Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
BMC Sports Sci Med Rehabil ; 14(1): 95, 2022 May 26.
Article in English | MEDLINE | ID: mdl-35619160

ABSTRACT

BACKGROUND: Endurance exercise training promotes the catabolism of branched-chain amino acids (BCAAs) in skeletal muscles. We have previously shown that mitochondrial DNA (mtDNA) haplogroups J and K are markers of low responders in endurance training. In this paper, we hypothesize that BCAA catabolism is a surrogate marker of lower respiratory chain activity attributed to these haplogroups. We evaluated whether exercise-induced changes in amino acid concentrations differ between subjects harbouring mtDNA haplogroups J or K and those with non-JK haplogroups. METHODS: Finnish male conscripts (N = 633) undertook the 12-min Cooper running test at the beginning and end of their military service. The intervention during the service mainly included endurance aerobic exercise and sports-related muscle training. Concentrations of seven amino acids were analysed in the serum using a high-throughput 1H NMR metabolomics platform. Total DNA was extracted from whole blood, and restriction fragment analysis was used to determine mtDNA haplogroups J and K. RESULTS: The concentrations of the seven amino acids were higher following the intervention, with the exception of phenylalanine; interestingly, the increase in the concentrations of three BCAAs was larger in subjects with haplogroup J or K than in subjects with non-JK haplogroups (p = 0.029). MtDNA haplogroups J and K share two common nonsynonymous variants. Structural analysis based on crystallographic data on bovine complexes I and III revealed that the Leu18 variant in cytochrome b encoded by m.14798T > C may interfere with ubiquinone binding at the Qi site in complex III. CONCLUSIONS: The increase in the concentrations of serum BCAAs following exercise intervention differs between subjects harbouring mtDNA haplogroup J or K and those harbouring non-JK haplogroups. Lower response in endurance training and difference in exercise-induced increase in the concentrations of serum BCAAs suggest decreased respiratory chain activity. Haplogroups J and K share m.14798T > C in MT-CYB, which may hamper the function of complex III.

2.
Antioxid Redox Signal ; 30(6): 857-874, 2019 02 20.
Article in English | MEDLINE | ID: mdl-29284289

ABSTRACT

SIGNIFICANCE: NAD+ and NADP+ are important cosubstrates in redox reactions and participate in regulatory networks operating in adjustment of metabolic pathways. Moreover, NAD+ is a cosubstrate in post-translational modification of proteins and is involved in DNA repair. NADPH is indispensable for reductive syntheses and the redox chemistry involved in attaining and maintaining correct protein conformation. Recent Advances: Within a couple of decades, a wealth of information has been gathered on NAD(H)+/NADP(H) redox imaging, regulatory role of redox potential in assembly of spatial protein structures, and the role of ADP-ribosylation of regulatory proteins affecting both gene expression and metabolism. All these have a bearing also on disease, healthy aging, and longevity. CRITICAL ISSUES: Knowledge of the signal propagation pathways of NAD+-dependent post-translational modifications is still fragmentary for explaining the mechanism of cellular stress effects and nutritional state on these actions. Evaluation of the cosubstrate and regulator roles of NAD(H) and NADP(H) still suffers from some controversies in experimental data. FUTURE DIRECTIONS: Activating or inhibiting interventions in NAD+-dependent protein modifications for medical purposes has shown promise, but restraining tumor growth by inhibiting DNA repair in tumors by means of interference in sirtuins is still in the early stage. The same is true for the use of this technology in improving health and healthy aging. New genetically encoded specific NAD and NADP probes are expected to modernize the research on redox biology.


Subject(s)
Metabolic Networks and Pathways , NADP/metabolism , NAD/metabolism , Signal Transduction , Animals , Humans , Oxidation-Reduction
3.
Mitochondrion ; 29: 53-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27223842

ABSTRACT

Due to the relative rarity of mitochondrial diseases, generating reference ranges is problematic in evaluation of respiratory chain activities particularly in pediatric cases. We determined the sample distribution of respiratory chain enzyme activities in skeletal muscle biopsies collected from pediatric patients suspected of neuromuscular disorders. Activities of NADH-ubiquinone reductase, NADH-cytochrome c reductase, succinate-cytochrome c reductase; ubiquinol-cytochrome c reductase and cytochrome c oxidase activities have log-normal distributions even when confirmed mitochondrial diseases were ruled out. Impact of the log-normal distribution of the respiratory chain enzyme activities on clinical diagnostics is discussed.


