Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circulation ; 150(8): 622-641, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38660786

ABSTRACT

BACKGROUND: Dysregulated metabolism of bioactive sphingolipids, including ceramides and sphingosine-1-phosphate, has been implicated in cardiovascular disease, although the specific species, disease contexts, and cellular roles are not completely understood. Sphingolipids are produced by the serine palmitoyltransferase enzyme, canonically composed of 2 subunits, SPTLC1 (serine palmitoyltransferase long chain base subunit 1) and SPTLC2 (serine palmitoyltransferase long chain base subunit 2). Noncanonical sphingolipids are produced by a more recently described subunit, SPTLC3 (serine palmitoyltransferase long chain base subunit 3). METHODS: The noncanonical (d16) and canonical (d18) sphingolipidome profiles in cardiac tissues of patients with end-stage ischemic cardiomyopathy and in mice with ischemic cardiomyopathy were analyzed by targeted lipidomics. Regulation of SPTLC3 by HIF1α under ischemic conditions was determined with chromatin immunoprecipitation. Transcriptomics, lipidomics, metabolomics, echocardiography, mitochondrial electron transport chain, mitochondrial membrane fluidity, and mitochondrial membrane potential were assessed in the cSPTLC3KO transgenic mice we generated. Furthermore, morphological and functional studies were performed on cSPTLC3KO mice subjected to permanent nonreperfused myocardial infarction. RESULTS: Herein, we report that SPTLC3 is induced in both human and mouse models of ischemic cardiomyopathy and leads to production of atypical sphingolipids bearing 16-carbon sphingoid bases, resulting in broad changes in cell sphingolipid composition. This induction is in part attributable to transcriptional regulation by HIF1α under ischemic conditions. Furthermore, cardiomyocyte-specific depletion of SPTLC3 in mice attenuates oxidative stress, fibrosis, and hypertrophy in chronic ischemia, and mice demonstrate improved cardiac function and increased survival along with increased ketone and glucose substrate metabolism utilization. Depletion of SPTLC3 mechanistically alters the membrane environment and subunit composition of mitochondrial complex I of the electron transport chain, decreasing its activity. CONCLUSIONS: Our findings suggest a novel essential role for SPTLC3 in electron transport chain function and a contribution to ischemic injury by regulating complex I activity.


Subject(s)
Cardiomyopathies , Electron Transport Complex I , Serine C-Palmitoyltransferase , Animals , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/genetics , Mice , Humans , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Sphingolipids/metabolism , Mice, Knockout , Myocardial Ischemia/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
2.
Am J Hum Genet ; 109(5): 961-966, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35397206

ABSTRACT

The well-established manifestation of mitochondrial mutations in functional cardiac disease (e.g., mitochondrial cardiomyopathy) prompted the hypothesis that mitochondrial DNA (mtDNA) sequence and/or copy number (mtDNAcn) variation contribute to cardiac defects in congenital heart disease (CHD). MtDNAcns were calculated and rare, non-synonymous mtDNA mutations were identified in 1,837 CHD-affected proband-parent trios, 116 CHD-affected singletons, and 114 paired cardiovascular tissue/blood samples. The variant allele fraction (VAF) of heteroplasmic variants in mitochondrial RNA from 257 CHD cardiovascular tissue samples was also calculated. On average, mtDNA from blood had 0.14 rare variants and 52.9 mtDNA copies per nuclear genome per proband. No variation with parental age at proband birth or CHD-affected proband age was seen. mtDNAcns in valve/vessel tissue (320 ± 70) were lower than in atrial tissue (1,080 ± 320, p = 6.8E-21), which were lower than in ventricle tissue (1,340 ± 280, p = 1.4E-4). The frequency of rare variants in CHD-affected individual DNA was indistinguishable from the frequency in an unaffected cohort, and proband mtDNAcns did not vary from those of CHD cohort parents. In both the CHD and the comparison cohorts, mtDNAcns were significantly correlated between mother-child, father-child, and mother-father. mtDNAcns among people with European (mean = 52.0), African (53.0), and Asian haplogroups (53.5) were calculated and were significantly different for European and Asian haplogroups (p = 2.6E-3). Variant heteroplasmic fraction (HF) in blood correlated well with paired cardiovascular tissue HF (r = 0.975) and RNA VAF (r = 0.953), which suggests blood HF is a reasonable proxy for HF in heart tissue. We conclude that mtDNA mutations and mtDNAcns are unlikely to contribute significantly to CHD risk.


Subject(s)
DNA, Mitochondrial , Heart Defects, Congenital , DNA Copy Number Variations/genetics , DNA, Mitochondrial/genetics , Heart Defects, Congenital/genetics , Humans , Mitochondria/genetics , Mutation/genetics
3.
Neuroimage ; 297: 120721, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38968977

ABSTRACT

Individuals with congenital heart disease (CHD) have an increased risk of neurodevelopmental impairments. Given the hypothesized complexity linking genomics, atypical brain structure, cardiac diagnoses and their management, and neurodevelopmental outcomes, unsupervised methods may provide unique insight into neurodevelopmental variability in CHD. Using data from the Pediatric Cardiac Genomics Consortium Brain and Genes study, we identified data-driven subgroups of individuals with CHD from measures of brain structure. Using structural magnetic resonance imaging (MRI; N = 93; cortical thickness, cortical volume, and subcortical volume), we identified subgroups that differed primarily on cardiac anatomic lesion and language ability. In contrast, using diffusion MRI (N = 88; white matter connectivity strength), we identified subgroups that were characterized by differences in associations with rare genetic variants and visual-motor function. This work provides insight into the differential impacts of cardiac lesions and genomic variation on brain growth and architecture in patients with CHD, with potentially distinct effects on neurodevelopmental outcomes.


Subject(s)
Brain , Heart Defects, Congenital , Magnetic Resonance Imaging , Humans , Heart Defects, Congenital/pathology , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/genetics , Female , Male , Child , Brain/diagnostic imaging , Brain/pathology , Adolescent , Young Adult , White Matter/diagnostic imaging , White Matter/pathology , Adult , Child, Preschool , Diffusion Magnetic Resonance Imaging , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/genetics
4.
Adv Exp Med Biol ; 1441: 397-416, 2024.
Article in English | MEDLINE | ID: mdl-38884722

ABSTRACT

Environmental factors have long been known to play a role in the pathogenesis of congenital heart disease (CHD), but this has not been a major focus of research in the modern era. Studies of human exposures and animal models demonstrate that demographics (age, race, socioeconomic status), diseases (e.g., diabetes, hypertension, obesity, stress, infection, high altitude), recreational and therapeutic drug use, and chemical exposures are associated with an increased risk for CHD. Unfortunately, although studies suggest that exposures to these factors may cause CHD, in most cases, the data are not strong, are inconclusive, or are contradictory. Although most studies concentrate on the effects of maternal exposure, paternal exposure to some agents can also modify this risk. From a mechanistic standpoint, recent delineation of signaling and genetic controls of cardiac development has revealed molecular pathways that may explain the effects of environmental signals on cardiac morphogenesis and may provide further tools to study the effects of environmental stimuli on cardiac development. For example, environmental factors likely regulate cellular signaling pathways, transcriptional and epigenetic regulation, proliferation, and physiologic processes that can control the development of the heart and other organs. However, understanding of the epidemiology and risk of these exposures and the mechanistic basis for any effects on cardiac development remains incomplete. Further studies defining the relationship between environmental exposures and human CHD and the mechanisms involved should reveal strategies to prevent, diagnose, and treat CHD induced by environmental signals.


Subject(s)
Environmental Exposure , Heart Defects, Congenital , Signal Transduction , Animals , Female , Humans , Pregnancy , Environmental Exposure/adverse effects , Heart/drug effects , Heart/physiopathology , Heart Defects, Congenital/epidemiology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/etiology , Maternal Exposure/adverse effects , Risk Factors
6.
PLoS Genet ; 16(11): e1009189, 2020 11.
Article in English | MEDLINE | ID: mdl-33216750

ABSTRACT

Although DNA methylation is the best characterized epigenetic mark, the mechanism by which it is targeted to specific regions in the genome remains unclear. Recent studies have revealed that local DNA methylation profiles might be dictated by cis-regulatory DNA sequences that mainly operate via DNA-binding factors. Consistent with this finding, we have recently shown that disruption of CTCF-binding sites by rare single nucleotide variants (SNVs) can underlie cis-linked DNA methylation changes in patients with congenital anomalies. These data raise the hypothesis that rare genetic variation at transcription factor binding sites (TFBSs) might contribute to local DNA methylation patterning. In this work, by combining blood genome-wide DNA methylation profiles, whole genome sequencing-derived SNVs from 247 unrelated individuals along with 133 predicted TFBS motifs derived from ENCODE ChIP-Seq data, we observed an association between the disruption of binding sites for multiple TFs by rare SNVs and extreme DNA methylation values at both local and, to a lesser extent, distant CpGs. While the majority of these changes affected only single CpGs, 24% were associated with multiple outlier CpGs within ±1kb of the disrupted TFBS. Interestingly, disruption of functionally constrained sites within TF motifs lead to larger DNA methylation changes at nearby CpG sites. Altogether, these findings suggest that rare SNVs at TFBS negatively influence TF-DNA binding, which can lead to an altered local DNA methylation profile. Furthermore, subsequent integration of DNA methylation and RNA-Seq profiles from cardiac tissues enabled us to observe an association between rare SNV-directed DNA methylation and outlier expression of nearby genes. In conclusion, our findings not only provide insights into the effect of rare genetic variation at TFBS on shaping local DNA methylation and its consequences on genome regulation, but also provide a rationale to incorporate DNA methylation data to interpret the functional role of rare variants.


