Review Article

How Can We Survive the Heart Failure Pandemic Era? Basic Research for Understanding the Pathophysiology of Heart Failure and Development of Novel Therapies

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

Heart failure is caused by a variety of factors, and although many new drug and non-drug therapies have been developed for its treatment in the past decades, the number of patients with heart failure continues to increase and its prognosis is still poor. In this era, it is necessary to continue to develop new therapies. For that purpose, basic research aimed at a deeper understanding of the pathogenesis of heart failure is essential. To that end, an investigation of the ischaemia-mediated molecular mechanisms of various processes, from adaptive cardiac hypertrophy to contractile failure and DNA damage, as a cause of heart failure, has been undertaken. A novel stratification of heart failure based on comprehensive mutation and gene expression analyses has also been carried out. This article reviews the recent advances in basic heart failure research and treatment. In the future, understanding of the diversity of this disease at the molecular level is expected to lead to the development of optimal therapies that are tailored to the individual patient.

Disclosure:MI has received grants from Agnes Inc. All other authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Issei Komuro, Department of Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan. E: komuro_tky2000@yahoo.co.jp

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Heart failure (HF) is a clinical syndrome that represents the final manifestation of many underlying cardiovascular disorders and is the most common cause of hospitalisation in the elderly population.1 The number of patients with HF has been increasing rapidly over the past few decades in Japan, mainly because of the ageing population.2,3 Epidemiological transition and population ageing are no exception in Asian countries, and HF is expected to become a larger epidemic in the near future.4

Despite decades of therapeutic development, cardiovascular disease remains the leading cause of morbidity and mortality globally, accounting for an estimated 18.6 million deaths annually in the US.5 In Japan, the 3-year mortality associated with cardiovascular death in hospitalised patients with HF is approximately 20%.6 In a study that compared the changes in the outcomes of Japanese patients with HF between the 2000s and the 2010s, the rate of rehospitalisation for patients with both HF with preserved ejection fraction (HFpEF) and HF with reduced ejection fraction (HFrEF) significantly decreased in the 2010s; however, the cardiac mortality rates were not significantly reduced in either HFpEF and HFrEF.7

In recent decades, numerous clinical trials for HF have revolutionised the management and understanding of its pathophysiology.8 In the 1980s, angiotensin-converting enzyme inhibitors (ACEIs) were proven to be effective for the treatment of various functional classes of HF, demonstrating the importance of neurohormone factors in HF. In the late 1990s, mineralocorticoid receptor antagonists (MRAs) and β-blockers were identified as two drug classes with a great impact on mortality in patients with HFrEF. The combination of ACEI, MRA, and β-blocker reigned as a cornerstone therapy for patients with HFrEF until the 2010s.

In the 2000s, implantable devices such as ICDs and CRT produced a reduction in mortality as well as an improvement in the quality of life in patients with HFrEF.9 And over the last two decades, mechanical circulatory support and left ventricular assist devices (LVADs) have altered the landscape of HF management and are now used as either a bridge to transplant (BTT) or as an alternative to heart transplantation (destination therapy: DT).10 Superior outcomes with the newest, third-generation LVAD (a fully magnetically levitated centrifugal-flow LVAD) have been observed, particularly with respect to blood compatibility-related adverse events, including pump thrombosis and stroke.11 In addition, the superior treatment effect of third-generation LVAD was similar in patients with BTT or DT.12

In the 2010s, new classes of drugs, such as angiotensin receptor–neprilysin inhibitor (ARNI), sodium-glucose cotransporter 2 inhibitors (SGLT2Is), and ivabradine have gained attention.13–15 The combination of ARNI, MRA, β-blocker and SGLT2I is now recognised as the ‘fantastic four’ or ‘foundation therapy’, which has been shown to improve the prognosis of HF compared with conventional therapy and is now incorporated into the guidelines of HF therapy.16–20 Recently, two other classes of drug have been tested in clinical trials. Vericiguat, a soluble guanylate cyclase stimulator, has been shown to reduce cardiovascular death and hospitalisation for HF in the Victoria trial, while omecamtiv mecarbil, a selective cardiac myosin activator, reduced the primary outcome in the GALACTIC-HF trial.21,22 Furthermore, drugs for specific heart diseases, such as mavacamten for hypertrophic obstructive cardiomyopathy or tafamidis for transthyretin amyloid cardiomyopathy, are also expected to reduce HF associated with these diseases.23–25