Subject(s)
Biopsy , Electron Transport Chain Complex Proteins/analysis , Mitochondrial Diseases/diagnosis , Mitochondrial Myopathies/diagnosis , Muscle, Skeletal/pathology , Nervous System Diseases/diagnosis , Adolescent , Child , Child, Preschool , Female , Human Activities , Humans , Infant , Infant, Newborn , Male
4.
J Inherit Metab Dis ; 39(1): 67-74, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26112752

ABSTRACT

Mitochondrial mutations impair glucose oxidation and increase glucose uptake in cell cultures and lead to cardiomyopathy in patients. Here we characterize cardiac glucose uptake in 14 patients with the m.3243A > G mutation in mitochondrial DNA. The 14 patients with m.3243A > G and 13 controls were similar in age, physical activity and body mass index. Ten patients had diabetes. Left ventricular glucose uptake per tissue mass (LVGU) was measured with 2-[(18) F]fluoro-2-deoxyglucose positron emission tomography during euglycemic hyperinsulinemia. Cardiac morphology and function were assessed with magnetic resonance imaging. We found that the LVGU was 25% lower in the patients than that in the controls (P = 0.029). LVGU was inversely correlated with mutation heteroplasmy, glycated haemoglobin and fasting lactate in patients. The seven patients with mutation heteroplasmy ≥ 49% had 44% lower LVGU than the seven patients with heteroplasmy < 49%. This difference remained significant after adjustment for concurrent free fatty acid concentration or glycated haemoglobin or glucose uptake in skeletal muscle or all (p < 0.048 [All]). Patients with m.3243A > G had a lower stroke volume and a higher heart rate than the controls, whereas cardiac output and work were similar. Myocardial glucose uptake is not increased but decreased with a threshold effect pattern in patients with the m.3243A > G mutation. The glucose hypometabolism adds to the impaired cardiac energetics and likely contributes to the progression of the mitochondrial cardiomyopathy.


Subject(s)
DNA, Mitochondrial/genetics , Glucose/metabolism , Mitochondria/genetics , Mutation/genetics , Myocardium/metabolism , Blood Glucose/metabolism , Case-Control Studies , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Epithelium/metabolism , Female , Glucose Tolerance Test/methods , Humans , Male , Middle Aged , Muscle, Skeletal/metabolism
5.
Mitochondrion ; 22: 23-30, 2015 May.
Article in English | MEDLINE | ID: mdl-25747201

ABSTRACT

Effects of Complex I mutations were studied by modeling in NuoH, NuoJ or NuoK subunits of Escherichia coli NDH-1 by simultaneous optical monitoring of deamino-NADH oxidation and proton translocation and fitting to the data a model equation of transmembrane proton transport. A homolog of the ND1-E24 LHON/MELAS mutation caused 95% inhibition of d-NADH oxidation and proton translocation. The NuoJ-Y59F replacement decreased proton translocation. The NuoK-E72Q mutation lowered the enzyme activity, but proton pumping could be rescued by the double mutation NuoK-E72Q/I39D. Moving the NuoK-E72/E36 pair one helix turn towards the periplasm did not affect redox activity but decreased proton pumping.


Subject(s)
Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Protons , Biological Transport , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Oxidation-Reduction
6.
J Mol Cell Cardiol ; 53(6): 906-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23036824

ABSTRACT

Reactive oxygen species (ROS) have been implicated in many aspects of tissue/cellular metabolic signaling and pathology, including cardioprotection against ischemia-reperfusion damage. Recent reports of enhanced ROS production under global or simulated ischemia in intact heart or isolated cardiomyocytes, respectively, and its decrease again upon reperfusion are paradoxical. Mechanisms for increasing ROS production with decreasing reactant (oxygen) concentration remain elusive, making it important to critically evaluate the experimental methods used to measure ROS production. In the present paper superoxide production in isolated perfused rat hearts was monitored by lucigenin chemiluminescence or dihydroethidine (DHE) oxidation product fluorescence in parallel with redox state of flavin and cytochrome oxidase. Lucigenin luminescence decreased in ischemia and increased again upon reperfusion, transiently reaching values eightfold the control value coincidently with an overshoot of mitochondrial oxygen concentration. Hypoxic perfusion decreased lucigenin chemiluminescence in spite of coronary flow increase, whereas change in lucigenin concentration in the perfusate had negligible effect. In contrast to lucigenin luminescence, the fluorescence of the DHE oxidation product increased continuously during a 30-min global ischemia and decreased precipitously upon reperfusion, this change is coincident with absorption changes of the oxygen-binding protein myoglobin. The time course of DHE oxidation product fluorescence during ischemia and reperfusion was similar to that of the mitochondrial membrane potential probe safranin as shown in perfused heart previously [Ylitalo KV, Ala-Rämi A, Liimatta EV, Peuhkurinen KJ, Hassinen IE. J Mol Cell Cardiol 2000;32:1223-38]. In solution under high oxygen partial pressure DHE was mainly oxidized to a product, whose fluorescence, absorbance and mass spectra were similar to ethidium, and this product behaved like a mitochondrial membrane potential probe in isolated mitochondria. As a membrane permeable cation it accumulates into the mitochondria when the membrane potential is high (high intramitochondrial concentration quenches fluorescence) and then is released (increased fluorescence) during hypoxia/ischemia. Upon reperfusion it is re-accumulated in the mitochondria as the membrane potential recovers. The non-specific oxidation of DHE makes this dye less suitable for superoxide detection in experiments on isolated perfused hearts that necessitate high oxygen partial pressure in the perfusate. The time course of lucigenin luminescence during ischemia/reperfusion is consistent with decreased ROS production during ischemia/hypoxia, while the oxygen concentration is decreased, followed by an overshoot when the heart tissue is reperfused and the oxygen pressures return to normal or above normal.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Superoxides/metabolism , Acridines , Animals , Coronary Circulation , Dicarbethoxydihydrocollidine/analogs & derivatives , Dicarbethoxydihydrocollidine/metabolism , Electron Transport Complex IV/metabolism , Flavoproteins/metabolism , In Vitro Techniques , Liver/metabolism , Luminescent Agents , Male , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , Mitochondria, Heart/metabolism , Myoglobin/metabolism , Oxidation-Reduction , Oxygen Consumption , Rats
7.
Biochim Biophys Acta ; 1817(2): 312-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22079202

ABSTRACT

Defects in complex I due to mutations in mitochondrial DNA are associated with clinical features ranging from single organ manifestation like Leber hereditary optic neuropathy (LHON) to multiorgan disorders like mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes (MELAS) syndrome. Specific mutations cause overlap syndromes combining several phenotypes, but the mechanisms of their biochemical effects are largely unknown. The m.3376G>A transition leading to p.E24K substitution in ND1 with LHON/MELAS phenotype was modeled here in a homologous position (NuoH-E36K) in the Escherichia coli enzyme and it almost totally abolished complex I activity. The more conservative mutation NuoH-E36Q resulted in higher apparent K(m) for ubiquinone and diminished inhibitor sensitivity. A NuoH homolog of the m.3865A>G transition, which has been found concomitantly in the overlap syndrome patient with the m.3376G>A, had only a minor effect. Consequences of a primary LHON-mutation m.3460G>A affecting the same extramembrane loop as the m.3376G>A substitution were also studied in the E. coli model and were found to be mild. The results indicate that the overlap syndrome-associated m.3376G>A transition in MTND1 is the pathogenic mutation and m.3865A>G transition has minor, if any, effect on presentation of the disease. The kinetic effects of the NuoH-E36Q mutation suggest its proximity to the putative ubiquinone binding domain in 49kD/PSST subunits. In all, m.3376G>A perturbs ubiquinone binding, a phenomenon found in LHON, and decreases the activity of fully assembled complex I as in MELAS.


Subject(s)
Electron Transport Complex I/genetics , Escherichia coli Proteins/chemistry , MELAS Syndrome/genetics , Membrane Proteins/chemistry , NADH Dehydrogenase/genetics , Optic Atrophy, Hereditary, Leber/genetics , Ubiquinone/metabolism , Amino Acid Sequence , Animals , Electron Transport Complex I/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/physiology , NADH Dehydrogenase/metabolism , Protein Binding/genetics , Protein Binding/physiology , Protein Subunits/genetics , Protein Subunits/metabolism , Sequence Homology, Amino Acid
8.
Mitochondrion ; 10(4): 358-61, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20197120

ABSTRACT

MtDNA sequence variation is presumed to be neutral in effect, but associations with diseases and mtDNA haplogroups have been reported. The aim here was to evaluate the functional consequences of m.4216T>C present in haplogroup J. Furthermore, we evaluated m.3866T>C in MT-ND1, a variant detected in a child belonging to haplogroup J and with an isolated complex I deficiency. Homologous substitutions were introduced into Escherichia coli. NADH dehydrogenase domain activity of NDH-1 with either one or both mutations was markedly decreased suggesting that m.4216T>C and m.3866T>C may have an effect on the structural integrity of complex I.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Escherichia coli/genetics , Mutagenesis , NADH Dehydrogenase/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Point Mutation
9.
J Biol Chem ; 285(18): 13646-57, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20185832

