Open access peer-reviewed chapter

Myocardial Remodeling with Ventricular Assist Devices

Written By

Buyan-Ochir Orgil, Neely R. Alberson, Jeffrey A. Towbin and Enkhsaikhan Purevjav

Submitted: 10 August 2022 Reviewed: 07 March 2023 Published: 29 March 2023

DOI: 10.5772/intechopen.110814

From the Edited Volume

Ventricular Assist Devices - Advances and Applications in Heart Failure

Edited by Enkhsaikhan Purevjav, Hugo Martinez, Jeffrey A. Towbin and Umar Boston

Chapter metrics overview

75 Chapter Downloads

View Full Metrics

Abstract

Most prominent functional abnormalities seen in the failing human heart are impaired contraction and slowed rates of relaxation of cardiac cells in the face of increased neurohormonal activation, sustained inflammation, mechanical and volume overload, and progressive maladaptive remodeling of the myocardium. Mechanical circulatory support devices (MCS) improve cardiac function and outcomes of patients with end-stage heart failure, allowing to bridge to heart transplantation and permitting the removal of MCS device as a bridge to recovery, in some patients with the sufficient recovery of heart function. Numerous reports have demonstrated favorable myocardial recovery and reverse remodeling after prolonged ventricular unloading by MCS. Ventricular unloading by MCS leads to a decreased concentration of peripheral natriuretic peptides in plasma, reduction in cardiac cytokines, kinases, collagens, and proteins involved in hypertrophy, fibrosis, programmed cell death, and necrosis in the heart. This chapter will summarize and review the effects and underlying mechanisms of myocardial remodeling during prolonged MCS in patients with end-stage heart failure. The mechanisms of myocardial recovery are multifactorial and remain to be further explored on cellular, organ, and systems levels.

Keywords

  • ventricular assist device
  • mechanical unloading
  • reverse remodeling
  • myocardial recovery
  • mechanical circulatory support devices (MCS)

1. Introduction

Cardiovascular diseases (CVDs) were responsible for an estimated 17.8 million deaths globally in 2017 and half of all people diagnosed with heart failure (HF) die within 5 years of diagnosis [1]. The major cause for CVD morbidity and mortality is HF, a complex clinical syndrome caused by many CV and other diseases that impairs the ability of the ventricle to fill with or eject blood. The key pathophysiological features involved in the development of HF are hypertrophy, fibrosis, apoptosis/necrosis, microvasculature and extracellular matrix (ECM) abnormalities, and disturbances in electrophysiologic, adrenergic, and angiotensin signaling. Currently, heart transplantation is the gold standard treatment of patients with end-stage HF and the current 10-year survival rates of heart transplant recipients reach 53% [2]. During the last decades, mechanical circulatory support (MCS) devices with ventricular assist devices (VAD) have improved the outcomes of patients with advanced and end-stage HF, becoming a cornerstone therapy to bridge those patients to heart transplantation or recovery [3, 4, 5, 6, 7]. The synopsis of structural and molecular changes in the heart underlying the improved cardiac function after VAD implantation is called “reverse cardiac remodeling.” Extensive investigations have been utilized to understand how the heart remodels to mechanical and volume unloading during MCS in a facet of stabilized neurohormonal and inflammatory responses [8, 9, 10]. MCS therapies lead to the improvement of HF symptoms with normalized cardiac size and shape with simultaneous biological remodeling on gene, molecular, cellular, and tissue levels [11, 12, 13]. Myocardial recovery is associated with improvements in structural, sarcomeric, sarcolemmal, and calcium handling-associated proteins expression and function [14, 15, 16]. Mechanical unloading has been shown to increase collagen cross-linking and myocardial stiffness [17], alter mitochondrial and metabolic processes [18], and promote repair and regeneration [19]. Moreover, studies focused on understanding the roles of biomarkers of neurohormonal activation, oxidative stress, and systemic inflammation pathways in patients with VAD support have identified a subset of vulnerable patients with risks of developing adverse events fostering the development of innovative applications of combined MSC and pharmacological agents [20, 21]. As a destination therapy MSC is critical in patients with the favorable restoration of cardiac function and this regenerative therapeutic strategy becomes a desirable alternative to heart transplantation [22]. Herein, we review and summarize research studies focused on understanding the roles of neurohormonal signaling, inflammation, signal transduction, cellular and subcellular remodeling, and transcriptional regulation in the failing human heart before and after MCS therapy.

Advertisement

2. Neurohormonal remodeling during LVAD therapy

HF is a highly complex clinical syndrome characterized by cardinal symptoms due to structural and/or functional abnormality of the heart, resulting in elevated intracardiac pressures and/or inadequate cardiac output [23]. The clinical symptoms of HF develop and progress through prolonged dyshomeostasis in the heart in response to various stressors, which include alterations of regulatory neurohormonal systems associated with the release of natriuretic peptides, proinflammatory cytokines as well as activations of the sympathetic nervous system (SNS), which in turn activates the renin-angiotensin-aldosterone system (RAAS) [8]. Some of these alterations appear to be reversible by VAD treatment in response to a decrease in cardiac pressure, volume overload, and ventricular wall tension and stretch [24]. These events lead to reduced cardiomyocyte hypertrophy, improved coronary perfusion, and decreased chronic ischemia in the heart [25]. Therefore, mechanical unloading of the failing heart by LVADs, coupled with neurohormonal and anti-inflammatory therapy, may further promote reverse remodeling and recovery of myocardial function.

2.1 Natriuretic peptides

The natriuretic peptide family consists of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP) [26]. Under normal conditions, ANP is produced in the atrium and BNP is synthesized primarily by the ventricles in response to cardiac mechanical stretch. Circulating in the plasma, ANP and BNP play compensatory diuretic roles by decreasing salt and water retention and inhibiting vasoconstrictor peptides. In contrast to ANP, levels of BNP are significantly elevated in plasma of HF patients in response to chronic volume overload and BNP concentration correlates with the status of ventricular dysfunction with high concentrations predicting poor long-term survival. Support with VAD in patients with end-stage HF reduces the myocardial wall stress and thereby may change BNP levels in the heart. Sodian et al. studied 21 patients with nonischemic cardiomyopathy on VAD support and demonstrated a significant decrease in BNP levels in plasma after initiation of MCS, reaching normal levels within the first week after VAD implantation [27]. Especially, an early decrease of BNP in plasma was indicative of cardiac function recovery during VAD support. The significant decrease in BNP serum concentration after VAD support coincides with a decrease in BNP messenger RNA (mRNA) and protein expression in the heart of patients with severe HF supported by VAD. They also showed a decrease in BNP production not only by cardiomyocytes, but also by endothelial cells, T cells, and macrophages infiltrating the heart [28].

ANP and BNP also exert local antihypertrophic, antifibrotic, and lusitropic effects in the heart via their interactions with guanylyl cyclase-A receptor (CG-AR) [29]. Comparative analysis of cardiac ANP and BNP mRNAs expression in patients with HF revealed normalization of ANP, BNP, and the NP-metabolizing NPR-C receptor after VAD support, while GC-AR mRNA expression levels remained intact, suggesting that reverse remodeling is associated with the local protective effects of ANP and BNP.

In chronic HF, expression of ANP and BNP serves as clinical markers of cardiac hypertrophy, decompensation, hypertension, and myocardial infarction. Acute coronary syndromes are linked with the expression of chromogranin A (CgA), CD56/NCAM (neural cell adhesion molecule), and endothelin-1 (ET-1) [30, 31]. Investigation of 33 paired myocardial and plasma samples demonstrated significantly increased ANP, BNP, and CgA in congestive HF (CHF) patients before LVAD support, and all of these indicators were significantly decreased by VAD support [32]. Concentrations of plasma ANP and BNP also depend on different types of devices and durations of MCS. The time courses of ANP and BNP concentration have been studied in patients supported by Thoratec (8 patients), TCI Heartmate (6 patients), Novacor (7 patients), and Lionheart (3 patients) by Milting et. al [33]. All patients supported with Novacor, and some patients supported by TCI Heartmate, showed a steady decrease in plasma BNP levels, reaching normal ranges at 30 to 50 days. In contrast, only few patients supported by Thoratec or Lionheart reached normal BNP plasma values during the entire duration of support, suggesting recognition of different time points in ANP and BNP decrease among various types of devices when weaning from MCS in patients without heart transplant is suggested.

In pediatric cohort, it has been demonstrated that BNP and N-terminal pro-BNP (NT-proBNP) were modified differently by MCS compared to adults, showing an increase up to 1 day after VAD implant with a subsequent decrease to the pre-VAD levels in one month. Another pediatric study found levels of BNP and NT-pro-BNP correlated with severity and unfavorable outcomes of acute decompensated HF and an incremental increase of those peptides within 48 hours of admission predicted the need for MCS [34]. Short-term VAD support in children with severe HF significantly decreased BNP levels in plasma from pre-VAD to post-VAD and reduced markers of apoptosis [35].

2.2 Renin angiotensin aldosterone system

Reduced blood supply causes renal hypoperfusion and stimulation of SNS and RAAS [36]. The key molecule that mediates RAAS activation is angiotensin II (Ang II), a potent vasoconstrictor. In early stages of HF, RAAS activation functions as a compensatory mechanism to increase cardiac output. However, with the HF progression, RAAS activation plays a detrimental role in myocardial ischemia, hypertrophy, and arrhythmia [37]. In end-stage HF, G-protein-coupled receptors (GPCRs) of RAAS, such as Ang receptors, AT1R and AT2R, are downregulated, while angiotensin-converting enzymes (ACE and ACE2), GPCR kinase (GRK), and β-arrestin are upregulated [38]. Following VAD support, a significant downregulation of Ang I, ACE2, GRK, and β-arrestin has been documented, while AT2R, JNK, and p38 were upregulated, indicating divergent and incomplete molecular reverse remodeling. Combined MCS with neurohormonal blockade drug therapy (NHBDT) improved survival and reduced adverse cardiac events in HF [39]. For example, ACE inhibition (ACEI) during VAD support was linked with decreased Ang II, cardiac collagen content, and myocardial stiffness [17, 40], demonstrating the pathophysiological benefits of combined therapy compared with VAD support alone. The ISHLT Mechanically Assisted Circulatory Support (IMACS) registry suggested positive effects for ACEI and angiotensin II receptor blockers (ARBs) therapy in adult HF patients with VAD implantation [41]. Among patients treated with an ACEI/ARB, significantly lower risk of cardiovascular death, gastrointestinal bleeding, and levels of creatinine has been demonstrated compared to those in patients treated with mineralocorticoid receptor antagonist (MRA).

Advertisement

3. Inflammation and cytokines

3.1 Tumor necrosis factor-alpha

Reports on inflammatory profiles in HF patients and LVAD recipients have recently been comprehensively summarized by Radley et al. [42]. Tumor necrosis factor-alpha (TNF-α) is a protein expressed in the myocardium that stimulates cardiac growth and cell death [43, 44]. High levels of TNF-α are found in patients with severe HF [45]. Expression of TNF-α mRNA and protein were both elevated in the heart and serum of VAD candidates with severe HF [46]. Moreover, interleukin 1 beta (IL-1β), IL-6, procaspase-9, and active caspase-9 were increased in the heart of those deteriorating patients who required VAD support. Torre-Amione et al. reported that prolonged MCS results in significant reductions in intracardiac TNF-α, with a greater reduction in myocardial TNF-α in VAD-treated patients with recovered cardiac function versus those who required cardiac transplantation [43].

3.2 Metallothionein

Metallothionein (MT) is a highly conserved cytokine-inducible protein whose role is the detoxification of heavy metals through the regulation of their metabolism [47]. High metal affinity to cadmium (Cd) of MT in renal tissue plays a major role in the kinetics and balance between CdMT and non-bound Cd, which is highly neurotoxic [48]. A study of heart transplant patients by Baba et al. demonstrated that MT expression correlated with IL-6 elevation in blood vessels and a decrease in plasma IL-2 [49]. Moreover, MT expression was associated with lower fractional shortening, increase in LV end-systolic diameter, and lower mean arterial pressure in the absence of rejection in transplant patients, implicating the role of MT in cellular stress response. Further immunohistochemical studies by the same group demonstrated a decrease of MT-positive cardiomyocytes and vessels in the subendocardial and subepicardial regions of the myocardium in 17 HF patients during prolonged VAD compared to pre-VAD state [50]. In addition, ventricular unloading leads to regression of cellular hypertrophy and a reversal of MT expression in the failing heart, suggesting the remodeling process with reduction of MT expression is due to diminished wall stress and improved blood supply. The authors also observed that MT reactivity was substantially lower in the hearts of patients supported longer than 88 days as compared to patients supported less than 88 days [50].

3.3 C-reactive protein and interleukins

C-reactive protein (CRP) is a protein that is produced in response to the release of pro-inflammatory cytokines when the body is in an inflammatory condition [51]. Patients with end-stage HF have almost 8-fold higher levels of circulating CRP (cCRP) in serum compared to normal references [52]. Batra et al. studied pre- and post-implant VAD patients and found that one-third of post-VAD patients have persistently high CRP levels. They concluded that high CRP levels are linked with high mortality risk and a higher possibility of having a stroke during VAD support. Longer VAD therapy (60 days after implantation) resulted in a 50% reduction of CRP levels compared to pre-VAD values, suggesting improved inflammatory status over time [53].

Interleukins (IL) are a group of small molecules and peptides secreted by a wide variety of body cells or cytokines that function in cellular signaling and communication. Serum levels of members of IL-1 family cytokines, IL-1β and IL-33, are highly elevated in HF and remained elevated after MCS [54]. Increased expression of IL-1β and correlated patterns of IL-1 receptors indicate enhanced IL-1β signaling in MCS patients, while expression of IL-33 correlates with CRP plasma levels in HF, but not in patients on MCS. Suppression of tumorigenicity 2 (ST2) is a receptor of IL-33 and coupling of IL-33 with its ST2 receptor (IL-33/ST2) triggers danger-associated cellular responses playing a pivotal role in tissue repair in many organs [55]. In the heart, IL-33 is expressed by activated cardiac fibroblasts and cardiomyocytes during cardiac stretch and then is released into the extracellular matrix (ECM), promoting cell survival by blocking pro-fibrotic intracellular signaling [56, 57]. A significant decrease in soluble ST2 (sST2) levels was observed in end-stage HF patients after VAD implantation, suggesting a lessening of fibrosis and inflammation [58]. Levels of other cytokines, including IL-6 and IL-8, were also linked to the severity of clinical course in end-stage HF patients and correlated with outcome after VAD implantation [59]. A significant correlation of those cytokines was also found with ET-1 and relaxin (RLX)-2, the vasoactive mediators involved in neurohormonal system responses in VAD-supported HF patients [60]. Elevated levels of galectin-3 (GAL-3) were associated with the severity of HF and dynamic changes in GAL-3 levels predicted post-VAD survival [61]. Although unloading with continuous-flow LVAD results in a decrease of GAL-3 levels early post-implant, GAL-3 levels become elevated after 6 months of VAD implantation [61, 62], suggesting that levels of GAL-3 may represent a higher risk of death in HF patients with long-term VAD support.

Advertisement

4. Myocardial remodeling during VAD support

The myocardium consists of cardiomyocytes, composing nearly 56% of the adult heart, fibroblasts (27%), endothelial cells (7%), smooth muscle cells (10%), and various immune cells that transiently reside in the ECM [63]. These cell types are important in preserving normal cardiac function and morphology. The cells interact with each other using reciprocally secreted auto and paracrine factors, secretion of which is regulated by numerous molecules-messengers involving integrins, ET-1, BMPs, PECAM-1, VE-cadherin, VEGF, and TGFβ [64, 65]. Engineered heart tissue (EHT), created in vitro by seeding decellularized porcine myocardial sections with primary cardiomyocytes and fibroblasts isolated from neonatal rat ventricular myocardium or with cardiomyocytes derived from human induced pluripotent stem cells (hiPSC), is a novel platform to study cardiac remodeling [66]. Characterization of EHTs demonstrated gradual normalization of stress-free tissue length after mechanical unloading and suggested that actomyosin contraction in cardiomyocytes and activity of fibroblasts may play crucial roles in reverse remodeling after mechanical unloading.

4.1 Cardiac fibroblasts and fibrosis

Cardiac fibrosis in the failing heart is a final product of a series of biomechanical, molecular, and cellular changes that causes an imbalanced increase in ECM production and decreased ECM degradation [67]. The resultant increase in ECM deposition is accompanied by inflammatory and fibrotic scar formation in the interstitial and perivascular areas of the myocardium, interfering with the normal array of cardiomyocytes along with the disturbing supply of oxygen and nutrients to the myocardium. Moreover, cardiac fibrosis triggers further pathological remodeling and functional decline of the heart [68]. According to Tseng et al., an increase in inflammation and fibrosis in the failing heart was associated with an increase in sST2 levels [58]. Synthesis and degradation of collagens I and III are highly regulated processes in human cardiac ECM. Collagen I is a major collagen component establishing the myocyte-collagen matrix, while collagen III contributes to elasticity, and changes in content may influence LV stiffness and size [69]. In HF, predominantly increased accumulation of collagens I and III in ECM results in cardiomyocyte injury, cardiac fibrosis, and the release of collagen-derived peptides into circulation [70]. Bruckner et al. recorded a significant decrease in intracardiac TNF-α, collagen I (by 66%), and collagen III (62%) in post-VAD myocardial samples of 18 patients compared to their pre-VAD levels [71]. They also found a decrease in cardiomyocyte size by 26% at post-VAD, demonstrating favorable reverse remodeling in cardiac hypertrophy.

Insulin-like growth factor I (IGF-1), released preferentially from cardiac fibroblasts, functions to negatively regulate atrophy and apoptosis, and stimulate cardiac repair by interacting with stromal cell-derived factor (SDF) [19]. SDF induces IGF-1 expression in cardiac myocytes in vitro. Patients with VAD support combined with β2-AR agonist clenbuterol have shown elevated IGF-1 mRNA at the time of VAD explantation relative to the time of LVAD implantation [72].

4.2 Extracellular matrix remodeling

Matrix metalloproteinases (MMPs) degrade the ECM, while tissue inhibitors of MMPs (TIMPs) prevent the ECM degradation during repair process of damaged tissues and cells. There are four variants of TIMPs that selectively inhibit different types of MMPs [73]. Typically, TIMP1 and MMP1 are increased in patients with deteriorating HF [74]. The increased ratio of MMP-1 to TIMP-1 in DCM has been shown to be almost normalized after LVAD, favoring decreased collagen degradation [17]. Felkin et al. found that high myocardial MMP1 and MMP8 expression is associated with high collagen content and increased IL-6 and IL-1β expression in HF patients requiring VAD support [75]. After VAD support, expression of MMP-2 mRNA and active MMP-2 protein has been shown to be significantly increased compared to pre-VAD (P < 0.01), which was associated with a reduction of collagen IV content in the cardiomyocyte basement membrane. Furthermore, this was associated with a decrease in the thickness of cardiomyocyte membrane as revealed by electron microscopy [76]. MCS support increases collagen cross-linking and the ratio of collagen I to III in the heart as a result of decreased tissue MMP-1-to-TIMP-1 ratio and increased myocardial Ang I and II levels that stimulate ECM synthesis [17]. Therapy with ACEI drugs decreased Ang II levels and myocardial collagen content, resulting in enhanced myocardial recovery during VAD support [40]. In elderly patients with end-stage HF, VAD therapy is associated with decreased collagen turnover and cross-linking and increased tissue Ang II, whereas combined VAD and ACEI therapy normalizes LV end-diastolic pressure-volume relationships [77].

