin 1 and nesprin 2 from murine myocardium results in cardiomyopathy, altered nuclear morphology and inhibition of the biomechanical gene response
.
PLoS Genet
2014
;
10
:
e1004114
.

127

Liu
Y
,
Drozdov
I
,
Shroff
R
,
Beltran
LE
,
Shanahan
CM
.
Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence-associated secretory phenotype in vascular smooth muscle cells
.
Circ Res
2013
;
112
:
e99
e109
.

128

Warren
DT
,
Tajsic
T
,
Porter
LJ
,
Minaisah
RM
,
Cobb
A
,
Jacob
A
,
Rajgor
D
,
Zhang
QP
,
Shanahan
CM
.
Nesprin-2-dependent ERK1/2 compartmentalisation regulates the DNA damage response in vascular smooth muscle cell ageing
.
Cell Death Differ
2015
;
22
:
1540
1550
.

129

Heffler
J
,
Shah
PP
,
Robison
P
,
Phyo
S
,
Veliz
K
,
Uchida
K
,
Bogush
A
,
Rhoades
J
,
Jain
R
,
Prosser
BL
.
A balance between intermediate filaments and microtubules maintains nuclear architecture in the cardiomyocyte
.
Circ Res
2020
;
126
:
e10
e26
.

130

Aydin
S
,
Uzun
H
,
Sozer
V
,
Altug
T
.
Effects of atorvastatin therapy on protein oxidation and oxidative DNA damage in hypercholesterolemic rabbits
.
Pharmacol Res
2009
;
59
:
242
247
.

131

Pernice
F
,
Floccari
F
,
Caccamo
C
,
Belghity
N
,
Mantuano
S
,
Pacile
ME
,
Romeo
A
,
Nostro
L
,
Barilla
A
,
Crasci
E
,
Frisina
N
,
Buemi
M
.
Chromosomal damage and atherosclerosis. A protective effect from simvastatin
.
Eur J Pharmacol
2006
;
532
:
223
229
.

132

Vilahur
G
,
Casani
L
,
Peña
E
,
Juan-Babot
O
,
Mendieta
G
,
Crespo
J
,
Badimon
L
.
HMG-CoA reductase inhibition prior reperfusion improves reparative fibrosis post-myocardial infarction in a preclinical experimental model
.
Int J Cardiol
2014
;
175
:
528
538
.

133

Abdel-Wahab
BA
,
Metwally
ME
,
El-khawanki
MM
,
Hashim
AM
.
Protective effect of captopril against clozapine-induced myocarditis in rats: role of oxidative stress, proinflammatory cytokines and DNA damage
.
Chem Biol Interact
2014
;
216
:
43
52
.

134

Keles
MS
,
Bayir
Y
,
Suleyman
H
,
Halici
Z
.
Investigation of effects of Lacidipine, Ramipril and Valsartan on DNA damage and oxidative stress occurred in acute and chronic periods following isoproterenol-induced myocardial infarct in rats
.
Mol Cell Biochem
2009
;
328
:
109
117
.

135

Brand
S
,
Amann
K
,
Mandel
P
,
Zimnol
A
,
Schupp
N
,
Bader
M
.
Oxidative DNA damage in kidneys and heart of hypertensive mice is prevented by blocking angiotensin II and aldosterone receptors
.
PLoS One
2014
;
9
:
e115715
.

136

Chen
YL
,
Chung
SY
,
Chai
HT
,
Chen
CH
,
Liu
CF
,
Chen
YL
,
Huang
TH
,
Zhen
YY
,
Sung
PH
,
Sun
CK
,
Chua
S
,
Lu
HI
,
Lee
FY
,
Sheu
JJ
,
Yip
HK
.
Early administration of carvedilol protected against doxorubicin-induced cardiomyopathy
.
J Pharmacol Exp Ther
2015
;
355
:
516
527
.

137

Lee
J
,
Lee
M
,
Kim
JU
,
Song
KI
,
Choi
YS
,
Cheong
SS
.
Carvedilol reduces plasma 8-hydroxy-2′-deoxyguanosine in mild to moderate hypertension: a pilot study
.
Hypertension
2005
;
45
:
986
990
.