ABSTRACT

Hypoxia-inducible factor (HIF) has a pivotal role in oxygen homeostasis and cardioprotection mediated by ischemic preconditioning. Its stability is regulated by HIF prolyl 4-hydroxylases (HIF-P4Hs), the inhibition of which is regarded as a promising strategy for treating diseases such as anemia and ischemia. We generated a viable Hif-p4h-2 hypomorph mouse line (Hif-p4h-2(gt/gt)) that expresses decreased amounts of wild-type Hif-p4h-2 mRNA: 8% in the heart; 15% in the skeletal muscle; 34-47% in the kidney, spleen, lung, and bladder; 60% in the brain; and 85% in the liver. These mice have no polycythemia and show no signs of the dilated cardiomyopathy or hyperactive angiogenesis observed in mice with broad spectrum conditional Hif-p4h-2 inactivation. We focused here on the effects of chronic Hif-p4h-2 deficiency in the heart. Hif-1 and Hif-2 were stabilized, and the mRNA levels of glucose transporter-1, several enzymes of glycolysis, pyruvate dehydrogenase kinase 1, angiopoietin-2, and adrenomedullin were increased in the Hif-p4h-2(gt/gt) hearts. When isolated Hif-p4h-2(gt/gt) hearts were subjected to ischemia-reperfusion, the recovery of mechanical function and coronary flow rate was significantly better than in wild type, while cumulative release of lactate dehydrogenase reflecting the infarct size was reduced. The preischemic amount of lactate was increased, and the ischemic versus preischemic [CrP]/[Cr] and [ATP] remained at higher levels in Hif-p4h-2(gt/gt) hearts, indicating enhanced glycolysis and an improved cellular energy state. Our data suggest that chronic stabilization of Hif-1alpha and Hif-2alpha by genetic knockdown of Hif-p4h-2 promotes cardioprotection by induction of many genes involved in glucose metabolism, cardiac function, and blood pressure.


Subject(s)
Dioxygenases/metabolism , Glucose/metabolism , Muscle Proteins/metabolism , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Acute Disease , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Coronary Circulation/genetics , Dioxygenases/genetics , Glucose/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Mice , Mice, Transgenic , Muscle Proteins/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/pathology , Organ Specificity/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
10.
Tissue Eng Part C Methods ; 16(3): 435-45, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19839730

ABSTRACT

Advanced therapies medicinal products (ATMPs) have introduced innovative cell-based products. However, the regulatory demands for characterization of ATMPs are currently unable to adequately address the safety of such products. As recent studies have emphasized the role of mitochondria in the osteogenic differentiation of human mesenchymal stem cells (hMSCs), we have studied in detail the viability and osteogenic differentiation potency of the hMSCs intended for use as ATMPs based on analyses of the mitochondrial inner membrane potential (DeltaPsi(m)). Flow cytometric measurement of 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolcarbocyanine iodide (JC-1), propidium iodide fluorescence, and AnnexinV was employed to determine DeltaPsi(m), plasma membrane integrity, and organization of phosphatidylserine in plasma membrane, respectively, in cultured hMSCs. Apoptosis was induced by incubating cells at critical concentration (20 muM) of menadione. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was used as an indicator for cell proliferation and alkaline phosphatase activity and calcium deposition as indicators of osteogenic differentiation. Based on JC-1 fluorescence, cell morphology, organization of phosphatidylserine, and plasma membrane integrity, we could sort cells into four categories that represented different cell quality. A strong correlation between JC-1 and osteogenic differentiation was demonstrated for the first time and thus this analytical tool is suitable not only to determine cell viability but also to predict osteogenic differentiation of hMSC.


Subject(s)
Bone and Bones/cytology , Mesenchymal Stem Cells/cytology , Mitochondria/physiology , Adolescent , Adult , Aged , Cell Differentiation , Child , Flow Cytometry , Humans , Membrane Potentials , Middle Aged , Young Adult
11.
Mitochondrion ; 9(6): 394-401, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19616643

ABSTRACT

Seven of the 45 subunits of mitochondrial NADH:ubiquinone oxidoreductase (complex I) are mitochondrially encoded and have been shown to harbor pathogenic mutations. We modeled the human disease-associated mutations A4136G/ND1-Y277C, T4160C/ND1-L285P and C4171A/ND1-L289M in a highly conserved region of the fourth matrix-side loop of the ND1 subunit by mutating homologous amino acids and surrounding conserved residues of the NuoH subunit of Escherichia coli NDH-1. Deamino-NADH dehydrogenase activity, decylubiquinone reduction kinetics, hexammineruthenium (HAR) reductase activity, and the proton pumping efficiency of the enzyme were assayed in cytoplasmic membrane preparations. Among the human disease-associated mutations, a statistically significant 22% decrease in enzyme activity was observed in the NuoH-L289C mutant and a 29% decrease in the double mutant NuoH-L289C/V297P compared with controls. The adjacent mutations NuoH-D295A and NuoH-R293M caused 49% and 39% decreases in enzyme activity, respectively. None of the mutations studied significantly affected the K(m) value of the enzyme for decylubiquinone or the amount of membrane-associated NDH-1 as estimated from the HAR reductase activity. In spite of the decrease in enzyme activity, all the mutant strains were able to grow on malate, which necessitates sufficient NDH-1 activity. The results show that in ND1/NuoH its fourth matrix-side loop is probably not directly involved in ubiquinone binding or proton pumping but has a role in modifying enzyme activity.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Membrane Proteins/genetics , Mutation, Missense , Amino Acid Sequence , Amino Acid Substitution , Escherichia coli/growth & development , Escherichia coli Proteins/metabolism , Humans , Kinetics , Malates/metabolism , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , NADH Dehydrogenase/metabolism , Protein Structure, Quaternary , Proton Pumps/metabolism , Ruthenium Compounds/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
12.
PLoS One ; 4(5): e5589, 2009.
Article in English | MEDLINE | ID: mdl-19440339

