bims-hafaim Biomed News
on Heart Failure Metabolism
Issue of 2021‒05‒02
twelve papers selected by
Kyle McCommis
Saint Louis University


  1. Am J Physiol Heart Circ Physiol. 2021 Apr 30.
      Heart failure presents as the leading cause of infant mortality in individuals with Barth syndrome (BTHS), a rare genetic disorder due to mutations in the tafazzin (TAZ) gene affecting mitochondrial structure and function. Investigations into the perturbed bioenergetics in the BTHS heart remain limited. Hence, our objective was to identify the potential alterations in myocardial energy metabolism and molecular underpinnings that may contribute to the early cardiomyopathy and heart failure development in BTHS. Cardiac function and myocardial energy metabolism were assessed via ultrasound echocardiography and isolated working heart perfusions, respectively, in a mouse model of BTHS (doxycycline-inducible Taz knockdown (TazKD) mice). In addition, we also performed mRNA/protein expression profiling for key regulators of energy metabolism in hearts from TazKD mice and their wild-type (WT) littermates. TazKD mice developed hypertrophic cardiomyopathy as evidenced by increased left ventricular anterior and posterior wall thickness, as well as increased cardiac myocyte cross sectional area, though no functional impairments were observed. Glucose oxidation rates were markedly reduced in isolated working hearts from TazKD mice compared to their WT littermates in the presence of insulin, which was associated with decreased pyruvate dehydrogenase activity. Conversely, myocardial fatty acid oxidation rates were elevated in TazKD mice, whereas no differences in glycolytic flux or ketone body oxidation rates were observed. Our findings demonstrate that myocardial glucose oxidation is impaired prior to the development of overt cardiac dysfunction in TazKD mice, and may thus represent a pharmacological target for mitigating the development of cardiomyopathy in BTHS.
    Keywords:  Barth Syndrome; cardiomyopathy; fatty acid oxidation; glucose oxidation; pyruvate dehydrogenase
    DOI:  https://doi.org/10.1152/ajpheart.00873.2020
  2. Cardiovasc Toxicol. 2021 Apr 30.
      Fatty acid-binding protein 5 (FABP5) is an important member of the FABP family and plays a vital role in the metabolism of fatty acids. However, few studies have examined the role of FABP5 in pathological cardiac remodeling and heart failure. The aim of this study was to explore the role of FABP5 in transverse aortic constriction (TAC)-induced pathological cardiac remodeling and dysfunction in mice. Quantitative RT-PCR (qRT-PCR) and western blotting (WB) analysis showed that the levels of FABP5 mRNA and protein, respectively, were upregulated in hearts of the TAC model. Ten weeks after TAC in FABP5 knockout and wild type control mice, echocardiography, histopathology, qRT-PCR, and WB demonstrated that FABP5 deficiency aggravated cardiac injury (both cardiac hypertrophy and fibrosis) and dysfunction. In addition, transmission electron microscopy, ATP detection, and WB revealed that TAC caused severe impairment to mitochondria in the hearts of FABP5-deficient mice compared with that in control mice. When FABP5 was downregulated by siRNA in primary mouse cardiac fibroblasts, FABP5 silencing increased oxidative stress, reduced mitochondrial respiration, and increased the expression of myofibroblast activation marker genes in response to treatment with transforming growth factor-β. Our findings demonstrate that FABP5 deficiency aggravates cardiac pathological remodeling and dysfunction by damaging cardiac mitochondrial function.
    Keywords:  Cardiac dysfunction; Cardiac fibroblasts; FABP5; Pathological cardiac remodeling
    DOI:  https://doi.org/10.1007/s12012-021-09653-2
  3. Circ Res. 2021 Apr 30.
      Rationale: Diabetic cardiomyopathy is accompanied by increased production of NADH, predominantly through oxidation of fatty acids and consequent increases in oxidative stress. The role of nicotinamide phosphoribosyltransferase (Nampt), the rate-limiting enzyme of the salvage pathway of NAD+ synthesis, in the development of diabetic cardiomyopathy is poorly understood. Objective: We investigated the role of endogenous and exogenous Nampt during the development of diabetic cardiomyopathy in response to high fat diet (HFD) consumption and in the context of oxidative stress. Methods and Results: HFD consumption upregulated endogenous Nampt, and HFD-induced cardiac diastolic dysfunction, fibrosis, apoptosis and pro-inflammatory signaling were alleviated in transgenic mice with cardiac-specific overexpression of Nampt. The alleviation of diastolic dysfunction observed in these mice was abolished by inhibition of NADP(H) production via NAD kinase (NADK) inhibition. Nampt overexpression decreased the GSSG/GSH ratio, oxidation of thioredoxin 1 (Trx1) targets, dityrosine, and the accumulation of toxic lipids, including ceramides and diglycerides, in the presence of HFD consumption. Nampt overexpression upregulated not only NAD+ but also NADP+ and NADPH in the heart and in cultured cardiomyocytes, which in turn stimulated the GSH and Trx1 systems and alleviated oxidative stress in the heart induced by HFD consumption. In cultured cardiomyocytes, Nampt-induced upregulation of NADPH was abolished in the presence of NADK knockdown, whereas that of NAD+ was not. Nampt overexpression attenuated H2O2-induced oxidative inhibition of Prdx1 and mTOR in an NADK-dependent manner in cultured cardiomyocytes. Nampt overexpression also attenuated H2O2-induced cell death, an effect that was partly abolished by inhibition of NADK, Trx1 or GSH synthesis. In contrast, oxidative stress and the development of diabetic cardiomyopathy in response to HFD consumption were exacerbated in Nampt+/- mice. Conclusions:Nampt-mediated production of NAD+ protects against oxidative stress in part through the NADPH-dependent reducing system, thereby alleviating the development of diabetic cardiomyopathy in response to HFD consumption.
    Keywords:  NAD; NAD kinase; NADPH; Nampt; thioredoxin
    DOI:  https://doi.org/10.1161/CIRCRESAHA.120.317943
  4. Int J Mol Sci. 2021 Apr 23. pii: 4427. [Epub ahead of print]22(9):
      ATPase inhibitory factor-1 (IF1) preserves cellular ATP under conditions of respiratory collapse, yet the function of IF1 under normal respiring conditions is unresolved. We tested the hypothesis that IF1 promotes mitochondrial dysfunction and pathological cardiomyocyte hypertrophy in the context of heart failure (HF). Methods and results: Cardiac expression of IF1 was increased in mice and in humans with HF, downstream of neurohumoral signaling pathways and in patterns that resembled the fetal-like gene program. Adenoviral expression of wild-type IF1 in primary cardiomyocytes resulted in pathological hypertrophy and metabolic remodeling as evidenced by enhanced mitochondrial oxidative stress, reduced mitochondrial respiratory capacity, and the augmentation of extramitochondrial glycolysis. Similar perturbations were observed with an IF1 mutant incapable of binding to ATP synthase (E55A mutation), an indication that these effects occurred independent of binding to ATP synthase. Instead, IF1 promoted mitochondrial fragmentation and compromised mitochondrial Ca2+ handling, which resulted in sarcoplasmic reticulum Ca2+ overloading. The effects of IF1 on Ca2+ handling were associated with the cytosolic activation of calcium-calmodulin kinase II (CaMKII) and inhibition of CaMKII or co-expression of catalytically dead CaMKIIδC was sufficient to prevent IF1 induced pathological hypertrophy. Conclusions: IF1 represents a novel member of the fetal-like gene program that contributes to mitochondrial dysfunction and pathological cardiac remodeling in HF. Furthermore, we present evidence for a novel, ATP-synthase-independent, role for IF1 in mitochondrial Ca2+ handling and mitochondrial-to-nuclear crosstalk involving CaMKII.
    Keywords:  CaMKII; calcium handling; cardiomyocyte hypertrophy; heart failure; mitochondria
    DOI:  https://doi.org/10.3390/ijms22094427
  5. Redox Biol. 2021 Mar 31. pii: S2213-2317(21)00108-7. [Epub ahead of print]43 101960
      Brief episodes of ischaemia and reperfusion render the heart resistant to subsequent prolonged ischaemic insult, termed ischaemic preconditioning. Here, we hypothesized that transient non-ischaemic stress by hypertrophic stimulation would induce endogenous cardioprotective signalling and enhance cardiac resistance to subsequent ischaemic damage. Transient transverse aortic constriction (TAC) or Ang-Ⅱ treatment was performed for 3-7 days in male mice and then withdrawn for several days by either aortic debanding or discontinuing Ang-Ⅱ treatment, followed by subsequent exposure to regional myocardial ischaemia by in situ coronary artery ligation. Following ischaemia/reperfusion (I/R) injury, myocardial infarct size and apoptosis were markedly reduced and contractile function was significantly improved in the TAC preconditioning group compared with that in the control group. Similar results were observed in mice receiving Ang-Ⅱ infusion. Mechanistically, TAC preconditioning enhanced ALDH2 activity, promoted AMPK activation and improved mitochondrial energy metabolism by increasing myocardial OXPHOS complex expression, elevating the mitochondrial ATP content and improving viable myocardium glucose uptake. Moreover, TAC preconditioning significantly mitigated I/R-induced myocardial iNOS/gp91phox activation, inhibited endoplasmic reticulum stress and ameliorated mitochondrial impairment. Using a pharmacological approach to inhibit AMPK signalling in the presence or absence of preconditioning, we demonstrated AMPK-dependent protective mechanisms of TAC preconditioning against I/R injury. Furthermore, treatment with adenovirus-encoded ALDH2 partially emulated the actions of hypertrophic preconditioning, as evidenced by improved mitochondrial metabolism, inhibited oxidative stress-induced mitochondrial damage and attenuated cell death through an AMPK-dependent mechanism, whereas genetic ablation of ALDH2 abrogated the aforementioned actions of TAC preconditioning. The present study demonstrates that preconditioning with hypertrophic stress protects the heart from I/R injury via mechanisms that improve mitochondrial metabolism, reduce oxidative/nitrative stress and inhibit apoptosis. ALDH2 is obligatorily required for the development of cardiac hypertrophic preconditioning and acts as the mediator of this process.
    Keywords:  ALDH2; Apoptosis; Ischaemia/reperfusion injury; Oxidative/nitrative stress; Preconditioning
    DOI:  https://doi.org/10.1016/j.redox.2021.101960
  6. J Mol Cell Cardiol. 2021 Apr 21. pii: S0022-2828(21)00083-3. [Epub ahead of print]157 31-44
      Essentially all biological processes fluctuate over the course of the day, manifesting as time-of-day-dependent variations with regards to the way in which organ systems respond to normal behaviors. For example, basic, translational, and epidemiologic studies indicate that temporal partitioning of metabolic processes governs the fate of dietary nutrients, in a manner in which concentrating caloric intake towards the end of the day is detrimental to both cardiometabolic and cardiovascular parameters. Despite appreciation that branched chain amino acids impact risk for obesity, diabetes mellitus, and heart failure, it is currently unknown whether the time-of-day at which dietary BCAAs are consumed influence cardiometabolic/cardiovascular outcomes. Here, we report that feeding mice a BCAA-enriched meal at the end of the active period (i.e., last 4 h of the dark phase) rapidly increases cardiac protein synthesis and mass, as well as cardiomyocyte size; consumption of the same meal at the beginning of the active period (i.e., first 4 h of the dark phase) is without effect. This was associated with a greater BCAA-induced activation of mTOR signaling in the heart at the end of the active period; pharmacological inhibition of mTOR (through rapamycin) blocked BCAA-induced augmentation of cardiac mass and cardiomyocyte size. Moreover, genetic disruption of the cardiomyocyte circadian clock abolished time-of-day-dependent fluctuations in BCAA-responsiveness. Finally, we report that repetitive consumption of BCAA-enriched meals at the end of the active period accelerated adverse cardiac remodeling and contractile dysfunction in mice subjected to transverse aortic constriction. Thus, our data demonstrate that the timing of BCAA consumption has significant implications for cardiac health and disease.
    Keywords:  Chronobiology; Hypertrophy; Nutrition; Protein synthesis; Signaling
    DOI:  https://doi.org/10.1016/j.yjmcc.2021.04.005
  7. Circ Res. 2021 Apr 30.
      Rationale: Diabetic hyperglycemia is associated with cardiac dysfunction and increased arrhythmia risk, and calcium/calmodulin-dependent protein kinase II (CaMKII) function has been implicated. CaMKII activity is promoted by both oxidation and O linked β-N-acetylglucosamine (O GlcNAc) of known CaMKII sites. Objective: To investigate which post-translational modifications occur in human diabetic hearts and how they alter electrophysiological and Ca2+ handling properties in hyperglycemia. Methods and Results: We assessed echocardiography, electrophysiology, Ca2+-handling, and protein expression in site-specific CaMKII mutant mice (O GlcNAc-resistant S280A and oxidation-resistant MM281/2VV knock-ins, and global and cardiac-specific knockouts), in myocytes subjected to acute hyperglycemia and angiotensin II (Ang-II) and mice after streptozotocin injections (to induce diabetes). Human patients with diabetes exhibit elevated CaMKII O GlcNAcylation but not oxidation. In mice, acute hyperglycemia increased spontaneous diastolic Ca2+ sparks and waves and arrhythmogenic action potential changes (prolongation, alternans and delayed afterdepolarizations), all of which required CaMKII-S280 O GlcNAcylation. Ang-II effects were dependent on NADPH oxidase 2 (NOX2)-mediated CaMKII MM281/2 oxidation. Diabetes led to much greater Ca2+ leak, RyR2 S2814 phosphorylation, electrophysiological remodeling, and increased susceptibility to in vivo arrhythmias, requiring CaMKII activation, predominantly via S280 O GlcNAcylation and less via MM281/2 oxidation. These effects were present in myocytes at normal glucose, but were exacerbated with the in-vivo high circulating glucose. Phospholamban (PLB) O-GlcNAcylation was increased and coincided with reduced PLB S16 phosphorylation in diabetes. Dantrolene, that reverses CaMKII-dependent proarrhythmic RyR-mediated Ca2+ leak, also prevented hyperglycemia-induced APD prolongation and delayed afterdepolarizations. Conclusions: We found that CaMKII-S280 O GlcNAcylation is required for increased arrhythmia susceptibility in diabetic hyperglycemia, which can be worsened by an additional angiotensin II-NOX2-CaMKII MM281/2 oxidation pathway. CaMKII-dependent RyR2 S2814 phosphorylation markedly increases proarrhythmic Ca2+ leak and PLB O-GlcNAcylation may limit SR Ca2+ reuptake, leading to impaired excitation-contraction coupling and arrhythmogenesis in diabetic hyperglycemia.
    Keywords:  Post-translational modifications; diabetic cardiomyopathy; electrophysiology
    DOI:  https://doi.org/10.1161/CIRCRESAHA.120.318402
  8. BMC Cardiovasc Disord. 2021 Apr 29. 21(1): 217
      BACKGROUND: Diabetes mellitus-related cardiomyopathy (DMCMP), defined as left ventricular (LV) dysfunction caused by hyperglycemia in the absence of coronary artery disease, leads to heart failure (HF). Previous studies have shown that treatment with sodium-glucose co-transporter 2 inhibitor (SGLT2i) reduces the risk of exacerbation of HF. The beneficial effects of SGLT2i on HF depend not only on indirect actions such as osmotic diuresis but also on direct actions on the myocardium, leading to improvements in LV function. However, it remains unclear whether SGLT2i treatment is equally effective in any phase of DMCMP. The aim of this observational study was to compare the efficacy of SGLT2i treatment on LV dysfunction between early and advanced DMCMP.METHODS: Thirty-five symptomatic non-ischemic HF patients with LV ejection fraction > 40% and type 2 diabetes mellitus (T2DM) treated with empagliflozin (EMPA group) and 20 controls treated without SGLT2i were enrolled. According to the myocardial extracellular volume fraction (ECV), a reliable marker of cardiac fibrosis quantified by cardiac magnetic resonance, the EMPA group was further divided into early DMCMP (n = 16, ECV ≤ 30%) and advanced DMCMP (n = 19, ECV > 30%) groups and followed up prospectively. Echocardiography was performed at baseline and after 12 months. LV function assessed as LV global longitudinal strain (LVGLS) and the ratio of early diastolic mitral inflow velocity to early diastolic mitral annular velocity (E/e') were compared.
    RESULTS: ECV was strongly correlated with T2DM duration (r2 = 0.65, p < 0.001). At baseline, each group had a similar background. After 12 months, the EMPA group, especially the early DMCMP group, showed remarkable improvements in LVGLS (ΔLVGLS: 2.9 ± 3.0% (EMPA) vs. 0.6 ± 2.2% (controls), p = 0.005, and 4.6 ± 1.5% (early DMCMP) vs. 1.6 ± 3.3% (advanced DMCMP), p = 0.003) and E/e' (ΔE/e': - 1.5 ± 4.7 vs. - 0.3 ± 3.0, p = 0.253, and - 3.4 ± 5.5 vs. - 0.1 ± 3.5, p = 0.043).
    CONCLUSIONS: The positive effects of empagliflozin on LV dysfunction were more remarkable in early than in advanced DMCMP. Early intervention of SGLT2i for DMCMP may be preferable.
    Keywords:  Diabetes mellitus-related cardiomyopathy; Heart failure; Left ventricular dysfunction; Left ventricular global longitudinal strain; Sodium–glucose co-transporter 2 inhibitor
    DOI:  https://doi.org/10.1186/s12872-021-02024-3
  9. Eur J Pharmacol. 2021 Apr 24. pii: S0014-2999(21)00234-X. [Epub ahead of print]902 174081
      Myocardial mitochondrial function and biogenesis are suppressed in diabetes, but the mechanisms are unclear. Increasing evidence suggests that asymmetric dimethylarginine (ADMA) is associated with diabetic cardiovascular complications. This study was to determine whether endogenous ADMA accumulation contributes to cardiac and mitochondrial dysfunctions of diabetic rats and elucidate the potential mechanisms. Diabetic rat was induced by single intraperitoneal injection of streptozotocin (50 mg/kg). N-acetylcysteine was given (250 mg/kg/d) by gavage for 12w. Cardiac function was detected by echocardiography. Left ventricle papillary muscles were isolated to examine myocardial contractility. Myocardial ATP and mitochondrial DNA contents were measured to evaluate mitochondrial function and biogenesis. Endogenous ADMA accumulation was augmented resulting in decreased nitric oxide (NO) production and increased oxidative stress, suggesting NO synthase (NOS) uncoupling in the myocardium of T1DM rats compared with control rats. ADMA augmentation was associated with cardiac and mitochondrial dysfunctions along with myocardial uncoupling protein-2 (UCP2) upregulation and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) downregulation in T1DM rats. Exogenous ADMA could directly inhibit myocardial contractility, mitochondrial function and biogenesis in parallel with decreasing NO content and PGC-1α expression while increasing oxidative stress and UCP2 expression in papillary muscles and cardiomyocytes. Treatment with antioxidant N-acetylcysteine, also an inhibitor of NOS uncoupling, either ameliorated ADMA-associated cardiac and mitochondrial dysfunctions or reversed ADMA-induced NO reduction and oxidative stress enhance in vivo and in vitro. These results indicate that myocardial ADMA accumulation precipitates cardiac and mitochondrial dysfunctions in T1DM rats. The underlying mechanism may be related to NOS uncoupling, resulting in NO reduction and oxidative stress increment, ultimate PGC-1α down-regulation and UCP2 up-regulation.
    Keywords:  Asymmetric dimethylarginine; Cardiac dysfunction; Diabetic cardiomyopathy; Mitochondrial dysfunction; N-acetylcysteine; Nitric oxide synthase uncoupling
    DOI:  https://doi.org/10.1016/j.ejphar.2021.174081
  10. Prim Care Diabetes. 2021 Apr 26. pii: S1751-9918(21)00049-8. [Epub ahead of print]
      AIM/OBJECTIVE: Recently, the glucagon-like peptide-1 receptor agonists (GLP-1RA) class showed a significant reduction in heart failure (HF) hospitalization in several meta-analyses of cardiovascular outcome trials (CVOTs). The objective of this systematic review is to summarize the real-world evidence regarding HF outcomes of GLP-1RAs.METHODS: We searched the PubMed and EMBASE databases for observational studies that investigated HF outcomes of GLP-1RAs.
    RESULTS: Our search yielded 10 observational studies. Of those, 7 were cohort studies, and 3 were nested case-control studies. The risk of HF was the outcome in four cohort studies. One study that compared exenatide and exenatide combined with insulin to insulin showed a reduction in HF risk in the exenatide and exenatide plus insulin groups (HR 0.34, 95% CI 0.22-0.52, p-value <0.001 and HR 0.40, 95% CI 0.32-0.50, p-value <0.001, respectively). The other three cohort studies did not show a statistically significant result. In the three cohort studies that investigated HF hospitalization as an outcome, two showed a lower rate of HF hospitalization [48 (16.7%) vs. 76 (28%), p-value <0.05 and HR 0.51, 95% CI 0.34-0.77, p = 0.002] in the GLP-1RA groups. Conversely, the remaining study showed a reduction of 14% in HF hospitalization in the dipeptidyl peptidase-4 inhibitors (DPP-4i) group compared to the GLP-1RA group (HR 0.86, 95% CI 0.83-0.90). In contrast to the cohort studies, the three nested case-control studies showed similar results of no association of GLP-1RA use and HF hospitalization with OR 0.67 (95% CI 0.32-1.42), HR 0.95 (95% CI 0.83-1.10), and OR 0.84 (95% CI 0.48-1.47), respectively.
    CONCLUSION: The real-world evidence regarding the reduction in HF risk and hospitalization in GLP-1RA users is conflicting. Further well-designed, large multicenter, observational studies are needed to show clearer evidence.
    Keywords:  Cardiovascular disease; Diabetes mellitus; Glucagon-like peptide-1 receptor agonist; Heart failure; Hospitalizations for heart failure; Observational study; Systematic review; Type 2 diabetes mellitus
    DOI:  https://doi.org/10.1016/j.pcd.2021.04.005
  11. J Clin Med. 2021 Apr 08. pii: 1581. [Epub ahead of print]10(8):
      Lower myocardial mechanic-energetic efficiency (MEEi), expressed as stroke volume/heart rate ratio (SV/HR) in mL/s/g of the left ventricular (LV) mass, is associated with the incidence of heart failure in subjects with cardiometabolic disorders. We explored the association of MEEi with LV systolic circumferential and longitudinal myocardial function in 480 subjects with increased body mass index (BMI) without known cardiovascular disease (mean age 47 ± 9 years, 61% women, 63% obese, 74% with hypertension) participating in the fat-associated cardiovascular dysfunction (FATCOR) study. Insulin resistance was assessed by the homeostasis model assessment insulin-resistance index (HOMA-IR). SV was calculated by Doppler echocardiography. The LV systolic circumferential myocardial function was evaluated by midwall fractional shortening (MFS) and longitudinal function by global longitudinal strain (GLS). Patients were grouped into MEEi quartiles. The lowest MEEi quartile (<0.41 mL/s per g) was considered low MEEi. The association of MEEi with MFS and GLS were tested in multivariable linear regression analyses. Patients with low MEEi were more frequently men, with obesity and hypertension, dyslipidemia and higher HOMA-IR index (all p for trend <0.05). In multivariable analyses, lower MEEi was associated with lower LV myocardial function by MFS and GLS independent of higher LV mass and clinical variables, including older age, male sex, presence of hypertension and a higher triglycerides level (all p < 0.05). In conclusion, in subjects with increased BMI without known cardiovascular disease participating in the FATCOR study, reduced MEEi was associated with lower LV myocardial function both in the circumferential and longitudinal direction, independent of cardiometabolic factors.
    Keywords:  global longitudinal strain; left ventricular mass; myocardial performance; triglycerides
    DOI:  https://doi.org/10.3390/jcm10081581