138

Azqueta
A
,
Collins
A
.
Polyphenols and DNA damage: a mixed blessing
.
Nutrients
2016
;
8
:
785
.

139

Riso
P
,
Klimis-Zacas
D
,
Del Bo
C
,
Martini
D
,
Campolo
J
,
Vendrame
S
,
Moller
P
,
Loft
S
,
De Maria
R
,
Porrini
M
.
Effect of a wild blueberry (Vaccinium angustifolium) drink intervention on markers of oxidative stress, inflammation and endothelial function in humans with cardiovascular risk factors
.
Eur J Nutr
2013
;
52
:
949
961
.

140

Botden
IP
,
Oeseburg
H
,
Durik
M
,
Leijten
FP
,
Van Vark-Van Der Zee
LC
,
Musterd-Bhaggoe
UM
,
Garrelds
IM
,
Seynhaeve
AL
,
Langendonk
JG
,
Sijbrands
EJ
,
Danser
AH
,
Roks
AJ
.
Red wine extract protects against oxidative-stress-induced endothelial senescence
.
Clin Sci (Lond)
2012
;
123
:
499
507
.

141

Serino
A
,
Salazar
G
.
Protective role of polyphenols against vascular inflammation, aging and cardiovascular disease
.
Nutrients
2019
;
11
:
53
.

142

Xia
N
,
Daiber
A
,
Förstermann
U
,
Li
H
.
Antioxidant effects of resveratrol in the cardiovascular system
.
Br J Pharmacol
2017
;
174
:
1633
1646
.

143

Sedeek
M
,
Hébert
RL
,
Kennedy
CR
,
Burns
KD
,
Touyz
RM
.
Molecular mechanisms of hypertension: role of Nox family NADPH oxidases
.
Curr Opin Nephrol Hypertens
2009
;
18
:
122
127
.

144

Münzel
T
,
Gori
T
,
Bruno
RM
,
Taddei
S
.
Is oxidative stress a therapeutic target in cardiovascular disease?
Eur Heart J
2010
;
31
:
2741
2748
.

145

Dietrich
M
,
Jacques
PF
,
Pencina
MJ
,
Lanier
K
,
Keyes
MJ
,
Kaur
G
,
Wolf
PA
,
D’Agostino
RB
,
Vasan
RS
.
Vitamin E supplement use and the incidence of cardiovascular disease and all-cause mortality in the Framingham Heart Study: does the underlying health status play a role?
Atherosclerosis
2009
;
205
:
549
553
.

146

Katsiki
N
,
Manes
C
.
Is there a role for supplemented antioxidants in the prevention of atherosclerosis?
Clin Nutr
2009
;
28
:
3
9
.

147

Donmez-Altuntas
H
,
Bayram
F
,
Coskun-Demirkalp
AN
,
Baspınar
O
,
Kocer
D
,
Toth
PP
.
Therapeutic effects of statins on chromosomal DNA damage of dyslipidemic patients
.
Exp Biol Med (Maywood)
2019
;
244
:
1089
1095
.

148

Spyridopoulos
I
,
Haendeler
J
,
Urbich
C
,
Brummendorf
TH
,
Oh
H
,
Schneider
MD
,
Zeiher
AM
,
Dimmeler
S
.
Statins enhance migratory capacity by upregulation of the telomere repeat-binding factor TRF2 in endothelial progenitor cells
.
Circulation
2004
;
110
:
3136
3142
.

149

Ostrau
C
,
Hülsenbeck
J
,
Herzog
M
,
Schad
A
,
Torzewski
M
,
Lackner
KJ
,
Fritz
G
.
Lovastatin attenuates ionizing radiation-induced normal tissue damage in vivo
.
Radiother Oncol
2009
;
92
:
492
499
.

150

Nübel
T
,
Damrot
J
,
Roos
WP
,
Kaina
B
,
Fritz
G
.
Lovastatin protects human endothelial cells from killing by ionizing radiation without impairing induction and repair of DNA double-strand breaks
.
Clin Cancer Res
2006
;
12
:
933
939
.