ABSTRACT

It has been recently recognized that mammalian mitochondria contain most, if not all, of the components of fatty acid synthesis type II (FAS II). Among the components identified is 2-enoyl thioester reductase/mitochondrial enoyl-CoA reductase (Etr1/Mecr), which catalyzes the NADPH-dependent reduction of trans-2-enoyl thioesters, generating saturated acyl-groups. Although the FAS type II pathway is highly conserved, its physiological role in fatty acid synthesis, which apparently occurs simultaneously with breakdown of fatty acids in the same subcellular compartment in mammals, has remained an enigma. To study the in vivo function of the mitochondrial FAS in mammals, with special reference to Mecr, we generated mice overexpressing Mecr under control of the mouse metallothionein-1 promoter. These Mecr transgenic mice developed cardiac abnormalities as demonstrated by echocardiography in vivo, heart perfusion ex vivo, and electron microscopy in situ. Moreover, the Mecr transgenic mice showed decreased performance in endurance exercise testing. Our results showed a ventricular dilatation behind impaired heart function upon Mecr overexpression, concurrent with appearance of dysmorphic mitochondria. Furthermore, the data suggested that inappropriate expression of genes of FAS II can result in the development of hereditary cardiomyopathy.


Subject(s)
Gene Expression , Heart Diseases/physiopathology , Mitochondrial Proteins/physiology , Myocardium/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Animals , Blotting, Northern , Blotting, Southern , Echocardiography , Exercise Test , Heart Diseases/genetics , In Situ Nick-End Labeling , Male , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Reverse Transcriptase Polymerase Chain Reaction , fas Receptor/genetics
13.
Circ Res ; 103(9): 1018-26, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18757827

ABSTRACT

Vascular endothelial growth factor (VEGF)-B is poorly angiogenic but prominently expressed in metabolically highly active tissues, including the heart. We produced mice expressing a cardiac-specific VEGF-B transgene via the alpha-myosin heavy chain promoter. Surprisingly, the hearts of the VEGF-B transgenic mice showed concentric cardiac hypertrophy without significant changes in heart function. The cardiac hypertrophy was attributable to an increased size of the cardiomyocytes. Blood capillary size was increased, whereas the number of blood vessels per cell nucleus remained unchanged. Despite the cardiac hypertrophy, the transgenic mice had lower heart rate and blood pressure than their littermates, and they responded similarly to angiotensin II-induced hypertension, confirming that the hypertrophy does not compromise heart function. Interestingly, the isolated transgenic hearts had less cardiomyocyte damage after ischemia. Significantly increased ceramide and decreased triglyceride levels were found in the transgenic hearts. This was associated with structural changes and eventual lysis of mitochondria, resulting in accumulation of intracellular vacuoles in cardiomyocytes and increased death of the transgenic mice, apparently because of mitochondrial lipotoxicity in the heart. These results suggest that VEGF-B regulates lipid metabolism, an unexpected function for an angiogenic growth factor.


Subject(s)
Cardiomegaly/metabolism , Cardiomyopathies/metabolism , Lipid Metabolism , Myocardium/metabolism , Vascular Endothelial Growth Factor B/metabolism , Ventricular Function, Left , Angiotensin II , Animals , Blood Pressure , Capillaries/metabolism , Capillaries/pathology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cell Size , Ceramides/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , Disease Models, Animal , Heart Rate , Humans , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Mice , Mice, Transgenic , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Neovascularization, Physiologic , Oxidation-Reduction , Promoter Regions, Genetic , Skin/blood supply , Skin/metabolism , Time Factors , Triglycerides/metabolism , Up-Regulation , Vascular Endothelial Growth Factor B/genetics , Ventricular Myosins/genetics
14.
Epilepsia ; 49(6): 1038-45, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18294203

