Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Trace Elem Med Biol ; 66: 126759, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33872833

ABSTRACT

BACKGROUND: Acquisition and distribution of zinc supports a number of biological processes. Various molecular factors are involved in zinc metabolism but not fully explored. BASIC PROCEDURES: Spontaneous mutants were generated in yeast with excess zinc culture followed by whole genome DNA sequencing to discover zinc metabolism related genes by bioinformatics. An identified mutant was characterized through metallomic and molecular biology methods. MAIN FINDINGS: Here we reported that MTM1 knockout cells displayed much stronger zinc tolerance than wild type cells on SC medium when exposed to excess zinc. Zn accumulation of mtm1Δ cells was dramatically decreased compared to wild type cells under excessive zinc condition due to MTM1 deletion reduced zinc uptake. ZRC1 mRNA level of mtm1Δ cells was significantly higher than that in the wild-type strain leading to increased vacuolar zinc accumulations in mtm1Δ cells. The mRNA levels of ZRT1 and ZAP1 decreased in mtm1Δ cells contributing to less Zn uptake. The zrc1Δmtm1Δ double knockout strain exhibited Zn sensitivity. MTM1 knockout did not afford resistance to excess zinc through an effect mediated through an influence on levels of ROS. Superoxide dismutase 2 (Sod2p) activity in mtm1Δ cells was severely impaired and not restored through Zn supplementation. Meanwhile, additional Zn showed no significant effect on the localization and expression of Mtm1p. PRINCIPAL CONCLUSIONS: Our study reveals the MTM1 gene plays an important role in the regulation of zinc homeostasis in yeast cells via changing zinc uptake and distribution. This discovery provides new insights for better understanding biochemical communication between vacuole and mitochondrial in relation to zinc-metabolism.


Subject(s)
Carrier Proteins/metabolism , Mitochondrial Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Zinc/metabolism , Carrier Proteins/genetics , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/genetics
2.
Mol Genet Metab ; 132(2): 146-153, 2021 02.
Article in English | MEDLINE | ID: mdl-33485800

ABSTRACT

TRMU is a nuclear gene crucial for mitochondrial DNA translation by encoding tRNA 5-methylaminomethyl-2-thiouridylate methyltransferase, which thiolates mitochondrial tRNA. Biallelic pathogenic variants in TRMU are associated with transient infantile liver failure. Other less common presentations such as Leigh syndrome, myopathy, and cardiomyopathy have been reported. Recent studies suggested that provision of exogenous L-cysteine or N-acetylcysteine may ameliorate the effects of disease-causing variants and improve the natural history of the disease. Here, we report six infants with biallelic TRMU variants, including four previously unpublished patients, all treated with exogenous cysteine. We highlight the first report of an affected patient undergoing orthotopic liver transplantation, the long-term effects of cysteine supplementation, and the ability of the initial presentation to mimic multiple inborn errors of metabolism. We propose that TRMU deficiency should be suspected in all children presenting with persistent lactic acidosis and hypoglycemia, and that combined N-acetylcysteine and L-cysteine supplementation should be considered prior to molecular diagnosis, as this is a low-risk approach that may increase survival and mitigate the severity of the disease course.


Subject(s)
Leigh Disease/therapy , Liver Failure/therapy , Mitochondrial Proteins/genetics , Protein Biosynthesis , tRNA Methyltransferases/genetics , Acetylcysteine/administration & dosage , Acetylcysteine/metabolism , Acidosis/genetics , Acidosis/metabolism , Cysteine/administration & dosage , Cysteine/metabolism , DNA, Mitochondrial/genetics , Female , Humans , Infant , Leigh Disease/genetics , Leigh Disease/metabolism , Leigh Disease/pathology , Liver Failure/genetics , Liver Failure/metabolism , Liver Failure/pathology , Liver Transplantation/methods , Male , Mitochondria/enzymology , Mitochondrial Proteins/deficiency , RNA, Transfer/genetics , tRNA Methyltransferases/deficiency
3.
Sci Rep ; 9(1): 12651, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477743