4.3 Endothelial and vasculature remodeling

A gene ontology (GO) analysis implicated endothelial to mesenchymal transition (EndoMT) and vice versa (MEndoT) pathways in human end-stage HF based on dual expressed VE-Cadherin endothelial and FSP-1 mesenchymal markers [78]. Gene expression analysis of 19 paired pre-VAD and post-VAD human heart samples by Hall et al. revealed differential expression of neuropilin-1, FGF9, Sprouty1, SDF1, and endomucin, the genes involved in the regulation of vascular networks [79]. In addition, a significant downregulation of GATA-4 binding protein, a critical mediator of myocyte hypertrophy, was observed in these heart samples following mechanical unloading. Drakos et al. observed an increased density of endothelial cells by 33% and decreased microvascular lumen area (36%) in post-VAD vs pre-VAD myocardial samples of patients with chronic HF (n = 15). This was associated with the activation of endothelial cells evidenced by ultrastructural and immunohistochemical analysis [80]. In agreement with these findings, a significant increase in interstitial and total collagen content without structural changes in cardiomyocytes was suggestive of increased fibrosis accompanied by regression of cardiomyocyte hypertrophy.

4.4 Reversal of cardiac hypertrophy

The myocardium is typically subjected to three types of mechanical loading during every heartbeat, including cyclic stretch, static stretch, and shear stress, generated by blood flow and an increase in chamber volume and pressure. Cardiomyocytes are sensitive to mechanical stress, which is transduced to molecular transduction signaling by biomechanical sensors. Comparative analysis of cardiomyocyte size in pre- and post-VAD patients demonstrated a decrease of 26% (33.1 ± 1.32 to 24.4 ± 1.64 μm, P < 0.001) in all patients studied [71]. Long-term VAD support resulted in a 28% reduction in myocyte volume, 20% reduction in cell length, 20% reduction in cell width, and 32% reduction in cell length-to-thickness ratio [81]. Another study examined the effects of continuous-flow VAD on cardiomyocyte size and demonstrated that cardiomyocyte cross-sectional area decreased after VAD, but not beyond that of normal donor hearts [82]. Electron microscopy, cardiac glycogen content, and echocardiographic assessment also did not suggest myocardial atrophy in post-VAD patients. Consistent with these findings, no upregulation of pro-atrophic genes and proteins of the ubiquitin-proteasome system (UPS) and no t-tubule pathologies have been demonstrated.

Myostatin (also called gdf-8) is a potent inhibitor of skeletal muscle growth from the TGF-β family and is secreted by cardiac muscle and adipocytes in response to pathological stress, such as myocardial infarction or obesity [83]. Myostatin has been shown to mediate the regression of cellular hypertrophy after unloading with LVAD support [84]. The nuclear factor (NF)-κB superfamily of transcription factors carries out broad functions by regulating immune cell maturation, cell survival, and inflammation in many cell types [85]. In the heart, NF-κB is shown to be cardioprotective during acute injury, however, prolonged activation of NF-κB enhances the release of TNF-α, IL-1, and IL-6 cytokines, triggering chronic inflammation, hypertrophy, and cell death [86, 87]. After VAD support, the NF-κB DNA-binding activity decreases in failing human hearts and this process has been associated with a decrease in cardiomyocyte diameter [88].

Several kinases such as mitogen-activated protein kinase (MAPK or MEK), ERK (extracellularly regulated kinase), AKT (protein kinase B, PKB), GSK-3b (glycogen synthase kinase-3 beta), JNK (c-Jun NH2-terminal kinase) and p38 are involved in the development of cardiac hypertrophy via kinase-mediated signal transduction pathways [89]. After VAD support, significantly decreased activities of ERKs and AKT were seen in failing hearts, while the activity of GSK-3β was increased [90]. These changes were associated with a decrease in TUNEL-positivity and myocyte diameter. The disparity in the regulation of MAPK activity with a concomitant decrease in ERK and JNK1/2 activities and an increase in p38 activity after VAD support has been also reported [91].

Osteopontin (OPN) is a pleiotropic extracellular signal-regulated bone sialoprotein. Expression and activity of OPN are increased in myocardial tissues in accordance with the severity of HF [92]. Levels of OPN mRNA in heart biopsy specimens decreased significantly after VAD support, while OPN protein remained intact [93]. Moreover, VAD support induced a decrease of OPN levels in the plasma of some patients with VAD support, whereas OPN plasma levels were reduced significantly in all patients after a heart transplant.

4.5 Cardiomyocyte apoptosis

While MCS improves the survival of end-stage HF patients by reversing many biological processes activated during progression of HF, the reports on modulation of apoptotic cell death in response to VAD remain controversial. Prescimone et al. found a significant increase of Bax (pro-apoptotic), Bcl-2 (pro-apoptotic), and Hsp72 (antiapoptotic) molecules and a mild increase in cardiac caspase (Casp)-3 activity in post-VAD hearts compared to pre-VAD, suggesting involvement of mitochondria in apoptotic signaling [94]. The authors also found an increase in Casp-1 after VAD implant in HF patients and lack of apoptotic nuclei [95]. Conversely, Francis et al. found Bcl-2 being downregulated after VAD implant [96]. Another study found no significant differences in Bcl-2, while autophagy markers such as beclin-1, autophagy-related gene 5 (Atg5), and microtubule-associated protein-1 light chain-3 (LC3) were all significantly decreased in response to unloading [97]. Moreover, Bedi et al.observed a highly variable expression of Fas among patients who had undergone MSC therapy [98]. Fas, also called Apo-1 or CD95, is a membrane receptor recognizing Fas ligand (Fas-L) and Fas/Fas-L coupling initiates an apoptotic cell death through the activation of caspase cascade in the heart [99]. Although apoptotic DNA fragmentation was attenuated in the myocardium, expression of antiapoptotic Bcl-XL and FasExo6Del/Fas genes was dependent on the duration of MCS [100]. Overall, no significant differences in number of TUNEL-positive cells between pre- and post-VAD samples have been reported by several groups [96, 97, 101, 102].

Abnormal Ca2+ cycling in HF triggers activation of UPS with an increase of binding immunoglobulin protein (BiP), eukaryotic initiation factor (eIF2α), and X-box binding protein 1 (XBP1) [103]. MCS support significantly decreases the levels of BiP and XBP1 and phosphorylation of eIF2α [104]. Moreover, a decrease in apoptosis observed during short-term VAD support has been associated with a decrease in phosphorylation of SMAD2 (mothers against decapentaplegic homolog 2), however, a long-term VAD support increased apoptosis and fibrosis in the heart via enhanced SMAD2 signaling and increased phosphorylation of HDAC4 (histone deacetylase 4) [101].

4.6 Cardiomyocyte regeneration

Diploid cardiomyocytes that are abundant in animal heart have a substantial capacity for cardiac repair and regeneration [105]. In human failing heart, polyploidy of cardiomyocytes is often observed as a precondition of heart hypertrophy [106], suggesting that cardiomyocyte polyploidization in HF may be associated with regeneration [107]. A study by Wohlschlaeger et al. demonstrated a marked reduction in the size of cardiomyocyte nuclei and in ratios between number of nuclei and cardiac myocytes after implantation of VAD [108]. They also reported a significant decrease in DNA content and reduction of polyploid cardiomyocytes in 23 myocardial samples studied after VAD, suggesting a decline in protein synthesis. On the contrary, an increase in the number of diploid cardiomyocytes was seen by other groups in post-VAD samples [108]. The decrease in polyploidy and increase in diploidy in response to MCS suggested an abundance of diploid cardiomyocytes going through cell cycle progression with the completion of mitosis or increase in stem cells. Prolonged MCS unloading increased the number of cardiomyocytes positive for phosphorylated histone H3 and Aurora B and this was associated with a decrease in cardiomyocyte size and mitochondrial content [109].

4.7 Transcriptional changes during VAD therapy

Accumulating evidence shows that the changes in transcriptome and metabolome profiles associated with HF persist in the reverse-remodeled myocardium despite apparent normalization on organ and cellular levels [110]. To identify transcriptional adaptations in failing and VAD-supported hearts, a comprehensive transcription analysis was performed in 199 human myocardial samples from nonfailing, failing, and VAD-supported human hearts. Although over 3088 transcripts exhibited alterations in HF samples, the number of differentially expressed genes (DEGs) with greater than or equal to a 2-fold difference was insignificant between HF and post-VAD samples, suggesting that many HF-associated transcriptional changes may have a limited role in regulating cardiac structure and function [111]. Significant elevation in myocardial arginine/glycine amidinotransferase (AGAT) expression is observed in HF patients and myocardial AGAT is one of the DEGs that had a significant decrease during recovery [112]. In HF patients recovering after combination therapy, levels of AGAT mRNA decreased by 4.3-fold [P < 0.001] and 2.7-fold [P < 0.005] in VAD combined and VAD alone groups compared to donors, respectively, and AGAT levels returned to normal after recovery. These data highlighted the involvement of elevated local creatine synthesis specific to HF and its reversal during recovery. The genetic response of pediatric myocardium to MCS is distinct with approximately 40% of DEGs compared to adult hearts with VAD support, highlighting the importance of understanding features of reverse remodeling specific to pediatric myocardium to improve clinical strategies and LVAD management in children [113].