ABSTRACT

PURPOSE: Polymerase gamma (POLG) is the sole enzyme in the replication of mitochondrial DNA (mtDNA). Numerous mutations in the POLG1 gene have been detected recently in patients with various phenotypes including a classic infantile-onset Alpers-Huttenlocher syndrome (AHS). Here we studied the molecular etiology of juvenile-onset AHS manifesting with status epilepticus and liver disease in three teenagers. PATIENTS AND METHODS: We examined 14- and 17-year-old female siblings (patients 1 and 2) and an unrelated 15-year-old girl (patient 3) with juvenile-onset AHS, sequenced POLG1, and the entire mtDNA, examined mtDNA deletions by amplification of the full-length mtDNA with the long PCR method and used real-time PCR to quantify mtDNA in the tissue samples. RESULTS: The initial manifestations were migraine-like headache and epilepsy, and the terminal manifestations status epilepticus and hepatic failure. A homozygous W748S mutation in POLG1 was detected in the three patients. No deletions or pathogenic point mutations were found in mtDNA, but all three patients had mtDNA depletion. CONCLUSIONS: POLG mutations should be considered in cases of teenagers and young adults with a sudden onset of intractable seizures or status epilepticus, and acute liver failure. The W748S POLG1 mutation seems to lead to tissue-specific, partial mtDNA depletion in patients with juvenile-onset Alpers syndrome. Valproic acid should be avoided in the treatment of epileptic seizures in these patients.


Subject(s)
DNA Mutational Analysis , DNA-Directed DNA Polymerase/genetics , Diffuse Cerebral Sclerosis of Schilder/genetics , Homozygote , Status Epilepticus/genetics , Adolescent , Brain/pathology , DNA Polymerase gamma , DNA, Mitochondrial/genetics , Diagnosis, Differential , Diffuse Cerebral Sclerosis of Schilder/diagnosis , Diffuse Cerebral Sclerosis of Schilder/pathology , Electroencephalography , Epilepsy, Tonic-Clonic/diagnosis , Epilepsy, Tonic-Clonic/genetics , Epilepsy, Tonic-Clonic/pathology , Fatal Outcome , Female , Humans , Liver/pathology , Liver Failure, Acute/diagnosis , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Migraine Disorders/diagnosis , Migraine Disorders/genetics , Migraine Disorders/pathology , Sequence Analysis, DNA , Status Epilepticus/diagnosis , Status Epilepticus/pathology
15.
Biochem J ; 409(1): 129-37, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17894548

ABSTRACT

LHON (Leber hereditary optic neuropathy) is a maternally inherited disease that leads to sudden loss of central vision at a young age. There are three common primary LHON mutations, occurring at positions 3460, 11778 and 14484 in the human mtDNA (mitochondrial DNA), leading to amino acid substitutions in mitochondrial complex I subunits ND1, ND4 and ND6 respectively. We have now examined the effects of ND6 mutations on the function of complex I using the homologous NuoJ subunit of Escherichia coli NDH-1 (NADH:quinone oxidoreductase) as a model system. The assembly level of the NDH-1 mutants was assessed using electron transfer from deamino-NADH to the 'shortcut' electron acceptor HAR (hexammine ruthenium), whereas ubiquinone reductase activity was determined using DB (decylubiquinone) as a substrate. Mutant growth in minimal medium with malate as the main carbon source was used for initial screening of the efficiency of energy conservation by NDH-1. The results indicated that NuoJ-M64V, the equivalent of the common LHON mutation in ND6, had a mild effect on E. coli NDH-1 activity, while nearby mutations, particularly NuoJ-Y59F, NuoJ-V65G and NuoJ-M72V, severely impaired the DB reduction rate and cell growth on malate. NuoJ-Met64 and NuoJ-Met72 position mutants lowered the affinity of NDH-1 for DB and explicit C-type inhibitors, whereas NuoJ-Y59C displayed substrate inhibition by oxidized DB. The results are compatible with the notion that the ND6 subunit delineates the binding cavity of ubiquinone substrate, but does not directly take part in the catalytic reaction. How these changes in the enzyme's catalytic properties contribute to LHON pathogenesis is discussed.


Subject(s)
Bacteria/metabolism , Electron Transport Complex I/genetics , Mitochondria/metabolism , Mutation , Optic Atrophy, Hereditary, Leber/genetics , Ubiquinone/chemistry , Catalysis , DNA, Mitochondrial/metabolism , Electron Transport Complex I/metabolism , Escherichia coli/metabolism , Female , Humans , Inhibitory Concentration 50 , Kinetics , Male , Mutagenesis
16.
J Biochem Biophys Methods ; 70(4): 547-54, 2007 Jun 10.
Article in English | MEDLINE | ID: mdl-17316820

ABSTRACT

Mitochondrial damage is the main source of cellular injury upon ischemia-reperfusion, and calcium loading has been implicated in this phenomenon. The use of optical probes for calcium monitoring of the intact heart is hampered by internal filter effects of intracellular hemoproteins, endogenous fluorescence, and their sensitivity to pH. We describe here a method for measurement of intracellular free calcium in isolated myoglobin-deficient perfused mouse hearts under conditions of large intracellular pH fluctuations by simultaneous fluorescence monitoring of the calcium-probe Fura-2 and the pH probe BCECF through dual wavelength excitation of both probes. In myoglobin-containing mouse heart endogenous chromophores interfere with Fura-2 fluorometry. It is shown that a paradoxical decrease in Fura-2 fluorescence occurs during ischemia in isolated mouse hearts. Simultaneous recording of BCECF fluorescence (calibrated against pH measurement with phosphorus NMR) and data reduction based on continual recalculation of the apparent dissociation constant of the calcium-probe complex revealed that a marked increase in intracellular free calcium occurs, and that the Fura-2 fluorescence decrease was caused by an increase in dissociation constant due to intracellular acidification. Intracellular free calcium rose almost linearly during a 20-min period of ischemia and returned to basal values rapidly upon the commencement of perfusion.