151

Sørensen
AL
,
Hasselbalch
HC
,
Nielsen
CH
,
Poulsen
HE
,
Ellervik
C
.
Statin treatment, oxidative stress and inflammation in a Danish population
.
Redox Biol
2019
;
21
:
101088
.

152

Kim
S
,
Iwao
H
.
Molecular and cellular mechanisms of angiotensin II-mediated cardiovascular and renal diseases
.
Pharmacol Rev
2000
;
52
:
11
34
.

153

Herbert
KE
,
Mistry
Y
,
Hastings
R
,
Poolman
T
,
Niklason
L
,
Williams
B
.
Angiotensin II-mediated oxidative DNA damage accelerates cellular senescence in cultured human vascular smooth muscle cells via telomere-dependent and independent pathways
.
Circ Res
2008
;
102
:
201
208
.

154

Khaper
N
,
Singal
PK
.
Modulation of oxidative stress by a selective inhibition of angiotensin II type 1 receptors in MI rats
.
J Am Coll Cardiol
2001
;
37
:
1461
1466
.

155

Brand
S
,
Amann
K
,
Schupp
N
.
Angiotensin II-induced hypertension dose-dependently leads to oxidative stress and DNA damage in mouse kidneys and hearts
.
J Hypertens
2013
;
31
:
333
344
.

156

Yu
X
,
Cheng
X
,
Xie
JJ
,
Liao
MY
,
Yao
R
,
Chen
Y
,
Ding
YJ
,
Tang
TT
,
Liao
YH
.
Poly (ADP-ribose) polymerase inhibition improves endothelial dysfunction induced by hyperhomocysteinemia in rats
.
Cardiovasc Drugs Ther
2009
;
23
:
121
127
.

157

Hans
CP
,
Zerfaoui
M
,
Naura
AS
,
Troxclair
D
,
Strong
JP
,
Matrougui
K
,
Boulares
AH
.
Thieno[2,3-c]isoquinolin-5-one, a potent poly(ADP-ribose) polymerase inhibitor, promotes atherosclerotic plaque regression in high-fat diet-fed apolipoprotein E-deficient mice: effects on inflammatory markers and lipid content
.
J Pharmacol Exp Ther
2009
;
329
:
150
158
.

158

Soriano
FG
,
Pacher
P
,
Mabley
J
,
Liaudet
L
,
Szabó
C
.
Rapid reversal of the diabetic endothelial dysfunction by pharmacological inhibition of poly(ADP-ribose) polymerase
.
Circ Res
2001
;
89
:
684
691
.

159

Szabo
C
,
Pacher
P
,
Zsengeller
Z
,
Vaslin
A
,
Komjati
K
,
Benko
R
,
Chen
M
,
Mabley
JG
,
Kollai
M
.
Angiotensin II-mediated endothelial dysfunction: role of poly(ADP-ribose) polymerase activation
.
Mol Med
2004
;
10
:
28
35
.

160

Song
ZF
,
Ji
XP
,
Li
XX
,
Wang
SJ
,
Wang
SH
,
Zhang
Y
.
Inhibition of the activity of poly (ADP-ribose) polymerase reduces heart ischaemia/reperfusion injury via suppressing JNK-mediated AIF translocation
.
J Cell Mol Med
2008
;
12
:
1220
1228
.

161

Song
ZF
,
Chen
DY
,
Du
B
,
Ji
XP
.
Poly (ADP-ribose) polymerase inhibitor reduces heart ischaemia/reperfusion injury via inflammation and Akt signalling in rats
.
Chin Med J (Engl)
2013
;
126
:
1913
1917
.

162

Morrow
DA
,
Brickman
CM
,
Murphy
SA
,
Baran
K
,
Krakover
R
,
Dauerman
H
,
Kumar
S
,
Slomowitz
N
,
Grip
L
,
McCabe
CH
,
Salzman
AL
.
A randomized, placebo-controlled trial to evaluate the tolerability, safety, pharmacokinetics, and pharmacodynamics of a potent inhibitor of poly(ADP-ribose) polymerase (INO-1001) in patients with ST-elevation myocardial infarction undergoing primary percutaneous coronary intervention: results of the TIMI 37 trial
.
J Thromb Thrombolysis
2009
;
27
:
359
364
.