Despite the long history of the development of therapeutic approaches for HF described above, significant knowledge gaps remain regarding robust and specific biomarkers for risk prediction, comprehensive understanding of the pathogenesis of HF, and critical regulators of HF development. A more comprehensive and systematic understanding of HF through basic research is indispensable for significant breakthroughs in its treatment.

Research on Heart Failure

Mechanism of Heart Failure

HF has many potential causes, including ischaemia, haemodynamic overload, ageing, inflammation and differentiation abnormalities. In many cases, an intricate combination of multiple risk factors triggers HF.26 Although the pathogenesis of HF is complex, we have elucidated some aspects of the underlying molecular mechanisms through basic research.

Heart Failure and Cardiac Hypertrophy

Cardiac hypertrophy is the primary sign of HF. Pathological cardiac hypertrophy causes HF owing to inadequate signalling, leading to unfavourable remodelling. The most common causes of cardiac hypertrophy are hypertension, MI, valvular disease, and genetic mutation (hypertrophic cardiomyopathy; HCM). As the disease progresses, cardiac contractility decreases, occasionally resulting in HF; however, the mechanism of collapse is still unclear.

Several pathways induce pathological hypertrophy. It has long been known that transforming growth factor-β/Smad signalling is involved in pathological hypertrophy.27 In addition, various factors, such as angiotensin II, endothelin-1, mechanical forces and catecholamines, can activate intracellular hypertrophic signalling pathways. The calcineurin–NFAT (nuclear factor of activated T-cells) pathway also causes hypertrophy by increasing fetal gene expression.28 Thus, the activation of various signalling pathways has been shown to be involved in the pathological response process, which provides evidence for the use of several clinical therapeutic agents that manipulate neurohormone factors regulating the signalling pathways responsible for HF.

We focused on elucidating the molecular mechanism underlying this transition from compensated to decompensated hypertrophy. One of the conventional features of decompensated hypertrophy is the occurrence of adverse changes in tissue oxygenation, such as the suppression of angiogenesis, endothelial dysfunction, and fibrosis of myocardial tissue. In hypertrophic hearts, increases in sarcomere synthesis and cardiomyocyte volume result in hypoxia of myocardial tissue due to the relative lack of vasculature and oxygen supply. Although hypoxia is usually ameliorated by induction of angiogenesis via activation of hypoxia-inducible factor (HIF), angiogenesis is suppressed in failing hearts by elevated expression of p53, which inhibits and degrades HIF. As a result, myocardial tissue is unable to evade ischaemia and contractility is reduced. These abnormalities are restored by inhibition of p53 (Figure 1).29

Figure 1: Molecular Mechanisms of HF via the DNA Damage–p53–Ischaemia Pathway

Article image

Subsequently, a number of studies have focused on the transition to pathological systolic dysfunction from hypertrophy and have reported the involvement of a variety of factors, including immune cells, hydrogen sulfide signalling, extracellular matrix remodelling, as well as hypoxia-related pathways.30–32 Many attempts are underway to identify signals, enzymes and molecules other than those targeted by existing therapeutics to prevent hypertrophy-related decompensatory failure.

Figure 2: DNA Damage Accumulation is a Cause and Prognostic Factor of Heart Failure

Article image

Heart Failure and DNA Damage

After determining the unfavourable effects of p53 activation in failing hearts, we focused on the association between the accumulation of DNA damage, a major cause of p53 activation, and cardiac dysfunction. In a subsequent study, we generated and analysed a mouse model of pressure overload HF, in which comet assays showed that DNA single-strand breaks accumulated in cardiomyocytes.33 Cardiomyocyte-specific knockout mice of Xrcc1, a DNA repair protein (αMHC-Cre tg/-; Xrcc1 flox/flox) had a higher degree of DNA damage accumulation in cardiomyocytes after pressure overload and a higher mortality rate (Figure 2A,B). Furthermore, when these mice were crossed with Atm knockout mice, an initiator of the DNA damage response (DDR), the expression of inflammatory cytokines was reduced and cardiac dysfunction was suppressed. These results indicate that the accumulation of DNA damage in the heart is the cause of HF and suggest the possibility of suppressing cardiac dysfunction through DDR inhibition (Figure 2C).

The accumulation of DNA damage is a major cause of cellular senescence. Furthermore, senescent cells acquire the ability to secrete specific cytokines, chemokines and matrix-remodelling proteases, known as senescence-associated secretory phenotypes, which negatively affect tissue function. Multiple reports indicate that the long-term accumulation of senescent cells impairs cardiac function and promotes heart disease.34 In contrast, removal of senescent cells has been shown to improve cardiac function in a continuous angiotensin II infusion model, suggesting that senescence due to DNA damage is a cause of HF.34

Recently, various groups have reported that accumulation of DNA damage in cardiomyocytes underlies the pathogenesis of HF. Zhang et al. reported that calmodulin-dependent kinase (CaMK) IIδ, a splice variant of CaMKII, is abundantly expressed in the heart and that this isotype protein causes DNA damage accumulation by inhibiting DNA damage repair through degradation of the ubiquitin-conjugating enzyme (UBE2T).35 Sato et al. showed that Caren, a cardiomyocyte-specific lncRNA, maintains cardiomyocyte function by suppressing DDR and activating mitochondrial respiration through the repression of Hint1 expression.36

Zhao et al. reported that the level of circulating resistin (Retn) in patients with HF is associated with miR-148b-3p expression, which suppresses the expression of Gadd45a and increases DNA damage, resulting in myocardial fibrosis, apoptosis and impaired cardiac function.37 Cao et al. narrowed down the genes associated with diastolic dysfunction in the HFpEF mouse heart transcriptome using a genome-wide association study of HF and identified Ascc2 as a DNA damage-reversal molecule. They showed that Ascc2 repression leads to Atm phosphorylation, increased Nppa expression and inflammatory responses, indicating that disruption of Ascc2-mediated DNA damage inhibition underlies the pathogenesis of HFpEF.38

Thus, this accumulating evidence reinforces the importance of DNA damage in HF and suggests the therapeutic potential of modifying this complex pathological process.

Heart Failure and Prognostic Factors

Brain natriuretic peptide (BNP) is probably the most clinically used marker for HF.39 Because HF is caused by a complex interplay of many pathological factors, including inflammation, oxidative stress and fibrosis, a variety of markers have been reported in addition to BNP. Interleukin 1 (IL-1), IL-6, tumour necrosis factor-α and highly sensitive C-reactive protein are known to be involved in inflammation, and for oxidative stress, oxidative low-density lipoprotein, myeloperoxidase and urinary biopyrrins have been reported. Cardiac troponin I and troponin T are useful markers of myocardial injury, which occurs in HF.40–42 In addition, cytokines and fibrosis markers, such as Igfbp7, resistin, osteoprotegerin and galectin 3, have also been noted as possible causes of HF.43–46

In our previous work, we also investigated whether we could predict the outcome of HF by assessing the degree of DNA damage in heart tissues using the DNA damage marker poly-ADP-ribose (PAR). Some patients recover from HF after adequate medical treatment, a phenomenon known as left ventricular reverse remodelling (LVRR). LVRR is correlated with a favourable prognosis for HF. We used myocardial biopsy specimens (collected at the time of diagnosis) from patients with HF due to dilated cardiomyopathy (DCM) and analysed the relationship between PAR positivity in cardiomyocytes and the incidence of LVRR. We found that patients with higher PAR positivity were less likely to develop LVRR and had poorer prognosis (Figure 2D). This indicates that DNA damage is also related to the reversibility and prognosis of HF in the clinical setting (Figure 2E).47

Genetics and Heart Failure

Advances in genetic engineering have enabled a better understanding of disease.48 In mice, knockout or overexpression of genes involved in cardiovascular diseases has enabled their function to be verified, thus advancing our understanding of the pathogenesis on a gene-by-gene basis. However, findings derived from experiments using mice are not always transferable to humans. Thus, by analysing the genomic DNA of patients and correlating genotypes with clinical and prognostic data, it is possible to deepen our understanding of the relationship between disease and genes to generate new stratifications of disease groups, and to predict outcomes. Expanding our knowledge by combining murine and human data is the key to medical genome science.49

Genetic Mutations and Heart Failure

Given that HF has various causes, there are only a limited number of cases in which a single molecular abnormality explains the pathophysiology. However, approximately 40% of DCM patients have a family history of DCM, suggesting the involvement of several gene factors.50 Although more than 100 genes have been reported to be causative factors for DCM, the relationship between mutation type and clinical course has not been fully elucidated.50 We performed panel sequencing of genetic variants in 120 Japanese patients with DCM and analysed the association between genetic variants and the clinical course.51 We found that titin-truncating variants (TTNtvs) were the most common (16.7%), and Lamin A/C (LMNA) mutations were the second most common (10.8%). Patients with TTNtvs responded to medical therapy and had a better prognosis, whereas those with LMNA mutations did not respond to existing treatments and had a poorer prognosis. The patient’s genetic information may be reflected in policy decisions for advanced therapies such as device implantation and heart transplantation. As the analysis of other HF causative diseases and the accumulation of genetic knowledge progresses, it is expected to lead to the realisation of precision medicine, in which genetic knowledge is applied to clinical decision-making.

Disease Stratification Based on Genetics

Gene sequencing technology has developed dramatically over the past two decades, enabling both genomic and transcriptomic information to be obtained inexpensively and rapidly, contributing to new biological insights.

Many previous studies have been limited to a few genes and intracellular signalling pathways that appear to be associated with disease phenotypes using gene expression data obtained from diseased animal organs or patient specimens. Recently, however, the reduction in the time and cost required to acquire transcriptome data has made it possible to follow the dynamics of comprehensive gene expression in organs at disease onset and during disease progression, even at the single-cell level. As a result, it is now possible to use longitudinal samples to infer the pathways of gene expression changes that lead to disease based on similarities in expression patterns.

Using a mouse model of HF, we investigated the gene expression patterns of cardiomyocytes by temporal single-cell analysis.52,53 Chronological analysis of murine hearts after pressure overload showed that pressure loading first caused compensatory hypertrophy of cardiomyocytes, followed by bidirectional bifurcation into compensated and failing states of cardiomyocytes, and myocardial remodelling during the development of HF. Analysis of the relationship between the degree of hypertrophy and gene expression in hypertrophied cardiomyocytes indicated activation of ERK1/2 and Nrf1/2 signals, which are involved in the hypertrophic response and antioxidant defence of the heart. We also analysed the gene network activated during the transition from hypertrophy to failure and found that p53 was an important regulator of cell fate conversion. This observation confirms that the cellular identity and morphological phenotypes of cardiomyocytes are encoded by a transcriptional program.53 Thus, by analysing gene expression at the turning points in cell fate, we can elucidate the molecular changes in tissues early in disease development, the heterogeneous processes of adaptation and disruption, and the factors that lead to a point of no return in disease progression.

A series of studies have been published on cardiac single-nuclei RNA sequencing (snRNA-seq) in patients with HF, which investigated the diversity of gene expression in patients, in disease aetiology, and in the heart of a single patient with HF. Koenig et al. performed snRNA-seq of DCM and healthy hearts and identified the major cardiac cell types and their specific transcriptional programs. They found cell type-specific transcriptional networks that regulate ageing and disease status. In the samples, the cardiomyocytes converged on a relatively common disease-related gene expression state, while fibroblasts and myeloid cells underwent dramatic disease-specific transcriptional diversification. In particular, the shift of fibroblasts to an activated state and the expression of CD14+ monocyte-derived inflammatory mediators were specific to failing hearts, suggesting that intervention in the cellular linkage network, including non-cardiomyocytes, may lead to novel therapeutic strategies.54

Chaffin et al. performed snRNA-seq of DCM, HCM and control human hearts, and described genetic alterations in failing hearts.55 In addition to constitutive changes in cell types between cardiomyopathy and non-failing hearts, they found common transcriptional pathways in cardiomyopathy patients, a decrease in proliferative CCR2-negative macrophages in the heart, and an increase in activated fibroblasts in the heart. Furthermore, in combination with CRISPR screening, they identified novel genes (PRELP and JAZF1) that contributed to fibroblast activation. This demonstrates the transcriptional diversity of healthy and diseased conditions in the human heart, as well as the shared molecular mechanisms involving non-cardiomyocytes and potential new therapeutic targets for HF.55

Reichart et al. used snRNA-seq in samples from patients with DCM and arrhythmogenic cardiomyopathy (ACM). They found that the cardiac cell atlas in DCM and ACM had different responses in the right and left ventricles, and that constitutive cardiac cell types, gene expressions, molecular signals and predicted intercellular communications differed according to the type of genetic mutation. This provides substantial evidence that genotype influences the pathological remodelling of cardiomyopathy and highlights heterogeneity in the cellular and molecular architecture of human HF. It reverses the common belief that HF converges on a common final pathway and provides guidelines for the future development of selectively targeted therapies to achieve personalised medicine.56

Conclusion

Many new drug and non-drug therapies have been developed for the treatment of HF in the past two decades. This process has been achieved through numerous clinical trials and post-marketing surveillance, and our field has been successful in further improving the prognosis of patients with HF. However, in order to address unmet needs, it is increasingly important to continue to update the existing clinical guidelines. One of the remaining challenges in HF management is the presence of non-responders to therapy. If the comprehensive genetic information-based practice described in this review becomes possible, diagnosis of the aetiology of HF may be more precisely subdivided, leading to the possibility of individualised and optimised treatment for each patient.

The other challenge is the achievement of preventive intervention at an earlier stage; prevention of HF requires interventions appropriate to each stage of disease progression. Such interventions include lifestyle modification to prevent the onset of the disease; treatment of lifestyle-related diseases such as hypertension and diabetes, which are established cardiovascular risk factors; appropriate treatment of underlying diseases such as ischaemic heart disease, arrhythmia and valvular disease; and maintenance therapy and rehabilitation to prevent transition to end-stage HF. As described above, we have proposed a new paradigm in the pathogenesis of HF, including the importance of myocardial ischaemia in the transition from cardiac hypertrophy to HF and the critical role of DNA damage and p53 activation in the progression of HF, focuses mainly on the treatment of the underlying disease and the prevention of transition to end-stage HF. Hence, the use of effective preventive measures at an earlier stage would be ideal from the standpoint of burden on medical resources. It is also desirable to broaden the scope of research in the fields of lifestyle modification and the treatment of lifestyle-related diseases in order to elucidate the mechanisms of HF even before its onset.

Basic research on HF is expected to improve the understanding of its pathogenesis and promote the development of new treatments for the next generation, thereby extending healthy life expectancies worldwide.

Clinical Perspective

  • The prognosis for heart failure (HF) has improved with the development of various therapies.
  • To combat the pandemic of HF in the growing elderly population, it is necessary to establish novel methods of treatment and prevention based on an accurate understanding of pathogenesis. Therefore, basic research in this area is important.
  • The accumulation of DNA damage and subsequent p53 activation play an important role in the transition from cardiac hypertrophy to functional failure, and these hallmarks are useful for predicting the therapeutic response.
  • Understanding the disease at the molecular level is important for precise and accurate diagnosis. With the development of nucleic acid sequencing technology, it is now possible to examine gene mutations and single-cell gene expression in patients with HF. Disease reclassification and treatment decisions based on this information are expected to advance in the near future.

References

  1. Mosterd A, Hoes AW. Clinical epidemiology of heart failure. Heart 2007;93:1137–46.
    Crossref | PubMed
  2. Okura Y, Ramadan MM, Ohno Y, et al. Impending epidemic: future projection of heart failure in Japan to the year 2055. Circ J 2008;72:489–91.
    Crossref | PubMed
  3. Yasuda S, Miyamoto Y, Ogawa H. Current status of cardiovascular medicine in the aging society of Japan. Circulation 2018;138:965–7.
    Crossref | PubMed
  4. Shimokawa H, Miura M, Nochioka K, Sakata Y. Heart failure as a general pandemic in Asia. Eur J Heart Fail 2015;17:884–92.
    Crossref | PubMed
  5. Virani SS, Alonso A, Aparicio HJ, et al. Heart disease and stroke statistics 2021 update: a report from the American Heart Association. Circulation 2021;143:e254–743.
    Crossref | PubMed
  6. Ide T, Kaku H, Matsushima S, et al. Clinical characteristics and outcomes of hospitalized patients with heart failure from the large-scale Japanese Registry of Acute Decompensated Heart Failure (JROADHF). Circ J 2021;85:1438–50.
    Crossref | PubMed
  7. Suzuki A, Shiga T, Kawashiro N, et al. Changes in characteristics and outcomes in Japanese patients with heart failure from the 2000s to the 2010s: the HIJ-HF cohorts. J Cardiol 2020;76:132–8.
    Crossref | PubMed
  8. Sharma A, Verma S, Bhatt DL, et al. Optimizing foundational therapies in patients with HFrEF: how do we translate these findings into clinical care? JACC Basic Transl Sci 2022;7:504–17.
    Crossref | PubMed
  9. Breitenstein A, Steffel J. Devices in heart failure patients: who benefits from ICD and CRT? Front Cardiovasc Med 2019;6:111.
    Crossref | PubMed
  10. Han JJ, Acker MA, Atluri P. Left ventricular assist devices. Circulation 2018;138:2841–51.
    Crossref | PubMed
  11. Mehra MR, Uriel N, Naka Y, et al. A fully magnetically levitated left ventricular assist device: final report. N Engl J Med 2019;380:1618–27.
    Crossref | PubMed
  12. Goldstein DJ, Naka Y, Horstmanshof D, et al. Association of clinical outcomes with left ventricular assist device use by bridge to transplant or destination therapy intent: the Multicenter Study of MagLev Technology in Patients Undergoing Mechanical Circulatory Support Therapy with HeartMate 3 (MOMENTUM 3) randomized clinical trial. JAMA Cardiol 2020;5:411–9.
    Crossref | PubMed
  13. McMurray JJ, Packer M, Desai AS, et al. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med 2014;371:993–1004.
    Crossref | PubMed
  14. Braunwald E. Gliflozins in the management of cardiovascular disease. N Engl J Med 2022;386:2024–34.
    Crossref | PubMed
  15. Swedberg K, Komajda M, Bohm M, et al. Ivabradine and outcomes in chronic heart failure (SHIFT): a randomised placebo-controlled study. Lancet 2010;376:875–85.
    Crossref | PubMed
  16. Bauersachs J. Heart failure drug treatment: the fantastic four. Eur Heart J 2021;42:681–3.
    Crossref | PubMed
  17. McMurray JJV, Docherty KF. Insights into foundational therapies for heart failure with reduced ejection fraction. Clin Cardiol 2022;45(Suppl 1):S26–30.
    Crossref | PubMed
  18. Vaduganathan M, Claggett BL, Jhund PS, et al. Estimating lifetime benefits of comprehensive disease-modifying pharmacological therapies in patients with heart failure with reduced ejection fraction: a comparative analysis of three randomised controlled trials. Lancet 2020;396:121–8.
    Crossref | PubMed
  19. Mebazaa A, Davison B, Chioncel O, et al. Safety, Tolerability and Efficacy of Up-titration of Guideline-directed Medical Therapies for Acute Heart Failure (STRONG-HF): a multinational, open-label, randomised, trial. Lancet 2022;400:1938–52.
    Crossref | PubMed
  20. Morris AA, Butler J. Updated heart failure guidelines: time for a refresh. Circulation 2022;145:1371–3.
    Crossref | PubMed
  21. Armstrong PW, Pieske B, Anstrom KJ, et al. Vericiguat in patients with heart failure and reduced ejection fraction. N Engl J Med 2020;382:1883–93.
    Crossref | PubMed
  22. Teerlink JR, Diaz R, Felker GM, et al. Cardiac myosin activation with omecamtiv mecarbil in systolic heart failure. N Engl J Med 2021;384:105–16.
    Crossref | PubMed
  23. Spertus JA, Fine JT, Elliott P, et al. Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (Explorer-HCM): health status analysis of a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2021;397:2467–75.
    Crossref | PubMed
  24. Hegde SM, Lester SJ, Solomon SD, et al. Effect of mavacamten on echocardiographic features in symptomatic patients with obstructive hypertrophic cardiomyopathy. J Am Coll Cardiol 2021;78:2518–32.
    Crossref | PubMed
  25. Maurer MS, Schwartz JH, Gundapaneni B, et al. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med 2018;379:1007–16.
    Crossref | PubMed
  26. Ziaeian B, Fonarow GC. Epidemiology and aetiology of heart failure. Nat Rev Cardiol 2016;13:368–78.
    Crossref | PubMed
  27. Dobaczewski M, Chen W, Frangogiannis NG. Transforming growth factor (TGF)-beta signaling in cardiac remodeling. J Mol Cell Cardiol 2011;51:600–6.
    Crossref | PubMed
  28. Nakamura M, Sadoshima J. Mechanisms of physiological and pathological cardiac hypertrophy. Nat Rev Cardiol 2018;15:387–407.
    Crossref | PubMed
  29. Sano M, Minamino T, Toko H, et al. p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload. Nature 2007;446:444–8.
    Crossref | PubMed
  30. Revelo XS, Parthiban P, Chen C, et al. Cardiac resident macrophages prevent fibrosis and stimulate angiogenesis. Circ Res 2021;129:1086–101.
    Crossref | PubMed
  31. Jackson MR, Cox KD, Baugh SDP, et al. Discovery of a first-in-class inhibitor of sulfide:quinone oxidoreductase that protects against adverse cardiac remodelling and heart failure. Cardiovasc Res 2022;118:1771–84.
    Crossref | PubMed
  32. El Hajj EC, El Hajj MC, Ninh VK, et al. Detrimental role of lysyl oxidase in cardiac remodeling. J Mol Cell Cardiol 2017;109:17–26.
    Crossref | PubMed
  33. Higo T, Naito AT, Sumida T, et al. DNA single-strand break-induced DNA damage response causes heart failure. Nat Commun 2017;8:15104.
    Crossref | PubMed
  34. Mehdizadeh M, Aguilar M, Thorin E, et al. The role of cellular senescence in cardiac disease: basic biology and clinical relevance. Nat Rev Cardiol 2022;19:250–64.
    Crossref | PubMed
  35. Zhang M, Gao H, Liu D, et al. CaMKII-delta9 promotes cardiomyopathy through disrupting UBE2T-dependent DNA repair. Nat Cell Biol 2019;21:1152–63.
    Crossref | PubMed
  36. Sato M, Kadomatsu T, Miyata K, et al. The lncRNA Caren antagonizes heart failure by inactivating DNA damage response and activating mitochondrial biogenesis. Nat Commun 2021;12:2529.
    Crossref | PubMed
  37. Zhao B, Bouchareb R, Lebeche D. Resistin deletion protects against heart failure injury by targetin_blankg DNA damage response. Cardiovasc Res 2022;118:1947–63.
    Crossref | PubMed
  38. Cao Y, Pan C, Wang YC, et al. Identification of DNA damage repair enzyme Ascc2 as causal for heart failure with preserved ejection fraction. Circulation 2022;145:1102–4.
    Crossref | PubMed
  39. Castiglione V, Aimo A, Vergaro G, et al. Biomarkers for the diagnosis and management of heart failure. Heart Fail Rev 2022;27:625–43.
    Crossref | PubMed
  40. Tsutsui T, Tsutamoto T, Wada A, et al. Plasma oxidized low-density lipoprotein as a prognostic predictor in patients with chronic congestive heart failure. J Am Coll Cardiol 2002;39:957–62.
    Crossref | PubMed
  41. Tang WH, Brennan ML, Philip K, et al. Plasma myeloperoxidase levels in patients with chronic heart failure. Am J Cardiol 2006;98:796–9.
    Crossref | PubMed
  42. Hokamaki J, Kawano H, Yoshimura M, et al. Urinary biopyrrins levels are elevated in relation to severity of heart failure. J Am Coll Cardiol 2004;43:1880–5.
    Crossref | PubMed
  43. Ahmed A, Ahmed S, Arvidsson M, et al. Elevated plasma sRAGE and IGFBP7 in heart failure decrease after heart transplantation in association with haemodynamics. ESC Heart Fail 2020;7:2340–53.
    Crossref | PubMed
  44. Frankel DS, Vasan RS, D’Agostino RB Sr, et al. Resistin, adiponectin, and risk of heart failure: The Framingham offspring study. J Am Coll Cardiol 2009;53:754–62.
    Crossref | PubMed
  45. di Giuseppe R, Biemann R, Wirth J, et al. Plasma osteoprotegerin, its correlates, and risk of heart failure: a prospective cohort study. Eur J Epidemiol 2017;32:113–23.
    Crossref | PubMed
  46. Ho JE, Liu C, Lyass A, et al. Galectin-3, a marker of cardiac fibrosis, predicts incident heart failure in the community. J Am Coll Cardiol 2012;60:1249–56.
    Crossref | PubMed
  47. Ko T, Fujita K, Nomura S, et al. 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;4:670–80.
    Crossref | PubMed
  48. Brown SDM. Advances in mouse genetics for the study of human disease. Hum Mol Genet 2021;30:R274–84.
    Crossref | PubMed
  49. Green ED, Gunter C, Biesecker LG, et al. Strategic vision for improving human health at the forefront of genomics. Nature 2020;586:683–92.
    Crossref | PubMed
  50. McNally EM, Mestroni L. Dilated cardiomyopathy: genetic determinants and mechanisms. Circ Res 2017;121:731–48.
    Crossref | PubMed
  51. Tobita T, Nomura S, Fujita T, et al. Genetic basis of cardiomyopathy and the genotypes involved in prognosis and left ventricular reverse remodeling. Sci Rep 2018;8:1998.
    Crossref | PubMed
  52. Yamada S, Nomura S. Review of single-cell RNA sequencing in the heart. Int J Mol Sci 2020;21:8345.
    Crossref | PubMed
  53. Nomura S, Satoh M, Fujita T, et al. Cardiomyocyte gene programs encoding morphological and functional signatures in cardiac hypertrophy and failure. Nat Commun 2018;9:4435.
    Crossref | PubMed
  54. Koenig AL, Shchukina I, Amrute J, et al. Single-cell transcriptomics reveals cell-type-specific diversification in human heart failure. Nat Cardiovasc Res 2022;1:263–80.
    Crossref | PubMed
  55. Chaffin M, Papangeli I, Simonson B, et al. Single-nucleus profiling of human dilated and hypertrophic cardiomyopathy. Nature 2022;608:174–80.
    Crossref | PubMed
  56. Reichart D, Lindberg EL, Maatz H, et al. Pathogenic variants damage cell composition and single cell transcription in cardiomyopathies. Science 2022;377:eabo1984.
    Crossref | PubMed