Subject(s)
Calcium/metabolism , Hydrogen-Ion Concentration , Mitochondria, Heart/metabolism , Myocardial Ischemia/metabolism , Myoglobin/metabolism , Animals , Calibration , Fluoresceins , Fluorescent Dyes , Fura-2 , Horses , Kinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoglobin/deficiency , Spectrometry, Fluorescence/methods
17.
J Biol Chem ; 282(7): 4524-4532, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17182618

ABSTRACT

The stability and transcriptional activity of the hypoxia-inducible factors (HIFs) are regulated by two oxygen-dependent events that are catalyzed by three HIF prolyl 4-hydroxylases (HIF-P4Hs) and one HIF asparaginyl hydroxylase (FIH). We have studied possible links between metabolic pathways and HIF hydroxylases by analyzing the abilities of citric acid cycle intermediates to inhibit purified human HIF-P4Hs and FIH. Fumarate and succinate were identified as in vitro inhibitors of all three HIF-P4Hs, fumarate having K(i) values of 50-80 microM and succinate 350-460 microM, whereas neither inhibited FIH. Oxaloacetate was an additional inhibitor of all three HIF-P4Hs with K(i) values of 400-1000 microM and citrate of HIF-P4H-3, citrate being the most effective inhibitor of FIH with a K(i) of 110 microM. Culturing of cells with fumarate diethyl or dimethyl ester, or a high concentration of monoethyl ester, stabilized HIF-1alpha and increased production of vascular endothelial growth factor and erythropoietin. Similar, although much smaller, changes were found in cultured fibroblasts from a patient with fumarate hydratase (FH) deficiency and upon silencing FH using small interfering RNA. No such effects were seen upon culturing of cells with succinate diethyl or dimethyl ester. As FIH was not inhibited by fumarate, our data indicate that the transcriptional activity of HIF is quite high even when binding of the coactivator p300 is prevented. Our data also support recent suggestions that the increased fumarate and succinate levels present in the FH and succinate dehydrogenase-deficient tumors, respectively, can inhibit the HIF-P4Hs with consequent stabilization of HIF-alphas and effects on tumor pathology.


Subject(s)
Citric Acid Cycle , Fibroblasts/enzymology , Gene Expression Regulation, Enzymologic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mixed Function Oxygenases/metabolism , Procollagen-Proline Dioxygenase/metabolism , Cell Line , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Erythropoietin/biosynthesis , Fibroblasts/chemistry , Fibroblasts/pathology , Fumarate Hydratase/chemistry , Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Neoplasms/chemistry , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/genetics , Succinate Dehydrogenase/chemistry , Succinate Dehydrogenase/deficiency , Succinate Dehydrogenase/metabolism , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A/biosynthesis , p300-CBP Transcription Factors/metabolism
18.
Hum Mol Genet ; 15(17): 2543-52, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16849371

ABSTRACT

The ND1 subunit gene of the mitochondrial NADH-ubiquinone oxidoreductase (complex I) is a hot spot for mutations causing Leber hereditary optic neuropathy and several mutations causing the mitochondrial encephalopathy, lactic acidosis and stroke-like episodes syndrome (MELAS). We have used Escherichia coli and Paracoccus denitrificans as model systems to study the effect of mutations 3946 and 3949, which change conserved residues in ND1 and cause MELAS. The vicinity of these mutations was also explored with a series of mutations in charged residues. The 3946 mutation results in E214K substitution in human ND1. Replacement of the equivalent residue in E. coli with lysine or glutamine detracted from enzyme assembly and the assembled enzyme was inactive. However, the equivalent E234Q mutant enzyme in P. denitrificans failed to assemble completely (or was rapidly degraded). Also the corresponding substitution with aspartate decreased the enzyme activity in P. denitrificans and E. coli. The 3949-equivalent substitution, Y229H in E. coli, lowered the catalytic activity by 30%. In addition, an activation of the enzyme during catalytic turnover was seen in this bacterial NDH-1, something that was even more pronounced in another mutant in the same loop, D213E. Several other mutations in this region decreased the enzyme activity. The studied MELAS mutations are situated in a matrix-side loop, which appears to be highly sensitive to structural perturbations. The results provide new information on the function of the region affected by the MELAS mutations 3946 and 3949 that is not obtainable from patient samples or current eukaryote models.


Subject(s)
Conserved Sequence , DNA, Mitochondrial , Electron Transport Complex I/genetics , Escherichia coli Proteins/chemistry , MELAS Syndrome/genetics , Paracoccus denitrificans/genetics , Point Mutation , Amino Acid Sequence , Catalysis , Cell Membrane/chemistry , Cell Membrane/genetics , Electron Transport Complex I/chemistry , Escherichia coli Proteins/genetics , Humans , Molecular Sequence Data , Paracoccus denitrificans/chemistry , Paracoccus denitrificans/enzymology , Protein Subunits/chemistry , Ubiquinone/chemistry
19.
Br J Pharmacol ; 147(4): 437-45, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16402039

ABSTRACT

1. Bisphosphonates are currently the most important class of antiresorptive drugs used for the treatment of diseases with excess bone resorption. On the basis of their molecular mechanism of action, bisphosphonates can be divided into two pharmacological classes; nitrogen-containing (N-BPs) and non-nitrogen-containing bisphosphonates (non-N-BP). Both classes induce apoptosis but they evoke it differently; N-BPs by inhibiting the intracellular mevalonate pathway and protein isoprenylation, and non-N-BPs via cytotoxic ATP analog-type metabolites. N-BPs are not metabolized to ATP analogs, but we report here that these bisphosphonates can induce formation of a novel ATP analog (ApppI) as a consequence of the inhibition of the mevalonate pathway in cells. We also investigated whether ApppI is involved in the apoptosis induced by N-BPs. 2. Mass spectrometry and NMR were used to identify ApppI in N-BP treated osteoclasts, macrophages and glioma cells. The potency of different bisphosphonates to promote ApppI production was tested in J774 macrophages. The effects of ApppI on ADP/ATP translocase in isolated mitochondria and its capability to induce apoptosis in osteoclasts were also studied. 3. ApppI production correlated well with the capacity of N-BPs to inhibit mevalonate pathway. ApppI inhibited the mitochondrial ADP/ATP translocase and caused apoptosis in osteoclasts. 4. In conclusion, these findings provide the basis for a new mechanism of action for N-BPs. Some of these very potent bisphosphonates, such as zoledronic acid, represent a third class of bisphosphonates that can act both via the inhibition of the mevalonate pathway and by the blockade of mitochondrial ADP/ATP translocase, which is known to be involved in the induction of apoptosis.


Subject(s)
Adenine Nucleotide Translocator 1/antagonists & inhibitors , Adenosine Triphosphate/analogs & derivatives , Apoptosis/drug effects , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Mitochondria, Liver/drug effects , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Diphosphonates/chemistry , Mitochondria, Liver/enzymology , Mitochondria, Liver/physiology , Nuclear Magnetic Resonance, Biomolecular , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoclasts/physiology , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization
20.
J Mol Med (Berl) ; 83(10): 786-94, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142472

ABSTRACT

Complex I has a vital role in the energy production of the cell, and the clinical spectrum of complex I deficiency varies from severe lactic acidosis in infants to muscle weakness in adults. It has been estimated that the cause of complex I deficiency, especially in children, is often a mutation in the nuclear-encoded genes and, more rarely, in the genes encoded by mitochondrial DNA. We sequenced nine complex I subunit coding genes, NDUFAB1, NDUFS1, NDUFS2, NDUFS3, NDUFS4, NDUFS7, NDUFS8, NDUFV1 and NDUFV2, in 13 children with defined complex I deficiency. Two novel substitutions were found: a synonymous replacement 201A>T in NDUFV2 and a non-synonymous base exchange 52C>T in NDUFS8. The 52C>T substitution produced the replacement Arg18Cys in the leading peptide of the TYKY subunit. This novel missense mutation was found as a heterozygote in one patient and her mother, but not among 202 healthy controls nor among 107 children with undefined encephalomyopathy. Bioinformatic analyses suggested that Arg18Cys could lead to marked changes in the physicochemical properties of the mitochondrial-targeting peptide of TYKY, but we could not see changes in the assembly or activity of complex I or in the transcription of NDUFS8 in the fibroblasts of our patient. We suggest that Arg18Cys in the leading peptide of the TYKY subunit is not solely pathogenic, and that other genetic factors contribute to the disease-causing potential of this mutation.


Subject(s)
Electron Transport Complex I/genetics , Genetic Variation , Mitochondrial Encephalomyopathies/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , Alleles , Amino Acid Substitution , Arginine/chemistry , Arginine/genetics , Child , Computational Biology , Conserved Sequence , Cysteine/chemistry , Cysteine/genetics , Electron Transport Complex I/deficiency , Humans , Mitochondrial Encephalomyopathies/enzymology , Mutation , NAD(P)H Dehydrogenase (Quinone)/deficiency , NADH Dehydrogenase , Protein Subunits/deficiency , Protein Subunits/genetics , Sequence Analysis, DNA , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...