163

Erbel
C
,
Achenbach
J
,
Akhavanpoor
M
,
Dengler
TJ
,
Lasitschka
F
,
Gleissner
CA
,
Bea
F
,
Katus
HA
,
Szabo
G
.
PARP inhibition in atherosclerosis and its effects on dendritic cells, T cells and auto-antibody levels
.
Eur J Med Res
2011
;
16
:
367
374
.

164

Beller
CJ
,
Radovits
T
,
Kosse
J
,
Gero
D
,
Szabo
C
,
Szabo
G
.
Activation of the peroxynitrite-poly(adenosine diphosphate-ribose) polymerase pathway during neointima proliferation: a new target to prevent restenosis after endarterectomy
.
J Vasc Surg
2006
;
43
:
824
830
.

165

Hu
F
,
Liu
C
,
Liu
H
,
Xie
L
,
Yu
L
.
Ataxia-Telangiectasia Mutated (ATM) protein signaling participates in development of pulmonary arterial hypertension in rats
.
Med Sci Monit
2017
;
23
:
4391
4400
.

166

Medunjanin
S
,
Daniel
JM
,
Weinert
S
,
Dutzmann
J
,
Burgbacher
F
,
Brecht
S
,
Bruemmer
D
,
Kahne
T
,
Naumann
M
,
Sedding
DG
,
Zuschratter
W
,
Braun-Dullaeus
RC
.
DNA-dependent protein kinase (DNA-PK) permits vascular smooth muscle cell proliferation through phosphorylation of the orphan nuclear receptor NOR1
.
Cardiovasc Res
2015
;
106
:
488
497
.

167

Satoh
K
,
Kikuchi
N
,
Kurosawa
R
,
Shimokawa
H
.
Checkpoint kinase 1 promotes the development of pulmonary arterial hypertension
.
Arterioscler Thromb Vasc Biol
2019
;
39
:
1504
1506
.

168

Wu
WH
,
Bonnet
S
,
Shimauchi
T
,
Toro
V
,
Grobs
Y
,
Romanet
C
,
Bourgeois
A
,
Vitry
G
,
Omura
J
,
Tremblay
E
,
Nadeau
V
,
Orcholski
M
,
Breuils-Bonnet
S
,
Martineau
S
,
Ferraro
P
,
Potus
F
,
Paulin
R
,
Provencher
S
,
Boucherat
O
.
Potential for inhibition of checkpoint kinases 1/2 in pulmonary fibrosis and secondary pulmonary hypertension
.
Thorax
2022
;
77
:
247
258
.

169

Liu
X
,
Chua
CC
,
Gao
J
,
Chen
Z
,
Landy
CL
,
Hamdy
R
,
Chua
BH
.
Pifithrin-α protects against doxorubicin-induced apoptosis and acute cardiotoxicity in mice
.
Am J Physiol Heart Circ Physiol
2004
;
286
:
H933
H939
.

170

Liu
J
,
Mao
W
,
Ding
B
,
Liang
CS
.
ERKs/p53 signal transduction pathway is involved in doxorubicin-induced apoptosis in H9c2 cells and cardiomyocytes
.
Am J Physiol Heart Circ Physiol
2008
;
295
:
H1956
H1965
.

171

Sardão
VA
,
Oliveira
PJ
,
Holy
J
,
Oliveira
CR
,
Wallace
KB
.
Doxorubicin-induced mitochondrial dysfunction is secondary to nuclear p53 activation in H9c2 cardiomyoblasts
.
Cancer Chemother Pharmacol
2009
;
64
:
811
827
.

172

Liu
P
,
Xu
B
,
Cavalieri
TA
,
Hock
CE
.
Pifithrin-alpha attenuates p53-mediated apoptosis and improves cardiac function in response to myocardial ischemia/reperfusion in aged rats
.
Shock
2006
;
26
:
608
614
.

173

Zhang
Y
,
Kohler
K
,
Xu
J
,
Lu
D
,
Braun
T
,
Schlitt
A
,
Buerke
M
,
Muller-Werdan
U
,
Werdan
K
,
Ebelt
H
.
Inhibition of p53 after acute myocardial infarction: reduction of apoptosis is counteracted by disturbed scar formation and cardiac rupture
.
J Mol Cell Cardiol
2011
;
50
:
471
478
.

174

Mazelin
L
,
Panthu
B
,
Nicot
AS
,
Belotti
E
,
Tintignac
L
,
Teixeira
G
,
Zhang
Q
,
Risson
V
,
Baas
D
,
Delaune
E
,
Derumeaux
G
,
Taillandier
D
,
Ohlmann
T
,
Ovize
M
,
Gangloff
YG
,
Schaeffer
L
.
mTOR inactivation in myocardium from infant mice rapidly leads to dilated cardiomyopathy due to translation defects and p53/JNK-mediated apoptosis
.
J Mol Cell Cardiol
2016
;
97
:
213
225
.

175

Gu
J
,
Wang
S
,
Guo
H
,
Tan
Y
,
Liang
Y
,
Feng
A
,
Liu
Q
,
Damodaran
C
,
Zhang
Z
,
Keller
BB
,
Zhang
C
,
Cai
L
.
Inhibition of p53 prevents diabetic cardiomyopathy by preventing early-stage apoptosis and cell senescence, reduced glycolysis, and impaired angiogenesis
.
Cell Death Dis
2018
;
9
:
82
.

176

Kraus
WL
,
Hottiger
MO
.
PARP-1 and gene regulation: progress and puzzles
.
Mol Aspects Med
2013
;
34
:
1109
1123
.

177

Ordog
K
,
Horvath
O
,
Eros
K
,
Bruszt
K
,
Toth
S
,
Kovacs
D
,
Kalman
N
,
Radnai
B
,
Deres
L
,
Gallyas
F
Jr
,
Toth
K
,
Halmosi
R
.
Mitochondrial protective effects of PARP-inhibition in hypertension-induced myocardial remodeling and in stressed cardiomyocytes
.
Life Sci
2021
;
268
:
118936
.

178

Feng
X
,
Wang
Y
,
Chen
W
,
Xu
S
,
Li
L
,
Geng
Y
,
Shen
A
,
Gao
H
,
Zhang
L
,
Liu
S
.
SIRT3 inhibits cardiac hypertrophy by regulating PARP-1 activity
.
Aging (Albany NY)
2020
;
12
:
4178
4192
.

179

Wang
H
,
Yang
X
,
Yang
Q
,
Gong
L
,
Xu
H
,
Wu
Z
.
PARP-1 inhibition attenuates cardiac fibrosis induced by myocardial infarction through regulating autophagy
.
Biochem Biophys Res Commun
2018
;
503
:
1625
1632
.

180

Liaudet
L
,
Szabo
E
,
Timashpolsky
L
,
Virag
L
,
Cziraki
A
,
Szabo
C
.
Suppression of poly (ADP-ribose) polymerase activation by 3-aminobenzamide in a rat model of myocardial infarction: long-term morphological and functional consequences
.
Br J Pharmacol
2001
;
133
:
1424
1430
.

181

Toth-Zsamboki
E
,
Horvath
E
,
Vargova
K
,
Pankotai
E
,
Murthy
K
,
Zsengeller
Z
,
Barany
T
,
Pek
T
,
Fekete
K
,
Kiss
RG
,
Preda
I
,
Lacza
Z
,
Gero
D
,
Szabo
C
.
Activation of poly(ADP-ribose) polymerase by myocardial ischemia and coronary reperfusion in human circulating leukocytes
.
Mol Med
2006
;
12
:
221
228
.

182

Khan
TA
,
Ruel
M
,
Bianchi
C
,
Voisine
P
,
Komjáti
K
,
Szabo
C
,
Sellke
FW
.
Poly(ADP-ribose) polymerase inhibition improves postischemic myocardial function after cardioplegia-cardiopulmonary bypass
.
J Am Coll Surg
2003
;
197
:
270
277
.

183

Xiao
CY
,
Chen
M
,
Zsengeller
Z
,
Szabo
C
.
Poly(ADP-ribose) polymerase contributes to the development of myocardial infarction in diabetic rats and regulates the nuclear translocation of apoptosis-inducing factor
.
J Pharmacol Exp Ther
2004
;
310
:
498
504
.

184

Roesner
JP
,
Mersmann
J
,
Bergt
S
,
Bohnenberg
K
,
Barthuber
C
,
Szabo
C
,
Nöldge-Schomburg
GEF
,
Zacharowski
K
.
Therapeutic injection of PARP inhibitor INO-1001 preserves cardiac function in porcine myocardial ischemia and reperfusion without reducing infarct size
.
Shock
2010
;
33
:
507
512
.

185

Henning
RJ
,
Bourgeois
M
,
Harbison
RD
.
Poly(ADP-ribose) polymerase (PARP) and PARP inhibitors: mechanisms of action and role in cardiovascular disorders
.
Cardiovasc Toxicol
2018
;
18
:
493
506
.

186

Shiloh
Y
,
Ziv
Y
.
The ATM protein kinase: regulating the cellular response to genotoxic stress, and more
.
Nat Rev Mol Cell Biol
2013
;
14
:
197
210
.

187

Weber
AM
,
Ryan
AJ
.
ATM and ATR as therapeutic targets in cancer
.
Pharmacol Ther
2015
;
149
:
124
138
.

188

Goodwin
JF
,
Knudsen
KE
.
Beyond DNA repair: DNA-PK function in cancer
.
Cancer Discov
2014
;
4
:
1126
1139
.

189

Zhou
H
,
Zhu
P
,
Wang
J
,
Toan
S
,
Ren
J
.
DNA-PKcs promotes alcohol-related liver disease by activating Drp1-related mitochondrial fission and repressing FUNDC1-required mitophagy
.
Signal Transduct Target Ther
2019
;
4
:
56
.

190

Zhou
H
,
Du
W
,
Li
Y
,
Shi
C
,
Hu
N
,
Ma
S
,
Wang
W
,
Ren
J
.
Effects of melatonin on fatty liver disease: the role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission, and mitophagy
.
J Pineal Res
2018
;
64
:
e12450
.

191

Bartunek
J
,
Vanderheyden
M
,
Knaapen
MW
,
Tack
W
,
Kockx
MM
,
Goethals
M
.
Deoxyribonucleic acid damage/repairproteins are elevated in the failing human myocardium due to idiopathic dilated cardiomyopathy
.
J Am Coll Cardiol
2002
;
40
:
1097
1103
.
discussion 1104–1095
.

192

Bartek
J
,
Lukas
J
.
Chk1 and Chk2 kinases in checkpoint control and cancer
.
Cancer Cell
2003
;
3
:
421
429
.

193

Oh
H
,
Wang
SC
,
Prahash
A
,
Sano
M
,
Moravec
CS
,
Taffet
GE
,
Michael
LH
,
Youker
KA
,
Entman
ML
,
Schneider
MD
.
Telomere attrition and Chk2 activation in human heart failure
.
Proc Natl Acad Sci U S A
2003
;
100
:
5378
5383
.

194

Bourgeois
A
,
Bonnet
S
,
Breuils-Bonnet
S
,
Habbout
K
,
Paradis
R
,
Tremblay
E
,
Lampron
MC
,
Orcholski
ME
,
Potus
F
,
Bertero
T
,
Peterlini
T
,
Chan
SY
,
Norris
KA
,
Paulin
R
,
Provencher
S
,
Boucherat
O
.
Inhibition of CHK 1 (Checkpoint Kinase 1) elicits therapeutic effects in pulmonary arterial hypertension
.
Arterioscler Thromb Vasc Biol
2019
;
39
:
1667
1681
.

195

Stewart
SA
,
Weinberg
RA
.
Telomeres: cancer to human aging
.
Annu Rev Cell Dev Biol
2006
;
22
:
531
557
.

196

Men
H
,
Cai
H
,
Cheng
Q
,
Zhou
W
,
Wang
X
,
Huang
S
,
Zheng
Y
,
Cai
L
.
The regulatory roles of p53 in cardiovascular health and disease
.
Cell Mol Life Sci
2021
;
78
:
2001
2018
.

Author notes

Conflict of interest: None declared.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)