ABSTRACT

Ethylmalonic encephalopathy protein 1 (ETHE1) and molybdenum cofactor (MoCo) deficiencies are hereditary disorders that affect the catabolism of sulfur-containing amino acids. ETHE1 deficiency is caused by mutations in the ETHE1 gene, while MoCo deficiency is due to mutations in one of three genes involved in MoCo biosynthesis (MOCS1, MOCS2 and GPHN). Patients with both disorders exhibit abnormalities of the mitochondrial respiratory chain, among other biochemical findings. However, the pathophysiology of the defects has not been elucidated. To characterize cellular derangements, mitochondrial bioenergetics, dynamics, endoplasmic reticulum (ER)-mitochondria communication, superoxide production and apoptosis were evaluated in fibroblasts from four patients with ETHE1 deficiency and one with MOCS1 deficiency. The effect of JP4-039, a promising mitochondrial-targeted antioxidant, was also tested on cells. Our data show that mitochondrial respiration was decreased in all patient cell lines. ATP depletion and increased mitochondrial mass was identified in the same cells, while variable alterations in mitochondrial fusion and fission were seen. High superoxide levels were found in all cells and were decreased by treatment with JP4-039, while the respiratory chain activity was increased by this antioxidant in cells in which it was impaired. The content of VDAC1 and IP3R, proteins involved in ER-mitochondria communication, was decreased, while DDIT3, a marker of ER stress, and apoptosis were increased in all cell lines. These data demonstrate that previously unrecognized broad disturbances of cellular function are involved in the pathophysiology of ETHE1 and MOCS1 deficiencies, and that reduction of mitochondrial superoxide by JP4-039 is a promising strategy for adjuvant therapy of these disorders.


Subject(s)
Carbon-Carbon Lyases/deficiency , Endoplasmic Reticulum/metabolism , Energy Metabolism , Fibroblasts/pathology , Homeostasis , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/deficiency , Nucleocytoplasmic Transport Proteins/deficiency , Adenosine Triphosphate/biosynthesis , Apoptosis , Carbon-Carbon Lyases/metabolism , Cell Line , Cell Respiration , DNA Mutational Analysis , Fibroblasts/metabolism , Humans , Mitochondrial Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Oxidation-Reduction , Oxygen Consumption , Superoxides/metabolism
4.
Hum Mutat ; 39(1): 69-79, 2018 01.
Article in English | MEDLINE | ID: mdl-29044765

ABSTRACT

Primary coenzyme Q10 (CoQ10 ; MIM# 607426) deficiencies are an emerging group of inherited mitochondrial disorders with heterogonous clinical phenotypes. Over a dozen genes are involved in the biosynthesis of CoQ10 , and mutations in several of these are associated with human disease. However, mutations in COQ5 (MIM# 616359), catalyzing the only C-methylation in the CoQ10 synthetic pathway, have not been implicated in human disease. Here, we report three female siblings of Iraqi-Jewish descent, who had varying degrees of cerebellar ataxia, encephalopathy, generalized tonic-clonic seizures, and cognitive disability. Whole-exome and subsequent whole-genome sequencing identified biallelic duplications in the COQ5 gene, leading to reduced levels of CoQ10 in peripheral white blood cells of all affected individuals and reduced CoQ10 levels in the only muscle tissue available from one affected proband. CoQ10 supplementation led to clinical improvement and increased the concentrations of CoQ10 in blood. This is the first report of primary CoQ10 deficiency caused by loss of function of COQ5, with delineation of the clinical, laboratory, histological, and molecular features, and insights regarding targeted treatment with CoQ10 supplementation.


Subject(s)
Biosynthetic Pathways/genetics , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/genetics , Methyltransferases/deficiency , Mitochondrial Encephalomyopathies/diagnosis , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Proteins/deficiency , Ubiquinone/analogs & derivatives , Biopsy , Cerebellar Ataxia/diet therapy , Cerebellar Ataxia/metabolism , DNA Copy Number Variations , Dietary Supplements , Electron Transport , Female , Fibroblasts/metabolism , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Leukocytes/metabolism , Methyltransferases/genetics , Mitochondrial Encephalomyopathies/diet therapy , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Proteins/genetics , Muscles/pathology , Oxygen Consumption , Pedigree , Polymorphism, Single Nucleotide , Siblings , Ubiquinone/biosynthesis
5.
Mol Genet Metab ; 121(3): 216-223, 2017 07.
Article in English | MEDLINE | ID: mdl-28552678

ABSTRACT

Coenzyme Q10 (CoQ10) is an essential cofactor of the mitochondrial oxidative phosphorylation (OXPHOS) system and its deficiency has important implications for several inherited metabolic disorders of childhood. The biosynthesis of CoQ10 is a complicated process, which involves at least 12 different enzymes. One of the metabolic intermediates that are formed during CoQ10 biosynthesis is the molecule 6-demethoxyubiquinone (6-DMQ). This CoQ precursor is processed at the level of COQ7 and COQ9. We selected this metabolite as a marker substance for metabolic analysis of cell lines with inherited genetic defects (COQ2, COQ4, COQ7 and COQ9) or siRNA knockdown in CoQ biosynthesis enzymes using ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS). In COQ4, COQ7 and COQ9 deficient cell lines, we detected significantly elevated levels of 6-DMQ. This suggests a functional interplay of these proteins. However, additional siRNA studies demonstrated that elevated 6-DMQ levels are not an exclusive marker of the COQ7/COQ9 enzymatic step of CoQ10 biosynthesis but constitute a more general phenomenon that occurs in disorders impairing the function or stability of the CoQ-synthome. To further investigate the interdependence of CoQ10 biosynthesis enzyme expression, we performed immunoblotting in various cell lines with CoQ10 deficiency, indicating that COQ4, COQ7 and COQ9 protein expression levels are highly regulated depending on the underlying defect. Supplementation of cell lines with synthetic CoQ precursor compounds demonstrated beneficial effects of 2,4-dihydroxybenzoic acid in COQ7 and COQ9 deficiency. Moreover, vanillic acid selectively stimulated CoQ10 biosynthesis and improved cell viability in COQ9 deficiency. However, compounds tested in this study failed to rescue COQ4 deficiency.


Subject(s)
Ataxia/metabolism , Mitochondrial Diseases/metabolism , Muscle Weakness/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Cell Line , Cell Survival/drug effects , Fibroblasts/drug effects , Humans , Hydroxybenzoates/pharmacology , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Tandem Mass Spectrometry , Ubiquinone/biosynthesis , Ubiquinone/metabolism , Vanillic Acid/pharmacology
6.
Metab Eng ; 43(Pt B): 198-207, 2017 09.
Article in English | MEDLINE | ID: mdl-27856334

ABSTRACT

The mitochondrial citrate transport protein (CTP), encoded by SLC25A1, accommodates bidirectional trafficking of citrate between the mitochondria and cytosol, supporting lipid biosynthesis and redox homeostasis. Genetic CTP deficiency causes a fatal neurodevelopmental syndrome associated with the accumulation of L- and D-2-hydroxyglutaric acid, and elevated CTP expression is associated with poor prognosis in several types of cancer, emphasizing the importance of this transporter in multiple human pathologies. Here we describe the metabolic consequences of CTP deficiency in cancer cells. As expected from the phenotype of CTP-deficient humans, somatic CTP loss in cancer cells induces broad dysregulation of mitochondrial metabolism, resulting in accumulation of lactate and of the L- and D- enantiomers of 2-hydroxyglutarate (2HG) and depletion of TCA cycle intermediates. It also eliminates mitochondrial import of citrate from the cytosol. To quantify the impact of CTP deficiency on metabolic flux, cells were cultured with a set of 13C-glucose and 13C-glutamine tracers with resulting data integrated by metabolic flux analysis (MFA). CTP-deficient cells displayed a major restructuring of central carbon metabolism, including suppression of pyruvate dehydrogenase (PDH) and induction of glucose-dependent anaplerosis through pyruvate carboxylase (PC). We also observed an unusual lipogenic pathway in which carbon from glucose supplies mitochondrial production of alpha-ketoglutarate (AKG), which is then trafficked to the cytosol and used to supply reductive carboxylation by isocitrate dehydrogenase 1 (IDH1). The resulting citrate is cleaved to produce lipogenic acetyl-CoA, thereby completing a novel pathway of glucose-dependent reductive carboxylation. In CTP deficient cells, IDH1 inhibition suppresses lipogenesis from either glucose or glutamine, implicating IDH1 as a required component of fatty acid synthesis in states of CTP deficiency.


Subject(s)
Anion Transport Proteins/deficiency , Fatty Acids/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Neoplasm Proteins , Neoplasms/metabolism , Cell Line, Tumor , Fatty Acids/genetics , Humans , Mitochondria/genetics , Mitochondria/pathology , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Organic Anion Transporters
7.
Mol Cell ; 63(4): 608-620, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499294

ABSTRACT

The UbiB protein kinase-like (PKL) family is widespread, comprising one-quarter of microbial PKLs and five human homologs, yet its biochemical activities remain obscure. COQ8A (ADCK3) is a mammalian UbiB protein associated with ubiquinone (CoQ) biosynthesis and an ataxia (ARCA2) through unclear means. We show that mice lacking COQ8A develop a slowly progressive cerebellar ataxia linked to Purkinje cell dysfunction and mild exercise intolerance, recapitulating ARCA2. Interspecies biochemical analyses show that COQ8A and yeast Coq8p specifically stabilize a CoQ biosynthesis complex through unorthodox PKL functions. Although COQ8 was predicted to be a protein kinase, we demonstrate that it lacks canonical protein kinase activity in trans. Instead, COQ8 has ATPase activity and interacts with lipid CoQ intermediates, functions that are likely conserved across all domains of life. Collectively, our results lend insight into the molecular activities of the ancient UbiB family and elucidate the biochemical underpinnings of a human disease.


Subject(s)
Behavior, Animal , Cerebellar Ataxia/enzymology , Cerebellum/enzymology , Mitochondrial Proteins/deficiency , Muscle, Skeletal/enzymology , Ubiquinone/deficiency , Animals , COS Cells , Cerebellar Ataxia/genetics , Cerebellar Ataxia/physiopathology , Cerebellar Ataxia/psychology , Cerebellum/physiopathology , Cerebellum/ultrastructure , Chlorocebus aethiops , Disease Models, Animal , Exercise Tolerance , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Lipid Metabolism , Male , Maze Learning , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Models, Molecular , Motor Activity , Muscle Strength , Muscle, Skeletal/physiopathology , Phenotype , Protein Binding , Protein Conformation , Proteomics/methods , Recognition, Psychology , Rotarod Performance Test , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Seizures/enzymology , Seizures/genetics , Seizures/physiopathology , Structure-Activity Relationship , Time Factors , Transfection , Ubiquinone/chemistry , Ubiquinone/genetics
8.
Clin Genet ; 90(2): 156-60, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26818466

ABSTRACT

Inherited ataxias are a group of heterogeneous disorders in children or adults but their genetic definition remains still undetermined in almost half of the patients. However, CoQ10 deficiency is a rare cause of cerebellar ataxia and ADCK3 is the most frequent gene associated with this defect. We herein report a 48 year old man, who presented with dysarthria and walking difficulties. Brain magnetic resonance imaging showed a marked cerebellar atrophy. Serum lactate was elevated. Tissues obtained by muscle and skin biopsies were studied for biochemical and genetic characterization. Skeletal muscle biochemistry revealed decreased activities of complexes I+III and II+III and a severe reduction of CoQ10 , while skin fibroblasts showed normal CoQ10 levels. A mild loss of maximal respiration capacity was also found by high-resolution respirometry. Molecular studies identified a novel homozygous deletion (c.504del_CT) in ADCK3, causing a premature stop codon. Western blot analysis revealed marked reduction of ADCK3 protein levels. Treatment with CoQ10 was started and, after 1 year follow-up, patient neurological condition slightly improved. This report suggests the importance of investigating mitochondrial function and, in particular, muscle CoQ10 levels, in patients with adult-onset cerebellar ataxia. Moreover, clinical stabilization by CoQ10 supplementation emphasizes the importance of an early diagnosis.


Subject(s)
Ataxia/genetics , Cerebellar Ataxia/genetics , Codon, Nonsense , Electron Transport Chain Complex Proteins/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscle Weakness/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Ataxia/complications , Ataxia/diagnosis , Ataxia/physiopathology , Cerebellar Ataxia/complications , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/physiopathology , Delayed Diagnosis , Electron Transport Chain Complex Proteins/deficiency , Fibroblasts/metabolism , Gene Expression , Homozygote , Humans , Lactic Acid/blood , Magnetic Resonance Imaging , Male , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/complications , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/physiopathology , Mitochondrial Proteins/deficiency , Muscle Weakness/complications , Muscle Weakness/diagnosis , Muscle Weakness/physiopathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Skin/metabolism , Ubiquinone/genetics
9.
Cell Rep ; 13(1): 8-14, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26411686

ABSTRACT

The hypothalamus has been implicated in skeletal metabolism. Whether hunger-promoting neurons of the arcuate nucleus impact the bone is not known. We generated multiple lines of mice to affect AgRP neuronal circuit integrity. We found that mice with Ucp2 gene deletion, in which AgRP neuronal function was impaired, were osteopenic. This phenotype was rescued by cell-selective reactivation of Ucp2 in AgRP neurons. When the AgRP circuitry was impaired by early postnatal deletion of AgRP neurons or by cell autonomous deletion of Sirt1 (AgRP-Sirt1(-/-)), mice also developed reduced bone mass. No impact of leptin receptor deletion in AgRP neurons was found on bone homeostasis. Suppression of sympathetic tone in AgRP-Sirt1(-/-) mice reversed osteopenia in transgenic animals. Taken together, these observations establish a significant regulatory role for AgRP neurons in skeletal bone metabolism independent of leptin action.


Subject(s)
Agouti-Related Protein/genetics , Bone Density/drug effects , Bone Diseases, Metabolic/metabolism , Femur/metabolism , Propranolol/pharmacology , Tibia/metabolism , Agouti-Related Protein/deficiency , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/pathology , Femur/drug effects , Femur/pathology , Gene Expression Regulation , Homeostasis , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Ion Channels/deficiency , Ion Channels/genetics , Leptin/genetics , Leptin/metabolism , Male , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Norepinephrine/metabolism , Phenotype , Receptors, Adrenergic, beta/genetics , Receptors, Adrenergic, beta/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction , Sirtuin 1/deficiency , Sirtuin 1/genetics , Tibia/drug effects , Tibia/pathology , Uncoupling Protein 2
10.
J Clin Invest ; 123(11): 4667-80, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24084737

ABSTRACT

Type 2 diabetes is characterized by insulin resistance and mitochondrial dysfunction in classical target tissues such as muscle, fat, and liver. Using a murine model of type 2 diabetes, we show that there is hypothalamic insulin resistance and mitochondrial dysfunction due to downregulation of the mitochondrial chaperone HSP60. HSP60 reduction in obese, diabetic mice was due to a lack of proper leptin signaling and was restored by leptin treatment. Knockdown of Hsp60 in a mouse hypothalamic cell line mimicked the mitochondrial dysfunction observed in diabetic mice and resulted in increased ROS production and insulin resistance, a phenotype that was reversed with antioxidant treatment. Mice with a heterozygous deletion of Hsp60 exhibited mitochondrial dysfunction and hypothalamic insulin resistance. Targeted acute downregulation of Hsp60 in the hypothalamus also induced insulin resistance, indicating that mitochondrial dysfunction can cause insulin resistance in the hypothalamus. Importantly, type 2 diabetic patients exhibited decreased expression of HSP60 in the brain, indicating that this mechanism is relevant to human disease. These data indicate that leptin plays an important role in mitochondrial function and insulin sensitivity in the hypothalamus by regulating HSP60. Moreover, leptin/insulin crosstalk in the hypothalamus impacts energy homeostasis in obesity and insulin-resistant states.


Subject(s)
Chaperonin 60/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypothalamus/metabolism , Insulin/metabolism , Leptin/metabolism , Mitochondrial Proteins/metabolism , Animals , Cell Line , Chaperonin 60/deficiency , Chaperonin 60/genetics , Female , Gene Knockdown Techniques , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Obesity/metabolism , Oxidative Stress , Signal Transduction
11.
Arterioscler Thromb Vasc Biol ; 33(9): 2154-61, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23825363

ABSTRACT

OBJECTIVE: On the basis of previous evidence that polymerase delta interacting protein 2 (Poldip2) increases reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (Nox4) activity in vascular smooth muscle cells, we hypothesized that in vivo knockdown of Poldip2 would inhibit reactive oxygen species production and alter vascular function. APPROACH AND RESULTS: Because homozygous Poldip2 deletion is lethal, Poldip2(+/-) mice were used. Poldip2 mRNA and protein levels were reduced by ≈50% in Poldip2(+/-) aorta, with no change in p22phox, Nox1, Nox2, and Nox4 mRNAs. NADPH oxidase activity was also inhibited in Poldip2(+/-) tissue. Isolated aortas from Poldip2(+/-) mice demonstrated impaired phenylephrine and potassium chloride-induced contractions, increased stiffness, and reduced compliance associated with disruption of elastic lamellae and excessive extracellular matrix deposition. Collagen I secretion was elevated in cultured vascular smooth muscle cells from Poldip2(+/-) mice and restored by H2O2 supplementation, suggesting that this novel function of Poldip2 is mediated by reactive oxygen species. Furthermore, Poldip2(+/-) mice were protected against aortic dilatation in a model of experimental aneurysm, an effect consistent with increased collagen secretion. CONCLUSIONS: Poldip2 knockdown reduces H2O2 production in vivo, leading to increases in extracellular matrix, greater vascular stiffness, and impaired agonist-mediated contraction. Thus, unaltered expression of Poldip2 is necessary for vascular integrity and function.


Subject(s)
Aorta/metabolism , Aortic Aneurysm/prevention & control , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Aorta/physiopathology , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Aortic Aneurysm/physiopathology , Blood Pressure , Cells, Cultured , Collagen Type I/metabolism , Cytochrome b Group/metabolism , Dilatation, Pathologic , Disease Models, Animal , Dose-Response Relationship, Drug , Elastic Tissue/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Genotype , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Myocytes, Smooth Muscle/metabolism , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Oxidants/pharmacology , Phenotype , RNA, Messenger/metabolism , Vascular Stiffness , Vasoconstrictor Agents/pharmacology , Vasodilation
12.
J Cell Biol ; 199(2): 215-24, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-23045551

ABSTRACT

Mclk1 (also known as Coq7) and Coq3 code for mitochondrial enzymes implicated in the biosynthetic pathway of ubiquinone (coenzyme Q or UQ). Mclk1(+/-) mice are long-lived but have dysfunctional mitochondria. This phenotype remains unexplained, as no changes in UQ content were observed in these mutants. By producing highly purified submitochondrial fractions, we report here that Mclk1(+/-) mice present a unique mitochondrial UQ profile that was characterized by decreased UQ levels in the inner membrane coupled with increased UQ in the outer membrane. Dietary-supplemented UQ(10) was actively incorporated in both mitochondrial membranes, and this was sufficient to reverse mutant mitochondrial phenotypes. Further, although homozygous Coq3 mutants die as embryos like Mclk1 homozygous null mice, Coq3(+/-) mice had a normal lifespan and were free of detectable defects in mitochondrial function or ubiquinone distribution. These findings indicate that MCLK1 regulates both UQ synthesis and distribution within mitochondrial membranes.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxygen Consumption/genetics , Ubiquinone/metabolism , Animals , Cell Respiration/genetics , Male , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/deficiency , Mixed Function Oxygenases , Submitochondrial Particles/metabolism , Ubiquinone/administration & dosage , Ubiquinone/genetics
13.
Am J Physiol Gastrointest Liver Physiol ; 302(3): G336-42, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22094601

ABSTRACT

Steatotic livers are sensitive to ischemic events and associated ATP depletion. Hepatocellular necrosis following these events may result from mitochondrial uncoupling protein-2 (UCP2) expression. To test this hypothesis, we developed a model of in vitro steatosis using primary hepatocytes from wild-type (WT) and UCP2 knockout (KO) mice and subjected them to hypoxia/reoxygenation (H/R). Using cultured hepatocytes treated with emulsified fatty acids for 24 h, generating a steatotic phenotype (i.e., microvesicular and broad-spectrum fatty acid accumulation), we found that the phenotype of the WT and UCP2 KO were the same; however, cellular viability was increased in the steatotic KO hepatocytes following 4 h of hypoxia and 24 h of reoxygenation; Hepatocellular ATP levels decreased during hypoxia and recovered after reoxygenation in the control and UCP2 KO steatotic hepatocytes but not in the WT steatotic hepatocytes; mitochondrial membrane potential in WT and UCP2 KO steatotic groups was less than control groups but higher than UCP2 KO hepatocytes. Following reoxygenation, lipid peroxidation, as measured by thiobarbituric acid reactive substances, increased in all groups but to a greater extent in the steatotic hepatocytes, regardless of UCP2 expression. These results demonstrate that UCP2 sensitizes steatotic hepatocytes to H/R through mitochondrial depolarization and ATP depletion but not lipid peroxidation.


Subject(s)
Cell Hypoxia/physiology , Fatty Liver , Hepatocytes/pathology , Ion Channels/deficiency , Mitochondrial Proteins/deficiency , Oxygen/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Emulsions/pharmacology , Fatty Acids/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred Strains , Mice, Knockout , Mice, Obese , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phospholipids/pharmacology , Soybean Oil/pharmacology , Uncoupling Protein 2
14.
Integr Biol (Camb) ; 3(11): 1135-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22005712

ABSTRACT

The aberrant expression and functional activity of proteins involved in ATP production pathways may cause a crisis in energy generation for cells and compromise their survival under stressful conditions such as excitation, starvation, pharmacological treatment or disease states. Under resting conditions such defects are often compensated for, and therefore masked by, alternative pathways which have significant spare capacity. Here we present a multiplexed 'cell energy budget' platform which facilitates metabolic assessment and cross-comparison of different cells and the identification of genes directly or indirectly involved in ATP production. Long-decay emitting O(2) and pH sensitive probes and time-resolved fluorometry are used to measure changes in cellular O(2) consumption, glycolytic and total extracellular acidification (ECA), along with the measurement of total ATP and protein content in multiple samples. To assess the extent of spare capacity in the main energy pathways, the cells are also analysed following double-treatment with carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone and oligomycin. The four-parametric platform operating in a high throughput format has been validated with two panels of transformed cells: mouse embryonic fibroblasts (MEFs) lacking the Krebs cycle enzyme fumarate hydratase (Fh1) and HeLa cells with reduced expression of pyrimidine nucleotide carrier 1. In both cases, a marked reduction in both respiration and spare respiratory capacity was observed, accompanied by a compensatory activation of glycolysis and consequent maintenance of total ATP levels. At the same time, in Fh1-deficient MEFs the contribution of non-glycolytic pathways to the ECA did not change.


Subject(s)
Energy Metabolism/physiology , Gene Knockout Techniques , RNA Interference/physiology , Adenosine Triphosphate/metabolism , Animals , Carbon Dioxide/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Respiration/drug effects , Cell Respiration/physiology , Citric Acid Cycle/physiology , Embryo, Mammalian/cytology , Energy Metabolism/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Fibroblasts/metabolism , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Gene Deletion , Glycolysis/physiology , HeLa Cells , Humans , Hydrogen-Ion Concentration/drug effects , Lactic Acid/metabolism , Mice , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Nucleotide Transport Proteins/genetics , Oligomycins/pharmacology , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , RNA, Small Interfering/genetics
15.
Transplant Proc ; 40(10): 3327-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19100382

ABSTRACT

Steatotic livers represent a growing proportion of marginal organs available for transplantation. These livers are highly prone to primary nonfunction following transplantation and are therefore routinely turned down for surgery. Given the elevated levels and sensitivity for reactive oxygen species (ROS) in these livers, we evaluated whether pretreatment with a targeted ROS scavenger, vitamin E succinate, increased survival and decreased injury after ischemia/reperfusion (I/R). For this study, ob/ob mice received 50 IU/d vitamin E succinate in supplemented vs control chow for 7 days, and were subjected to 15 minutes of total hepatic ischemia and 24 hours of reperfusion. Treatment resulted in a 5-fold decrease in serum alanine aminotransferase (ALT) levels after reperfusion, mirrored by significant decreases in hepatocellular necrosis. These results suggested that targeted antioxidants such as vitamin E succinate may prove to be highly applicable for the pretreatment of steatotic donor livers, increasing their tolerance for I/R and the transplantation process.


Subject(s)
Liver/pathology , Reperfusion Injury/prevention & control , Tocopherols/therapeutic use , Animals , Ion Channels/deficiency , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mitochondrial Proteins/deficiency , Necrosis , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Uncoupling Protein 2
16.
Am J Physiol Endocrinol Metab ; 294(3): E600-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18089762

ABSTRACT

Uncoupling protein 2 (UCP2) is a possible target molecule for energy dissipation. Many dietary fats, including safflower oil and lard, induce obesity in C57BL/6 mice, whereas fish oil does not. Fish oil increases UCP2 expression in hepatocytes and may enhance UCP2 activity by activating the UCP2 molecule or altering the lipid bilayer environment. To examine the role of liver UCP2 in obesity, we created transgenic mice that overexpressed human UCP2 in hepatocytes and examined whether UCP2 transgenic mice showed less obesity when fed a high-fat diet (safflower oil or lard). In addition, we examined whether fish oil had antiobesity effects in UCP2 knockout mice. UCP2 transgenic and wild-type mice fed a high-fat diet (safflower oil or lard) developed obesity to a similar degree. UCP2 knockout and wild-type mice fed fish oil had lower rates of obesity than mice fed safflower oil. Remarkably, safflower oil did not induce obesity in female UCP2 knockout mice, an unexpected phenotype for which we presently have no explanation. However, this unexpected effect was not observed in male UCP2 knockout mice or in UCP2 knockout mice fed a high-lard diet. These data indicate that liver UCP2 is not essential for fish oil-induced decreases in body fat.


Subject(s)
Anti-Obesity Agents/therapeutic use , Fish Oils/therapeutic use , Ion Channels/physiology , Mitochondrial Proteins/physiology , Obesity/therapy , Animals , Dietary Fats/administration & dosage , Female , Gene Expression , Ion Channels/deficiency , Ion Channels/genetics , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Obesity/etiology , RNA, Messenger/analysis , Safflower Oil/administration & dosage , Sex Characteristics , Uncoupling Protein 2
17.
Cell Metab ; 3(6): 417-27, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16753577

ABSTRACT

Uncoupling protein 2 (UCP2) negatively regulates insulin secretion. UCP2 deficiency (by means of gene knockout) improves obesity- and high glucose-induced beta cell dysfunction and consequently improves type 2 diabetes in mice. In the present study, we have discovered that the small molecule, genipin, rapidly inhibits UCP2-mediated proton leak. In isolated mitochondria, genipin inhibits UCP2-mediated proton leak. In pancreatic islet cells, genipin increases mitochondrial membrane potential, increases ATP levels, closes K(ATP) channels, and stimulates insulin secretion. These actions of genipin occur in a UCP2-dependent manner. Importantly, acute addition of genipin to isolated islets reverses high glucose- and obesity-induced beta cell dysfunction. Thus, genipin and/or chemically modified variants of genipin are useful research tools for studying biological processes thought to be controlled by UCP2. In addition, these agents represent lead compounds that comprise a starting point for the development of therapies aimed at treating beta cell dysfunction.


Subject(s)
Glucose/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/pharmacology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Membrane Transport Proteins/drug effects , Mitochondrial Proteins/drug effects , Obesity/metabolism , Pyrans/pharmacology , Adenosine Triphosphate/metabolism , Aldehydes/antagonists & inhibitors , Aldehydes/metabolism , Animals , Drugs, Chinese Herbal/pharmacology , Glucose/pharmacology , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/chemistry , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/physiology , Ion Channels , Iridoid Glycosides , Iridoids , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Mice, Obese , Mitochondria/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/metabolism , Molecular Conformation , Potassium Channels/drug effects , Potassium Channels/metabolism , Protons , Pyrans/chemistry , Uncoupling Protein 2
18.
J Org Chem ; 70(21): 8417-23, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16209586

ABSTRACT

A new class of highly fluorescent (phi(F) 0.3-0.8) low molecular weight water-soluble cholephilic compounds has been synthesized in two steps from dipyrrinones. The dipyrrinone nitrogens are first bridged by reaction with 1,1'-carbonyldiimidazole to form an N,N'-carbonyldipyrrinone (3H,5H-dipyrrolo[1,2-c:2',1'-f]pyrimidine-3,5-dione) nucleus, and a sulfonic acid group is then introduced at C(8) by reaction with concd H(2)SO(4). The resulting sulfonated N,N'-carbonyl-bridged dipyrrinones ("sulfoglows") are isolated as their sodium salts. When the alkyl substituents of the lactam ring are lengthened from ethyl to decyl, sulfoglows become increasingly lipophilic while maintaining water solubility. Low molecular weight sulfoglows were rapidly excreted intact in both bile and urine after intravenous infusion into rats, but higher molecular weight sulfoglows were excreted more selectively in bile. Hepatobiliary excretion of sulfoglows was partially, but not completely, blocked in mutant rats deficient in the multidrug-resistance associated transport protein Mrp2 (ABCC2). These observations point to the feasibility of developing simple sulfoglows with clinical diagnostic potential that are normally excreted in bile but appear in urine when hepatic elimination is impaired by cholestatic liver disease.


Subject(s)
Colchicine/analysis , Fluorescent Dyes/chemical synthesis , Liver Diseases/diagnosis , Liver/metabolism , Pyrimidinones/chemical synthesis , Pyrimidinones/pharmacokinetics , Pyrroles/chemical synthesis , Pyrroles/pharmacokinetics , Sulfonic Acids/chemical synthesis , Sulfonic Acids/pharmacokinetics , Animals , Bile/chemistry , Bile/metabolism , Bilirubin/analogs & derivatives , Bilirubin/chemistry , Colchicine/chemistry , Drug Evaluation, Preclinical , Feasibility Studies , Fluorescent Dyes/chemistry , Liver/drug effects , Liver Diseases/genetics , Liver Diseases/metabolism , Male , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Pyrimidinones/analysis , Pyrroles/analysis , Pyrroles/chemistry , Rats , Rats, Gunn , Rats, Sprague-Dawley , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Spectrometry, Fluorescence , Sulfonic Acids/analysis , Urine/chemistry
19.
Clin Lab ; 51(5-6): 289-306, 2005.
Article in English | MEDLINE | ID: mdl-15991803

ABSTRACT

Fatty acids are a major fuel for the body and fatty acid oxidation is particularly important during fasting, sustained aerobic exercise & stress. The myocardium and resting skeletal muscle utilise long-chain fatty acids as a major source of energy. Inherited disorders of fatty acid oxidation seriously compromise the function of muscle and other highly energy-dependent tissues such as brain, nerve, heart, kidney & liver. Defects of fatty acid oxidation lead to a range of neuromyopathic disease in both adults and children. Such defects encompass a wide spectrum of clinical disease, presenting in the neonatal period or infancy with recurrent hypoketotic hypoglycaemic encephalopathy, liver dysfunction and hyperammonaemia with neurosensory deficits secondary to the acute onset. In addition, there may be cardiac arrhythmias and/or progressive cardiomyopathy, which may give rise to secondary hypoxic-ischaemic encephalopathy. In older children, adolescence or adults there is often exercise intolerance with episodic myalgia or rhabdomyolysis in association with prolonged aerobic exercise or other exacerbating factors. Some disorders are particularly associated with toxic metabolites that may contribute to encephalopathy, polyneuropathy, axonopathy and pigmentary retinopathy. Diagnosis is through clinical suspicion with appropriate investigations in blood and urine taken during crisis. Definitive diagnosis is usually by fibroblast assay. Treatment is generally through avoidance of fasting, frequent carbohydrate rich feeds and in long-chain defects MCT supplementation. Novel treatments include the use of D,L-3-hydroxybutyrate and the potential use of fibrates to increase mutant protein levels in mild disorders.


Subject(s)
Brain Diseases, Metabolic/etiology , Fatty Acids/metabolism , Acyl-CoA Dehydrogenase/deficiency , Brain Diseases, Metabolic/therapy , Carnitine/biosynthesis , Carnitine/deficiency , Diet Therapy , Humans , Mitochondrial Proteins/deficiency , Oxidation-Reduction
20.
Mol Genet Metab ; 77(3): 202-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12409267

ABSTRACT

In an infant who suffered from prolonged icterus and hepatocellular dysfunction we detected an increase of citrulline and dibasic amino acids in plasma and urine. The amino acid levels along with all the abnormal liver tests normalized upon replacing breast-milk by formula feeding; there was no relapse after human milk was tentatively reintroduced. A novel mutation, a approximately 9.5-kb genomic duplication, was identified in the citrin gene (SLC25A13) resulting in the insertion of exon 15. No mutation was detected in the CAT2A specific exon of the SLC7A2 gene which encodes for the liver transporter of cationic amino acids. This is the first report of infantile citrin deficiency in non-Asian patients.


Subject(s)
Amino Acids, Diamino/metabolism , Citrullinemia/etiology , Fibroblasts/metabolism , Membrane Transport Proteins/deficiency , Mitochondrial Proteins/deficiency , DNA, Complementary , Humans , In Vitro Techniques , Infant , Male , Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL