Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Sci Rep ; 11(1): 21649, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737337

ABSTRACT

The E2 component of the mitochondrial pyruvate dehydrogenase complex (PDC) is the key autoantigen in primary biliary cholangitis (PBC) and STAT3 is an inflammatory modulator that participates in the pathogenesis of many liver diseases. This study investigated whether PDC-E2 interacts with STAT3 in human cholangiocytes (NHC) and hepatocytes (Hep-G2) under cholestatic conditions induced by glyco-chenodeoxycholic acid (GCDC). GCDC induced PDC-E2 expression in the cytoplasmic and nuclear fraction of NHC, whereas in Hep-G2 cells PDC-E2 expression was induced only in the cytoplasmic fraction. GCDC-treatment stimulated phosphorylation of STAT3 in the cytoplasmic fraction of NHC. siRNA-mediated gene silencing of PDC-E2 reduced the expression of pY-STAT3 in NHC but not in HepG2 cells. Immunoprecipitation and a proximity ligation assay clearly demonstrated that GCDC enhanced pY-STAT3 binding to PDC-E2 in the nuclear and cytoplasmic fraction of NHC cells. Staining with Mitotracker revealed mitochondrial co-localization of PDC-E2/pS-STAT3 complexes in NHC and Hep-G2 cells. In cirrhotic PBC livers the higher expression of both PDC-E2 and pY-STAT3 was observed. The immunoblot analysis demonstrated the occurrence of double bands of PDC-E2 protein in control livers, which was associated with a lower expression of pY-STAT3. Our data indicate the interaction between PDC-E2 and phosphorylated STAT3 under cholestatic conditions, which may play a role in the development of PBC.


Subject(s)
Autoantigens/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Mitochondrial Proteins/metabolism , Pyruvate Dehydrogenase Complex/metabolism , STAT3 Transcription Factor/metabolism , Autoantigens/physiology , Bile Ducts/pathology , Cell Line , Dihydrolipoyllysine-Residue Acetyltransferase/physiology , Epithelial Cells/metabolism , Glycochenodeoxycholic Acid/pharmacology , Hep G2 Cells , Hepatocytes/metabolism , Humans , Immunoblotting/methods , Immunoprecipitation/methods , Liver/pathology , Liver Cirrhosis, Biliary/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/physiology , Pyruvate Dehydrogenase Complex/physiology , STAT3 Transcription Factor/physiology
2.
NMR Biomed ; 34(4): e4471, 2021 04.
Article in English | MEDLINE | ID: mdl-33458907

ABSTRACT

The diabetic heart has a decreased ability to metabolize glucose. The anti-ischemic drug meldonium may provide a route to counteract this by reducing l-carnitine levels, resulting in improved cardiac glucose utilization. Therefore, the aim of this study was to use the novel technique of hyperpolarized magnetic resonance to investigate the in vivo effects of treatment with meldonium on cardiac metabolism and function in control and diabetic rats. Thirty-six male Wistar rats were injected either with vehicle, or with streptozotocin (55 mg/kg) to induce a model of type 1 diabetes. Daily treatment with either saline or meldonium (100 mg/kg/day) was undertaken for three weeks. in vivo cardiac function and metabolism were assessed with CINE MRI and hyperpolarized magnetic resonance respectively. Isolated perfused hearts were challenged with low-flow ischemia/reperfusion to assess the impact of meldonium on post-ischemic recovery. Meldonium had no significant effect on blood glucose concentrations or on baseline cardiac function. However, hyperpolarized magnetic resonance revealed that meldonium treatment elevated pyruvate dehydrogenase flux by 3.1-fold and 1.2-fold in diabetic and control animals, respectively, suggesting an increase in cardiac glucose oxidation. Hyperpolarized magnetic resonance further demonstrated that meldonium reduced the normalized acetylcarnitine signal by 2.1-fold in both diabetic and control animals. The increase in pyruvate dehydrogenase flux in vivo was accompanied by an improvement in post-ischemic function ex vivo, as meldonium elevated the rate pressure product by 1.3-fold and 1.5-fold in the control and diabetic animals, respectively. In conclusion, meldonium improves in vivo pyruvate dehydrogenase flux in the diabetic heart, contributing to improved cardiac recovery after ischemia.


Subject(s)
Diabetes Mellitus, Experimental/complications , Magnetic Resonance Spectroscopy/methods , Methylhydrazines/therapeutic use , Myocardial Ischemia/drug therapy , Pyruvate Dehydrogenase Complex/physiology , Animals , Glucose/metabolism , Male , Metabolomics , Methylhydrazines/pharmacology , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Rats , Rats, Wistar , Streptozocin
3.
J Physiol ; 596(15): 3357-3369, 2018 08.
Article in English | MEDLINE | ID: mdl-29383727

ABSTRACT

KEY POINTS: The cardiac metabolic reprogramming seen in heart diseases such as myocardial infarction and hypertrophy shares similarities with that seen in chronic hypoxia, but understanding of how the hypoxic heart responds to further hypoxic challenge - hypoxic tolerance - is limited. The pyruvate dehydrogenase complex serves to control irreversible decarboxylation of pyruvate within mitochondria, and is a key regulator of substrate metabolism, potentially regulating hypoxic tolerance. Acute activation of the pyruvate dehydrogenase complex did not improve cardiac function during acute hypoxia; however, simultaneous activation of the pyruvate dehydrogenase complex during chronic hypoxic exposure improved tolerance to subsequent acute hypoxia. Activation of the pyruvate dehydrogenase complex during chronic hypoxia stockpiled cardiac acetylcarnitine, and this was used during acute hypoxia. This maintained cardiac ATP and glycogen, and improved hypoxic tolerance as a result. These findings demonstrate that pyruvate dehydrogenase complex activation can improve cardiac function under hypoxia. ABSTRACT: The pattern of metabolic reprogramming in chronic hypoxia shares similarities with that following myocardial infarction or hypertrophy; however, the response of the chronically hypoxic heart to subsequent acute injury, and the role of metabolism is not well understood. Here, we determined the myocardial tolerance of the chronically hypoxic heart to subsequent acute injury, and hypothesised that activation of a key regulator of myocardial metabolism, the pyruvate dehydrogenase complex (PDC), could improve hypoxic tolerance. Mouse hearts, perfused in Langendorff mode, were exposed to 30 min of hypoxia, and lost 80% of pre-hypoxic function (P = 0.001), with only 51% recovery of pre-hypoxic function with 30 min of reoxygenation (P = 0.046). Activation of the PDC with infusion of 1 mm dichloroacetate (DCA) during hypoxia and reoxygenation did not alter function. Acute hypoxic tolerance was assessed in hearts of mice housed in hypoxia for 3 weeks. Chronic hypoxia reduced cardiac tolerance to subsequent acute hypoxia, with recovery of function 22% of pre-acute hypoxic levels vs. 39% in normoxic control hearts (P = 0.012). DCA feeding in chronic hypoxia (per os, 70 mg kg-1  day-1 ) doubled cardiac acetylcarnitine content, and this fell following acute hypoxia. This acetylcarnitine use maintained cardiac ATP and glycogen content during acute hypoxia, with hypoxic tolerance normalised. In summary, chronic hypoxia renders the heart more susceptible to acute hypoxic injury, which can be improved by activation of the PDC and pooling of acetylcarnitine. This is the first study showing functional improvement of the chronically hypoxic heart with activation of the PDC, and offers therapeutic potential in cardiac disease with a hypoxic component.


Subject(s)
Heart/physiology , Hypoxia/physiopathology , Pyruvate Dehydrogenase Complex/physiology , Adaptation, Physiological , Animals , Male , Mice
4.
Nat Genet ; 50(2): 219-228, 2018 02.
Article in English | MEDLINE | ID: mdl-29335542

ABSTRACT

The mechanisms by which mitochondrial metabolism supports cancer anabolism remain unclear. Here, we found that genetic and pharmacological inactivation of pyruvate dehydrogenase A1 (PDHA1), a subunit of the pyruvate dehydrogenase complex (PDC), inhibits prostate cancer development in mouse and human xenograft tumor models by affecting lipid biosynthesis. Mechanistically, we show that in prostate cancer, PDC localizes in both the mitochondria and the nucleus. Whereas nuclear PDC controls the expression of sterol regulatory element-binding transcription factor (SREBF)-target genes by mediating histone acetylation, mitochondrial PDC provides cytosolic citrate for lipid synthesis in a coordinated manner, thereby sustaining anabolism. Additionally, we found that PDHA1 and the PDC activator pyruvate dehydrogenase phosphatase 1 (PDP1) are frequently amplified and overexpressed at both the gene and protein levels in prostate tumors. Together, these findings demonstrate that both mitochondrial and nuclear PDC sustain prostate tumorigenesis by controlling lipid biosynthesis, thus suggesting this complex as a potential target for cancer therapy.


Subject(s)
Cell Compartmentation/physiology , Lipogenesis , Prostatic Neoplasms/metabolism , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase Complex/physiology , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cells, Cultured , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/pathology , Humans , Lipogenesis/genetics , Male , Mice , Mice, Knockout , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Pyruvate Dehydrogenase Complex/metabolism
5.
Biochim Biophys Acta Rev Cancer ; 1868(2): 359-371, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28757126

ABSTRACT

Metabolism is essential to all living organisms that provide cells with energy, regulators, building blocks, enzyme cofactors and signaling molecules, and is in tune with nutritional conditions and the function of cells to make the appropriate developmental decisions or maintain homeostasis. As a fundamental biological process, metabolism state affects the production of multiple metabolites and the activation of various enzymes that participate in regulating gene expression, cell apoptosis, cancer progression and immunoreactions. Previous studies generally focus on the function played by the metabolic enzymes in the cytoplasm and mitochondrion. In this review, we conclude the role of them in the nucleus and their implications for cancer progression, immunity and metastasis.


Subject(s)
Cell Nucleus/metabolism , Immunity , Neoplasm Metastasis , Neoplasms/etiology , ATP Citrate (pro-S)-Lyase/physiology , Active Transport, Cell Nucleus , Animals , Carrier Proteins/physiology , Gene Expression Regulation , Humans , Membrane Proteins/physiology , Protein Transport , Pyruvate Dehydrogenase Complex/physiology , Thyroid Hormones/physiology , Thyroid Hormone-Binding Proteins
6.
Free Radic Biol Med ; 97: 501-512, 2016 08.
Article in English | MEDLINE | ID: mdl-27394173

ABSTRACT

Pyruvate dehydrogenase (Pdh) and 2-oxoglutarate dehydrogenase (Ogdh) are vital for Krebs cycle metabolism and sources of reactive oxygen species (ROS). O2(·-)/H2O2 formation by Pdh and Ogdh from porcine heart were compared when operating under forward or reverse electron transfer conditions. Comparisons were also conducted with liver and cardiac mitochondria. During reverse electron transfer (RET) from NADH, purified Ogdh generated ~3-3.5× more O2(·-)/H2O2 in comparison to Pdh when metabolizing 0.5-10µM NADH. Under forward electron transfer (FET) conditions Ogdh generated ~2-4× more O2(·-)/H2O2 than Pdh. In both liver and cardiac mitochondria, Ogdh displayed significantly higher rates of ROS formation when compared to Pdh. Ogdh was also a significant source of ROS in liver mitochondria metabolizing 50µM and 500µM pyruvate or succinate. Finally, we also observed that DTT directly stimulated O2(·-)/H2O2 formation by purified Pdh and Ogdh and in cardiac or liver mitochondria in the absence of substrates and cofactors. Taken together, Ogdh is a more potent source of ROS than Pdh in liver and cardiac tissue. Ogdh is also an important ROS generator regardless of whether pyruvate or succinate serve as the sole source of carbon. Our observations provide insight into the ROS generating capacity of either complex in cardiac and liver tissue. The evidence presented herein also indicates DTT, a reductant that is routinely added to biological samples, should be avoided when assessing mitochondrial O2(·-)/H2O2 production.


Subject(s)
Hydrogen Peroxide/metabolism , Ketoglutarate Dehydrogenase Complex/physiology , Pyruvate Dehydrogenase Complex/physiology , Superoxides/metabolism , Animals , Male , Mice, Inbred C57BL , Mitochondria, Heart/enzymology , Mitochondria, Liver/enzymology , Succinic Acid/metabolism
7.
PLoS One ; 11(3): e0150454, 2016.
Article in English | MEDLINE | ID: mdl-26930489

ABSTRACT

It is generally accepted that ATP regulates mitochondrial function through the AMPK signaling pathway. However, the AMPK-independent pathway remains largely unknown. In this study, we investigated ATP surplus in the negative regulation of mitochondrial function with a focus on pyruvate dehydrogenase (PDH) phosphorylation and protein acetylation. PDH phosphorylation was induced by a high fat diet in the liver of obese mice, which was associated with ATP elevation. In 1c1c7 hepatoma cells, the phosphorylation was induced by palmitate treatment through induction of ATP production. The phosphorylation was associated with a reduction in mitochondria oxygen consumption after 4 h treatment. The palmitate effect was blocked by etomoxir, which inhibited ATP production through suppression of fatty acid ß-oxidation. The PDH phosphorylation was induced by incubation of mitochondrial lysate with ATP in vitro without altering the expression of PDH kinase 2 (PDK2) and 4 (PDK4). In addition, acetylation of multiple mitochondrial proteins was induced by ATP in the same conditions. Acetyl-CoA exhibited a similar activity to ATP in induction of the phosphorylation and acetylation. These data suggest that ATP elevation may inhibit mitochondrial function through induction of the phosphorylation and acetylation of mitochondrial proteins. The results suggest an AMPK-independent mechanism for ATP regulation of mitochondrial function.


Subject(s)
Adenosine Triphosphate/physiology , Mitochondria, Liver/physiology , Mitochondrial Proteins/metabolism , Protein Processing, Post-Translational/physiology , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/physiology , Acetylation , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Diet, High-Fat , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mitochondria, Liver/metabolism , Mitochondrial Proteins/physiology , Phosphorylation , Pyruvate Dehydrogenase Complex/metabolism , Pyruvate Dehydrogenase Complex/physiology , Signal Transduction/physiology
8.
Neurochem Int ; 82: 33-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25684072

ABSTRACT

Glucose is essentially the sole fuel for the adult brain and the mapping of its metabolism has been extensive in the adult but not in the neonatal brain, which is believed to rely mainly on ketone bodies for energy supply. However, glucose is absolutely indispensable for normal development and recent studies have shed light on glycolysis, the pentose phosphate pathway and metabolic interactions between astrocytes and neurons in the 7-day-old rat brain. Appropriately (13)C labeled glucose was used to distinguish between glycolysis and the pentose phosphate pathway during development. Experiments using (13)C labeled acetate provided insight into the GABA-glutamate-glutamine cycle between astrocytes and neurons. It could be shown that in the neonatal brain the part of this cycle that transfers glutamine from astrocytes to neurons is operating efficiently while, in contrast, little glutamate is shuttled from neurons to astrocytes. This lack of glutamate for glutamine synthesis is compensated for by anaplerosis via increased pyruvate carboxylation relative to that in the adult brain. Furthermore, compared to adults, relatively more glucose is prioritized to the pentose phosphate pathway than glycolysis and pyruvate dehydrogenase activity. The reported developmental differences in glucose metabolism and neurotransmitter synthesis may determine the ability of the brain at various ages to resist excitotoxic insults such as hypoxia-ischemia.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Glucose/metabolism , Neurons/metabolism , Adult , Animals , Animals, Newborn , Astrocytes/cytology , Brain/cytology , Brain/growth & development , Citric Acid Cycle , Glutamic Acid/metabolism , Glutamine/biosynthesis , Glycolysis , Humans , Infant, Newborn , Ketone Bodies/metabolism , Milk/chemistry , Neurons/cytology , Pentose Phosphate Pathway , Pyruvate Dehydrogenase Complex/physiology , Rats
9.
Stroke ; 46(2): 492-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25563647

ABSTRACT

BACKGROUND AND PURPOSE: Ischemic stroke induces metabolic disarray. A central regulatory site, pyruvate dehydrogeanse complex (PDHC) sits at the cross-roads of 2 fundamental metabolic pathways: aerobic and anaerobic. In this study, we combined ethanol (EtOH) and normobaric oxygen (NBO) to develop a novel treatment to modulate PDHC and its regulatory proteins, namely pyruvate dehydrogenase phosphatase and pyruvate dehydrogenase kinase, leading to improved metabolism and reduced oxidative damage. METHODS: Sprague-Dawley rats were subjected to transient (2, 3, or 4 hours) middle cerebral artery occlusion followed by 3- or 24-hour reperfusion, or permanent (28 hours) middle cerebral artery occlusion without reperfusion. At 2 hours after the onset of ischemia, rats received either an intraperitoneal injection of saline, 1 dose of EtOH (1.5 g/kg) for 2- and 3-hour middle cerebral artery occlusion, 2 doses of EtOH (1.5 g/kg followed by 1.0 g/kg in 2 hours) in 4 hours or permanent middle cerebral artery occlusion, and EtOH+95% NBO (at 2 hours after the onset of ischemia for 6 hours) in permanent stroke. Infarct volumes and neurological deficits were examined. Oxidative metabolism and stress were determined by measuring ADP/ATP ratio and reactive oxygen species levels. Protein levels of PDHC, pyruvate dehydrogenase kinase, and pyruvate dehydrogenase phosphatase were assessed. RESULTS: EtOH induced dose-dependent neuroprotection in transient ischemia. Compared to EtOH or NBO alone, NBO+EtOH produced the best outcomes in permanent ischemia. These therapies improved brain oxidative metabolism by decreasing ADP/ATP ratios and reactive oxygen species levels, in association with significantly raised levels of PDHC and pyruvate dehydrogenase phosphatase, as well as decreased pyruvate dehydrogenase kinase. CONCLUSIONS: Both EtOH and EtOH+NBO treatments conferred neuroprotection in severe stroke by affecting brain metabolism. The treatment may modulate the damaging cascade of metabolic events by bringing the PDHC activity back to normal metabolic levels.


Subject(s)
Ethanol/therapeutic use , Ischemic Attack, Transient/therapy , Oxygen Inhalation Therapy/methods , Pyruvate Dehydrogenase Complex/physiology , Severity of Illness Index , Stroke/therapy , Animals , Ischemic Attack, Transient/enzymology , Male , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Stroke/enzymology
10.
PLoS One ; 8(6): e67473, 2013.
Article in English | MEDLINE | ID: mdl-23840713

ABSTRACT

UNLABELLED: Pyruvate dehydrogenase (PDH) complex (PDC) deficiency is an inborn error of pyruvate metabolism causing a variety of neurologic manifestations. Systematic analyses of development of affected brain structures and the cellular processes responsible for their impairment have not been performed due to the lack of an animal model for PDC deficiency. METHODS: In the present study we investigated a murine model of systemic PDC deficiency by interrupting the X-linked Pdha1 gene encoding the α subunit of PDH to study its role on brain development and behavioral studies. RESULTS: Male embryos died prenatally but heterozygous females were born. PDC activity was reduced in the brain and other tissues in female progeny compared to age-matched control females. Immunohistochemical analysis of several brain regions showed that approximately 40% of cells were PDH(-). The oxidation of glucose to CO2 and incorporation of glucose-carbon into fatty acids were reduced in brain slices from 15 day-old PDC-deficient females. Histological analyses showed alterations in several structures in white and gray matters in 35 day-old PDC-deficient females. Reduction in total cell number and reduced dendritic arbors in Purkinje neurons were observed in PDC-deficient females. Furthermore, cell proliferation, migration and differentiation into neurons by newly generated cells were reduced in the affected females during pre- and postnatal periods. PDC-deficient mice had normal locomotor activity in a novel environment but displayed decreased startle responses to loud noises and there was evidence of abnormal pre-pulse inhibition of the startle reflex. CONCLUSIONS: The results show that a reduction in glucose metabolism resulting in deficit in energy production and fatty acid biosynthesis impairs cellular differentiation and brain development in PDC-deficient mice.


Subject(s)
Brain Diseases/pathology , Brain/abnormalities , Disease Models, Animal , Pyruvate Dehydrogenase Complex Deficiency Disease/complications , Pyruvate Dehydrogenase Complex/physiology , Animals , Brain/metabolism , Brain/pathology , Brain Diseases/etiology , Carbohydrate Metabolism , Female , Lipogenesis/physiology , Male , Mice , Mice, Knockout , Pyruvate Dehydrogenase Complex Deficiency Disease/physiopathology
11.
Cardiovasc Res ; 95(1): 69-76, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22593200

ABSTRACT

AIMS: The aim of this work was to use hyperpolarized carbon-13 ((13)C) magnetic resonance (MR) spectroscopy and cine MR imaging (MRI) to assess in vivo cardiac metabolism and function in the 15-week-old spontaneously hypertensive rat (SHR) heart. At this time point, the SHR displays hypertension and concentric hypertrophy. One of the cellular adaptations to hypertrophy is a reduction in ß-oxidation, and it has previously been shown that in response to hypertrophy the SHR heart switches to a glycolytic/glucose-oxidative phenotype. METHODS AND RESULTS: Cine-MRI (magnetic resonance imaging) was used to assess cardiac function and degree of cardiac hypertrophy. Wistar rats were used as controls. SHRs displayed functional changes in stroke volume, heart rate, and late peak-diastolic filling alongside significant hypertrophy (a 56% increase in left ventricular mass). Using hyperpolarized [1-(13)C] and [2-(13)C]pyruvate, an 85% increase in (13)C label flux through pyruvate dehydrogenase (PDH) was seen in the SHR heart and (13)C label incorporation into citrate, acetylcarnitine, and glutamate pools was elevated in proportion to the increase in PDH flux. These findings were confirmed using biochemical analysis of PDH activity and protein expression of PDH regulatory enzymes. CONCLUSIONS: Functional and structural alterations in the SHR heart are consistent with the hypertrophied phenotype. Our in vivo work indicates a preference for glucose metabolism in the SHR heart, a move away from predominantly fatty acid oxidative metabolism. Interestingly, (13)C label flux into lactate was unchanged, indicating no switch to an anaerobic glycolytic phenotype, but rather an increased reliance on glucose oxidation in the SHR heart.


Subject(s)
Hypertension/metabolism , Myocardium/metabolism , Adenosine Triphosphate/metabolism , Animals , Bicarbonates/metabolism , Carbon Dioxide/metabolism , Cardiomegaly/etiology , Citric Acid Cycle , Hydrogen-Ion Concentration , Hypertension/complications , Magnetic Resonance Imaging, Cine , Male , Pyruvate Dehydrogenase Complex/physiology , Rats , Rats, Inbred SHR , Rats, Wistar
12.
Alcohol Alcohol ; 46(3): 233-8, 2011.
Article in English | MEDLINE | ID: mdl-21349883

ABSTRACT

AIMS: To clarify the role of acetate in neurochemical mechanisms of the initial (inborn) tolerance to ethanol. METHODS: Rats with low and high inborn tolerance to hypnotic effect of ethanol were used. In the brain region homogenates (frontal and parietal cortex, hypothalamus, striatum, medulla oblongata) and brain cortex synaptosomes, the levels of acetate, acetyl-CoA, acetylcholine (AcH), the activity of pyruvate dehydrogenase (PDG) and acetyl-CoA synthetase were examined. RESULTS: It has been found that brain cortex of rats with high tolerance to hypnotic effect of ethanol have higher level of acetate and activity of acetyl-CoA synthetase, but lower level of acetyl-СCoA and activity of PDG. In brain cortex synaptosomes of tolerant rats, the pyruvate oxidation rate as well as the content of acetyl-CoA and AcH synthesis were lower when compared with intolerant animals. The addition of acetate into the medium significantly increased the AcH synthesis in synaptosomes of tolerant, but not of intolerant animals. Calcium ions stimulated the AcH release from synaptosomes twice as high in tolerant as in intolerant animals. Acetate eliminated the stimulating effect of calcium ions upon the release of AcH in synaptosomes of intolerant rats, but not in tolerant animals. As a result, the quantum release of AcH from synaptosomes in the presence of acetate was 6.5 times higher in tolerant when compared with intolerant rats. CONCLUSION: The brain cortex of rats with high inborn tolerance to hypnotic effect of ethanol can better utilize acetate for the acetyl-CoA and AcH synthesis, as well as being resistant to inhibitory effect of acetate to calcium-stimulated release of AcH. It indicates the metabolic and cholinergic mechanisms of the initial tolerance to ethanol.


Subject(s)
Acetates/metabolism , Adaptation, Physiological/genetics , Alcohol-Related Disorders/genetics , Central Nervous System Depressants/metabolism , Ethanol/metabolism , Synaptosomes/drug effects , Acetyl Coenzyme A/drug effects , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/physiology , Acetylcholine/analysis , Acetylcholine/genetics , Acetylcholine/physiology , Adaptation, Physiological/physiology , Alcohol-Related Disorders/metabolism , Animals , Brain/metabolism , Central Nervous System Depressants/pharmacology , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Ethanol/pharmacology , Humans , Hypothalamus/metabolism , Male , Medulla Oblongata/metabolism , Pyruvate Dehydrogenase Complex/drug effects , Pyruvate Dehydrogenase Complex/genetics , Pyruvate Dehydrogenase Complex/physiology , Rats , Rats, Wistar , Synaptosomes/enzymology
13.
FASEB J ; 24(2): 627-36, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19812372

ABSTRACT

The physiology of two metabolites of vitamin A is understood in substantial detail: retinaldehyde functions as the universal chromophore in the vertebrate and invertebrate eye; retinoic acid regulates a set of vertebrate transcription factors, the retinoic acid receptor superfamily. The third member of this retinoid triumvirate is retinol. While functioning as the precursor of retinaldehyde and retinoic acid, a growing body of evidence suggests a far more fundamental role for retinol in signal transduction. Here we show that retinol is essential for the metabolic fitness of mitochondria. When cells were deprived of retinol, respiration and ATP synthesis defaulted to basal levels. They recovered to significantly higher energy output as soon as retinol was restored to physiological concentration, without the need for metabolic conversion to other retinoids. Retinol emerged as an essential cofactor of protein kinase Cdelta (PKCdelta), without which this enzyme failed to be activated in mitochondria. Furthermore, retinol needed to physically bind PKCdelta, because mutation of the retinol binding site rendered PKCdelta unresponsive to Rol, while retaining responsiveness to phorbol ester. The PKCdelta/retinol complex signaled the pyruvate dehydrogenase complex for enhanced flux of pyruvate into the Krebs cycle. The baseline response was reduced in vitamin A-deficient lecithin:retinol acyl transferase-knockout mice, but this was corrected within 3 h by intraperitoneal injection of vitamin A; this suggests that vitamin A is physiologically important. These results illuminate a hitherto unsuspected role of vitamin A in mitochondrial bioenergetics of mammals, acting as a nutritional sensor. As such, retinol is of fundamental importance for energy homeostasis. The data provide a mechanistic explanation to the nearly 100-yr-old question of why vitamin A deficiency causes so many pathologies that are independent of retinoic acid action.


Subject(s)
Energy Metabolism/physiology , Mitochondria/metabolism , Protein Kinase C-delta/metabolism , Pyruvate Dehydrogenase Complex/physiology , Vitamin A/physiology , Animals , Homeostasis/drug effects , Homeostasis/physiology , Humans , Jurkat Cells , Male , Mice , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/drug effects , Mitochondrial Proton-Translocating ATPases/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Pyruvate Dehydrogenase Complex/drug effects , Retinoids/pharmacology , Signal Transduction , Vitamin A Deficiency/metabolism
14.
Biochim Biophys Acta ; 1787(11): 1334-41, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19481532

ABSTRACT

The heart is capable of balancing the rate of mitochondrial ATP production with utilization continuously over a wide range of activity. This results in a constant phosphorylation potential despite a large change in metabolite turnover. The molecular mechanisms responsible for generating this energy homeostasis are poorly understood. The best candidate for a cytosolic signaling molecule reflecting ATP hydrolysis is Ca(2+). Since Ca(2+) initiates and powers muscle contraction as well as serves as the primary substrate for SERCA, Ca(2+) is an ideal feed-forward signal for priming ATP production. With the sarcoplasmic reticulum to cytosolic Ca(2+) gradient near equilibrium with the free energy of ATP, cytosolic Ca(2+) release is exquisitely sensitive to the cellular energy state providing a feedback signal. Thus, Ca(2+) can serve as a feed-forward and feedback regulator of ATP production. Consistent with this notion is the correlation of cytosolic and mitochondrial Ca(2+) with work in numerous preparations as well as the localization of mitochondria near Ca(2+) release sites. How cytosolic Ca(2+) signaling might regulate oxidative phosphorylation is a focus of this review. The relevant Ca(2+) sensitive sites include several dehydrogenases and substrate transporters together with a post-translational modification of F1-FO-ATPase and cytochrome oxidase. Thus, Ca(2+) apparently activates both the generation of the mitochondrial membrane potential as well as utilization to produce ATP. This balanced activation extends the energy homeostasis observed in the cytosol into the mitochondria matrix in the never resting heart.


Subject(s)
Adenosine Triphosphate/biosynthesis , Calcium Signaling/physiology , Heart/physiology , Mitochondria/metabolism , Animals , Electron Transport Complex IV/physiology , Humans , NAD/metabolism , Oxidative Phosphorylation , Proton-Translocating ATPases/physiology , Pyruvate Dehydrogenase Complex/physiology
16.
Am J Physiol Heart Circ Physiol ; 295(3): H946-H952, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18586888

ABSTRACT

Pyruvate dehydrogenase complex (PDC) plays an important role in energy homeostasis in the heart by catalyzing the oxidative decarboxylation of pyruvate derived primarily from glucose and lactate. Because various pathophysiological states can markedly alter cardiac glucose metabolism and PDC has been shown to be altered in response to chronic ischemia, cardiac physiology of a mouse model with knockout of the alpha-subunit of the pyruvate dehydrogenase component of PDC in heart/skeletal muscle (H/SM-PDCKO) was investigated. H/SM-PDCKO mice did not show embryonic lethality and grew normally during the preweaning period. Heart and skeletal muscle of homozygous male mice had very low PDC activity (approximately 5% of wild-type), and PDC activity in these tissues from heterozygous females was approximately 50%. Male mice did not survive for >7 days after weaning on a rodent chow diet. However, they survived on a high-fat diet and developed left ventricular hypertrophy and reduced left ventricular systolic function compared with wild-type male mice. The changes in the heterozygote female mice were of lesser severity. The deficiency of PDC in H/SM-PDCKO male mice greatly compromises the ability of the heart to oxidize glucose for the generation of energy (and hence cardiac function) and results in cardiac pathological changes. This mouse model demonstrates the importance of glucose oxidation in cardiac energetics and function under basal conditions.


Subject(s)
Cardiomegaly/pathology , Death, Sudden/pathology , Pyruvate Dehydrogenase Complex/physiology , Animals , Body Weight/physiology , Cell Size , Dietary Fats/pharmacology , Electrocardiography , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Glucose/metabolism , Male , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/pathology , Organ Size/physiology , Oxidation-Reduction , Pyruvate Dehydrogenase Complex/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology
17.
Appl Environ Microbiol ; 73(7): 2079-84, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17293513

ABSTRACT

Corynebacterium glutamicum was engineered for the production of L-valine from glucose by deletion of the aceE gene encoding the E1p enzyme of the pyruvate dehydrogenase complex and additional overexpression of the ilvBNCE genes encoding the L-valine biosynthetic enzymes acetohydroxyacid synthase, isomeroreductase, and transaminase B. In the absence of cellular growth, C. glutamicum DeltaaceE showed a relatively high intracellular concentration of pyruvate (25.9 mM) and produced significant amounts of pyruvate, L-alanine, and L-valine from glucose as the sole carbon source. Lactate or acetate was not formed. Plasmid-bound overexpression of ilvBNCE in C. glutamicum DeltaaceE resulted in an approximately 10-fold-lower intracellular pyruvate concentration (2.3 mM) and a shift of the extracellular product pattern from pyruvate and L-alanine towards L-valine. In fed-batch fermentations at high cell densities and an excess of glucose, C. glutamicum DeltaaceE(pJC4ilvBNCE) produced up to 210 mM L-valine with a volumetric productivity of 10.0 mM h(-1) (1.17 g l(-1) h(-1)) and a maximum yield of about 0.6 mol per mol (0.4 g per g) of glucose.


Subject(s)
Corynebacterium glutamicum/metabolism , Pyruvate Dehydrogenase Complex/physiology , Valine/biosynthesis , Alanine/biosynthesis , Fermentation , Isoleucine/biosynthesis , Lysine/biosynthesis , Pyruvate Dehydrogenase Complex/genetics , Pyruvic Acid/metabolism
18.
Endocrine ; 29(1): 169-73, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16622307

ABSTRACT

AIMS: The aim of this study was to examine the chronic effects of different non-esterified fatty acids (NEFA) on insulin secretion by pancreatic islets of normal Wistar rats in vitro. METHODS: Pancreatic islets were isolated from normal Wistar rats, and were incubated with 0.2, 0.4, or 0.8 mmol/L palmitate (C16:0), stearate (C18:0), oleate (C18:1), or linoleate (C18:2) for 24 h, then the insulin secretion and pyruvate dehydrogenase (PDH) activity were examined. RESULTS: Neither islet insulin content nor islet DNA content differed among islets incubated with each kind of NEFA. Compared with control, linoleate significantly inhibited glucose-stimulated insulin secretion (GSIS) and PDH activity at each concentration (p < 0.05), while others inhibited GSIS and PDH activity significantly only at 0.4 and 0.8 mmol/L (p < 0.05). There was no significant difference in GSIS and PDH activity among islets pretreated by palmitate, stearate, and oleate at the same concentration (p > 0.05). However, linoleate decreased GSIS more than others at the same concentration (p < 0.05), while linoleate (0.4 or 0.8 mmol/L) inhibited PDH activity more than others at the same concentration (p < 0.05). CONCLUSIONS: Elevation of palmitate, stearate, oleate or linoleate decreases the beta-cell secretory response to glucose, through inhibiting PDH activity. Linoleate exerts more negative effect on GSIS than other NEFA.


Subject(s)
Fatty Acids, Nonesterified/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Animals , Cells, Cultured , DNA/analysis , Glucose/pharmacology , Insulin/analysis , Insulin Secretion , Islets of Langerhans/chemistry , Islets of Langerhans/physiology , Male , Pyruvate Dehydrogenase Complex/analysis , Pyruvate Dehydrogenase Complex/antagonists & inhibitors , Pyruvate Dehydrogenase Complex/physiology , Rats , Rats, Wistar
19.
Neurochem Int ; 48(6-7): 531-40, 2006.
Article in English | MEDLINE | ID: mdl-16504342

ABSTRACT

The aim of the present study was to identify the distinguishing metabolic characteristics of brain tissue salvaged by reperfusion following focal cerebral ischemia. Rats were subjected to 120 min of middle cerebral artery occlusion followed by 120 min of reperfusion. The rats received an intravenous bolus injection of [1-(13)C]glucose plus [1,2-(13)C]acetate. Subsequently two brain regions considered to represent penumbra and ischemic core, i.e. the frontoparietal cortex and the lateral caudoputamen plus lower parietal cortex, respectively, were analyzed with (13)C NMRS and HPLC. The results demonstrated four metabolic events that distinguished the reperfused penumbra from the ischemic core. (1) Improved astrocytic metabolism demonstrated by increased amounts of [4,5-(13)C]glutamine and improved acetate oxidation. (2) Neuronal mitochondrial activity was better preserved although the flux of glucose via pyruvate dehydrogenase into the tricarboxylic acid (TCA) cycle in glutamatergic and GABAergic neurons was halved. However, NAA content was at control level. (3) Glutamatergic and GABAergic neurons used relatively more astrocytic metabolites derived from the pyruvate carboxylase pathway. (4) Lactate synthesis was not increased despite decreased glucose metabolism in the TCA cycle via pyruvate dehydrogenase. In the ischemic core both neuronal and astrocytic TCA cycle activity declined significantly despite reperfusion. The utilization of astrocytic precursors originating from the pyruvate carboxylase pathway was markedly reduced compared the pyruvate dehydrogenase pathway in glutamate, and completely stopped in GABA. The NAA level fell significantly and lactate accumulated. The results demonstrate that preservation of astrocytic metabolism is essential for neuronal survival and a predictor for recovery.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neurons/pathology , gamma-Aminobutyric Acid/metabolism , Acetic Acid/metabolism , Animals , Astrocytes/pathology , Caudate Nucleus/metabolism , Caudate Nucleus/pathology , Cell Survival , Citric Acid Cycle , Frontal Lobe/metabolism , Frontal Lobe/pathology , Glucose/metabolism , Glutamine/metabolism , Infarction, Middle Cerebral Artery/pathology , Lactic Acid/biosynthesis , Male , Neurons/metabolism , Parietal Lobe/metabolism , Parietal Lobe/pathology , Putamen/metabolism , Putamen/pathology , Pyruvate Carboxylase/physiology , Pyruvate Dehydrogenase Complex/physiology , Rats , Rats, Wistar , Reperfusion
20.
J Appl Physiol (1985) ; 100(6): 2041-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16469933

ABSTRACT

Our laboratory recently showed that six sessions of sprint interval training (SIT) over 2 wk increased muscle oxidative potential and cycle endurance capacity (Burgomaster KA, Hughes SC, Heigenhauser GJF, Bradwell SN, and Gibala MJ. J Appl Physiol 98: 1895-1900, 2005). The present study tested the hypothesis that short-term SIT would reduce skeletal muscle glycogenolysis and lactate accumulation during exercise and increase the capacity for pyruvate oxidation via pyruvate dehydrogenase (PDH). Eight men [peak oxygen uptake (VO2 peak)=3.8+/-0.2 l/min] performed six sessions of SIT (4-7x30-s "all-out" cycling with 4 min of recovery) over 2 wk. Before and after SIT, biopsies (vastus lateralis) were obtained at rest and after each stage of a two-stage cycling test that consisted of 10 min at approximately 60% followed by 10 min at approximately 90% of VO2 peak. Subjects also performed a 250-kJ time trial (TT) before and after SIT to assess changes in cycling performance. SIT increased muscle glycogen content by approximately 50% (main effect, P=0.04) and the maximal activity of citrate synthase (posttraining: 7.8+/-0.4 vs. pretraining: 7.0+/-0.4 mol.kg protein -1.h-1; P=0.04), but the maximal activity of 3-hydroxyacyl-CoA dehydrogenase was unchanged (posttraining: 5.1+/-0.7 vs. pretraining: 4.9+/-0.6 mol.kg protein -1.h-1; P=0.76). The active form of PDH was higher after training (main effect, P=0.04), and net muscle glycogenolysis (posttraining: 100+/-16 vs. pretraining: 139+/-11 mmol/kg dry wt; P=0.03) and lactate accumulation (posttraining: 55+/-2 vs. pretraining: 63+/-1 mmol/kg dry wt; P=0.03) during exercise were reduced. TT performance improved by 9.6% after training (posttraining: 15.5+/-0.5 vs. pretraining: 17.2+/-1.0 min; P=0.006), and a control group (n=8, VO2 peak=3.9+/-0.2 l/min) showed no change in performance when tested 2 wk apart without SIT (posttraining: 18.8+/-1.2 vs. pretraining: 18.9+/-1.2 min; P=0.74). We conclude that short-term SIT improved cycling TT performance and resulted in a closer matching of glycogenolytic flux and pyruvate oxidation during submaximal exercise.


Subject(s)
Carbohydrate Metabolism/physiology , Exercise/physiology , Glycogenolysis/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Physical Endurance/physiology , Running/physiology , Adult , Case-Control Studies , Citrate (si)-Synthase/analysis , Citrate (si)-Synthase/physiology , Exercise Test , Glycogen/analysis , Glycogen/metabolism , Humans , Lactates/analysis , Lactates/metabolism , Male , Mitochondria, Muscle/enzymology , Muscle, Skeletal/chemistry , Oxygen Consumption/physiology , Pyruvate Dehydrogenase Complex/analysis , Pyruvate Dehydrogenase Complex/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...