Review Article

Molecular Mechanisms of the Failing Heart: A Fatal Regression?

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 (HF) is one of the most common causes of death, and the number of HF patients is increasing worldwide due to population ageing. The pathogenesis of HF has been extensively studied by many researchers with a focus on cardiomyocytes, but its complex pathophysiology has yet to be elucidated. Non-cardiomyocytes account for >70% of the cells that comprise the heart, and there is close communication between non-cardiomyocytes and cardiomyocytes, suggesting that non-cardiomyocytes might play a pivotal role in the development of HF. Neurohumoral factors, such as the autonomic nerves and hormones, regulate the heart’s function. Conversely, the heart affects many other organs through blood perfusion, underscoring the importance of interorgan communication. This review discusses the role of non-cardiomyocytes and interorgan communication between the heart and other organs in the development of HF, a topic that has not been extensively explored.

Disclosure:IK has received honoraria from AstraZeneca and Novo Nordisk, payments from Idorsia, Sanofi and Daiichi Sankyo, and IK is a member of endowment program of Idorsia, Novo Nordisk, Toa Eiyo, Takara Bio, Nippon Boehringer Ingelheim and BioStream. All other authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Issei Komuro, International University of Health and Welfare, 4-1-26 Akasaka, Minato, Tokyo 107-8402, Japan. E: komuro-tky@umin.ac.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.

The number of patients with heart failure (HF) is increasing worldwide due to ageing, and HF is one of the most common causes of death.1 Although there have been many studies on the mechanisms of HF, including abnormal calcium handling, adrenergic receptor overactivation, apoptosis and DNA damage in cardiomyocytes, the complex pathophysiology remains incompletely understood.2–6 The pathogenesis of HF has been extensively studied by many researchers with a focus on cardiomyocytes, but cardiomyocytes constitute only 20–30% of cells forming the heart, with >70% being non-cardiomyocytes, such as endothelial cells, fibroblasts, smooth muscle cells, pericytes and blood cells.7 This highlights the potentially pivotal role of non-cardiomyocytes in HF development.8

A failing heart is not an isolated entity. Haemodynamically, hypoperfusion and congestion induce dysfunction of extracardiac organs, such as the lung, liver and kidney. Elevated liver transaminase levels are associated with an increased risk of mortality, and the presence of chronic kidney disease is a potent poor prognostic factor for patients with HF.9,10 Renal dysfunction is well-known to be intimately related to HF, and albuminuria alone could provoke incident HF.11 Albuminuria is not only reflective of endothelial damage, but also precedes neurohormonal activation, including the renin–angiotensin–aldosterone system, and autonomous nervous system, which disturbs cardiac homeostasis. These results suggest that interorgan communications are deeply involved in the pathological conditions of HF.

The Roles of Non-cardiomyocytes in the Pathogenesis of Heart Failure

Endothelial Cells

The role of endothelial cells in cardiac function has been widely recognised.12 Endothelial cells form a single layer covering the blood vessels’ inner lining and perform many functions, such as the regulation of vasodilation, thromboprophylaxis, anti-inflammatory effects, antioxidant effects and neovascularisation. These functions are impaired in many diseases, including hypertension, obesity, diabetes and hyperlipidaemia, and in ageing individuals and smokers.13 Dysfunction of endothelial cells due to injury is known to be partially reversible, necessitating early therapeutic intervention through pharmacotherapy and lifestyle modification.

Heart Failure and Dysfunction of Endothelial Cells

Since the late 1980s, the concept that endothelial dysfunction in the heart is one of the causes of HF has gradually gained acceptance. Endothelial dysfunction refers primarily to abnormalities in the regulation of vasodilation and contraction, vascular permeability regulation, and inflammatory processes. These endothelial dysfunctions are assessed through peripheral blood flow-dependent vasodilation during congestion and through coronary flow reserve. In patients with cardiovascular diseases or those at risk, abnormal coronary flow reserve is strongly associated with increased risks of death and major adverse cardiac events.14

Endothelial dysfunction is believed to be deeply involved in the pathophysiology of cardiovascular diseases (CVD).15 One of the most crucial molecules responsible for endothelial function is nitric oxide (NO).16 NO is biosynthesised from arginine by the enzyme NO synthase (NOS). There are three isoforms of NOS: endothelial NOS, inducible NOS and neuronal NOS. Each is predominantly expressed in different tissues: endothelial NOS in vascular endothelial cells; inducible NOS in inflammatory cells, such as monocytes, macrophages and neutrophils, and neuronal NOS in the central nervous system. Normal endothelial cells release several relaxants, notably NO, which play a vital role in vascular regulation through guanylyl cyclase-cyclic guanosine monophosphate. Vasodilation is maintained by the continuous synthesis and release of NO, and vasodilation ability is reduced in patients with HF.17

Endothelial dysfunction is caused by an imbalance between endothelium-derived vasodilatory and vasoconstrictive actions, leading to reduced flow-mediated vasodilation and an inability to meet the oxygen demands of the heart. Additionally, endothelial dysfunction impairs antithrombotic activity and induces inflammation. There have been reports showing cardiac dysfunction in endothelial NOS knockout mice.18 In 2020, a phase III trial of the guanylyl cyclase stimulator, vericiguat, was conducted, showing a lower incidence of cardiovascular death and hospitalisation due to HF in high-risk patients treated with vericiguat.19 While the effects of vericiguat on various cells in the heart are known to be multifaceted, at least the improvement in vascular regulatory ability in endothelial cells is thought to contribute to these results.

Heart Failure and Senescence of Endothelial Cells

The relationship between vascular endothelial cells and ageing has been the subject of many studies.20 It has been reported that expression levels of p53 increase in the murine heart after pressure overload, and that p53 plays a critical role in the development of HF.5,21 In mouse models of diabetes, inhibition of p53 has been reported to prevent early-stage myocardial apoptosis and suppress diabetes-induced cellular senescence.22 Additionally, endothelial cell-specific knockout of p53 has been reported to improve cardiac function in a mouse model of pressure-overload HF, with one mechanism being the prevention of apoptosis.23 Another study has shown that heterozygous knockout of p21 in endothelial cells increases angiogenesis without increasing apoptotic cells.24 In mouse models subjected to ischaemia-reperfusion injury, the administration of the senolytic drug, navitoclax, reduced the area of MI.25 Furthermore, we have recently observed that administering CDK4/6 inhibitors to a mouse model of pressure-overload HF improves cardiac function by inhibiting excessive cell proliferation and senescence of endothelial cells. In conclusion, the senescence of endothelial cells is deeply linked to HF, and preventing vascular senescence may become a new therapeutic intervention for HF.

Figure 1: The Crosstalk Between Endothelial Cells and Cardiac Myocytes

Article image

Heart Failure and Intercellular Communication

Substances that mediate intercellular communication are crucial for the crosstalk between endothelial cells and cardiac myocytes (Figure 1). Apart from NO, humoral factors, including angiotensin II, endothelin-1 and neuregulin-1, play pivotal roles in the crosstalk.26 Under various stimuli, such as mechanical stresses and chemical stimuli, endothelin-1 is secreted from endothelial cells, and induces hypertrophy of cardiac myocytes by activating the protein kinase C and mitogen-activated protein kinase kinase/extracellular-regulated kinase pathways, leading to the development of HF.27 Neuregulin-1, which is upregulated when NO levels decrease, induces cardiomyocyte hypertrophy, but protects cardiomyocytes by activating the erythroblastic leukaemia viral oncogene homologue pathway.28 These factors secreted from endothelial cells directly affect cardiomyocytes and are critically involved in the development of HF.

Thus, endothelial cells play pivotal roles in the pathogenesis of HF, not only by forming blood vessels that carry oxygen and nutrients, but also by secreting various factors. This suggests that targeting endothelial cells may offer a potential therapeutic avenue for HF. Various drugs are delivered to organs through the blood vessels, and since endothelial cells are located in the closest proximity to the blood, they are an organ that is easy to target for drug delivery. Given the ongoing poor prognosis associated with HF, the role of endothelial cells is increasingly viewed as a key target for the innovation of new treatments for HF.

Cardiac Fibroblasts

Cardiac fibroblasts constitute 10–15% of cardiac cells. In general, cardiac fibroblasts play a crucial role in fibrosis during wound healing in various organs and tissues by producing various extracellular proteins including collagens, which form the extracellular matrix.29 Cardiac fibroblasts also meticulously generate and arrange collagen to maintain extracellular matrix structure, creating a robust foundation in various situations, such as MI and mechanical stress. They play a vital role both structurally and mechanically in maintaining cardiac function. Due to the scarcity of reliable cardiac fibroblast markers, research on the role of cardiac fibroblasts was primarily confined to their mechanical functions. While markers, such as double data rate 2, ferroptosis suppressor protein 1, Thy-1 (CD90) and vimentin, were employed for fibrotic characterisation, their expression was also found in fibroblasts of other tissues besides the heart.

MI and Cardiac Fibroblasts

The identification of specific markers, such as Tcf21 and Pdgfrα, has propelled advancements in cardiac fibroblast research.30 The primary source of cardiac fibroblasts was identified as the epicardium during embryonic heart development. In hearts affected by MI and hypertrophy, however, the origin of activated fibroblasts was traced back to resident fibroblasts in the heart.31 Furthermore, the identification of periostin as the activated fibroblast marker has facilitated experiments with genetically engineered mice. These studies revealed transformations in cardiac fibroblasts, where resident fibroblasts differentiate into activated fibroblasts in the context of MI or HF. Subsequently, these activated fibroblasts further differentiate into myofibroblasts, contributing to a robust induction of fibrosis.32

Ultimately, in the setting of MI, cardiac fibroblasts undergo differentiation into matrifibrocytes, which lose their proliferative capacity and generate specific extracellular matrix constituent proteins, contributing to the stabilisation of the scar tissue (Figure 2). These findings suggest that cardiac fibroblasts alter their gene expression and morphology in MI to safeguard the heart.33 As mentioned earlier, research on cardiac fibroblasts has predominantly focused on the MI mouse model due to its notable phenotypic changes, and the role of cardiac fibroblasts in other heart diseases remains unclear. The relationship between cardiac dysfunction or the pathophysiology of HF and cardiac fibroblasts is not fully understood.

Figure 2: The Crosstalk Between Fibroblasts and Cardiac Myocytes

Article image

Heart Failure and Cardiac Fibroblasts

In the mouse model of HF induced by pressure overload through transverse aortic constriction, myofibroblasts were absent in the perivascular region, and only 15% of the total cardiac fibroblasts were found in the interstitial region 4 weeks after transverse aortic constriction.33 This result suggests that the function of cardiac fibroblasts in failing hearts differs from their role in stabilising scars by producing collagens in hearts with MI. In failing hearts, fibrosis is recognised to advance from the epicardium to the endocardium, driven by pressure overload-induced wall stress on the heart, microcirculatory disturbances and other factors.33

There are two patterns of fibrosis: one involves the accumulation of extracellular matrix around blood vessels, and the other entails the construction of fibrotic tissue among cardiomyocytes. While research on cardiac fibrosis has traditionally concentrated on mechanical changes, some recent studies have reported its involvement in the pathophysiology of HF. Although it is well-known that cardiac fibrosis is associated with left ventricular diastolic dysfunction, González et al. reported that an excess of fibrosis induces the remodelling of collagen fibres and cardiomyocytes, resulting in left ventricular systolic dysfunction.34 The same study also suggested that cardiac fibrosis could be a causative factor for arrhythmias, and that the accumulation of cardiac fibrosis might impede the delivery of oxygen to cardiomyocytes, leading to ischaemia.34 Another study indicated that eliminating cardiac fibrosis using chimeric antigen receptor T-cells resulted in improved cardiac function.35

While cell populations were once thought to be homogeneous and analysed collectively, the ability to analyse at the single-cell level has revealed significant heterogeneity within cell populations, even among cells of the same type.36 Using single-cell analysis, Ko et al. reported that high-temperature requirement factor A3 in cardiac fibroblasts regulates the amount of transforming growth factor-β (TGF-β), and that deletion of high-temperature requirement factor A3 induces not only marked fibrosis, but also cardiac dysfunction by activating TGF-β signalling and inducing DNA damage in cardiac myocytes.37 This study indicated that cardiac fibroblasts affect cardiac myocytes and play critical roles in the pathogenesis of HF, as well as fibrosis, and suggests that the interaction between cardiac fibroblasts and myocytes is a novel target for HF.

Blood Cells

Growing evidence has shown a relationship between CVD and the immune system.38 The advent of next-generation sequencing has enabled the analysis of acquired mutations in blood cells that were previously not assessable by other techniques, such as flow cytometry or fluorescence microscopy. This has drawn attention to the association between somatic mutation cells and CVD, including HF. Here, we focus on the role of somatic mutations in haematopoietic stem cells in the onset and exacerbation of HF (Figure 3).

Figure 3: The Crosstalk Between Blood Cells and Cardiac Myocytes

Article image

Clonal Haematopoiesis of Indeterminate Potential

Clonal haematopoiesis of indeterminate potential (CHIP), a premalignant state with somatic mutations of haematopoietic stem cells in the genes related to haematological malignancies, is associated with mortality due to various chronic diseases.39 Among them, CVD contributes significantly to the increased mortality from CHIP. The attention to the fact that CHIP worsens the prognosis of CVD began with a large study reported in 2014, which performed a whole exon analysis of approximately 17,000 patients.40,41 In this study, approximately 10% of patients aged >70 years had CHIP, indicating that CHIP is an independent risk factor for ischaemic heart disease and ischaemic stroke. Since then, many studies have examined the association between CHIP and CVD, and many basic studies have shown that CHIP induces inflammatory activation. For example, TET2 mutations induce secretion of interleukin (IL)-1β, a major proinflammatory cytokine, through activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 inflammasome and caspase-1, while DNMT3A and JAK2 mutations also induce excessive secretion of inflammatory cytokines, such as IL-6.42

In approximately 57,000 patients without established HF drawn from five European and US databases, a 25% increased risk of new onset of HF was reported in patients with CHIP compared with those without it.43 The study also showed that this risk was even higher in cases with larger clone sizes, and that the contribution of CHIP was dose dependent. In addition, this study also examined the differential contribution depending on which gene the mutation was in, and found that ASXL1 mutation-positive patients had significantly more left ventricular systolic dysfunction. In contrast, no such trend was observed for DNMT3A or TET2, suggesting that the significance may vary depending on the driver gene. More recent preliminary data have shown that different bases at the same locus of the DNMT3A mutation contribute differently to the development of CVD.44 Future studies that address the differences in effect size by gene and mutation are desirable.

Mosaic Loss of Y Chromosome

In males possessing one X chromosome and one Y chromosome, the gradual loss of the Y chromosome with ageing is referred to as mosaic loss of Y (mLOY). This phenomenon is primarily observed in blood cells, and its association with various chronic diseases has been reported in the literature.45,46 In cardiovascular diseases, it has been reported as an adverse prognostic factor in some conditions, such as cerebral infarction and post-transcatheter aortic valve replacement.47,48 Although studies on the association and mechanisms of mLOY with the development of HF are limited, Sano et al. suggested the following possibilities.49 The mice model with mLOY showed myocardial fibrosis, and the myocardial macrophages, induced by the loss of Y, were found to be in an activated inflammatory state. Additionally, inhibiting TGF-β1 was shown to alleviate myocardial damage, indicating the potential role of mLOY to induce HF through inflammation. Furthermore, using data from the UK Biobank, they demonstrated that men with mLOY had approximately a 30% increased mortality rate due to cardiovascular diseases over a tracking period of about 11 years. This suggests that mLOY could be a useful indicator for clinical stratification. Future studies are anticipated to investigate the role and effect size of mLOY in various CVDs.

Skewed X Chromosome Inactivation

X-chromosome inactivation (XCI) refers to the phenomenon in which one of the X chromosomes is silenced in each cell to regulate the expression levels of genes on the X chromosome.50 This is a phenomenon specific to females who possess two X chromosomes. Originally, XCI was believed to exhibit a 1:1 distribution in a cell population occurring randomly to maintain physiological homeostasis. However, it has become apparent that there is variation in some individuals.51 This phenomenon is referred to as skewed XCI. Roberts et al. analysed skewed XCI using whole blood samples from the TwinsUK registry, focusing on 1,575 women.52 This study quantified XCI, and demonstrated that XCI is an independent predictor of atherosclerotic CVD risk. Atherosclerotic CVD stands as a leading cause of HF, and activated inflammation has been reported to contribute to its pathogenesis. XCI has also been shown to induce an increase in monocytes and a decrease in interleukin-10 with anti-inflammatory properties, suggesting potential involvement in the progression of atherosclerotic CVD through inflammation. Additionally, Roberts et al. indicated that XCI serves as a risk factor for the onset of malignant tumours, implying potential involvement in major societal issues, such as HF and cancer.

The analysis of somatic mutations is expected to contribute to the realisation of further precision in medical care. Despite numerous studies conducted in recent years, it is important to note that these findings may undergo quantitative changes over time. Therefore, future research is eagerly awaited, particularly with the execution of longitudinal studies.

The Roles of Other Organs in the Pathogenesis of Heart Failure

Intercommunication Between the Heart and Kidney

Beneficial potentials of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and mineralocorticoid receptor antagonists for both HF with reduced ejection fraction and chronic kidney disease have been widely accepted. In addition, mounting evidence that sodium-glucose cotransporter 2 inhibitor (SGLT2i) improves the prognosis of both diseases has been building consensus.53 These findings indicate the existence of common pathophysiological mechanisms for HF and kidney disease. SGLT2i sustains favourable effects on failing hearts beyond short-term osmotic diuretic action. SGLT2i-induced ketogenesis promotes nutrient deprivation signalling, including activation of adenosine monophosphate-activated protein kinase and repression of mammalian target of rapamycin, which activates sirtuins and increases the expression of peroxisome proliferator-activated receptor γ coactivator-1α.54 Consequent enhancement of autophagic flux suppresses the development of cardiomyopathies.

SGLT2i also exerts direct cellular effects on cardiomyocytes and renal parenchymal cells regardless of the expression of SGLT2 by unknown mechanisms. Several papers reported the sympatholytic effects of SGLT2i presented as a repressed expression of tyrosine hydroxylase in kidneys.55,56

Regarding the renal sympathetic nerve traffic, the increased efferent signal in the setting of HF leads to inappropriate renal sympathetic tone, and upregulates renin–angiotensin–aldosterone activity and neprilysin expression in kidneys to cause excessive fluid retention, while the afferent sympathetic signal deteriorates cardiometabolic diseases in various ways.57 Augmented input signals from mechanoreceptors and chemoreceptors of injured kidneys are transmitted to the subfornical organ of the brain, which contains angiotensin II receptor type 1a-positive neurons regulating thirst and salt-appetite sensations.58 Subfornical organ neurons partly have connections with the paraventricular nucleus neurons projecting to the rostral ventrolateral medulla, a sympathetic nerve center.59 Kidney injury upregulates angiotensin II receptor type 1a signalling in the subfornical organ and subsequently activates the paraventricular nucleus and the rostral ventrolateral medulla.

Centrally integrated signals through the communications in the brainstem result in an increased sympathetic tone directed towards several organs and detrimental effects on failing hearts.60 However, sympathetic stimulation in kidneys also exerts an adaptive neuroimmune response for failing hearts. Efferent nerves stimulate renal collecting duct epithelial cells, which leads to the secretion of the cytokine, colony-stimulating factor 2.60 Circulating colony-stimulating factor 2 is essential for the maintenance of cardiac tissue macrophages. These tight cooperative communications among the heart, brain and kidney complicate the pathophysiology of a failing heart, and it could be attributed to inconclusive early studies of catheter-based renal denervation, although recent meta-analysis exhibited a beneficial potential of renal denervation for HF (Figure 4).61,62

Figure 4: Neuronal Networks Between the Brain and Kidneys in the Setting of Heart Failure.

Article image

Intercommunication Between the Heart and Gut

Bowel wall oedema due to HF impairs intestinal barrier function, and a leaky gut allows bacterial translocation and the entry of damage-associated molecular patterns and pathogen-associated molecular patterns. The correlations of circulating endotoxin levels and intestinal epithelial cell injury markers with the severity of HF verify the gut hypothesis of HF.63,64 Failing hearts affect the diversity of gut microbial composition. Dysbiosis, a status of decreased intra-individual taxonomic diversity in HF, results in a decreased abundance of Roseburia, Faecalibacterium and Eubacterium rectale, known as butyrate-producing commensal bacteria.65 Short-chain fatty acids, including butyrate and acetate, do not only upregulate the expression of epithelial barrier genes, but also promote immune tolerance; thus, the alteration of microbiota in HF exacerbates inflammatory responses of leaky gut.

Regarding the metabolites produced by microbiota, trimethylamine N-oxide (TMAO) is also noteworthy. Dietary sources rich in choline, phosphatidylcholine and L-carnitine can be metabolised by gut microbiota to trimethylamine (TMA). TMA enters the portal circulation and is oxidised by hepatic flavin-containing monooxygenase into TMAO. While plasma TMAO activates platelets and causes endothelial dysfunction to facilitate atherogenesis, TMAO can directly induce cardiac dysfunction. TMAO activates TGF-β1 signalling to progress cardiac fibrosis.66 TMAO exacerbates mitochondrial function in failing cardiomyocytes via inhibition of the activity of pyruvate dehydrogenase.67 Mounting evidence supports the notion that elevated TMAO levels predict the poor prognosis of patients with HF, although the underlying mechanism has not been fully elucidated, and the clinical efficacy of reduction intervention remains elusive.67

Bacterial translocation and endotoxaemia unsurprisingly heighten inflammatory responses of the host immune system, and chronic low-grade inflammation contributes to the pathogenesis and progression of HF.68 Damage-associated molecular patterns and pathogen-associated molecular patterns stimulate pattern recognition receptors of innate immune cells and even cardiomyocytes to produce various inflammatory cytokines, and activate inflammasome to induce the secretion of IL-1β. IL-1β inhibits L-type calcium channels and desensitises β adrenergic signalling in cardiomyocytes, thereby weakening myocardial contractility, whereas IL-1 signalling decreases calcium reuptake into the sarcoplasmic reticulum to impair diastolic function.69 These mechanistic hypotheses are supported by the evidence that canakinumab, an IL-1β inhibitor, reduced hospitalisation and mortality of decompensated HF patients.70

Meanwhile, recent reports indicate that enhanced immune responses with accompanying epigenetic modulation and metabolic alteration in haematopoietic cells would be a central mechanism driving long-lasting inflammation.71 This trained immunity, or termed innate immune memory depending on the context, is implicated in the development of various CVDs, and especially centrally trained immunity at the level of haematopoietic stem cells (HSCs) in the bone marrow niche would be noticeable. Trained immunity in the setting of aging, referred to as inflammaging, is also triggered by translocations of gut microbiome-derived compounds, which stimulate myeloid cells in the bone marrow to sustain increased IL-1 secretion.72 IL-1 signalling drives preferential expansion of myeloid-skewing HSCs and imprints myeloid differentiation bias of HSCs. Periodontitis, which is also a risk of HF, permits microbial challenge in the circulation, and also upregulates IL-1β-mediated signalling in HSCs.73 Maladaptively trained HSCs could induce myelopoiesis and increased susceptibility to inflammatory comorbid conditions. Myelopoiesis and sustained inflammation could trigger incident HF, as exemplified by the association of CHIP and mLOY with a greater risk of HF, as discussed before.

Although the pathogenesis of HF has been extensively studied with a focus on cardiomyocytes, >70% of the cells in the heart are non-cardiomyocytes, and the interactions between many cell types, including cardiomyocytes and noncardiomyocytes, have been elucidated. HF is also a culminating status of miscellaneous heart diseases, and failing heart is closely intertwined with the pathophysiology of extracardiac organs. Therefore, this review was put together from a slightly different perspective than the more common traditional reviews, namely, the linkage between non-cardiomyocytes and cardiomyocytes, and between organs other than the heart and the heart. The approaches from these novel aspects may open new avenues for developing new treatments for HF.

Clinical Perspective

  • Heart failure is one of the most common causes of death.
  • Non-cardiomyocytes play a pivotal role in the development of heart failure.
  • Interorgan communication between the heart and other organs is important for heart failure.

References

  1. Braunwald E. The war against heart failure: the Lancet lecture. Lancet 2015;385:812–24. 
    Crossref | PubMed
  2. Komuro I, Kurabayashi M, Shibazaki Y, et al. Molecular cloning and characterization of a Ca2+ + Mg2+-dependent adenosine triphosphatase from rat cardiac sarcoplasmic reticulum. Regulation of its expression by pressure overload and developmental stage. J Clin Invest 1989;83:1102–8. 
    Crossref | PubMed
  3. Mudd JO, Kass DA. Tackling heart failure in the twenty-first century. Nature 2008;451:919–28. 
    Crossref | PubMed
  4. Williams RS. Apoptosis and heart failure. N Engl J Med 1999;341:759–60. 
    Crossref | PubMed
  5. 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
  6. 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
  7. Pinto AR, Ilinykh A, Ivey MJ, et al. Revisiting cardiac cellular composition. Circ Res 2016;118:400–9. 
    Crossref | PubMed
  8. Kamo T, Akazawa H, Komuro I. Cardiac nonmyocytes in the hub of cardiac hypertrophy. Circ Res 2015;117:89–98. 
    Crossref | PubMed
  9. Suzuki K, Claggett B, Minamisawa M, et al. Liver function and prognosis, and influence of sacubitril/valsartan in patients with heart failure with reduced ejection fraction. Eur J Heart Fail 2020;22:1662–71. 
    Crossref | PubMed
  10. Damman K, Valente MAE, Voors AA, et al. Renal impairment, worsening renal function, and outcome in patients with heart failure: an updated meta-analysis. Eur Heart J 2014;35:455–69. 
    Crossref | PubMed
  11. Khan MS, Shahid I, Anker SD, et al. Albuminuria and heart failure: JACC state-of-the-art review. J Am Coll Cardiol 2023;81:270–82. 
    Crossref | PubMed
  12. Allbritton-King JD, Garcia-Cardena G. Endothelial cell dysfunction in cardiac disease: driver or consequence? Front Cell Dev Biol 2023;11:1278166. 
    Crossref | PubMed
  13. Minamino T, Miyauchi H, Yoshida T, et al. Vascular cell senescence and vascular aging. J Mol Cell Cardiol 2004;36:175–83. 
    Crossref | PubMed
  14. Kelshiker MA, Seligman H, Howard JP, et al. Coronary flow reserve and cardiovascular outcomes: a systematic review and meta-analysis. Eur Heart J 2022;43:1582–93. 
    Crossref | PubMed
  15. Zuchi C, Tritto I, Carluccio E, et al. Role of endothelial dysfunction in heart failure. Heart Fail Rev 2020;25:21–30. 
    Crossref | PubMed
  16. Farah C, Michel LYM, Balligand JL. Nitric oxide signalling in cardiovascular health and disease. Nat Rev Cardiol 2018;15:292–316. 
    Crossref | PubMed
  17. Kubo SH, Rector TS, Bank AJ, et al. Endothelium-dependent vasodilation is attenuated in patients with heart-failure. Circulation 1991;84:1589–96. 
    Crossref | PubMed
  18. Musicante M, Kim HH, Chen Y, et al. Regulation of endothelial nitric oxide synthase in cardiac remodeling. Int J Cardiol 2022;364:96–101. 
    Crossref | PubMed
  19. 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
  20. Minamino T, Komuro I. Vascular aging: insights from studies on cellular senescence, stem cell aging, and progeroid syndromes. Nat Clin Pract Cardiovasc Med 2008;5:637–48. 
    Crossref | PubMed
  21. 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
  22. Gu J, Wang S, Guo H, et al. 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. 
    Crossref | PubMed
  23. Gogiraju R, Xu X, Bochenek ML, et al. Endothelial p53 deletion improves angiogenesis and prevents cardiac fibrosis and heart failure induced by pressure overload in mice. J Am Heart Assoc 2015;4:e001770. 
    Crossref | PubMed
  24. Bruhl T, Heeschen C, Aicher A, et al. P21Cip1 levels differentially regulate turnover of mature endothelial cells, endothelial progenitor cells, and in vivo neovascularization. Circ Res 2004;94:686–92. 
    Crossref | PubMed
  25. Dookun E, Walaszczyk A, Redgrave R, et al. Clearance of senescent cells during cardiac ischemia-reperfusion injury improves recovery. Aging Cell 2020;19:e13249. 
    Crossref | PubMed
  26. Nakagawa A, Naito AT, Sumida T, et al. Activation of endothelial β-catenin signaling induces heart failure. Sci Rep 2016;6:25009. 
    Crossref | PubMed
  27. Segers VFM, Brutsaert DL, De Keulenaer GW. Cardiac remodeling: endothelial cells have more to say than just NO. Front Physiol 2018;9:382. 
    Crossref | PubMed
  28. Wang Y, Wei J, Zhang P, et al. Neuregulin-1, a potential therapeutic target for cardiac repair. Front Pharmacol 2022;13:945206. 
    Crossref | PubMed
  29. Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020;587:555–66. 
    Crossref | PubMed
  30. Smith CL, Baek ST, Sung CY, Tallquist MD. Epicardial-derived cell epithelial-to-mesenchymal transition and fate specification require PDGF receptor signaling. Circ Res 2011;108:e15–26. 
    Crossref | PubMed
  31. Acharya A, Baek ST, Huang G, et al. The bHLH transcription factor Tcf21 is required for lineage specific EMT of cardiac fibroblast progenitors. Development 2012;139:2139–49. 
    Crossref | PubMed
  32. Tallquist MD, Molkentin JD. Redefining the identity of cardiac fibroblasts. Nat Rev Cardiol 2017;14:484–91. 
    Crossref | PubMed
  33. Fu X, Khalil H, Kanisicak O, et al. Specialized fibroblast differentiated states underlie scar formation in the infarcted mouse heart. J Clin Invest 2018;128:2127–43. 
    Crossref | PubMed
  34. González A, Schelbert EB, Díez J, Butler J. Myocardial interstitial fibrosis in heart failure: biological and translational perspectives. J Am Coll Cardiol 2018;71:1696–706. 
    Crossref | PubMed
  35. Rurik JG, Tombácz I, Yadegari A, et al. CAR T cells produced in vivo to treat cardiac injury. Science 2022;375:91–6. 
    Crossref | PubMed
  36. Macosko EZ, Basu A, Satija R, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 2015;161:1202–14. 
    Crossref | PubMed
  37. Ko T, Nomura S, Yamada S, et al. Cardiac fibroblasts regulate the development of heart failure via Htra3-TGF-beta-IGFBP7 axis. Nat Commun 2022;13:3275. 
    Crossref | PubMed
  38. Swirski FK, Nahrendorf M. Cardioimmunology: the immune system in cardiac homeostasis and disease. Nat Rev Immunol 2018;18:733–44. 
    Crossref | PubMed
  39. Evans MA, Walsh K. Clonal hematopoiesis, somatic mosaicism, and age-associated disease. Physiol Rev 2023;103:649–716. 
    Crossref | PubMed
  40. Jaiswal S, Natarajan P, Silver AJ, et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med 2017;377:111–21. 
    Crossref | PubMed
  41. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014;371:2488–98. 
    Crossref | PubMed
  42. Yura Y, Sano S, Walsh K. Clonal hematopoiesis: a new step linking inflammation to heart failure. JACC Basic Transl Sci 2020;5:196–207. 
    Crossref | PubMed
  43. Yu B, Roberts MB, Raffield LM, et al. Association of clonal hematopoiesis with incident heart failure. J Am Coll Cardiol 2021;78:42–52. 
    Crossref | PubMed
  44. Silver AJ, Brown DJ, Vlasschaert C, et al. DNMT3A R882H exhibits greater inflammatory potential than R882C in primary hematopoietic stem and progenitor cell knock-in model and population data. Blood 2023;142(Suppl 1):815. 
    Crossref
  45. Forsberg LA, Rasi C, Malmqvist N, et al. Mosaic loss of chromosome Y in peripheral blood is associated with shorter survival and higher risk of cancer. Nat Genet 2014;46:624–8. 
    Crossref | PubMed
  46. Dumanski JP, Rasi C, Lönn M, et al. Mutagenesis. Smoking is associated with mosaic loss of chromosome Y. Science 2015;347:81–3. 
    Crossref | PubMed
  47. Dorvall M, Pedersen A, Dumanski JP, et al. Mosaic loss of chromosome Y is associated with functional outcome after ischemic stroke. Stroke 2023;54:2434–7. 
    Crossref | PubMed
  48. Mas-Peiro S, Abplanalp WT, Rasper T, et al. Mosaic loss of Y chromosome in monocytes is associated with lower survival after transcatheter aortic valve replacement. Eur Heart J 2023;44:1943–52. 
    Crossref | PubMed
  49. Sano S, Horitani K, Ogawa H, et al. Hematopoietic loss of Y chromosome leads to cardiac fibrosis and heart failure mortality. Science 2022;377:292–7. 
    Crossref | PubMed
  50. Lyon MF. Gene action in the X-chromosome of the mouse (Mus musculus L.). Nature 1961;190:372–3. 
    Crossref | PubMed
  51. Tukiainen T, Villani AC, Yen A, et al. Landscape of X chromosome inactivation across human tissues. Nature 2017;550:244–8. 
    Crossref | PubMed
  52. Roberts AL, Morea A, Amar A, et al. Age acquired skewed X chromosome inactivation is associated with adverse health outcomes in humans. eLife 2022;11:e78263. 
    Crossref | PubMed
  53. Vaduganathan M, Docherty KF, Claggett BL, et al. SGLT-2 inhibitors in patients with heart failure: a comprehensive meta-analysis of five randomised controlled trials. Lancet 2022;400:757–67. 
    Crossref | PubMed
  54. Packer M. Critical reanalysis of the mechanisms underlying the cardiorenal benefits of SGLT2 inhibitors and reaffirmation of the nutrient deprivation signaling/autophagy hypothesis. Circulation 2022;146:1383–405. 
    Crossref | PubMed
  55. Gueguen C, Burke SL, Barzel B, et al. Empagliflozin modulates renal sympathetic and heart rate baroreflexes in a rabbit model of diabetes. Diabetologia 2020;63:1424–34. 
    Crossref | PubMed
  56. Herat LY, Matthews J, Azzam O, et al. Targeting features of the metabolic syndrome through sympatholytic effects of SGLT2 inhibition. Curr Hypertens Rep 2022;24:67–74. 
    Crossref | PubMed
  57. Patel KP, Katsurada K, Zheng H. Cardiorenal syndrome: the role of neural connections between the heart and the kidneys. Circ Res 2022;130:1601–17. 
    Crossref | PubMed
  58. Matsuda T, Hiyama TY, Niimura F, et al. Distinct neural mechanisms for the control of thirst and salt appetite in the subfornical organ. Nat Neurosci 2017;20:230–41. 
    Crossref | PubMed
  59. Cao W, Yang Z, Liu X, et al. A kidney-brain neural circuit drives progressive kidney damage and heart failure. Signal Transduct Target Ther 2023;8:184. 
    Crossref | PubMed
  60. Fujiu K, Shibata M, Nakayama Y, et al. A heart-brain-kidney network controls adaptation to cardiac stress through tissue macrophage activation. Nat Med 2017;23:611–22. 
    Crossref | PubMed
  61. Bhatt DL, Kandzari DE, O’Neill WW, et al. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014;370:1393–401. 
    Crossref | PubMed
  62. Fukuta H, Goto T, Wakami K, et al. Effects of catheter-based renal denervation on heart failure with reduced ejection fraction: a meta-analysis of randomized controlled trials. Heart Fail Rev 2022;27:29–36. 
    Crossref | PubMed
  63. Niebauer J, Volk HD, Kemp M, et al. Endotoxin and immune activation in chronic heart failure: a prospective cohort study. Lancet 1999;353:1838–42. 
    Crossref | PubMed
  64. Nendl A, Raju SC, Broch K, et al. Intestinal fatty acid binding protein is associated with cardiac function and gut dysbiosis in chronic heart failure. Front Cardiovasc Med 2023;10:1160030. 
    Crossref | PubMed
  65. Tang WHW, Li DY, Hazen SL. Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol 2019;16:137–54. 
    Crossref | PubMed
  66. Zhen J, Zhou Z, He M, et al. The gut microbial metabolite trimethylamine N-oxide and cardiovascular diseases. Front Endocrinol (Lausanne) 2023;14:1085041. 
    Crossref | PubMed
  67. Li X, Fan Z, Cui J, et al. Trimethylamine N-oxide in heart failure: a meta-analysis of prognostic value. Front Cardiovasc Med 2022;9:817396. 
    Crossref | PubMed
  68. Mann DL. Innate immunity and the failing heart: the cytokine hypothesis revisited. Circ Res 2015;116:1254–68. 
    Crossref | PubMed
  69. Murphy SP, Kakkar R, McCarthy CP, Januzzi JL. Inflammation in heart failure: JACC state-of-the-art review. J Am Coll Cardiol 2020;75:1324–40. 
    Crossref | PubMed
  70. Everett BM, Cornel JH, Lainscak M, et al. Anti-inflammatory therapy with canakinumab for the prevention of hospitalization for heart failure. Circulation 2019;139:1289–99. 
    Crossref | PubMed
  71. Riksen NP, Bekkering S, Mulder WJM, Netea MG. Trained immunity in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2023;20:799–811. 
    Crossref | PubMed
  72. Kovtonyuk LV, Caiado F, Garcia-Martin S, et al. IL-1 mediates microbiome-induced inflammaging of hematopoietic stem cells in mice. Blood 2022;139:44–58. 
    Crossref | PubMed
  73. Li X, Wang H, Yu X, et al. Maladaptive innate immune training of myelopoiesis links inflammatory comorbidities. Cell 2022;185:1709–1727.e18. 
    Crossref | PubMed