Subject(s)
CpG Islands/genetics , DNA Methylation , Epigenesis, Genetic , Genome, Human/genetics , Transcription Factors/metabolism , Adolescent , Adult , Binding Sites/genetics , Child , Child, Preschool , Chromatin Immunoprecipitation Sequencing , Cohort Studies , Female , Heart Defects, Congenital/blood , Heart Defects, Congenital/genetics , Humans , Infant , Infant, Newborn , Male , Middle Aged , Polymorphism, Single Nucleotide , Whole Genome Sequencing , Young Adult
7.
J Extra Corpor Technol ; 54(3): 203-211, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36742212

ABSTRACT

Conservation of mitochondrial adenosine triphosphate (ATP) synthase proteins during ischemia is critical to preserve ATP supply and ventricular function. Following myocardial ischemia in adults, higher order ATP synthase tetramer proteins disassemble into simpler monomer units, reducing the efficiency of ATP production. However, it is unknown if myocardial ischemia following the use of cardioplegia results in tetramer disassembly in neonates, and whether it can be mitigated by cardioplegia if it does occur. We investigated myocardial ATP synthase tetramer disassembly in both a neonatal lamb cardiac surgery model and in neonatal children requiring cardiac surgery for the repair of congenital heart disease. Neonatal lambs (Ovis aries) were placed on cardiopulmonary bypass (CPB) and underwent cardioplegic arrest using a single dose of 30 mL/kg antegrade blood-based potassium cardioplegia (n = 4) or a single dose of 30 mL/kg antegrade del Nido cardioplegia (n = 6). Right ventricular biopsies were taken at baseline on CPB (n = 10) and after approximately 60 minutes of cardioplegic arrest before the cross clamp was released (n = 10). Human right ventricular biopsies (n = 3) were taken following 40.0 ± 23.1 minutes of ischemia after a single dose of antegrade blood-based cardioplegia. Protein complexes were separated on clear native gels and the tetramer to monomer ratio quantified. From the neonatal lamb model regardless of the cardioplegia strategy, the tetramer:monomer ratio decreased significantly during ischemia from baseline measurements (.6 ± .2 vs. .5 ± .1; p = .03). The del Nido solution better preserved the tetramer:monomer ratio when compared to the blood-based cardioplegia (Blood .4 ± .1 vs. del Nido .5 ± .1; p = .05). The tetramer:monomer ratio following the use of blood-based cardioplegia in humans aligned with the lamb data (tetramer:monomer .5 ± .2). These initial results suggest that despite cardioprotection, ischemia during neonatal cardiac surgery results in tetramer disassembly which may be limited when using the del Nido solution.


Subject(s)
Cardiac Surgical Procedures , Coronary Artery Disease , Myocardial Ischemia , Animals , Humans , Cardioplegic Solutions/therapeutic use , Heart Arrest, Induced/methods , Mitochondrial Proton-Translocating ATPases , Myocardial Ischemia/drug therapy , Retrospective Studies , Sheep
8.
J Biol Chem ; 293(18): 6925-6941, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29540484

ABSTRACT

Cardiac energy demands during early embryonic periods are sufficiently met through glycolysis, but as development proceeds, the oxidative phosphorylation in mitochondria becomes increasingly vital. Adrenergic hormones are known to stimulate metabolism in adult mammals and are essential for embryonic development, but relatively little is known about their effects on metabolism in the embryonic heart. Here, we show that embryos lacking adrenergic stimulation have ∼10-fold less cardiac ATP compared with littermate controls. Despite this deficit in steady-state ATP, neither the rates of ATP formation nor degradation was affected in adrenergic hormone-deficient hearts, suggesting that ATP synthesis and hydrolysis mechanisms were fully operational. We thus hypothesized that adrenergic hormones stimulate metabolism of glucose to provide chemical substrates for oxidation in mitochondria. To test this hypothesis, we employed a metabolomics-based approach using LC/MS. Our results showed glucose 1-phosphate and glucose 6-phosphate concentrations were not significantly altered, but several downstream metabolites in both glycolytic and pentose-phosphate pathways were significantly lower compared with controls. Furthermore, we identified glyceraldehyde-3-phosphate dehydrogenase and glucose-6-phosphate dehydrogenase as key enzymes in those respective metabolic pathways whose activity was significantly (p < 0.05) and substantially (80 and 40%, respectively) lower in adrenergic hormone-deficient hearts. Addition of pyruvate and to a lesser extent ribose led to significant recovery of steady-state ATP concentrations. These results demonstrate that without adrenergic stimulation, glucose metabolism in the embryonic heart is severely impaired in multiple pathways, ultimately leading to insufficient metabolic substrate availability for successful transition to aerobic respiration needed for survival.


Subject(s)
Heart/embryology , Metabolomics , Mitochondria, Heart/metabolism , Myocardium/metabolism , Pentose Phosphate Pathway , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/metabolism , Animals , Epinephrine/metabolism , Female , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphates/metabolism , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/metabolism , Glycolysis , Hydrolysis , Ketone Oxidoreductases/metabolism , Male , Mice, Inbred C57BL , Norepinephrine/metabolism , Phosphorylation , Pregnancy
9.
Arch Biochem Biophys ; 662: 177-189, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30571965

ABSTRACT

We have previously demonstrated that inorganic polyphosphate (polyP) is a potent activator of the mitochondrial permeability transition pore (mPTP) in cardiac myocytes. PolyP depletion protected against Ca2+-induced mPTP opening, however it did not prevent and even exacerbated cell death during ischemia-reperfusion (I/R). The central goal of this study was to investigate potential molecular mechanisms underlying these dichotomous effects of polyP on mitochondrial function. We utilized a Langendorff-perfused heart model of I/R to monitor changes in polyP size and chain length at baseline, 20 min no-flow ischemia, and 15 min reperfusion. Freshly isolated cardiac myocytes and mitochondria from C57BL/6J (WT) and cyclophilin D knock-out (CypD KO) mice were used to measure polyP uptake, mPTP activity, mitochondrial membrane potential, respiration and ATP generation. We found that I/R induced a significant decrease in polyP chain length. We, therefore, tested, the ability of synthetic polyPs with different chain length to accumulate in mitochondria and induce mPTP. Both short and long chain polyPs accumulated in mitochondria in oligomycin-sensitive manner implicating potential involvement of mitochondrial ATP synthase in polyP transport. Notably, only short-chain polyP activated mPTP in WT myocytes, and this effect was prevented by mPTP inhibitor cyclosprorin A and absent in CypD KO myocytes. To the contrary, long-chain polyP suppressed mPTP activation, and enhanced ADP-linked respiration and ATP production. Our data indicate that 1) effect of polyP on cardiac function strongly depends on polymer chain length; and 2) short-chain polyPs (as increased in ischemia-reperfusion) induce mPTP and mitochondrial uncoupling, while long-chain polyPs contribute to energy generation and cell metabolism.


Subject(s)
Energy Metabolism/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Myocytes, Cardiac/drug effects , Polyphosphates/pharmacology , Animals , Inorganic Chemicals/pharmacology , Mice , Mice, Inbred C57BL , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism
10.
Am J Physiol Renal Physiol ; 315(5): F1271-F1282, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30110571

ABSTRACT

To better understand the role of the inward-rectifying K channel Kir4.1 (KCNJ10) in the distal nephron, we initially studied a global Kir4.1 knockout mouse (gKO), which demonstrated the hypokalemia and hypomagnesemia seen in SeSAME/EAST syndrome and was associated with reduced Na/Cl cotransporter (NCC) expression. Lethality by ~3 wk, however, limits the usefulness of this model, so we developed a kidney-specific Kir4.1 "knockdown" mouse (ksKD) using a cadherin 16 promoter and Cre-loxP methodology. These mice appeared normal and survived to adulthood. Kir4.1 protein expression was decreased ~50% vs. wild-type (WT) mice by immunoblotting, and immunofluorescence showed moderately reduced Kir4.1 staining in distal convoluted tubule that was minimal or absent in connecting tubule and cortical collecting duct. Under control conditions, the ksKD mice showed metabolic alkalosis and relative hypercalcemia but were normokalemic and mildly hypermagnesemic despite decreased NCC expression. In addition, the mice had a severe urinary concentrating defect associated with hypernatremia, enlarged kidneys with tubulocystic dilations, and reduced aquaporin-3 expression. On a K/Mg-free diet for 1 wk, however, ksKD mice showed marked hypokalemia (serum K: 1.5 ± 0.1 vs. 3.0 ± 0.1 mEq/l for WT), which was associated with renal K wasting (transtubular K gradient: 11.4 ± 0.8 vs. 1.6 ± 0.4 in WT). Phosphorylated-NCC expression increased in WT but not ksKD mice on the K/Mg-free diet, suggesting that loss of NCC adaptation underlies the hypokalemia. In conclusion, even modest reduction in Kir4.1 expression results in impaired K conservation, which appears to be mediated by reduced expression of activated NCC.


Subject(s)
Nephrons/metabolism , Potassium Channels, Inwardly Rectifying/deficiency , Potassium, Dietary/blood , Renal Reabsorption , Alkalosis/blood , Alkalosis/genetics , Alkalosis/physiopathology , Animals , Aquaporin 3/metabolism , Gene Knockdown Techniques , Genotype , Hypercalcemia/blood , Hypercalcemia/genetics , Hypercalcemia/physiopathology , Hyperkalemia/blood , Hyperkalemia/genetics , Hyperkalemia/physiopathology , Hypernatremia/blood , Hypernatremia/genetics , Hypernatremia/physiopathology , Kidney Concentrating Ability , Mice, Inbred C57BL , Mice, Knockout , Nephrons/physiopathology , Phenotype , Phosphorylation , Potassium Channels, Inwardly Rectifying/genetics , Solute Carrier Family 12, Member 3/metabolism
11.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L846-L859, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29345197

ABSTRACT

Supplemental oxygen given to preterm infants has been associated with permanently altering postnatal lung development. Now that these individuals are reaching adulthood, there is growing concern that early life oxygen exposure may also promote cardiovascular disease through poorly understood mechanisms. We previously reported that adult mice exposed to 100% oxygen between postnatal days 0 and 4 develop pulmonary hypertension, defined pathologically by capillary rarefaction, dilation of arterioles and veins, cardiac failure, and a reduced lifespan. Here, Affymetrix Gene Arrays are used to identify early transcriptional changes that take place in the lung before pulmonary capillary rarefaction. We discovered neonatal hyperoxia reduced expression of cardiac muscle genes, including those involved in contraction, calcium signaling, mitochondrial respiration, and vasodilation. Quantitative RT-PCR, immunohistochemistry, and genetic lineage mapping using Myh6CreER; Rosa26RmT/mG mice revealed this reflected loss of pulmonary vein cardiomyocytes. The greatest loss of cadiomyocytes was seen within the lung followed by a graded loss beginning at the hilum and extending into the left atrium. Loss of these cells was seen by 2 wk of age in mice exposed to ≥80% oxygen and was attributed, in part, to reduced proliferation. Administering mitoTEMPO, a scavenger of mitochondrial superoxide during neonatal hyperoxia prevented loss of these cells. Since pulmonary vein cardiomyocytes help pump oxygen-rich blood out of the lung, their early loss following neonatal hyperoxia may contribute to cardiovascular disease seen in these mice, and perhaps in people who were born preterm.


Subject(s)
Biomarkers/metabolism , Hyperoxia/physiopathology , Hypertension, Pulmonary/pathology , Mitochondria/chemistry , Myocytes, Cardiac/pathology , Oxygen/metabolism , Pulmonary Veins/pathology , Animals , Animals, Newborn , Cells, Cultured , Gene Expression Profiling , Hypertension, Pulmonary/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Pulmonary Veins/metabolism
12.
J Bioenerg Biomembr ; 49(1): 13-25, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26868013

ABSTRACT

Neurons experience high metabolic demand during such processes as synaptic vesicle recycling, membrane potential maintenance and Ca2+ exchange/extrusion. The energy needs of these events are met in large part by mitochondrial production of ATP through the process of oxidative phosphorylation. The job of ATP production by the mitochondria is performed by the F1FO ATP synthase, a multi-protein enzyme that contains a membrane-inserted portion, an extra-membranous enzymatic portion and an extensive regulatory complex. Although required for ATP production by mitochondria, recent findings have confirmed that the membrane-confined portion of the c-subunit of the ATP synthase also houses a large conductance uncoupling channel, the mitochondrial permeability transition pore (mPTP), the persistent opening of which produces osmotic dysregulation of the inner mitochondrial membrane, uncoupling of oxidative phosphorylation and cell death. Recent advances in understanding the molecular components of mPTP and its regulatory mechanisms have determined that decreased uncoupling occurs in states of enhanced mitochondrial efficiency; relative closure of mPTP therefore contributes to cellular functions as diverse as cardiac development and synaptic efficacy.


Subject(s)
Ion Channels/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cell Death , Humans , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Oxidative Phosphorylation
13.
Pediatr Res ; 81(6): 932-941, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28141792

ABSTRACT

BACKGROUND: In embryonic myocytes, closure of the mitochondrial permeability transition pore (PTP) drives mitochondrial maturation and cardiac myocyte differentiation. Since neonatal cardiac myocytes remain relatively immature, we hypothesized that inducing PTP closure at this age, by inhibiting the PTP regulator, cyclophilin D (CyPD), genetically or with Cyclosporin A (CsA) and NIM811, would increase cardiac function by increasing mitochondrial maturation and myocyte differentiation. METHODS: Cultured neonatal myocytes or neonatal mice were treated for 5 d with vehicle, CsA or NIM811. Mitochondrial function and structure were measured in vitro. Myocyte differentiation was assessed by immunolabeling for contractile proteins. Cardiac function was determined using echocardiography. RESULTS: The probability of PTP opening was high in WT neonatal myocytes. Treatment with CsA or NIM811 in vitro increased mitochondrial structural complexity and membrane potential, decreased reactive oxygen species levels, and increased myocyte differentiation. WT mice treated with either CsA or NIM811 in vivo for the first 5 d of life had higher ejection fractions. Deleting CyPD had similar effects as CsA and NIM811 on all parameters. CONCLUSIONS: It may be feasible to inhibit the PTP using available drugs to increase mitochondrial maturation, myocyte differentiation, and cardiac function in neonates.


Subject(s)
Cell Differentiation , Mitochondrial Membrane Transport Proteins/physiology , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cells, Cultured , Heart Function Tests , Mice , Mice, Inbred C57BL , Mitochondrial Permeability Transition Pore
14.
Handb Exp Pharmacol ; 240: 21-46, 2017.
Article in English | MEDLINE | ID: mdl-27590224

ABSTRACT

Mitochondrial ATP generation by oxidative phosphorylation combines the stepwise oxidation by the electron transport chain (ETC) of the reducing equivalents NADH and FADH2 with the generation of ATP by the ATP synthase. Recent studies show that the ATP synthase is not only essential for the generation of ATP but may also contribute to the formation of the mitochondrial permeability transition pore (PTP). We present a model, in which the PTP is located within the c-subunit ring in the Fo subunit of the ATP synthase. Opening of the PTP was long associated with uncoupling of the ETC and the initiation of programmed cell death. More recently, it was shown that PTP opening may serve a physiologic role: it can transiently open to regulate mitochondrial signaling in mature cells, and it is open in the embryonic mouse heart. This review will discuss how the ATP synthase paradoxically lies at the center of both ATP generation and cell death.


Subject(s)
Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Proton-Translocating ATPases/physiology , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis , Electron Transport , Energy Metabolism , Humans , Mitochondrial Permeability Transition Pore
15.
Proc Natl Acad Sci U S A ; 111(29): 10580-5, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-24979777

ABSTRACT

Mitochondria maintain tight regulation of inner mitochondrial membrane (IMM) permeability to sustain ATP production. Stressful events cause cellular calcium (Ca(2+)) dysregulation followed by rapid loss of IMM potential known as permeability transition (PT), which produces osmotic shifts, metabolic dysfunction, and cell death. The molecular identity of the mitochondrial PT pore (mPTP) was previously unknown. We show that the purified reconstituted c-subunit ring of the FO of the F1FO ATP synthase forms a voltage-sensitive channel, the persistent opening of which leads to rapid and uncontrolled depolarization of the IMM in cells. Prolonged high matrix Ca(2+) enlarges the c-subunit ring and unhooks it from cyclophilin D/cyclosporine A binding sites in the ATP synthase F1, providing a mechanism for mPTP opening. In contrast, recombinant F1 beta-subunit applied exogenously to the purified c-subunit enhances the probability of pore closure. Depletion of the c-subunit attenuates Ca(2+)-induced IMM depolarization and inhibits Ca(2+) and reactive oxygen species-induced cell death whereas increasing the expression or single-channel conductance of the c-subunit sensitizes to death. We conclude that a highly regulated c-subunit leak channel is a candidate for the mPTP. Beyond cell death, these findings also imply that increasing the probability of c-subunit channel closure in a healthy cell will enhance IMM coupling and increase cellular metabolic efficiency.


Subject(s)
Ion Channels/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Protein Subunits/metabolism , Proton-Translocating ATPases/metabolism , Animals , Calcium/pharmacology , Cell Death/drug effects , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Liposomes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Mutation/genetics , Protein Conformation , Proton-Translocating ATPases/chemistry , Rats , Reactive Oxygen Species/metabolism
16.
Pharmacol Res ; 99: 382-92, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25956324

ABSTRACT

Ion transport across the mitochondrial inner and outer membranes is central to mitochondrial function, including regulation of oxidative phosphorylation and cell death. Although essential for ATP production by mitochondria, recent findings have confirmed that the c-subunit of the ATP synthase also houses a large conductance uncoupling channel, the mitochondrial permeability transition pore (mPTP), the persistent opening of which produces osmotic dysregulation of the inner mitochondrial membrane and cell death. This review will discuss recent advances in understanding the molecular components of mPTP, its regulatory mechanisms and how these contribute directly to its physiological as well as pathological roles.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Death/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Animals , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore
17.
Am J Physiol Heart Circ Physiol ; 306(12): H1602-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24748594

ABSTRACT

Ischemia-reperfusion (IR) injury is significantly worse in aged hearts, but the underlying mechanisms are poorly understood. Age-related damage to mitochondria may be a critical feature, which manifests in an exacerbation of IR injury. Silent information regulator of transcription 3 (SIRT3), the major mitochondrial NAD(+)-dependent lysine deacetylase, regulates a variety of functions, and its inhibition may disrupt mitochondrial function to impact recovery from IR injury. In this study, the role of SIRT3 in mediating the response to cardiac IR injury was examined using an in vitro model of SIRT3 knockdown (SIRT3(kd)) in H9c2 cardiac-derived cells and in Langendorff preparations from adult (7 mo old) wild-type (WT) and SIRT3(+/-) hearts and aged (18 mo old) WT hearts. SIRT3(kd) cells were more vulnerable to simulated IR injury and exhibited a 46% decrease in mitochondrial complex I (Cx I) activity with low O2 consumption rates compared with controls. In the Langendorff model, SIRT3(+/-) adult hearts showed less functional recovery and greater infarct vs. WT, which recapitulates the in vitro results. In WT aged hearts, recovery from IR injury was similar to SIRT3(+/-) adult hearts. Mitochondrial protein acetylation was increased in both SIRT3(+/-) adult and WT aged hearts (relative to WT adult), suggesting similar activities of SIRT3. Also, enzymatic activities of two SIRT3 targets, Cx I and MnSOD, were similarly and significantly inhibited in SIRT3(+/-) adult and WT aged cardiac mitochondria. In conclusion, decreased SIRT3 may increase the susceptibility of cardiac-derived cells and adult hearts to IR injury and may contribute to a greater level of IR injury in the aged heart.


Subject(s)
Aging/metabolism , Myocardial Reperfusion Injury/epidemiology , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Sirtuin 3/deficiency , Animals , Disease Models, Animal , Down-Regulation , Heart Rate , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Risk Factors , Sirtuin 3/genetics , Ventricular Function, Left
18.
Circ Res ; 111(9): 1222-36, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23065345

ABSTRACT

Ischemic heart disease is a significant cause of morbidity and mortality in Western society. Although interventions, such as thrombolysis and percutaneous coronary intervention, have proven efficacious in ischemia and reperfusion injury, the underlying pathological process of ischemic heart disease, laboratory studies suggest further protection is possible, and an expansive research effort is aimed at bringing new therapeutic options to the clinic. Mitochondrial dysfunction plays a key role in the pathogenesis of ischemia and reperfusion injury and cardiomyopathy. However, despite promising mitochondria-targeted drugs emerging from the laboratory, very few have successfully completed clinical trials. As such, the mitochondrion is a potential untapped target for new ischemic heart disease and cardiomyopathy therapies. Notably, there are a number of overlapping therapies for both these diseases, and as such novel therapeutic options for one condition may find use in the other. This review summarizes efforts to date in targeting mitochondria for ischemic heart disease and cardiomyopathy therapy and outlines emerging drug targets in this field.


Subject(s)
Cardiomyopathies/physiopathology , Cardiovascular Agents/pharmacology , Mitochondria, Heart/drug effects , Myocardial Ischemia/physiopathology , Animals , Cardiomyopathies/drug therapy , Cardiovascular Agents/therapeutic use , Disease Models, Animal , Humans , Mitochondria, Heart/physiology , Myocardial Ischemia/drug therapy , Treatment Outcome
19.
Redox Biol ; 76: 103351, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39276392

ABSTRACT

Diastolic dysfunction is increasingly common in preterm infants exposed to supplemental oxygen (hyperoxia). Previous studies in neonatal mice showed hyperoxia suppresses fatty acid synthesis genes required for proliferation and survival of atrial cardiomyocytes. The loss of atrial cardiomyocytes creates a hypoplastic left atrium that inappropriately fills the left ventricle during diastole. Here, we show that hyperoxia stimulates adenosine monophosphate-activated kinase (AMPK) and peroxisome proliferator activated receptor-gamma (PPARγ) signaling in atrial cardiomyocytes. While both pathways can regulate lipid homeostasis, PPARγ was the primary pathway by which hyperoxia inhibits fatty acid gene expression and inhibits proliferation of mouse atrial HL-1 cells. It also enhanced the toxicity of hyperoxia by increasing expression of activating transcription factor (ATF) 5 and other mitochondrial stress response genes. Silencing PPARγ signaling restored proliferation and survival of HL-1 cells as well as atrial cardiomyocytes in neonatal mice exposed to hyperoxia. Our findings reveal PPARγ enhances the toxicity of hyperoxia on atrial cardiomyocytes, thus suggesting inhibitors of PPARγ signaling may prevent diastolic dysfunction in preterm infants.


Subject(s)
Animals, Newborn , Heart Atria , Hyperoxia , Myocytes, Cardiac , PPAR gamma , Signal Transduction , Animals , PPAR gamma/metabolism , PPAR gamma/genetics , Myocytes, Cardiac/metabolism , Mice , Hyperoxia/metabolism , Heart Atria/metabolism , Heart Atria/pathology , Mitochondria/metabolism , Cell Proliferation , AMP-Activated Protein Kinases/metabolism , Humans
20.
Free Radic Biol Med ; 217: 141-156, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38552927

ABSTRACT

Current studies indicate that pathological modifications of tau are associated with mitochondrial dysfunction, synaptic failure, and cognitive decline in neurological disorders and aging. We previously showed that caspase-3 cleaved tau, a relevant tau form in Alzheimer's disease (AD), affects mitochondrial bioenergetics, dynamics and synaptic plasticity by the opening of mitochondrial permeability transition pore (mPTP). Also, genetic ablation of tau promotes mitochondrial function boost and increased cognitive capacities in aging mice. However, the mechanisms and relevance of these alterations for the cognitive and mitochondrial abnormalities during aging, which is the primary risk factor for AD, has not been explored. Therefore, in this study we used aging C57BL/6 mice (2-15 and 28-month-old) to evaluate hippocampus-dependent cognitive performance and mitochondrial function. Behavioral tests revealed that aged mice (15 and 28-month-old) showed a reduced cognitive performance compared to young mice (2 month). Concomitantly, isolated hippocampal mitochondria of aged mice showed a significant decrease in bioenergetic-related functions including increases in reactive oxygen species (ROS), mitochondrial depolarization, ATP decreases, and calcium handling defects. Importantly, full-length and caspase-3 cleaved tau were preferentially present in mitochondrial fractions of 15 and 28-month-old mice. Also, aged mice (15 and 28-month-old) showed an increase in cyclophilin D (CypD), the principal regulator of mPTP opening, and a decrease in Opa-1 mitochondrial localization, indicating a possible defect in mitochondrial dynamics. Importantly, we corroborated these findings in immortalized cortical neurons expressing mitochondrial targeted full-length (GFP-T4-OMP25) and caspase-3 cleaved tau (GFP-T4C3-OMP25) which resulted in increased ROS levels and mitochondrial fragmentation, along with a decrease in Opa-1 protein expression. These results suggest that tau associates with mitochondria and this binding increases during aging. This connection may contribute to defects in mitochondrial bioenergetics and dynamics which later may conduce to cognitive decline present during aging.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Mice , Animals , Reactive Oxygen Species/metabolism , Caspase 3/metabolism , Mice, Inbred C57BL , Alzheimer Disease/metabolism , Cognitive Dysfunction/pathology , Aging/genetics , Mitochondria/metabolism , Hippocampus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL