bims-hafaim Biomed News
on Heart failure metabolism
Issue of 2022‒05‒15
fourteen papers selected by
Kyle McCommis
Saint Louis University


  1. Curr Heart Fail Rep. 2022 May 14.
      PURPOSE OF REVIEW: We review the clinical benefits of altering myocardial substrate metabolism in heart failure.RECENT FINDINGS: Modulation of cardiac substrates (fatty acid, glucose, or ketone metabolism) offers a wide range of therapeutic possibilities which may be applicable to heart failure. Augmenting ketone oxidation seems to offer great promise as a new therapeutic modality in heart failure. The heart has long been recognized as metabolic omnivore, meaning it can utilize a variety of energy substrates to maintain adequate ATP production. The adult heart uses fatty acid as a major fuel source, but it can also derive energy from other substrates including glucose and ketone, and to some extent pyruvate, lactate, and amino acids. However, cardiomyocytes of the failing heart endure remarkable metabolic remodeling including a shift in substrate utilization and reduced ATP production, which account for cardiac remodeling and dysfunction. Research to understand the implication of myocardial metabolic perturbation in heart failure has grown in recent years, and this has raised interest in targeting myocardial substrate metabolism for heart failure therapy. Due to the interdependency between different pathways, the main therapeutic metabolic approaches include inhibiting fatty acid uptake/fatty acid oxidation, reducing circulating fatty acid levels, increasing glucose oxidation, and augmenting ketone oxidation.
    Keywords:  Cardiac metabolism; Fatty acid; Glucose; Heart failure; Ketone bodies
    DOI:  https://doi.org/10.1007/s11897-022-00554-1
  2. FASEB J. 2022 May;36 Suppl 1
      BACKGROUND: Disturbances in cardiac metabolism are increasingly being recognized as crucial drivers of the development and progression of heart disease and ageing. In particular, lipid overload due to Western life style or obesity eventually elicits cardiac insulin resistance and contractile dysfunction (referred to as diabetic cardiomyopathy). As a result, there is an urgent unmet clinical need for treatments that can directly target metabolic defects in heart failure. The transmembrane protein CD36 both facilitates myocardial fatty acid uptake and regulates this process by its subcellular recycling between endosomes and the sarcolemma. In turn, CD36 recycling was recently found to be regulated by instantaneous changes in activity of vacuolar-type H+ -ATPase (v-ATPase) in the endosomes, as controlled by rapid assembly and disassembly of V0 and V1 subunits of v-ATPase.HYPOTHESIS: Regulation of subcellular recycling of CD36 by v-ATPase is an effective target for metabolic modulation therapy in diabetic cardiomyopathy.
    OBJECTIVE: To investigate whether manipulating v-ATPase activity, and thereby CD36 recycling, rescues insulin resistance and contractile dysfunction of the diabetic heart.
    METHODS: Isolated rat cardiomyocytes and human (iPSC-derived and differentiated) cardiomyocytes were treated with bafilomycin (v-ATPase inhibitor) or energy substrates, i.e., fatty acids, glucose, amino acids (AAs) or b-hydroxy butyrate (HB). In addition, rats in vivo were treated with AAs. v-ATPase activity (assembly status), substrate transporter localization (CD36 and glucose transporter GLUT4) and fatty acid and glucose uptake rates (in absence or presence of insulin) were measured.
    RESULTS: In rat and human cardiomyocytes both bafilomycin, excess palmitate and HB inhibited v-ATPase activity, increased sarcolemmal CD36 content, increased fatty acid uptake and lipid accumulation, and elicited contractile dysfunction. However, the latter could be rescued by excess glucose or AAs, concomitant with in each case a similar increase of v-ATPase activity and CD36 reinternalization, while glucose uptake was increased. Hence, v-ATPase activity/assembly is required for retention of CD36 in endosomes. In case of AAs, v-ATPase (re)assembly was dependent on mTORC1 activation. In high fat diet-fed rats, AAs had a similar beneficial action at the myocellular level.
    CONCLUSIONS: These novel data suggest a pivotal role for endosomal v-ATPase activity and subcellular CD36 recycling in determining myocardial substrate preference, in particular fatty acids versus glucose. The molecular mechanism involves assembly/disassembly of the V0 and V1 subunits of v-ATPase, allowing a rapid change in activity. Lipid accumulation and contractile dysfunction in diabetic cardiomyopathy is aggravated by HB but resolved by AAs.
    PERSPECTIVE: Specific amino acids acting through v-ATPase reassembly may be an effective nutraceutical therapy in diabetic cardiomyopathy, but ketone bodies should not be used as these may aggravate lipid accumulation and contractile dysfunction.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R4670
  3. FASEB J. 2022 May;36 Suppl 1
      INTRODUCTION: Heart failure (HF) is still a major cause of mortality worldwide. HF pathogenesis involves disturbances in cardiac energy metabolism. Studies have shown that preventive treatment with a diet supplemented with vitamins B3 or B9/B12 in experimental models of HF, exhibits beneficial effects on cardiac and mitochondrial functions and improves survival. Therefore, the overall objective of our pilot study is to evaluate the curative benefit of a synthetic diet enriched with vitamins B3 (nicotinamide riboside), B9 and B12 (BVit) in a mouse model of HF based on the hypothesis that this enriched diet is beneficial for cardiac function via an improvement of cardiac metabolism.METHOD: Pressure overload was induced by transverse aortic constriction (TAC) in 8-week-old male and female C57Bl6/N mice. Four weeks post-TAC and according to their cardiac function evaluated by echocardiography, mice were randomised to a BVit-enriched diet or not and compared to SHAM animals. The criteria of selection for randomisation were as follows: 10% decrease in ejection fraction (EF), 30% increase in left ventricular (LV) mass and an average pressure gradient of 60mmHg. Mice survival was evaluated and cardiac function was assessed by echocardiography every 4 weeks. Blood samples were taken at 8 weeks of treatment in fasted mice (5 hours) for non-targeted lipidomics mass spectrometry-based analysis.
    RESULTS: Our pilot study show that survival is not improved in males while mortality is reduced in females from 12 weeks of treatment. In males, the reduced EF in the TAC group is not improved by BVit treatment and myocardial hypertrophy, as illustrated by the left ventricular mass (+20% in TAC), is exacerbated with BVit treatment. In contrast, in females, both EF and cardiac hypertrophy were improved (+20% and -13% respectively) after 12 weeks of BVit treatment. To better understand the sexual dimorphism in the response of BVit, non-targeted lipidomics was used on plasma from mice following 8 weeks of treatment. In TAC-females, compared to their SHAM controls, a decrease in triglycerides (TG) and an increase in choline phospholipids were observed while the lipid profile was normalized with BVit treatment. In TAC-males vs SHAM, however, TGs were significantly increased and the treatment with BVit exacerbated the lipidomic profile both in terms of TGs and additional lipid species (identification in progress).
    CONCLUSION: Our pilot study suggests a sexual dimorphism in the response to BVit treatment in experimental HF. Indeed, the benefit of BVit treatment is only shown in females with a delay in the mortality rate associated with improved cardiac function and normalisation of lipid disturbances. To explore and understand the mechanisms underlying this sexual dimorphism, the lipid profile hypothesis appears as a relevant avenue to explore in the future.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0R232
  4. Front Cardiovasc Med. 2022 ;9 821317
      Objective: Neuraminidase 1 (NEU1) participates in the response to multiple receptor signals and regulates various cellular metabolic behaviors. Importantly, it is closely related to the occurrence and progression of cardiovascular diseases. Because ischemic heart disease is often accompanied by impaired mitochondrial energy metabolism and oxidative stress. The purpose of this study was to investigate the functions and possible mechanisms of NEU1 in myocardial remodeling and mitochondrial metabolism induced by myocardial infarction (MI).Methods: In this study, the MI-induced mouse mode, hypoxia-treated H9C2 cells model, and hypoxia-treated neonatal rat cardiomyocytes (NRCMs) model were constructed. Echocardiography and histological analysis were adopted to evaluate the morphology and function of the heart at the whole heart level. Western blot was adopted to determine the related expression level of signaling pathway proteins and mitochondria. Mitochondrial energy metabolism and oxidative stress were detected by various testing kits.
    Results: Neuraminidase 1 was markedly upregulated in MI cardiac tissue. Cardiomyocyte-specific NEU1 deficiency restored cardiac function, cardiac hypertrophy, and myocardial interstitial fibrosis. What is more, cardiomyocyte-specific NEU1 deficiency inhibited mitochondrial dysfunction and oxidative stress induced by MI. Further experiments found that the sirtuin-1/peroxisome proliferator-activated receptor γ coactivator α (SIRT1/PGC-1α) protein level in MI myocardium was down-regulated, which was closely related to the above-mentioned mitochondrial changes. Cardiomyocyte-specific NEU1 deficiency increased the expression of SIRT1, PGC-1α, and mitochondrial transcription factor A (TFAM); which improved mitochondrial metabolism and oxidative stress. Inhibition of SIRT1 activity or PGC-1α activity eliminated the beneficial effects of cardiomyocyte-specific NEU1 deficiency. PGC-1α knockout mice experiments verified that NEU1 inhibition restored cardiac function induced by MI through SIRT1/PGC-1α signaling pathway.
    Conclusion: Cardiomyocyte-specific NEU1 deficiency can alleviate MI-induced myocardial remodeling, oxidative stress, and mitochondrial energy metabolism disorder. In terms of mechanism, the specific deletion of NEU1 may play a role by enhancing the SIRT1/PGC-1α signaling pathway. Therefore, cardiomyocyte-specific NEU1 may provide an alternative treatment strategy for heart failure post-MI.
    Keywords:  SIRT1; mitochondrial metabolism; myocardial infarction; neuraminidase 1; oxidative stress
    DOI:  https://doi.org/10.3389/fcvm.2022.821317
  5. FASEB J. 2022 May;36 Suppl 1
      Healthy hearts rely more on fatty acid (FA) rather than glucose utilization. It remains unclear whether diabetic cardiomyopathy (DbCM) is accounted for by glucotoxicity or lipotoxicity. Previously, we discovered that either insulin deficiency or insulin resistance causes FOXO1-KLF5 activation in human and murine hearts, which drives cardiac lipotoxicity and oxidative stress. Now, we investigate how glucose activates cardiac KLF5 and causes DbCM. We hypothesized that higher cardiac glucose content in diabetes potentiates FOXO1-KLF5 activation and causes glucolipotoxicity. We mimicked Type-1 diabetes (T1D) in C57BL/6 mice via 5 daily intraperitoneal injections of streptozotocin (STZ). In contrast to late T1D (12 Wks post-STZ), cardiac KLF5 expression levels are not increased in the early T1D (4 Wks post-STZ). However, mice developed cardiac dysfunction in early T1D. Seahorse analysis in adult cardiomyocytes isolated from mice with early T1D showed suppression of FA dependence in the expense of higher glucose dependence compared to non-diabetic mice. To confirm whether hyperglycemia or higher cardiac glucose content accounts for cardiac dysfunction in early T1D, we applied anti-hyperglycemia treatment (Dapagliflozin, DAPA, SGLT2 inhibitor) or GLUT1 inhibition (STF-31). Either of the two treatments restored cardiac FA dependence and prevented both glucose preference and cardiac dysfunction in early T1D. GC-MS analysis in hearts of mice with late T1D, which have shifted back to increased FA dependence, showed increased cardiac glucose content -presumably unused glucose- and subsequent increase of cardiac KLF5, as previously shown, which exacerbates cardiac dysfunction. New data revealed that GLUT1 levels were increased in late T1D compared to early T1D, as well as that this is driven by KLF5 activation. The lack of activation of KLF5 expression in early T1D, which reverses in late T1D, is mirrored by FOXO1 transcriptional activity as shown by expression of FOXO1 targets and lower FOXO1 acetylation in late T1D, which is controlled by Sirtuin-1. Analysis of hearts from mice with late T1D and a human cardiomyocyte cell line (AC16) that was treated with high glucose indicated higher Sirtuin-1 expression and Sirtuin-1 binding on FOXO1. Accordingly, prevention of glucose transport to hearts of diabetic mice via treatment with either DAPA or STF-31 for 12 Wks lowered cardiac expression of KLF5 and its targets and improved cardiac function. Conclusively, DbCM begins in early T1D with lower mitochondrial FA utilization that is compensated by higher glucose utilization and is exacerbated in late T1D with activation of SIRT1-FOXO1-KLF5 axis that causes combined lipotoxicity and glucotoxicity. Inhibition of GLUT1 alleviates DbCM via prevention of both early glucose dependence and late KLF5 activation.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R4341
  6. FASEB J. 2022 May;36 Suppl 1
      OBJECTIVE: Heart failure is often preceded by distinct metabolic alterations. However, this shift cannot be sustained indefinitely and typically leads to cardiac dysfunction and arrest. These alterations are thought to be caused by mitochondrial dysfunction leading to the accumulation of acyl-CoAs. To compensate, the injured heart shifts from oxidation of long chain fatty acids (C16) to metabolism of substrates that can replenish the TCA cycle. This is because these fatty acids are restricted to catabolism to acetyl-CoA, for which the enzymes are saturated. By contrast, odd chain fatty acids are metabolized to propionate. The enzymes specific for propionate are freely available for metabolism to succinate leading to TCA cycle anaplerosis. Propionate conversion to propionyl-CoA is the first and major step for its sequestration and metabolism. Moreover, the mitochondrial acyl-CoA synthetase (ACSS) responsible for this conversion is highly expressed in the heart. Hence, we hypothesize that 2-[18 F]-fluoropropionate ([18 F]-FPA) would significantly accumulate in the failing heart and can be imaged by positron emission tomography (PET). We addressed our hypothesis by studying the substrate behavior of FPA for ACSS, and by metabolic profiling of HepG2 cells incubated with FPA. We also successfully imaged heart uptake of [18 F]-FPA in healthy rats by PET.METHODS: The ability of acetate, propionate, and FPA to act as ACSS substrates was determined by a colorimetric pyrophosphate assay. Metabolites were determined by LC/MS in lysates of HepG2 cells incubated with basal media, 10 mM propionate or 10 mM FPA. Racemic 18 F-FPA was synthesized from ethyl-2-bromopropionate by standard nucleophilic substitution. For PET, Sprague-Dawley rats were injected with 250-500 µCi (9.25-18.5 MBq) of [18 F]-FPA, and a 60-minute dynamic PET scan was immediately acquired.
    RESULTS: The kinetic characteristics for the reaction of ACSS acting on acetate (Km = 4.94 mM, Vmax = 0.25 nmols/sec), propionate (Km = 24.20 mM, Vmax = 0.19 nmols/sec), and 2-fluoropropionoate (V = 0.05 nmols/sec) were determined. We also observed a reduction of free CoA in HepG2 cells incubated in 10 mM propionate (4.4 ± 1.2-fold) or 10 mM FPA (3.09 ± 0.96-fold). The kinetics of 18 F-FPA uptake into the healthy heart was assessed by PET and determined to be maximal at 10 minutes post-injection.
    CONCLUSIONS: We identified [18 F]-FPA, and potentially other odd chain fatty acids, as promising metabolic tracers for imaging anaplerosis in the injured heart. FPA effectively decreases free CoA in HepG2 cells, which indicates the formation of acyl-CoAs. Moreover, FPA exhibits some substrate activity for ACSS, representing a major conduit by which it is metabolized in the heart. The poor substrate activity of FPA is ideal as it precludes metabolism by the highly active and specific ACSS enzymes in the liver. Albeit we expect FPA to accumulate in the heart due to its high ACSS expression which is induced in energy deficient states that are characteristic of heart failure. In support of this idea, we demonstrated that [18 F]-FPA is taken up by the healthy heart. We expect the metabolic alterations resulting from ensuing heart failure are sufficient to increase the uptake of [18 F]-FPA in these tissues. Our long-term aim is to develop 18 F-labeled short chain fatty acids, such as [18 F]-FPA, that can be used to diagnose at-risk patients. The early detection of heart failure by non-invasive means is critical for the development of interventions to reduce the severity and aid recovery from cardiac disease.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R2394
  7. FASEB J. 2022 May;36 Suppl 1
      The progression of cardiac diseases is often accompanied by disturbances in substrate metabolism. Extracellular flux analysis has become a standard tool to investigate metabolic alterations in cell lines. However, the enzymatic digestion of the heart to isolate adult cardiomyocytes as well as the cultivation procedure that is required for cell attachment to the cell culture plates might affect metabolism. Therefore, we developed a flux analyser-based method to measure substrate metabolism of intact cardiac tissue slices. Furthermore, we tested this method in a proof-of-principle approach in remote myocardium after myocardial infarction. To yield cardiac tissue pieces of comparable size, mouse cardiac tissue was sliced (150 µm) using a vibratome, and tissue pieces (diameter 1.9 mm) of these slices were punched out. Using "islet capture plates" in a Seahorse XFe 24 analyser, oxygen consumption rates (OCR) were measured at baseline and after FCCP-induced uncoupling in palmitate, glucose (Glc) and glutamine (Gln) enriched medium. To determine long-chain fatty acid metabolism, CPT1 was inhibited by etomoxir, and Glc/Gln metabolism by inhibition of mitochondrial pyruvate carrier (MPC) and glutaminase (Gls) with UK5099/BPTES. Optical mapping of membrane potential was used to assess action potentials in tissue slices as indicator of cellular integrity. Finally, the developed method was used to analyse substrate metabolism in the remote myocardium at day 3 after myocardial ischemia and reperfusion (n=7) in comparison to sham mice (n=8). Data are mean±SD; unpaired two-sample t-test. Basal OCR was 53±8 pmol/min, and FCCP increased OCR to 92±18 pmol/min. Both etomoxir and UK5099/BPTES reduced OCR indicating that both palmitate and Glc/Gln are metabolised. Optical mapping of tissue slices showed regular action potential characteristics and propagation. After myocardial infarction, CPT1 inhibition caused a smaller reduction of uncoupled mitochondrial OCR in I/R animals compared to sham (40±13 vs. 52±4%, p<0.05). This effect was caused by an increased metabolism of Glc/Glu (37±13 vs. 24±4 pmol/min, p<0.05), whilst the effect of CPT1 was not different. Here, we describe a new method to analyse cardiac metabolism using cardiac tissue slices that metabolise fatty acids as well as glucose, and show high functional integrity. Therefore, this method has the potential to expand the methodological alternatives to investigate cardiac substrate metabolism. In a proof-of-principle approach, the analysis of cardiac substrate metabolism of the remote myocardium after I/R showed an augmented glucose/glutamine metabolism.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R2966
  8. Circ Heart Fail. 2022 May 11. CIRCHEARTFAILURE121009521
      BACKGROUND: Defects in energetics are thought to be central to the pathophysiology of hypertrophic cardiomyopathy (HCM); yet, the determinants of ATP availability are not known. The purpose of this study is to ascertain the nature and extent of metabolic reprogramming in human HCM, and its potential impact on contractile function.METHODS: We conducted proteomic and targeted, quantitative metabolomic analyses on heart tissue from patients with HCM and from nonfailing control human hearts.
    RESULTS: In the proteomic analysis, the greatest differences observed in HCM samples compared with controls were increased abundances of extracellular matrix and intermediate filament proteins and decreased abundances of muscle creatine kinase and mitochondrial proteins involved in fatty acid oxidation. These differences in protein abundance were coupled with marked reductions in acyl carnitines, byproducts of fatty acid oxidation, in HCM samples. Conversely, the ketone body 3-hydroxybutyrate, branched chain amino acids, and their breakdown products, were all significantly increased in HCM hearts. ATP content, phosphocreatine, nicotinamide adenine dinucleotide and its phosphate derivatives, NADP and NADPH, and acetyl CoA were also severely reduced in HCM compared with control hearts. Functional assays performed on human skinned myocardial fibers demonstrated that the magnitude of observed reduction in ATP content in the HCM samples would be expected to decrease the rate of cross-bridge detachment. Moreover, left atrial size, an indicator of diastolic compliance, was inversely correlated with ATP content in hearts from patients with HCM.
    CONCLUSIONS: HCM hearts display profound deficits in nucleotide availability with markedly reduced capacity for fatty acid oxidation and increases in ketone bodies and branched chain amino acids. These results have important therapeutic implications for the future design of metabolic modulators to treat HCM.
    Keywords:  cardiomyopathy, hypertrophic; mitochondrial proteins; phosphate; phosphocreatine; proteomics
    DOI:  https://doi.org/10.1161/CIRCHEARTFAILURE.121.009521
  9. FASEB J. 2022 May;36 Suppl 1
      INTRODUCTION: Heart failure is the major cause of death characterized by changes in myocardial energetics, including a reduced level of creatine. In organs with high energy demand such as the heart, creatine is taken up by cardiomyocytes via creatine transporter with subsequent transformation into phosphocreatine, which controls by creatine kinase. This system functions as shuttles high-energy phosphates from mitochondria to contractile elements such as the myofibril, which utilize 70 % of total produced energy by mitochondria. For the past decades, many studies tried to understand whether a reduced level of creatine affects the myocardial function leading to the failing heart. However, most of the results from these studies have been contradictory. The present study describes the detailed morphologic changes of cardiomyocytes of the left ventricle (LV) of mice with whole-body creatine deficiency due to knockout of the biosynthetic enzyme (guanidinoacetate N-methyltransferase, GAMT).METHODS & RESULTS: The present study used GAMT-/- mice model with the unique metabolic fingerprint of zero creatine similar to those in human GAMT deficiency. The histological and transmission electron microscopy (TEM) studies of LV myocardium in GAMT-/- mice showed cardiac hypertrophy and increased collagen accumulation in the myocardial interstitium compared to the wild-type (WT) mice. The TEM examination showed irreversible destructive ultrastructure changes of cardiomyocytes in conjunction with insufficient adaptive protein synthesis accompanied by focal and diffuse lysis of myofilaments in cardiac myocytes. The mitochondrial changes were characterized by cristae homogenization and fragmentation in association with cluster formation. The volume of density of mitochondria (VvMt), myofibrils (VvMf), and their ratios (VvMt/VvMf), along with the coefficient of energy efficiency of mitochondria (CEEM) have been examined in GAMT-/- mice in comparison to WT. We established that VvMt, VvMf, VvMt/VvMf, and CEEM were all significantly reduced. Further, destructive changes in the nucleus and nuclei, focal degradation of sarcoplasm, increases autophagy, are all evidence of inadequate adaptive protein synthesis in cardiac myocytes.
    CONCLUSIONS: These studies of GAMT-/- mice demonstrated that creatine deficiency promotes decreased VvMt/VvMf and CEEM in LV myocytes, which may contribute to the development progression of heart failure and in conjunction with insufficient adaptive protein synthesis contribute to irreversible ultrastructure restructuring of cardiomyocytes with loss contractile myocardial function.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R6306
  10. FASEB J. 2022 May;36 Suppl 1
      INTRODUCTION: Pyruvate kinase (PKM1) directs pyruvate to the Krebs cycle for oxidative metabolism in the healthy heart. Our lab described a hypoxia-mediated switch to the alternatively spliced isoform PKM2, enhancing pyruvate to lactate conversion. Recently, we have also found that Pkm2 knockout (KO) mice had profound depletion of basal glucose in the heart compared to control mice. Pkm2 has also been shown to reduce oxidative damage and promote cardiomyocyte cell proliferation after myocardial infarction (MI). We hypothesize that upregulation of PKM2 can alter metabolic pathways by promoting glycolysis, and that after injury, this can preserve ATP production, protecting the heart from the stresses of hypoxia and injury.METHODS: Global Pkm2 KO mice were subjected to permanent ligation of the left anterior descending coronary artery to mimic an MI. RNA-seq analysis of left ventricles from control (n=8) and Pkm2 KO mice (n=8) before and 3 days after sham or MI surgery was performed. Semi-quantitative real-time PCR was used to confirm changes in selected genes of interest.
    RESULTS: Loss of Pkm2 moderately altered gene expression at baseline (q<0.05, FDR<0.05). Notably, the mitochondrial gene COX3 was downregulated in Pkm2 KO hearts. 68 genes were differentially expressed in Pkm2 KO hearts after MI, not observed in control MI hearts. MI of Pkm2 KO hearts resulted in considerable reduction of transcripts of enzymes in the insulin signaling pathway, mitochondrial oxidative phosphorylation, mitochondrial uncoupling, fatty acid metabolism, and increase in transcripts encoding enzymes in the pentose phosphate pathway, response to oxidative stress, and apoptotic signaling. Semi-quantitative PCR of selected genes involved in glucose metabolism confirmed RNA-seq results.
    CONCLUSIONS: RNA-seq analysis of Pkm2 KO hearts demonstrated that loss of Pkm2 altered gene expression of metabolic and mitochondrial enzymes. Pkm2 KO hearts also showed increased abundance of pro-apoptotic markers which may be a result of increased oxidative stress.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0R496
  11. FASEB J. 2022 May;36 Suppl 1
      The incidence of cardiovascular diseases (CVD) is highly associated with type 2 diabetes (T2D). Women at an early age show reduced incidence of CVD and improved insulin sensitivity compared to men, but these disparities disappear during postmenopausal. Thus, ovary and ovarian hormone, such as estrogen, are expected to protect from these diseases. This study aimed to investigate the role of ovaries and ovarian hormone estrogen in cardiac function and energy metabolism upon loss-of-function of cardiac IRS1 and IRS2 in mice and investigate the underlying mechanisms. We found that all male heart-specific IRS1 and IRS2 double knock-out (H-DKO) mice died of heart failure (HF) at 6-8 weeks with severely impaired energy metabolism. However, all female H-DKO mice survived more than 1 year and showed improvement in energy metabolism. Removal of ovaries in H-DKO female mice led to cardiac dysfunction, disruption of energy metabolism, and ultimately death. However, E2 supplement significantly improved cardiac function, enhanced energy metabolism, and prolonged the lifespan in both male and OVX female H-DKO mice. Finally, we provided evidence that the protective effect of E2 is partially mediated through activation of Akt-Foxo1 signaling in H-DKO heart. These results show that estrogen protects mice from heart insulin resistance induced cardiomyopathy. This study indicates that estrogen signaling could be a potential target to prevent insulin resistance induced heart dysfunction in humans. Estrogen signaling is mediated through estrogen receptors, such as estrogen receptor (ER) α, ERβ, and membrane estrogen receptor (GPER). In the future, we will further decipher the role of estrogen receptors in estrogen mediated protection against cardiac dysfunction during insulin resistance.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.L7593
  12. FASEB J. 2022 May;36 Suppl 1
      Ketone bodies have been identified as an important fuel source during physiological and pathological stress. During exercise, augmenting ketone body metabolism in the skeletal muscle may enhance exercise performance while targeting ketone body metabolism in cardiac disease may improve cardiac function. These provocative, yet still debated, findings may promote the consumption of the ketogenic diet (KD) as a method to increase ketone body availability. Since the KD is a high-fat, low carbohydrate diet, this dietary strategy has the potential to have untoward effects on the heart. Therefore, the purpose of this study was to evaluate the effects of short-term consumption of the KD on cardiac mitochondrial function. A secondary objective was to evaluate potential sex differences in response to the KD. Male (n=9) and female (n=11) mice at four-months of age were randomly assigned to a standard chow or KD (90% fat, 9% protein, 1% carbohydrates) group for 6 weeks. Body weight (BW) was obtained weekly. Glucose and ketone bodies were measured at the end of the dietary intervention via hand-held meters. After 6 weeks, mitochondria were isolated from all hearts and respiration was assessed with an oxygen electrode system. During the first week, Male-KD lost 2.5±1.5% of initial BW compared to an increase of 0.3±1.5% of initial BW for Male-Chow. By the third week, both Male-KD and Male-Chow gained BW at a similar rate until the end of the study. In contrast, Female-KD mice gained BW throughout the 6 weeks and the relative increased compared to baseline was approximately 1.7-2.0-fold greater than Female-chow. Serum ketone bodies were significantly increased in both Male-KD and Female-KD mice compared to chow fed groups (Male-chow, 0.33±0.05 vs. Male-KD, 0.66±0.10 mM, P<0.05; Female-chow, 0.33±0.02 vs. Female-KD, 0.66±0.11 mM, P<0.05). There were no significant sex differences in serum ketone bodies or glucose. State 3 respiration with succinate + ADP was ~35% lower in both male and female KD groups (Male-chow, 236±14 vs. Male-KD, 148±11 nmol/min/mg, P<0.05; Female-chow, 186±22 vs. Female-KD, 133±10 nmol/min/mg, P=0.08). State 3 respiration with pyruvate/malate + ADP were ~30% lower in both Male and female KD groups (Male-chow, 157±18 vs. Male-KD, 105±10 nmol/min/mg, P=NS; Female-chow, 138±22 vs. Female-KD, 81±8 nmol/min/mg, P=0.06). No statistically significant sex differences in mitochondria function were noted. In summary, 6-weeks of KD results in sex differences in patterns of weight gain as male mice experience a transient weight loss and females gain weight throughout the study period. Despite the difference in weight gain, hearts from both males and females fed the KD have similar decrements in mitochondrial function. In conclusion, these findings suggest that the KD may negatively affect cardiac mitochondria and caution against the short-term use of KD.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R5954
  13. Case Rep Cardiol. 2022 ;2022 5529355
      Cardiac involvement may accompany various inborn errors of metabolism (IEM) including fatty acid oxidation (FAO) disorders, presenting as rhythm disturbances, conduction abnormalities, cardiomyopathies, pericardial effusion, and sudden cardiac death. FAO disorders are rare mitochondrial diseases with variable organ involvements and clinical presentations. Very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a FAO disorder with diverse clinical presentations. We report two VLCADD patients with cardiac involvement and diverse presentations. The first patient represents with cardiogenic shock and dilated cardiomyopathy (DCM) at childhood. The second patient represents with suspicious sepsis at early infancy and hypertrophic cardiomyopathy (HCM) at further evaluation. IEM should be thought of in every individual case with suspicious sepsis or cardiac failure regardless of age or previous history.
    DOI:  https://doi.org/10.1155/2022/5529355
  14. FASEB J. 2022 May;36 Suppl 1
      INTRODUCTION: Cardiorespiratory capacity (pVO2 ) is an important prognostic indicator in patients with HCM. Individuals with HCM that have a lower pVO2 possess a greater risk for heart failure (HF) and mortality. High intensity exercise (HIE) can increase pVO2 to a greater degree compared to other training modalities, including when used for cardiac rehabilitation for in cardiac disease states (e.g., HF and Cardiovascular disease). The effects of HIE on HCM, however, are largely unknown. Our study sought to evaluate the effects of High Intensity Interval Training (HIIT) on pVO2 , as well as cardiac structural and functional parameters, in a preclinical mouse model of HCM.METHODS: Female and male C57BL/6J non-transgenic nTG (n=27) and TG (n=32) mice were subjected to HIIT protocol that mirrored methods used clinically in cardiac rehabilitation. A single treadmill training session included 4-4 minute high intensity intervals (~80% preVO2 max speed) interspersed by 5-3 minute recovery intervals (~50% preVO2 max speed) for 31 total minutes. Exercise training sessions were repeated 3 times per week for 6 weeks. Pre and post pVO2 and murine echocardiography were measured and analyzed by a blinded technician.
    RESULTS: Following 6 weeks of the HIIT, nTG female (+13.5 mL/kg/min ± 3.6, p<0.01), nTG male (+13.5 mL/kg/min ± 3.6, p<0.01) and TG female (+7.06 ± 3.3 mL/kg/min, p<0.05) mice demonstrated an average increase in pVO2. A change in pVO2 observed in TG male mice did not reach statistical significance (+4.34 ± p=0.12). Sedentary female (-15.9 mL/kg/min ± 3.3, p<0.001) and male (-13.1 mL/kg/min ± 3.9, p<0.01) TG control mice experienced a significant decrease in pVO2 following the same period, while no change was observed in nTG sedentary controls. Independent of sex, nTG and TG HIIT mice had a significantly greater change in pVO2 compared to sedentary counterparts. HIIT training did not result in changes to the following echocardiography measures in either nTG or TG mice: LV mass, LA volume, cardiac output, or stroke volume.
    CONCLUSION: Our results indicate that a translationally derived HIIT protocol improved cardiorespiratory capacity in a preclinical HCM mouse model, absent of adverse events or worsening of organ level disease pathology.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.L8091