In long-term analysis of gene expression, data of patients studied for an average of 3.8 years post-explant revealed a significant association of integrin signaling and its downstream EPAC2 (exchange protein activated by cyclic-AMP2) during recovery of ventricular function by combined LVAD and clenbuterol therapy [20]. Downregulation of EPAC2 that regulates calcium involving cAMP pathway was associated with improvements in cardiac contractility and metabolism [114].

4.8 miRNAs in response to LVAD therapy

MicroRNAs (miRNAs) are small, endogenous noncoding RNAs that regulate posttranscriptional processes by repressing the translation of targeted protein-coding genes via binding to the 3’ UTRs of mRNAs [115]. Therefore, cardiac miRNAs and circulating miRNAs (c-miRNAs) are promising biomarkers for HF diagnosis and prognosis [116]. Comprehensive microarray profiling of miRNAs and mRNAs, comparing myocardial specimens from adults with end stage HF with VAD and nonfailing hearts, showed upregulation of 28 miRNAs with almost normalization of miRNA profiles by VAD treatment [117]. Cardiac miRNAs have also been compared in 13 HF children at pre-VAD and at the moment of heart transplant (post-VAD) by next-generation sequencing [118]. The investigators found hsa-miR-199b-5p, hsa-miR-19a-3p, and hsa-miR-1246 being differentially expressed at post-VAD compared to that at pre-VAD. The candidate targets of those differentially expressed miRNAs were sarcomeric troponins showing significantly higher post-VAD when compared with pre-VAD values, suggesting that miRNAs can be therapeutically targeted to improve heart function in pediatric HF. Levels of nine c-miRNAs were downregulated and four c-miRNAs were upregulated in the post-VAD samples vs pre-VAD levels [119]. In particular, the c-miR-409-3p has been shown to regulate coagulation factor 7 (F7) and F2, suggesting a role of c-miRNA-409-3p in thrombotic events during MCS.

4.9 Beta-adrenergic receptor remodeling

Myocardial beta-adrenergic receptor (β-AR) signaling is severely diminished in failing heart due to increase in phosphorylation of agonist-occupied β-ARs by GRK2 [120, 121]. In chronic HF, VAD support leads to the restoration of cardiac β-AR signaling via the reduction of myocardial GRK2 expression and activity [122]. Unloading with VAD normalizes the ability of cardiac muscle to respond to SNS stimulation, reversing the downregulation of β-ARs [123]. Both types of devices, continuous-flow and pulsatile, decreased the expression and activity of GRK2 and normalized neurohormonal homeostasis disturbed with HF [124]. In pediatric HF, VAD treatment also resulted in the recovery of total β-AR and β1-AR expressions and reversal of several pathologic processes in the heart [125].

4.10 Cyclic guanosine monophosphate

Cyclic guanosine monophosphate (cGMP) is a cyclic nucleotide derived from GTP (guanosine triphosphate) that acts as a second messenger for activation of intracellular protein kinases in response to the binding of membrane-impermeable hormones to the cell membrane [126]. The important components of cGMP signaling include cGMP-dependent protein kinase G (PKG), ANP, BNP, natriuretic peptide receptor A and C (NPR-A and NPR-C, respectively), neprilysin, NOS3, soluble guanylyl cyclase (sGC), and PDE5 [127]. The cGMP-PKG cascade can decrease the level of calcium and alter the expression of glycoprotein IIb/IIIa. Both fluctuations impact the aggregation of platelets within the body [128]. cGMP levels were found to be higher in patients with implanted VAD compared to healthy individuals. According to Grosman-Rimon et al., cGMP was associated with an elevated risk of gastrointestinal (GI) bleeding during LVAD support [128]. The researchers presume that the association between elevated GI bleeding and higher cGMP levels could be due to platelet abnormalities. The study also found significant alterations of the cGMP-PKG pathway (downregulation of ANP, NPR-C, and cGMP) in patients with dilated cardiomyopathy after VAD implant, while the duration of VAD support negatively correlated with expression differences of PKG I, PDE5, and sGC in patients with ischemic cardiomyopathy.

Advertisement

5. Cardiomyocyte intracellular remodeling

5.1 Remodeling of cytoskeletal and sarcomeric proteins

Cytoskeletal proteins are essential for the structure and function of the cardiac myocyte. Stetson et al. reported ventricular unloading in humans dynamically changes not only myocardial TNF-α, total collagen, and myocyte size, but also remodels the expression of structural proteins [129]. To understand if myocardial recovery was associated with changes in sarcomeric, nonsarcomeric, and membrane-associated proteins, microarray analysis has been performed on the paired HF samples before and after VAD [16]. Significant increase of lamin A/C, spectrin and integrins (α5 and β5), and decrease of integrins β1, β6, and α7 has been observed at VAD explantation compared to pre-LVAD. Expression of sarcomeric proteins such as β-actin, α-tropomyosin, actinin-α1, and filamin A increased, while troponin T3 and actinin-α2 decreased. Vinculin expression decreased 4.1-fold in the recovered group. Despite decreased cardiomyocyte size post-VAD, severe structural damage in cardiomyocytes persisted with partial improvement in the expression of actin, tropomyosin, troponin C, troponin T, and titin [130]. In pediatric HF, MCS increased the expression of structural proteins, including dystrophin and actin [35]. Furthermore, expression of genes involved in calcium homeostasis, cell differentiation, and growth, including CNNA1, CDK2B, CSF2, E2F1, EGR1, and EGR2, were normalized after VAD therapy, suggesting an active reverse remodeling process after MCS in pediatric HF.

5.2 Dystrophin remodeling

Dystrophin is a rod-shaped protein encoded by the DMD gene located on the X chromosome, the largest gene of 2.4 megabases (Mb) in the human genome [131]. Dystrophin connects the actin and cytoskeleton of muscle fibers to the myocyte membrane at its N-terminus. At the C-terminus, it connects the sarcolemmal complex known as the dystrophin-associated protein complex (DAPC) to the ECM, providing structural support for myocytes. Mutations in DMD cause Duchenne and Becker muscular dystrophies [132, 133]. Mutations in genes encoding cytoskeletal and sarcolemmal proteins provide the genetic basis for dilation and contractile dysfunction via “final common pathway.” Abnormalities in DMD such as mutations in the N-terminus of dystrophin or in the cardiac-specific promoter, preferentially affecting cardiac function are associated with X-linked cardiomyopathy [134]. Vatta et al. investigated the integrity and response of dystrophin in end-stage dilated or ischemic cardiomyopathy HF patients to VAD therapy and identified disruption of N-terminal dystrophin in 18 HF patients studied [135]. This disruption was shown to be reversible in four patients after VAD support.

5.3 Remodeling in calcium cycling

Regulation of Ca2+ cycling is a versatile signaling process that regulates cellular homeostasis in different cell types, including cardiac myocytes [136]. Reduced rates of relaxation and impaired contractile reserve are the major abnormalities seen in the failing heart as a result of disturbances in Ca2+ transients [137]. The proteins that regulate cardiomyocyte Ca2+ cycling include sarcoplasmic reticulum (SR) Ca2+ ATPase (SERCA), ryanodine receptor 2 (RyR2), phospholamban (PLB), and the sarcolemmal Na+/Ca2+ exchanger (NCX) [138, 139, 140]. Chaudhary et al. demonstrated that improvement in cardiac function during LVAD support was associated with a favorable balance between SERCA and NCX, resulting from the isolated decrease in NCX without an increase in SERCA [141]. Reverse remodeling of SERCA2a expression has been shown to be completed by about 20 days of VAD support, while hearts supported by VAD for longer than 40 days have significantly increased relative collagen content [142]. Post-VAD recovery increased SR calcium content and shortened action potential duration due to rapid inactivation in L-type Ca2+ current [15]. Short-term VAD support recovered post-rest potentiation (PRP) response to a level close to that in nonfailing hearts, but recovery of impaired SR Ca2+ cycling was dependent on duration MCS [143]. Chronic unloading with recovery of contractile function demonstrated upregulation of SERCA2a, RyR2, and NCX genes after MCS [144]. Recovery of rate-dependent contractility in failing human hearts during early VAD support was associated with faster decay of Ca2+ transients, while long-term MCS triggered abnormal Ca2+ cycling [101, 143]. Moreover, long-term MCS resulted in significantly increased SMAD2 activity with downstream phosphorylation of Ca2+/calmodulin-dependent protein kinase type-IIδ (CaMKIIδ), myocyte enhancer factor 2 (MEF2), and myostatin. Improvements in the Ca2+ handling also depended on the severity of myocardial fibrosis, and ECM pathologies and excessive fibrosis limited the ability to recover [13].

5.4 Mitochondria and metabolism remodeling

Unloading with VAD has been shown to contribute to reverse remodeling of mitochondria and recovery of energy metabolism of the failing heart. In healthy adult hearts, the generation of ATP as a source of energy relies on the oxidation of fatty acids, glucose and lactate in mitochondria, and fatty acid oxidation provides the majority (> 70%) of total ATP [145]. The balance between lactate production and consumption by lactate dehydrogenase (LDH) that converts it to pyruvate, which is then transported by mitochondrial pyruvate carrier (MPC) into the mitochondrial tricarboxylic acid (TCA) cycle is important in producing plentiful ATP. The MPC expression is lower in patients with HF compared to those of non-failing cohorts [146]. Thus, the failing heart runs increased glycolysis and decreased fatty acid oxidation for ATP production and the proportion of glucose oxidation to fatty acid oxidation depends on the severity of HF [147]. The generation of ATP is disturbed in HF with an increased glycolytic pyruvate-derived lactate and a simultaneous decrease in lactate utilization [148]. In addition, the opening of mitochondrial permeability transition pore (mPTP) in HF disrupts the mitochondrial membrane potential and disturbs oxidative phosphorylation pathways for ATP production, causing mitochondrial swelling and inducing apoptotic and necrotic cell death.

MCS improves systemic and cardiac metabolism via improvements in fatty acid oxidation, insulin resistance, and reductions in myocardial lipotoxicity through improved activation of the insulin/PI3K/AKT signaling cascade [149]. Significant decrease in long-chain acylcarnitines levels was consistent with improved fatty acid oxidation and utilization during long-term VAD support [150]. Diakos et al. reported induction of glycolysis through TCA without a subsequent increase in pyruvate oxidation in post-VAD patients [148], which may be attributed to the poor post-VAD recovery of mitochondrial oxidative capacity. Recently, the same group reported the beneficial cardioprotective effects of induced glycolysis as a result of an increase in rate-limiting enzymes of the pentose-phosphate pathway and 1-carbon metabolism in post-VAD patients [151]. All these have been associated with significantly reduced reactive oxygen species (ROS) and improved mitochondrial density [151, 152]. These metabolic improvements enhanced the glycosylation of α-dystroglycan, which maintains integrity between cytoskeleton and ECM [18]. Moreover, using high-resolution respirometry, a reduction in mitochondrial ROS up to 40% [153] and increased MPC1 abundance and glucose and glucose-6-phosphate levels, particularly, in mechanically unloaded hearts of ischemic HF patients has been demonstrated [154].

Levels of Ca2+ in the mitochondrial matrix regulate the activity of kinases and phosphatases involved in ATP production and mitochondrial quality control [155, 156]. In HF, the opening of mPTP not only disrupts the mitochondrial membrane potential but also reduces Ca2+ uptake, alters pH, and induces inflammation, leading to necrosis and death of cardiac myocytes [157]. Impaired mitochondrial Ca2+ uptake is the result of reduced Ca2+ release from SR and stimulates Ca2+ −sensitive dehydrogenases of the Krebs cycle [158].

About 20% of the total lipid composition on the mitochondrial inner membrane is constituted by cardiolipin and loss of cardiolipin and tetralinoleoyl-cardiolipin in HF is linked to excessive ROS production and cardiomyocyte apoptosis [159]. During mechanical unloading with LVAD, cardiolipin arrangement normalizes, which in turn, improves mitochondrial coupling [160]. Cellular proteases, such as cathepsins, are involved in the progression of HF. Parallel activation of cathepsins and their inhibitors was observed after VAD support. The expression of cathepsins and their inhibitors was significantly higher in pre-VAD compared to the heart transplant group and VAD induced a further increase in the cathepsin system. Significant positive correlations were observed between cardiac expression of cathepsins and their inhibitors as well as inflammatory cytokines [59, 161].

5.5 Cardiomyocyte signal transduction pathways and signaling

5.5.1 Mitogen-activated protein kinases

There are several cell signal-transduction pathways regulated in the heart in direct response to changes in mechanical loading and stress. The family of MAPKs, such as ERKs, p38, and JNK1/2, are well-characterized signal-transduction pathways [162]. These kinases are involved in the regulation of cell growth, cardiac hypertrophy, and cell death [163, 164]. They are upregulated in patients with HF secondary to ischemic heart disease and cardiomyopathy [165, 166]. The ERKs activity regulates adaptive hypertrophy and prevention of cell death during the early phase of chronic pressure overload in response to stimulation of GPCRs and integrin activation [167]. Mechanical unloading with VAD support resulted in differential regulation of MAPK activity with a significant decrease in the activity of p44/42 ERK and JNK1/2 along with a subsequent increase in p38 activity after LVAD support [91]. The authors explained a decrease in ERK activity is likely due to its decreased phosphorylation at p44/42, while a combination of decreased phosphorylation and expression of JNK1/2 is responsible for decreased JNK1/2 activity in VAD-supported hearts. Activation of AKT regulates cardiac physiological hypertrophy, glucose metabolism, cell death, and angiogenesis [168]. In failing human hearts, a high grade of kinase phosphorylation in all 3 MAPKs and AKT have been observed [166]. After VAD support, ERKs and AKT activities were dramatically decreased in failing hearts, while GSK-3β activities were increased [89].

Advertisement

6. Conclusions

Neurohormonal imbalance, inflammation, apoptosis, and abnormal inter and intracellular signaling and remodeling on molecular and genetic levels are critical processes contributing to adverse events in HF patients. This chapter provides a comprehensive overview of reverse remodeling on neurohormonal, myocardial, and cardiomyocyte intracellular levels in response to MCS in patients with HF. Knowledge about molecular mechanisms of underlying effects of VAD support aid to understand the adverse effects of myocardial unloading and deterioration of patients undergoing VAD therapy. While cardiac reverse remodeling has been correlated with clinical recovery in most post-VAD patients, many of those patients have deteriorated back to the original HF phenotype after LVAD explantation, suggesting the importance of considering a higher degree of myocardial recovery that may persist after device removal [169]. We found a significantly lower number of pediatric reports on clinical and pathological features of reverse remodeling compared to adult HF patients undergoing VAD support. Hence, there is a need for research on pediatric patients with VAD support, so we can better understand features of reverse remodeling specific to pediatric myocardium.

Advertisement

Conflict of interest

None.

References

  1. 1. Group, W.C.R.C.W. World Health Organization cardiovascular disease risk charts: Revised models to estimate risk in 21 global regions. The Lancet Global Health. 2019;7(10):e1332-e1345
  2. 2. Dolgner SJ et al. Long-term adult congenital heart disease survival after heart transplantation: A restricted mean survival time analysis. The Journal of Heart and Lung Transplantation. 2021;40(7):698-706
  3. 3. Miller LW et al. Use of a continuous-flow device in patients awaiting heart transplantation. The New England Journal of Medicine. 2007;357(9):885-896
  4. 4. Slaughter MS et al. Advanced heart failure treated with continuous-flow left ventricular assist device. The New England Journal of Medicine. 2009;361(23):2241-2251
  5. 5. Rose EA et al. Long-term use of a left ventricular assist device for end-stage heart failure. The New England Journal of Medicine. 2001;345(20):1435-1443
  6. 6. Dandel M et al. Long-term results in patients with idiopathic dilated cardiomyopathy after weaning from left ventricular assist devices. Circulation. 2005;112(9 Suppl):I37-I45
  7. 7. Frazier OH, Myers TJ. Left ventricular assist system as a bridge to myocardial recovery. The Annals of Thoracic Surgery. 1999;68(2):734-741
  8. 8. Liem DA et al. Molecular- and organelle-based predictive paradigm underlying recovery by left ventricular assist device support. Circulation. Heart Failure. 2014;7(2):359-366
  9. 9. Drakos SG et al. Left ventricular assist device unloading effects on myocardial structure and function: Current status of the field and call for action. Current Opinion in Cardiology. 2011;26(3):245-255
  10. 10. Klotz S, Jan Danser AH, Burkhoff D. Impact of left ventricular assist device (LVAD) support on the cardiac reverse remodeling process. Progress in Biophysics and Molecular Biology. 2008;97(2-3):479-496
  11. 11. Mann DL, Barger PM, Burkhoff D. Myocardial recovery and the failing heart: Myth, magic, or molecular target? Journal of the American College of Cardiology. 2012;60(24):2465-2472
  12. 12. Ambardekar AV, Buttrick PM. Reverse remodeling with left ventricular assist devices: A review of clinical, cellular, and molecular effects. Circulation. Heart Failure. 2011;4(2):224-233
  13. 13. Birks EJ. Molecular changes after left ventricular assist device support for heart failure. Circulation Research. 2013;113(6):777-791
  14. 14. Hall JL et al. Clinical, molecular, and genomic changes in response to a left ventricular assist device. Journal of the American College of Cardiology. 2011;57(6):641-652
  15. 15. Terracciano CM et al. Clinical recovery from end-stage heart failure using left-ventricular assist device and pharmacological therapy correlates with increased sarcoplasmic reticulum calcium content but not with regression of cellular hypertrophy. Circulation. 2004;109(19):2263-2265
  16. 16. Birks EJ et al. Gene profiling changes in cytoskeletal proteins during clinical recovery after left ventricular-assist device support. Circulation. 2005;112(9 Suppl):I57-I64
  17. 17. Klotz S et al. Mechanical unloading during left ventricular assist device support increases left ventricular collagen cross-linking and myocardial stiffness. Circulation. 2005;112(3):364-374
  18. 18. Jiang M et al. Mitochondrial metabolism in myocardial Remodeling and mechanical unloading: Implications for ischemic heart disease. Frontiers in Cardiovascular Medicine. 2021;8:789267
  19. 19. Bhavsar PK et al. Clenbuterol induces cardiac myocyte hypertrophy via paracrine signalling and fibroblast-derived IGF-1. Journal of Cardiovascular Translational Research. 2010;3(6):688-695
  20. 20. Hall JL et al. Molecular signature of recovery following combination left ventricular assist device (LVAD) support and pharmacologic therapy. European Heart Journal. 2007;28(5):613-627
  21. 21. Burkhoff D et al. Reverse Remodeling with left ventricular assist devices. Circulation Research. 2021;128(10):1594-1612
  22. 22. Miyagawa S et al. Building a bridge to recovery: The pathophysiology of LVAD-induced reverse modeling in heart failure. Surgery Today. 2016;46(2):149-154
  23. 23. McDonagh TA et al. 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. European Heart Journal. 2021;42(36):3599-3726
  24. 24. Sciaccaluga C et al. Biomarkers in patients with left ventricular assist device: An insight on current evidence. Biomolecules. 2022;12(2):334
  25. 25. Wohlschlaeger J et al. Reverse remodeling following insertion of left ventricular assist devices (LVAD): A review of the morphological and molecular changes. Cardiovascular Research. 2005;68(3):376-386
  26. 26. Abassi Z et al. Implications of the natriuretic peptide system in the pathogenesis of heart failure: Diagnostic and therapeutic importance. Pharmacology & Therapeutics. 2004;102(3):223-241
  27. 27. Sodian R et al. Decreased plasma concentration of brain natriuretic peptide as a potential indicator of cardiac recovery in patients supported by mechanical circulatory assist systems. Journal of the American College of Cardiology. 2001;38(7):1942-1949
  28. 28. Bruggink AH et al. Brain natriuretic peptide is produced both by cardiomyocytes and cells infiltrating the heart in patients with severe heart failure supported by a left ventricular assist device. The Journal of Heart and Lung Transplantation. 2006;25(2):174-180
  29. 29. Kuhn M et al. Left ventricular assist device support reverses altered cardiac expression and function of natriuretic peptides and receptors in end-stage heart failure. Cardiovascular Research. 2004;64(2):308-314
  30. 30. Angelone T, Mazza R, Cerra MC. Chromogranin-a: A multifaceted cardiovascular role in health and disease. Current Medicinal Chemistry. 2012;19(24):4042-4050
  31. 31. Pieroni M et al. Myocardial production of chromogranin a in human heart: A new regulatory peptide of cardiac function. European Heart Journal. 2007;28(9):1117-1127
  32. 32. Wohlschlaeger J et al. Decreased myocardial chromogranin a expression and colocalization with brain natriuretic peptide during reverse cardiac remodeling after ventricular unloading. The Journal of Heart and Lung Transplantation. 2008;27(4):442-449
  33. 33. Milting H et al. The time course of natriuretic hormones as plasma markers of myocardial recovery in heart transplant candidates during ventricular assist device support reveals differences among device types. The Journal of Heart and Lung Transplantation. 2001;20(9):949-955
  34. 34. Wong DT et al. Effectiveness of serial increases in amino-terminal pro-B-type natriuretic peptide levels to indicate the need for mechanical circulatory support in children with acute decompensated heart failure. The American Journal of Cardiology. 2011;107(4):573-578
  35. 35. Mohapatra B et al. Short-term mechanical unloading and reverse remodeling of failing hearts in children. The Journal of Heart and Lung Transplantation. 2010;29(1):98-104
  36. 36. Schmieder RE et al. Renin-angiotensin system and cardiovascular risk. Lancet. 2007;369(9568):1208-1219
  37. 37. Chaggar PS et al. Neuroendocrine effects on the heart and targets for therapeutic manipulation in heart failure. Cardiovascular Therapeutics. 2009;27(3):187-193
  38. 38. Messmann R et al. Alterations of the renin angiotensin system in human end-stage heart failure before and after mechanical cardiac unloading by LVAD support. Molecular and Cellular Biochemistry. 2020;472(1-2):79-94
  39. 39. Grupper A et al. Effect of Neurohormonal blockade drug therapy on outcomes and left ventricular function and structure after left ventricular assist device implantation. The American Journal of Cardiology. 2016;117(11):1765-1770
  40. 40. Klotz S et al. The impact of angiotensin-converting enzyme inhibitor therapy on the extracellular collagen matrix during left ventricular assist device support in patients with end-stage heart failure. Journal of the American College of Cardiology. 2007;49(11):1166-1174
  41. 41. Brinkley DM Jr et al. Impact of renin-angiotensin-aldosterone system inhibition on morbidity and mortality during long-term continuous-flow left ventricular assist device support: An IMACS report. The Journal of Heart and Lung Transplantation. 2021;40(12):1605-1613
  42. 42. Radley G et al. The inflammatory response to ventricular assist devices. Frontiers in Immunology. 2018;9:2651
  43. 43. Torre-Amione G et al. Decreased expression of tumor necrosis factor-alpha in failing human myocardium after mechanical circulatory support : A potential mechanism for cardiac recovery. Circulation. 1999;100(11):1189-1193
  44. 44. Bryant D et al. Cardiac failure in transgenic mice with myocardial expression of tumor necrosis factor-alpha. Circulation. 1998;97(14):1375-1381
  45. 45. Torre-Amione G et al. Proinflammatory cytokine levels in patients with depressed left ventricular ejection fraction: A report from the studies of left ventricular dysfunction (SOLVD). Journal of the American College of Cardiology. 1996;27(5):1201-1206
  46. 46. Birks EJ et al. Quantitative myocardial cytokine expression and activation of the apoptotic pathway in patients who require left ventricular assist devices. Circulation. 2001;104(12 Suppl 1):I233-I240
  47. 47. Kagi JH, Schaffer A. Biochemistry of metallothionein. Biochemistry. 1988;27(23):8509-8515
  48. 48. Jin T, Lu J, Nordberg M. Toxicokinetics and biochemistry of cadmium with special emphasis on the role of metallothionein. Neurotoxicology. 1998;19(4-5):529-535
  49. 49. Baba HA et al. Metallothionein: Localization in human transplant endomyocardium, relation to cytokines and allograft function. The Journal of Heart and Lung Transplantation. 1999;18(10):963-971
  50. 50. Baba HA et al. Reversal of metallothionein expression is different throughout the human myocardium after prolonged left-ventricular mechanical support. The Journal of Heart and Lung Transplantation. 2000;19(7):668-674
  51. 51. Batra J et al. C-reactive protein levels predict outcomes in continuous-flow left ventricular assist device patients: An INTERMACS analysis. ASAIO Journal. 2021;67(8):884-890
  52. 52. Syeda T et al. Pre- and post-operative values of serum CRP in patients undergoing surgery for brain tumour. The Journal of the Pakistan Medical Association. 2014;64(3):271-274
  53. 53. Ahmad T et al. Effects of left ventricular assist device support on biomarkers of cardiovascular stress, fibrosis, fluid homeostasis, inflammation, and renal injury. JACC Heart Fail. 2015;3(1):30-39
  54. 54. Niazy N et al. Altered mRNA expression of Interleukin-1 receptors in myocardial tissue of patients with left ventricular assist device support. Journal of Clinical Medicine. 2021;10(21):4856
  55. 55. Kotsiou OS, Gourgoulianis KI, Zarogiannis SG. IL-33/ST2 Axis in organ fibrosis. Frontiers in Immunology. 2018;9:2432
  56. 56. Millar NL et al. Wounds that heal and wounds that don't - the role of the IL-33/ST2 pathway in tissue repair and tumorigenesis. Seminars in Cell & Developmental Biology. 2017;61:41-50
  57. 57. Tseng CCS et al. The Interleukin-33/ST2 pathway is expressed in the failing human heart and associated with pro-fibrotic Remodeling of the myocardium. Journal of Cardiovascular Translational Research. 2018;11(1):15-21
  58. 58. Tseng CCS et al. Soluble ST2 in end-stage heart failure, before and after support with a left ventricular assist device. European Journal of Clinical Investigation. 2018;48(3):e12886
  59. 59. Caselli C et al. IL-33/ST2 pathway and classical cytokines in end-stage heart failure patients submitted to left ventricular assist device support: A paradoxic role for inflammatory mediators? Mediators of Inflammation. 2013;2013:498703
  60. 60. Cabiati M et al. Transcriptional evaluation of relaxin and endothelin-1 axis in heart failure patients: First evidence of its involvement during left ventricular assist device support. International Journal of Cardiology. 2020;306:109-115
  61. 61. Coromilas E et al. Dynamics and prognostic role of galectin-3 in patients with advanced heart failure, during left ventricular assist device support and following heart transplantation. BMC Cardiovascular Disorders. 2016;16:138
  62. 62. Lok SI et al. Myocardial fibrosis and pro-fibrotic markers in end-stage heart failure patients during continuous-flow left ventricular assist device support. European Journal of Cardio-Thoracic Surgery. 2015;48(3):407-415
  63. 63. Banerjee I et al. Determination of cell types and numbers during cardiac development in the neonatal and adult rat and mouse. American Journal of Physiology. Heart and Circulatory Physiology. 2007;293(3):H1883-H1891
  64. 64. Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nature Reviews. Molecular Cell Biology. 2006;7(8):589-600
  65. 65. Harvey PA, Leinwand LA. The cell biology of disease: Cellular mechanisms of cardiomyopathy. The Journal of Cell Biology. 2011;194(3):355-365
  66. 66. Shen S, Sewanan LR, Campbell SG. Evidence for synergy between sarcomeres and fibroblasts in an in vitro model of myocardial reverse remodeling. Journal of Molecular and Cellular Cardiology. 2021;158:11-25
  67. 67. Ma ZG et al. Cardiac fibrosis: New insights into the pathogenesis. International Journal of Biological Sciences. 2018;14(12):1645-1657
  68. 68. Nielsen SH et al. Understanding cardiac extracellular matrix remodeling to develop biomarkers of myocardial infarction outcomes. Matrix Biology. 2019;75-76:43-57
  69. 69. Bishop JE et al. Enhanced deposition of predominantly type I collagen in myocardial disease. Journal of Molecular and Cellular Cardiology. 1990;22(10):1157-1165
  70. 70. Nikolov A, Popovski N. Extracellular matrix in heart disease: Focus on circulating collagen type I and III derived peptides as biomarkers of myocardial fibrosis and their potential in the prognosis of heart failure: A concise review. Metabolites. 2022;12(4):297
  71. 71. Bruckner BA et al. Regression of fibrosis and hypertrophy in failing myocardium following mechanical circulatory support. The Journal of Heart and Lung Transplantation. 2001;20(4):457-464
  72. 72. Barton PJ et al. Myocardial insulin-like growth factor-I gene expression during recovery from heart failure after combined left ventricular assist device and clenbuterol therapy. Circulation. 2005;112(9 Suppl):I46-I50
  73. 73. Brew K, Nagase H. The tissue inhibitors of metalloproteinases (TIMPs): An ancient family with structural and functional diversity. Biochimica et Biophysica Acta. 2010;1803(1):55-71
  74. 74. Barton PJ et al. Increased expression of extracellular matrix regulators TIMP1 and MMP1 in deteriorating heart failure. The Journal of Heart and Lung Transplantation. 2003;22(7):738-744
  75. 75. Felkin LE et al. A quantitative gene expression profile of matrix metalloproteinases (MMPS) and their inhibitors (TIMPS) in the myocardium of patients with deteriorating heart failure requiring left ventricular assist device support. The Journal of Heart and Lung Transplantation. 2006;25(12):1413-1419
  76. 76. Bruggink AH et al. Type IV collagen degradation in the myocardial basement membrane after unloading of the failing heart by a left ventricular assist device. Laboratory Investigation. 2007;87(11):1125-1137
  77. 77. Butler CR, Jugdutt BI. The paradox of left ventricular assist device unloading and myocardial recovery in end-stage dilated cardiomyopathy: Implications for heart failure in the elderly. Heart Failure Reviews. 2012;17(4-5):615-633
  78. 78. Wang G et al. Role of endothelial and mesenchymal cell transitions in heart failure and recovery thereafter. Frontiers in Genetics. 2020;11:609262
  79. 79. Hall JL et al. Genomic profiling of the human heart before and after mechanical support with a ventricular assist device reveals alterations in vascular signaling networks. Physiological Genomics. 2004;17(3):283-291
  80. 80. Drakos SG et al. Impact of mechanical unloading on microvasculature and associated central remodeling features of the failing human heart. Journal of the American College of Cardiology. 2010;56(5):382-391
  81. 81. Zafeiridis A et al. Regression of cellular hypertrophy after left ventricular assist device support. Circulation. 1998;98(7):656-662
  82. 82. Diakos NA et al. Myocardial atrophy and chronic mechanical unloading of the failing human heart: Implications for cardiac assist device-induced myocardial recovery. Journal of the American College of Cardiology. 2014;64(15):1602-1612
  83. 83. Breitbart A et al. Myostatin from the heart: Local and systemic actions in cardiac failure and muscle wasting. American Journal of Physiology. Heart and Circulatory Physiology. 2011;300(6):H1973-H1982
  84. 84. George I et al. Myostatin activation in patients with advanced heart failure and after mechanical unloading. European Journal of Heart Failure. 2010;12(5):444-453
  85. 85. Gordon JW, Shaw JA, Kirshenbaum LA. Multiple facets of NF-kappaB in the heart: To be or not to NF-kappaB. Circulation Research. 2011;108(9):1122-1132
  86. 86. Zelarayan L et al. NF-kappaB activation is required for adaptive cardiac hypertrophy. Cardiovascular Research. 2009;84(3):416-424
  87. 87. Hamid T et al. Divergent tumor necrosis factor receptor-related remodeling responses in heart failure: Role of nuclear factor-kappaB and inflammatory activation. Circulation. 2009;119(10):1386-1397
  88. 88. Grabellus F et al. Reversible activation of nuclear factor-kappaB in human end-stage heart failure after left ventricular mechanical support. Cardiovascular Research. 2002;53(1):124-130
  89. 89. Baba HA et al. Dynamic regulation of MEK/Erks and Akt/GSK-3beta in human end-stage heart failure after left ventricular mechanical support: Myocardial mechanotransduction-sensitivity as a possible molecular mechanism. Cardiovascular Research. 2003;59(2):390-399
  90. 90. Razeghi P et al. Mechanical unloading of the failing human heart fails to activate the protein kinase B/Akt/glycogen synthase kinase-3beta survival pathway. Cardiology. 2003;100(1):17-22
  91. 91. Flesch M et al. Differential regulation of mitogen-activated protein kinases in the failing human heart in response to mechanical unloading. Circulation. 2001;104(19):2273-2276
  92. 92. Stawowy P et al. Increased myocardial expression of osteopontin in patients with advanced heart failure. European Journal of Heart Failure. 2002;4(2):139-146
  93. 93. Schipper ME et al. Osteopontin: A potential biomarker for heart failure and reverse remodeling after left ventricular assist device support. The Journal of Heart and Lung Transplantation. 2011;30(7):805-810
  94. 94. Prescimone T et al. Cardiac molecular markers of programmed cell death are activated in end-stage heart failure patients supported by left ventricular assist device. Cardiovascular Pathology. 2014;23(5):272-282
  95. 95. Prescimone T et al. Caspase-1 transcripts in failing human heart after mechanical unloading. Cardiovascular Pathology. 2015;24(1):11-18
  96. 96. Francis GS et al. Apoptosis, Bcl-2, and proliferating cell nuclear antigen in the failing human heart: Observations made after implantation of left ventricular assist device. Journal of Cardiac Failure. 1999;5(4):308-315
  97. 97. Kassiotis C et al. Markers of autophagy are downregulated in failing human heart after mechanical unloading. Circulation. 2009;120(11 Suppl):S191-S197
  98. 98. Bedi MS et al. Myocardial Fas and cytokine expression in end-stage heart failure: Impact of LVAD support. Clinical and Translational Science. 2008;1(3):245-248
  99. 99. Purevjav E et al. Myocardial Fas ligand expression increases susceptibility to AZT-induced cardiomyopathy. Cardiovascular Toxicology. 2007;7(4):255-263
  100. 100. Bartling B et al. Myocardial gene expression of regulators of myocyte apoptosis and myocyte calcium homeostasis during hemodynamic unloading by ventricular assist devices in patients with end-stage heart failure. Circulation. 1999;100(19 Suppl):216-223
  101. 101. Castillero E et al. Structural and functional cardiac profile after prolonged duration of mechanical unloading: Potential implications for myocardial recovery. American Journal of Physiology. Heart and Circulatory Physiology. 2018;315(5):H1463-H1476
  102. 102. de Jonge N et al. Cardiomyocyte death in patients with end-stage heart failure before and after support with a left ventricular assist device: Low incidence of apoptosis despite ubiquitous mediators. The Journal of Heart and Lung Transplantation. 2003;22(9):1028-1036
  103. 103. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nature Reviews. Molecular Cell Biology. 2007;8(7):519-529
  104. 104. Castillero E et al. Attenuation of the unfolded protein response and endoplasmic reticulum stress after mechanical unloading in dilated cardiomyopathy. American Journal of Physiology. Heart and Circulatory Physiology. 2015;309(3):H459-H470
  105. 105. Yuan X, Braun T. Multimodal regulation of cardiac myocyte proliferation. Circulation Research. 2017;121(3):293-309
  106. 106. Sandritter W, Adler CP. Polyploidization of heart muscle nuclei as a prerequisite for heart growth and numerical hyperplasia in heart hypertrophy. Recent Advances in Studies on Cardiac Structure and Metabolism. 1976;12:115-127
  107. 107. Gonzalez-Rosa JM et al. Myocardial Polyploidization creates a barrier to heart regeneration in zebrafish. Developmental Cell. 2018;44(4):433-446 e7
  108. 108. Wohlschlaeger J et al. Hemodynamic support by left ventricular assist devices reduces cardiomyocyte DNA content in the failing human heart. Circulation. 2010;121(8):989-996
  109. 109. Canseco DC et al. Human ventricular unloading induces cardiomyocyte proliferation. Journal of the American College of Cardiology. 2015;65(9):892-900
  110. 110. Kim GH, Uriel N, Burkhoff D. Reverse remodelling and myocardial recovery in heart failure. Nature Reviews. Cardiology. 2018;15(2):83-96
  111. 111. Margulies KB et al. Mixed messages: Transcription patterns in failing and recovering human myocardium. Circulation Research. 2005;96(5):592-599
  112. 112. Cullen ME et al. Myocardial expression of the arginine:Glycine amidinotransferase gene is elevated in heart failure and normalized after recovery: Potential implications for local creatine synthesis. Circulation. 2006;114(1 Suppl):I16-I20
  113. 113. Weia BC, Adachi I, Jacot JG. Clinical and molecular comparison of Pediatric and adult reverse Remodeling with ventricular assist devices. Artificial Organs. 2015;39(8):691-700
  114. 114. Sugawara K et al. Structure and functional roles of Epac2 (Rapgef4). Gene. 2016;575(2 Pt 3):577-583
  115. 115. Saunders MA, Liang H, Li WH. Human polymorphism at microRNAs and microRNA target sites. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(9):3300-3305
  116. 116. Peterlin A et al. The role of microRNAs in heart failure: A systematic review. Frontiers in Cardiovascular Medicine. 2020;7:161
  117. 117. Matkovich SJ et al. Reciprocal regulation of myocardial microRNAs and messenger RNA in human cardiomyopathy and reversal of the microRNA signature by biomechanical support. Circulation. 2009;119(9):1263-1271
  118. 118. Ragusa R et al. Epigenetic regulation of cardiac troponin genes in Pediatric patients with heart failure supported by ventricular assist device. Biomedicine. 2021;9(10):1409
  119. 119. Ragusa R et al. Variations of circulating miRNA in paediatric patients with heart failure supported with ventricular assist device: A pilot study. Scientific Reports. 2020;10(1):5905
  120. 120. Bristow MR et al. Decreased catecholamine sensitivity and beta-adrenergic-receptor density in failing human hearts. The New England Journal of Medicine. 1982;307(4):205-211
  121. 121. Ungerer M et al. Altered expression of beta-adrenergic receptor kinase and beta 1-adrenergic receptors in the failing human heart. Circulation. 1993;87(2):454-463
  122. 122. Pandalai PK et al. Restoration of myocardial beta-adrenergic receptor signaling after left ventricular assist device support. The Journal of Thoracic and Cardiovascular Surgery. 2006;131(5):975-980
  123. 123. Ogletree-Hughes ML et al. Mechanical unloading restores beta-adrenergic responsiveness and reverses receptor downregulation in the failing human heart. Circulation. 2001;104(8):881-886
  124. 124. Akhter SA et al. Reversal of impaired myocardial beta-adrenergic receptor signaling by continuous-flow left ventricular assist device support. The Journal of Heart and Lung Transplantation. 2010;29(6):603-609
  125. 125. Medina E et al. Molecular changes in children with heart failure undergoing left ventricular assist device therapy. The Journal of Pediatrics. 2017;182:184-189 e1
  126. 126. Francis SH, Corbin JD. Cyclic nucleotide-dependent protein kinases: Intracellular receptors for cAMP and cGMP action. Critical Reviews in Clinical Laboratory Sciences. 1999;36(4):275-328
  127. 127. Persoon S et al. Cardiac unloading by LVAD support differentially influences components of the cGMP-PKG signaling pathway in ischemic and dilated cardiomyopathy. Heart and Vessels. 2018;33(8):948-957
  128. 128. Grosman-Rimon L et al. Increased cyclic guanosine monophosphate levels and continuous-flow left-ventricular assist devices: Implications for gastrointestinal bleeding. The Journal of Thoracic and Cardiovascular Surgery. 2016;151(1):219-227
  129. 129. Stetson SJ et al. Improved myocardial structure following LVAD support: Effect of unloading on dystrophin expression. The Journal of Heart and Lung Transplantation. 2001;20(2):240
  130. 130. de Jonge N et al. Left ventricular assist device in end-stage heart failure: Persistence of structural myocyte damage after unloading. An immunohistochemical analysis of the contractile myofilaments. Journal of the American College of Cardiology. 2002;39(6):963-969
  131. 131. Tennyson CN, Klamut HJ, Worton RG. The human dystrophin gene requires 16 hours to be transcribed and is cotranscriptionally spliced. Nature Genetics. 1995;9(2):184-190
  132. 132. Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: The protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51(6):919-928
  133. 133. Le Rumeur E. Dystrophin and the two related genetic diseases, Duchenne and Becker muscular dystrophies. Bosnian Journal of Basic Medical Sciences. 2015;15(3):14-20
  134. 134. Towbin JA et al. X-linked dilated cardiomyopathy. Molecular genetic evidence of linkage to the Duchenne muscular dystrophy (dystrophin) gene at the Xp21 locus. Circulation. 1993;87(6):1854-1865
  135. 135. Vatta M et al. Molecular remodelling of dystrophin in patients with end-stage cardiomyopathies and reversal in patients on assistance-device therapy. Lancet. 2002;359(9310):936-941
  136. 136. Berridge MJ. Calcium signalling remodelling and disease. Biochemical Society Transactions. 2012;40(2):297-309
  137. 137. Smith GL, Eisner DA. Calcium buffering in the heart in health and disease. Circulation. 2019;139(20):2358-2371
  138. 138. Mishra S et al. Molecular mechanisms of reduced sarcoplasmic reticulum Ca(2+) uptake in human failing left ventricular myocardium. The Journal of Heart and Lung Transplantation. 2002;21(3):366-373
  139. 139. Schillinger W et al. Influence of SR Ca(2+)-ATPase and Na(+)-Ca(2+)-exchanger on the force-frequency relation. Basic Research in Cardiology. 1998;93(Suppl. 1):38-45
  140. 140. Pieske B et al. Ca2+ handling and sarcoplasmic reticulum Ca2+ content in isolated failing and nonfailing human myocardium. Circulation Research. 1999;85(1):38-46
  141. 141. Chaudhary KW et al. Altered myocardial Ca2+ cycling after left ventricular assist device support in the failing human heart. Journal of the American College of Cardiology. 2004;44(4):837-845
  142. 142. Madigan JD et al. Time course of reverse remodeling of the left ventricle during support with a left ventricular assist device. The Journal of Thoracic and Cardiovascular Surgery. 2001;121(5):902-908
  143. 143. Ogletree ML et al. Duration of left ventricular assist device support: Effects on abnormal calcium cycling and functional recovery in the failing human heart. The Journal of Heart and Lung Transplantation. 2010;29(5):554-561
  144. 144. Heerdt PM et al. Chronic unloading by left ventricular assist device reverses contractile dysfunction and alters gene expression in end-stage heart failure. Circulation. 2000;102(22):2713-2719
  145. 145. Huss JM, Kelly DP. Mitochondrial energy metabolism in heart failure: A question of balance. The Journal of Clinical Investigation. 2005;115(3):547-555
  146. 146. Cluntun AA et al. The pyruvate-lactate axis modulates cardiac hypertrophy and heart failure. Cell Metabolism. 2021;33(3):629-648 e10
  147. 147. Jaswal JS et al. Targeting fatty acid and carbohydrate oxidation--a novel therapeutic intervention in the ischemic and failing heart. Biochimica et Biophysica Acta. 2011;1813(7):1333-1350
  148. 148. Diakos NA et al. Evidence of glycolysis up-regulation and pyruvate mitochondrial oxidation mismatch during mechanical unloading of the failing human heart: Implications for cardiac reloading and conditioning. JACC Basic to Translational Science. 2016;1(6):432-444
  149. 149. Chokshi A et al. Ventricular assist device implantation corrects myocardial lipotoxicity, reverses insulin resistance, and normalizes cardiac metabolism in patients with advanced heart failure. Circulation. 2012;125(23):2844-2853
  150. 150. Ahmad T et al. Prognostic implications of long-chain Acylcarnitines in heart failure and reversibility with mechanical circulatory support. Journal of the American College of Cardiology. 2016;67(3):291-299
  151. 151. Badolia R et al. The role of nonglycolytic glucose metabolism in myocardial recovery upon mechanical unloading and circulatory support in chronic heart failure. Circulation. 2020;142(3):259-274
  152. 152. Lee SH et al. Improvement of myocardial mitochondrial function after hemodynamic support with left ventricular assist devices in patients with heart failure. The Journal of Thoracic and Cardiovascular Surgery. 1998;116(2):344-349
  153. 153. Scheiber D et al. Reduced myocardial mitochondrial ROS production in mechanically unloaded hearts. Journal of Cardiovascular Translational Research. 2019;12(2):107-115
  154. 154. Shahinian JH et al. Impact of left ventricular assist device therapy on the cardiac proteome and metabolome composition in ischemic cardiomyopathy. Artificial Organs. 2020;44(3):257-267
  155. 155. Boyman L, Karbowski M, Lederer WJ. Regulation of mitochondrial ATP production: Ca(2+) Signaling and quality control. Trends in Molecular Medicine. 2020;26(1):21-39
  156. 156. McCormack JG, Halestrap AP, Denton RM. Role of calcium ions in regulation of mammalian intramitochondrial metabolism. Physiological Reviews. 1990;70(2):391-425
  157. 157. Ramachandra CJA et al. Mitochondria in acute myocardial infarction and cardioprotection. eBioMedicine. 2020;57:102884
  158. 158. Kohlhaas M, Maack C. Adverse bioenergetic consequences of Na+-Ca2+ exchanger-mediated Ca2+ influx in cardiac myocytes. Circulation. 2010;122(22):2273-2280
  159. 159. Dolinsky VW et al. Cardiac mitochondrial energy metabolism in heart failure: Role of cardiolipin and sirtuins. Biochimica et Biophysica Acta. 2016;1861(10):1544-1554
  160. 160. Heerdt PM et al. Disease-specific remodeling of cardiac mitochondria after a left ventricular assist device. The Annals of Thoracic Surgery. 2002;73(4):1216-1221
  161. 161. D'Amico A et al. Uncovering the cathepsin system in heart failure patients submitted to left ventricular assist device (LVAD) implantation. Journal of Translational Medicine. 2014;12:350
  162. 162. Rose BA, Force T, Wang Y. Mitogen-activated protein kinase signaling in the heart: Angels versus demons in a heart-breaking tale. Physiological Reviews. 2010;90(4):1507-1546
  163. 163. Communal C, Colucci WS, Singh K. p38 mitogen-activated protein kinase pathway protects adult rat ventricular myocytes against beta -adrenergic receptor-stimulated apoptosis. Evidence for Gi-dependent activation. The Journal of Biological Chemistry. 2000;275(25):19395-19400
  164. 164. Wang Y et al. Cardiac muscle cell hypertrophy and apoptosis induced by distinct members of the p38 mitogen-activated protein kinase family. The Journal of Biological Chemistry. 1998;273(4):2161-2168
  165. 165. Cook SA, Sugden PH, Clerk A. Activation of c-Jun N-terminal kinases and p38-mitogen-activated protein kinases in human heart failure secondary to ischaemic heart disease. Journal of Molecular and Cellular Cardiology. 1999;31(8):1429-1434
  166. 166. Haq S et al. Differential activation of signal transduction pathways in human hearts with hypertrophy versus advanced heart failure. Circulation. 2001;103(5):670-677
  167. 167. Gallo S et al. ERK: A key player in the pathophysiology of cardiac hypertrophy. International Journal of Molecular Sciences. 2019;20(9):2164
  168. 168. Abeyrathna P, Su Y. The critical role of Akt in cardiovascular function. Vascular Pharmacology. 2015;74:38-48
  169. 169. Marinescu KK et al. Left ventricular assist device-induced reverse remodeling: it's not just about myocardial recovery. Expert Review of Medical Devices. 2017;14(1):15-26

Written By

Buyan-Ochir Orgil, Neely R. Alberson, Jeffrey A. Towbin and Enkhsaikhan Purevjav

Submitted: 10 August 2022 Reviewed: 07 March 2023 Published: 29 March 2023