Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Biochem J ; 478(3): 463-486, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33544126

ABSTRACT

NaCT/SLC13A5 is a Na+-coupled transporter for citrate in hepatocytes, neurons, and testes. It is also called mINDY (mammalian ortholog of 'I'm Not Dead Yet' in Drosophila). Deletion of Slc13a5 in mice leads to an advantageous phenotype, protecting against diet-induced obesity, and diabetes. In contrast, loss-of-function mutations in SLC13A5 in humans cause a severe disease, EIEE25/DEE25 (early infantile epileptic encephalopathy-25/developmental epileptic encephalopathy-25). The difference between mice and humans in the consequences of the transporter deficiency is intriguing but probably explainable by the species-specific differences in the functional features of the transporter. Mouse Slc13a5 is a low-capacity transporter, whereas human SLC13A5 is a high-capacity transporter, thus leading to quantitative differences in citrate entry into cells via the transporter. These findings raise doubts as to the utility of mouse models to evaluate NaCT biology in humans. NaCT-mediated citrate entry in the liver impacts fatty acid and cholesterol synthesis, fatty acid oxidation, glycolysis, and gluconeogenesis; in neurons, this process is essential for the synthesis of the neurotransmitters glutamate, GABA, and acetylcholine. Thus, SLC13A5 deficiency protects against obesity and diabetes based on what the transporter does in hepatocytes, but leads to severe brain deficits based on what the transporter does in neurons. These beneficial versus detrimental effects of SLC13A5 deficiency are separable only by the blood-brain barrier. Can we harness the beneficial effects of SLC13A5 deficiency without the detrimental effects? In theory, this should be feasible with selective inhibitors of NaCT, which work only in the liver and do not get across the blood-brain barrier.


Subject(s)
Symporters/deficiency , Animals , Blood-Brain Barrier , Bone and Bones/metabolism , Citric Acid/metabolism , Citric Acid Cycle/genetics , Dental Enamel/metabolism , Diabetes Mellitus/metabolism , Dicarboxylic Acid Transporters/antagonists & inhibitors , Dicarboxylic Acid Transporters/deficiency , Dicarboxylic Acid Transporters/physiology , Disease Models, Animal , Drosophila Proteins/physiology , Fatty Liver/metabolism , Female , Germ Cells/metabolism , Hepatocytes/metabolism , Humans , Infant, Newborn , Ion Transport , Longevity/genetics , Male , Mice , Mice, Knockout , Mutation , Neoplasms/metabolism , Neurons/metabolism , Protein Conformation , Spasms, Infantile/genetics , Species Specificity , Symporters/antagonists & inhibitors , Symporters/genetics , Symporters/physiology
2.
JCI Insight ; 6(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33491666

ABSTRACT

Reduced expression of the plasma membrane citrate transporter INDY (acronym I'm Not Dead, Yet) extends life span in lower organisms. Deletion of the mammalian Indy (mIndy) gene in rodents improves metabolism via mechanisms akin to caloric restriction, known to lower blood pressure (BP) by sympathoadrenal inhibition. We hypothesized that mIndy deletion attenuates sympathoadrenal support of BP. Continuous arterial BP and heart rate (HR) were reduced in mINDY-KO mice. Concomitantly, urinary catecholamine content was lower, and the decreases in BP and HR by mIndy deletion were attenuated after autonomic ganglionic blockade. Catecholamine biosynthesis pathways were reduced in mINDY-KO adrenals using unbiased microarray analysis. Citrate, the main mINDY substrate, increased catecholamine content in pheochromocytoma cells, while pharmacological inhibition of citrate uptake blunted the effect. Our data suggest that deletion of mIndy reduces sympathoadrenal support of BP and HR by attenuating catecholamine biosynthesis. Deletion of mIndy recapitulates beneficial cardiovascular and metabolic responses to caloric restriction, making it an attractive therapeutic target.


Subject(s)
Blood Pressure/genetics , Blood Pressure/physiology , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acid Transporters/physiology , Sympathoadrenal System/physiology , Symporters/genetics , Symporters/physiology , Adrenal Glands/anatomy & histology , Adrenal Glands/physiology , Animals , Caloric Restriction , Catecholamines/biosynthesis , Cell Line , Chromaffin Cells/physiology , Dicarboxylic Acid Transporters/deficiency , Gene Expression , Heart Rate/genetics , Heart Rate/physiology , Longevity/genetics , Longevity/physiology , Malates/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Cardiovascular , Motor Activity/genetics , Motor Activity/physiology , Pyridines/pharmacology , Symporters/deficiency
3.
Psychiatr Genet ; 31(1): 32-35, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33290383

ABSTRACT

SLC13A5/NaCT is a sodium-coupled citrate transporter expressed in the plasma membrane of the liver, testis, and brain. In these tissues, SLC13A5 has important functions in the synthesis of fatty acids, cholesterol, and neurotransmitters. In recent years, patients homozygous for recessive mutations in SLC13A5, known as SLC13A5 deficiency [early infantile epileptic encephalopathy-25 (EIEE-25)], exhibit severe global developmental delay, early-onset intractable seizures, spasticity, and amelogenesis imperfecta affecting tooth development. Although the pathogenesis of SLC13A5 deficiency remains not clearly understood, cytoplasmic citrate deficits, decreased energy status in neurons, and citrate-zinc chelation are hypothesized to explain the neurological deficits. However, no study has examined the possibility of specific pharmacological drugs and/or lifestyle changes synergizing with heterozygosity of SLC13A5 deficiency to increase the risk of EIEE-25 clinical phenotype. Here, we report on a heterozygous SLC13A5-deficient patient who demonstrated evidence of pharmaco-synergistic heterozygosity upon administration of metformin, valproic acid, and starvation. The report illustrates the importance of careful consideration of the potential adverse effects of specific pharmacological treatments in patients with heterozygosity for disease-causing recessive mutations in SLC13A5.


Subject(s)
Epilepsy/genetics , Metformin/adverse effects , Symporters/deficiency , Valproic Acid/adverse effects , Adult , Amino Acid Substitution , Ammonia/blood , Animals , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Autistic Disorder/genetics , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Citrates/blood , Dicarboxylic Acid Transporters/physiology , Drosophila Proteins/physiology , Epilepsy/blood , Epilepsy/chemically induced , Epilepsy/etiology , Female , Food Deprivation , Heterozygote , Humans , Lactates/blood , Longevity/genetics , Metformin/therapeutic use , Mice , Mutation, Missense , Point Mutation , Psychotic Disorders/drug therapy , Psychotic Disorders/genetics , Psychotropic Drugs/therapeutic use , Pyruvates/blood , Recurrence , Spasms, Infantile/genetics , Spasms, Infantile/metabolism , Symporters/genetics , Symporters/physiology , Tooth Abnormalities/genetics , Valproic Acid/therapeutic use
4.
J Invest Dermatol ; 140(10): 2032-2040.e1, 2020 10.
Article in English | MEDLINE | ID: mdl-32119868

ABSTRACT

Hyperpigmentary conditions can arise when melanogenesis in the epidermis is misregulated. Understanding the pathways underlying melanogenesis is essential for the development of effective treatments. Here, we report that a group of metabolites called polyamines are important in the control of melanogenesis in human skin. Polyamines are cationic molecules present in all cells and are essential for cellular function. We report that polyamine regulator ODC1 is upregulated in melanocytes from melasma lesional skin. We report that the polyamine putrescine can promote pigmentation in human skin explants and primary normal human epidermal melanocytes through induction of tyrosinase which is rate-limiting for the synthesis of melanin. Putrescine supplementation on normal human epidermal melanocytes results in the activation of polyamine catabolism, which results in increased intracellular H2O2. Polyamine catabolism is also increased in human skin explants that have been treated with putrescine. We further report that inhibition of polyamine catabolism prevents putrescine-induced promotion of tyrosinase levels and pigmentation in normal human epidermal melanocytes, showing that polyamine catabolism is responsible for the putrescine induction of melanogenesis. Our data showing that putrescine promotes pigmentation has important consequences for hyperpigmented and hypopigmented conditions. Further understanding of how polyamines control epidermal pigmentation could open the door for the development of new therapeutics.


Subject(s)
Epidermis/drug effects , Melanins/biosynthesis , Putrescine/pharmacology , Biogenic Polyamines/metabolism , Cells, Cultured , Dicarboxylic Acid Transporters/physiology , Epidermis/metabolism , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Middle Aged , Mitochondrial Membrane Transport Proteins/physiology , Putrescine/analogs & derivatives , Skin Pigmentation/drug effects
5.
J Biosci Bioeng ; 125(5): 505-512, 2018 May.
Article in English | MEDLINE | ID: mdl-29395959

ABSTRACT

Enterobacter aerogenes, a gram-negative, rod-shaped bacterium, is an effective producer of succinate from glucose via the reductive tricarboxylic acid cycle under anaerobic conditions. However, to date, succinate-exporter genes have not been identified in E. aerogenes, although succinate exporters have a large impact on fermentative succinate production. Recently, we genetically identified yjjP and yjjB, as genes encoding a succinate transporter in Escherichia coli. Evaluation of the yjjPB homologs in E. aerogenes (EayjjPB genes) showed that succinate accumulation increased from 4.1 g L-1 to 9.1 g L-1 when the EayjjPB genes were expressed under aerobic conditions. Under anaerobic conditions, succinate yield increased from 53% to 60% by EayjjPB expression and decreased to 48% by deletion of EayjjPB. Furthermore, the production levels of fumarate and malate, which are intermediates of the succinate-biosynthesis pathway, were also increased by EayjjPB expression. A complementation assay conducted in Corynebacterium glutamicum strain AJ110655ΔsucE1 demonstrated that both EaYjjP and EaYjjB are required for the restoration of succinate production. Taken together, these results suggest that EaYjjPB function as a dicarboxylate transporter in E. aerogenes and that the products of both genes are required for dicarboxylate transport.


Subject(s)
Bacteriological Techniques/methods , Cloning, Molecular/methods , Dicarboxylic Acid Transporters/genetics , Enterobacter aerogenes/genetics , Enterobacter aerogenes/metabolism , Succinic Acid/metabolism , Aerobiosis/genetics , Anaerobiosis/genetics , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/metabolism , Dicarboxylic Acid Transporters/isolation & purification , Dicarboxylic Acid Transporters/physiology , Enterobacter aerogenes/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Fermentation , Metabolic Engineering , Metabolic Networks and Pathways/genetics , Organisms, Genetically Modified
6.
Mol Metab ; 5(11): 1072-1082, 2016 11.
Article in English | MEDLINE | ID: mdl-27818933

ABSTRACT

OBJECTIVE: Non-alcoholic fatty liver disease is a world-wide health concern and risk factor for cardio-metabolic diseases. Citrate uptake modifies intracellular hepatic energy metabolism and is controlled by the conserved sodium-dicarboxylate cotransporter solute carrier family 13 member 5 (SLC13A5, mammalian homolog of INDY: mINDY). In Drosophila melanogaster and Caenorhabditis elegans INDY reduction decreased whole-body lipid accumulation. Genetic deletion of Slc13a5 in mice protected from diet-induced adiposity and insulin resistance. We hypothesized that inducible hepatic mINDY inhibition should prevent the development of fatty liver and hepatic insulin resistance. METHODS: Adult C57BL/6J mice were fed a Western diet (60% kcal from fat, 21% kcal from carbohydrate) ad libitum. Knockdown of mINDY was induced by weekly injection of a chemically modified, liver-selective siRNA for 8 weeks. Mice were metabolically characterized and the effect of mINDY suppression on glucose tolerance as well as insulin sensitivity was assessed with an ipGTT and a hyperinsulinemic-euglycemic clamp. Hepatic lipid accumulation was determined by biochemical measurements and histochemistry. RESULTS: Within the 8 week intervention, hepatic mINDY expression was suppressed by a liver-selective siRNA by over 60%. mINDY knockdown improved hepatic insulin sensitivity (i.e. insulin-induced suppression of endogenous glucose production) of C57BL/6J mice in the hyperinsulinemic-euglycemic clamp. Moreover, the siRNA-mediated mINDY inhibition prevented neutral lipid storage and triglyceride accumulation in the liver, while we found no effect on body weight. CONCLUSIONS: We show that inducible mINDY inhibition improved hepatic insulin sensitivity and prevented diet-induced non-alcoholic fatty liver disease in adult C57BL6/J mice. These effects did not depend on changes of body weight or body composition.


Subject(s)
Dicarboxylic Acid Transporters/physiology , Insulin Resistance , Lipid Metabolism , Non-alcoholic Fatty Liver Disease , RNA Interference , Symporters/physiology , Animals , Citrates , Citric Acid , Diet , Mice , Mice, Inbred C57BL
7.
J Pharmacol Exp Ther ; 355(2): 247-54, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26324167

ABSTRACT

The citric acid cycle intermediate citrate plays a crucial role in metabolic processes such as fatty acid synthesis, glucose metabolism, and ß-oxidation. Citrate is imported from the circulation across the plasma membrane into liver cells mainly by the sodium-dependent citrate transporter (NaCT; SLC13A5). Deletion of NaCT from mice led to metabolic changes similar to caloric restriction; therefore, NaCT has been proposed as an attractive therapeutic target for the treatment of obesity and type 2 diabetes. In this study, we expressed mouse and human NaCT into Xenopus oocytes and examined some basic functional properties of those transporters. Interestingly, striking differences were found between mouse and human NaCT with respect to their sensitivities to citric acid cycle intermediates as substrates for these transporters. Mouse NaCT had at least 20- to 800-fold higher affinity for these intermediates than human NaCT. Mouse NaCT is fully active at physiologic plasma levels of citrate, but its human counterpart is not. Replacement of extracellular sodium by other monovalent cations revealed that human NaCT was markedly less dependent on extracellular sodium than mouse NaCT. The low sensitivity of human NaCT for citrate raises questions about the translatability of this target from the mouse to the human situation and raises doubts about the validity of this transporter as a therapeutic target for the treatment of metabolic diseases in humans.


Subject(s)
Citric Acid Cycle , Dicarboxylic Acid Transporters/physiology , Symporters/physiology , Animals , Cations, Monovalent , Choline/metabolism , Dicarboxylic Acid Transporters/genetics , Female , Humans , Lithium/metabolism , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Sodium/metabolism , Substrate Specificity , Symporters/genetics , Xenopus laevis
8.
PLoS One ; 10(8): e0134764, 2015.
Article in English | MEDLINE | ID: mdl-26241473

ABSTRACT

Enhanced sodium excretion is associated with intrarenal oxidative stress. The present study evaluated whether oxidative stress caused by high sodium (HS) may be involved in calcium oxalate crystal formation. Male rats were fed a sodium-depleted diet. Normal-sodium and HS diets were achieved by providing drinking water containing 0.3% and 3% NaCl, respectively. Rats were fed a sodium-depleted diet with 5% hydroxyl-L-proline (HP) for 7 and 42 days to induce hyperoxaluria and/or calcium oxalate deposition. Compared to normal sodium, HS slightly increased calcium excretion despite diuresis; however, the result did not reach statistical significance. HS did not affect the hyperoxaluria, hypocalciuria or supersaturation caused by HP; however, it increased calcium oxalate crystal deposition soon after 7 days of co-treatment. Massive calcium oxalate formation and calcium crystal excretion in HS+HP rats were seen after 42 days of treatment. HP-mediated hypocitraturia was further exacerbated by HS. Moreover, HS aggravated HP-induced renal injury and tubular damage via increased apoptosis and oxidative stress. Increased urinary malondialdehyde excretion, in situ superoxide production, NAD(P)H oxidase and xanthine oxidase expression and activity, and decreased antioxidant enzyme expression or activity in the HS+HP kidney indicated exaggerated oxidative stress. Interestingly, this redox imbalance was associated with reduced renal osteopontin and Tamm-Horsfall protein expression (via increased excretion) and sodium-dependent dicarboxylate cotransporter NaDC-1 upregulation. Collectively, our results demonstrate that a HS diet induces massive crystal formation in the hyperoxaluric kidney; this is not due to increased urinary calcium excretion but is related to oxidative injury and loss of anticrystallization defense.


Subject(s)
Calcium Oxalate/chemistry , Hyperoxaluria/metabolism , Kidney Calculi/etiology , Kidney Tubules/metabolism , Natriuresis/physiology , Oxidative Stress/drug effects , Sodium, Dietary/pharmacology , Animals , Antioxidants/metabolism , Biomarkers , Citrates/urine , Creatinine/urine , Crystallization , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acid Transporters/physiology , Diet, Sodium-Restricted , Diuresis/drug effects , Enzyme Induction , Gene Expression Regulation , Hydroxyproline/toxicity , Hyperoxaluria/chemically induced , Hyperoxaluria/genetics , Kidney Calculi/metabolism , Kidney Calculi/urine , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/physiology , Osteopontin/genetics , Osteopontin/physiology , Rats , Rats, Wistar , Sodium, Dietary/administration & dosage , Sodium, Dietary/toxicity , Superoxides/metabolism , Symporters/genetics , Symporters/physiology , Uromodulin/genetics , Uromodulin/physiology
9.
IUBMB Life ; 66(7): 462-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25045044

ABSTRACT

The citrate carrier (CiC), characteristic of animals, and the dicarboxylate-tricarboxylate carrier (DTC), characteristic of plants and protozoa, belong to the mitochondrial carrier protein family whose members are responsible for the exchange of metabolites, cofactors, and nucleotides between the cytoplasm and the mitochondrial matrix. Most of the functional data on these transporters are obtained from the studies performed with the protein purified from rat, eel yeast, and maize mitochondria or recombinant proteins from different sources incorporated into phospholipid vesicles (liposomes). The functional data indicate that CiC is responsible for the efflux of acetyl-CoA from the mitochondria to the cytosol in the form of citrate, the primer for fatty acid, cholesterol synthesis, and histone acetylation. Like the CiC, the citrate exported by DTC from the mitochondria to the cytosol in exchange for oxaloacetate can be cleaved by citrate lyase to acetyl-CoA and oxaloacetate and used for fatty acid elongation and isoprenoid synthesis. In addition to its role in fatty acid synthesis, CiC is involved in other processes such as gluconeogenesis, insulin secretion, inflammation, and cancer progression, whereas DTC is involved in the production of glycerate, nitrogen assimilation, ripening of fruits, ATP synthesis, and sustaining of respiratory flux in fruit cells. This review provides an assessment of the current understanding of CiC and DTC structural and biochemical characteristics, underlying the structure-function relationship of these carriers. Furthermore, a phylogenetic relationship between CiC and DTC is proposed.


Subject(s)
Carrier Proteins/physiology , Dicarboxylic Acid Transporters/physiology , Animals , Base Sequence , Humans , Mitochondria/metabolism , Mitochondrial Proteins/physiology , Molecular Sequence Data , Organ Specificity , Phylogeny , Promoter Regions, Genetic
10.
J Membr Biol ; 246(9): 705-15, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23979173

ABSTRACT

The SdcF transporter from Bacillus licheniformis (gene BL02343) is a member of the divalent anion sodium symporter (DASS)/SLC13 family that includes Na⁺/dicarboxylate transporters from bacteria to humans. SdcF was functionally expressed in Escherichia coli (BL21) and assayed in right side out membrane vesicles. ScdF catalyzed the sodium-coupled transport of succinate and α-ketoglutarate. Succinate transport was strongly inhibited by malate, fumarate, tartrate, oxaloacetate and L-aspartate. Similar to the other DASS transporters, succinate transport by SdcF was inhibited by anthranilic acids, N-(p-amylcinnamoyl) anthranilic acid and flufenamate. SdcF transport was cation-dependent, with a K0.5 for sodium of ~1.5 mM and a K0.5 for Li⁺ of ~40 mM. Succinate transport kinetics by SdcF were sigmoidal, suggesting that SdcF may contain two cooperative substrate binding sites. The results support an ordered binding mechanism for SdcF in which sodium binds first and succinate binds last. We conclude that SdcF is a secondary active transporter for four- and five-carbon dicarboxylates that can use Na⁺ or Li⁺ as a driving cation.


Subject(s)
Bacillus/metabolism , Bacterial Proteins/physiology , Dicarboxylic Acid Transporters/physiology , Amino Acid Sequence , Bacterial Proteins/chemistry , Biological Transport , Dicarboxylic Acid Transporters/chemistry , Escherichia coli , Kinetics , Lithium/chemistry , Molecular Sequence Data , Sequence Homology, Amino Acid , Sodium Chloride/chemistry , Substrate Specificity , Succinic Acid/metabolism
11.
Biochim Biophys Acta ; 1808(6): 1454-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21073858

ABSTRACT

Citric acid cycle intermediates, including succinate and citrate, are absorbed across the apical membrane by the NaDC1 Na+/dicarboxylate cotransporter located in the kidney and small intestine. The secondary structure model of NaDC1 contains 11 transmembrane helices (TM). TM7 was shown previously to contain determinants of citrate affinity, and Arg-349 at the extracellular end of the helix is required for transport. The present study involved cysteine scanning mutagenesis of 26 amino acids in TM7 and the associated loops. All of the mutants were well expressed on the plasma membrane, but many had low or no transport activity: 6 were inactive and 7 had activity less than 25% of the parental. Three of the mutants had notable changes in functional properties. F336C had increased transport activity due to an increased Vmax for succinate. The conserved residue F339C had very low transport activity and a change in substrate selectivity. G356C in the putative extracellular loop was the only cysteine mutant that was affected by the membrane-impermeant cysteine reagent, MTSET. However, direct labeling of G356C with MTSEA-biotin gave a weak signal, indicating that this residue is not readily accessible to more bulky reagents. The results suggest that the amino acids of TM7 are functionally important because their replacement by cysteine had large effects on transport activity. However, most of TM7 does not appear to be accessible to the extracellular fluid and is likely to be an outer helix in contact with the lipid bilayer.


Subject(s)
Dicarboxylic Acid Transporters/chemistry , Organic Anion Transporters, Sodium-Dependent/chemistry , Protein Structure, Secondary , Symporters/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Biological Transport/drug effects , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acid Transporters/physiology , HeLa Cells , Humans , Immunoblotting , Kinetics , Mesylates , Molecular Sequence Data , Mutation , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/physiology , Rabbits , Sequence Homology, Amino Acid , Succinic Acid/metabolism , Sulfhydryl Reagents/pharmacology , Symporters/genetics , Symporters/physiology
12.
J Nephrol ; 23 Suppl 16: S49-56, 2010.
Article in English | MEDLINE | ID: mdl-21170889

ABSTRACT

Hypocitraturia is a known risk factor for kidney stone formation. By forming soluble complexes with calcium, citrate prevents crystal nucleation, aggregation and growth; therefore, the presence of citrate in the urine reduces the risk for calcium stone formation. Ingested citrate is rapidly metabolized, and plasma citrate levels vary little, so changes in filtered load do not significantly influence urinary citrate excretion. Changes in urinary citrate excretion are predominantly influenced by the rate of citrate absorption from the glomerular filtrate and metabolism by the proximal tubule cell. The former is mediated by the apical membrane cotransporter NaDC1, and the latter is mediated by both cytoplasmic and mitochondrial metabolism. Acid-base status is the most important physiological determinant of urinary citrate excretion, by modulating the activities of NaDC1 and cytoplasmic (ATP citrate lyase) and mitochondrial (m-aconitase) enzymes involved in citrate metabolism. Following an acid load, both the transport and metabolic processes are up-regulated leading to hypocitraturia; in contrast, an alkaline load increases citrate excretion, by regulating only the mitochondrial metabolic process.


Subject(s)
Citric Acid/metabolism , Kidney/metabolism , Animals , Citric Acid/urine , Dicarboxylic Acid Transporters/physiology , Humans , Mitochondria/metabolism , Organic Anion Transporters, Sodium-Dependent/physiology , Symporters/physiology
13.
Am J Physiol Cell Physiol ; 299(2): C497-505, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538765

ABSTRACT

Glutathione transport into mitochondria is mediated by oxoglutarate (OGC) and dicarboxylate carrier (DIC) in the kidney and liver. However, transport mechanisms in brain mitochondria are unknown. We found that both carriers were expressed in the brain. Using cortical mitochondria incubated with physiological levels of glutathione, we found that butylmalonate, a DIC inhibitor, reduced mitochondrial glutathione to levels similar to those seen in mitochondria incubated without extramitochondrial glutathione (59% of control). In contrast, phenylsuccinate, an OGC inhibitor, had no effect (97% of control). Additional experiments with DIC and OGC short hairpin RNA in neuronal-like PC12 cells resulted in similar findings. Significantly, DIC inhibition resulted in increased reactive oxygen species (ROS) content in and H(2)O(2) release from mitochondria. It also led to decreased membrane potential, increased basal respiration rates, and decreased phosphorus-to-oxygen (P/O) ratios, especially when electron transport was initiated from complex I. Accordingly, we found that DIC inhibition impaired complex I activity, but not those for complexes II and III. This impairment was not associated with dislodgment of complex subunits. These results suggest that DIC is the main glutathione transporter in cortical mitochondria and that DIC-mediated glutathione transport is essential for these mitochondria to maintain ROS homeostasis and normal respiratory functions.


Subject(s)
Brain/metabolism , Dicarboxylic Acid Transporters/physiology , Glutathione/metabolism , Homeostasis/physiology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Animals, Newborn , Biological Transport/physiology , Cell Respiration/physiology , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , PC12 Cells , Rats , Rats, Sprague-Dawley
14.
Infect Immun ; 78(6): 2782-92, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20368342

ABSTRACT

In addition to their classical roles as carbon or nitrogen sources, amino acids can be used for bacterial virulence, colonization, or stress resistance. We found that original deamidase-transport systems impact colonization by Helicobacter pylori, a human pathogen associated with gastric pathologies, including adenocarcinoma. We demonstrated that l-asparaginase (Hp-AnsB) and gamma-glutamyltranspeptidase (Hp-gammaGT) are highly active periplasmic deamidases in H. pylori, producing ammonia and aspartate or glutamate from asparagine and glutamine, respectively. Hp-GltS was identified as a sole and specialized transporter for glutamate, while aspartate was exclusively imported by Hp-DcuA. Uptake of Gln and Asn strictly relies on indirect pathways following prior periplasmic deamidation into Glu and Asp. Hence, in H. pylori, the coupled action of periplasmic deamidases with their respective transporters enables the acquisition of Glu and Asp from Gln and Asn, respectively. These systems were active at neutral rather than acidic pH, suggesting their function near the host epithelial cells. We showed that Hp-DcuA, the fourth component of these novel deamidase-transport systems, was as crucial as Hp-gammaGT, Hp-AnsB, and Hp-GltS for animal model colonization. In conclusion, the pH-regulated coupled amino acid deamidase-uptake system represents an original optimized system that is essential for in vivo colonization of the stomach environment by H. pylori. We propose a model in which these two nonredundant systems participate in H. pylori virulence by depleting gastric or immune cells from protective amino acids such as Gln and producing toxic ammonia close to the host cells.


Subject(s)
Amino Acid Transport Systems, Acidic/physiology , Asparaginase/physiology , Bacterial Proteins/physiology , Dicarboxylic Acid Transporters/physiology , Helicobacter pylori/enzymology , Helicobacter pylori/pathogenicity , Virulence Factors/physiology , gamma-Glutamyltransferase/physiology , Amino Acid Transport Systems, Acidic/metabolism , Amino Acids/metabolism , Ammonia/metabolism , Ammonia/toxicity , Animals , Asparaginase/metabolism , Aspartic Acid/metabolism , Bacterial Proteins/metabolism , Colony Count, Microbial , Dicarboxylic Acid Transporters/metabolism , Glutamic Acid/metabolism , Humans , Hydrogen-Ion Concentration , Mice , Models, Biological , Stomach/microbiology , Virulence , Virulence Factors/metabolism , gamma-Glutamyltransferase/metabolism
15.
Proc Natl Acad Sci U S A ; 106(7): 2277-82, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19164521

ABSTRACT

Decreased Indy activity extends lifespan in D. melanogaster without significant reduction in fecundity, metabolic rate, or locomotion. To understand the underlying mechanisms leading to lifespan extension in this mutant strain, we compared the genome-wide gene expression changes in the head and thorax of adult Indy mutant with control flies over the course of their lifespan. A signature enrichment analysis of metabolic and signaling pathways revealed that expression levels of genes in the oxidative phosphorylation pathway are significantly lower in Indy starting at day 20. We confirmed experimentally that complexes I and III of the electron transport chain have lower enzyme activity in Indy long-lived flies by Day 20 and predicted that reactive oxygen species (ROS) production in mitochondria could be reduced. Consistently, we found that both ROS production and protein damage are reduced in Indy with respect to control. However, we did not detect significant differences in total ATP, a phenotype that could be explained by our finding of a higher mitochondrial density in Indy mutants. Thus, one potential mechanism by which Indy mutants extend life span could be through an alteration in mitochondrial physiology leading to an increased efficiency in the ATP/ROS ratio.


Subject(s)
Dicarboxylic Acid Transporters/metabolism , Drosophila Proteins/metabolism , Reactive Oxygen Species , Symporters/metabolism , Adenosine Triphosphate/metabolism , Animals , Dicarboxylic Acid Transporters/physiology , Drosophila Proteins/physiology , Drosophila melanogaster , Electron Transport , Genome , Male , Mitochondria/metabolism , Models, Biological , Mutation , Oxidative Stress , Oxygen/chemistry , Phenotype , Phosphorylation , Symporters/physiology
16.
Biochem J ; 415(1): 11-9, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18778247

ABSTRACT

In higher plants, light is crucial for regulation of nitrate uptake, translocation and assimilation into organic compounds. Part of this metabolism is tightly coupled to photosynthesis because the enzymes involved, nitrite reductase and glutamate synthase, are localized to the chloroplasts and receive reducing power from photosynthetic electron transport. However, important enzymes in nitrate acquisition and reduction are localized to cellular compartments other than chloroplasts and are also up-regulated by light, i.e. transporters in cell and organellar membranes and nitrate reductase in the cytosol. This review describes the different light-dependent signalling cascades regulating nitrate metabolism at the transcriptional as well as post-transcriptional level, and how reactions in different compartments of the cell are co-ordinated. Essential players in this network are phytochrome and HY5 (long hypocotyls 5)/HYH (HY5 homologue)-dependent signalling pathways, the energy-related AMPK (AMP-activated protein kinase) protein kinase homologue SNRK1 (sucrose non-fermenting kinase 1-related kinase), chloroplastic thioredoxins and the prokaryotically originated PII protein. A complex light-dependent network of regulation emerges, which appears to be necessary for optimal nitrogen assimilation and for avoiding the accumulation of toxic intermediates and side products, such as nitrite and reactive oxygen compounds.


Subject(s)
Nitrates/metabolism , Phytochrome/physiology , Plant Physiological Phenomena/radiation effects , Signal Transduction/radiation effects , AMP-Activated Protein Kinases , Arabidopsis/physiology , Arabidopsis Proteins/physiology , Basic-Leucine Zipper Transcription Factors/physiology , Carrier Proteins/physiology , Chloroplast Thioredoxins/metabolism , Chloroplasts/metabolism , Circadian Rhythm , DNA-Binding Proteins , Dicarboxylic Acid Transporters/physiology , Genes, Plant/radiation effects , Glutamate-Ammonia Ligase/physiology , Light , Multienzyme Complexes/physiology , Nitrate Reductase/physiology , Nitrite Reductases/physiology , Nitrites/metabolism , Nuclear Proteins/physiology , PII Nitrogen Regulatory Proteins/physiology , Phosphoprotein Phosphatases/physiology , Protein Serine-Threonine Kinases/physiology
17.
J Bacteriol ; 190(19): 6458-66, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18658264

ABSTRACT

Many bacteria can utilize C(4)-carboxylates as carbon and energy sources. However, Corynebacterium glutamicum ATCC 13032 is not able to use tricarboxylic acid cycle intermediates such as succinate, fumarate, and l-malate as sole carbon sources. Upon prolonged incubation, spontaneous mutants which had gained the ability to grow on succinate, fumarate, and l-malate could be isolated. DNA microarray analysis showed higher mRNA levels of cg0277, which subsequently was named dccT, in the mutants than in the wild type, and transcriptional fusion analysis revealed that a point mutation in the promoter region of dccT was responsible for increased expression. The overexpression of dccT was sufficient to enable the C. glutamicum wild type to grow on succinate, fumarate, and l-malate as the sole carbon sources. Biochemical analyses revealed that DccT, which is a member of the divalent anion/Na(+) symporter family, catalyzes the effective uptake of dicarboxylates like succinate, fumarate, L-malate, and likely also oxaloacetate in a sodium-dependent manner.


Subject(s)
Bacterial Proteins/physiology , Corynebacterium glutamicum/metabolism , Dicarboxylic Acid Transporters/physiology , Dicarboxylic Acids/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Corynebacterium glutamicum/genetics , Corynebacterium glutamicum/growth & development , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acid Transporters/metabolism , Fumarates/metabolism , Malates/metabolism , Mutation , Oligonucleotide Array Sequence Analysis , Oxaloacetic Acid/metabolism , Sodium/metabolism , Succinic Acid/metabolism , Symporters/genetics , Symporters/metabolism , Symporters/physiology , Transcription, Genetic
18.
Glia ; 56(9): 990-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18442087

ABSTRACT

The excitatory amino acid transporters (EAATs) underlie the so-called "high affinity" uptake of glutamate, which is well characterized. In contrast, the "low-affinity" uptake of glutamate remains poorly defined, and it has been discussed whether it may represent a mere in vitro artifact. Here we have visualized "low-affinity" excitatory amino acid uptake sites by incubating rat hippocampal slices with the glutamate analogue D-aspartate in the presence of PMB-TBOA, which blocks the EAATs. After fixation of the slices, D-aspartate taken up into the tissue was localized with the use of light microscopic immunoperoxidase and electron microscopic immunogold methods, exploiting highly specific antibodies against D-aspartate. PMB-TBOA blocked uptake of both low and high exogenous D-aspartate concentrations (0.01-1.0 mM) into nerve terminals, as well as the uptake of 0.01 mM D-aspartate into astrocytes. Interestingly, there was a residual PMB-TBOA insensitive uptake of D-aspartate in astrocytes at higher exogenous D-aspartate concentrations (0.05-1.0 mM), strongly suggesting that astrocytes have "low-affinity" uptake sites for excitatory amino acid. The PMB-TBOA insensitive D-aspartate uptake in astrocytes was sodium dependent and inhibited by succinate and to certain extent by homocysteate, but not by cystine or DIDS. We suggest that excitatory amino acid is transported into astrocytes in a "low-affinity" fashion by sodium/dicarboxylate transporters.


Subject(s)
Astrocytes/metabolism , Dicarboxylic Acid Transporters/physiology , Excitatory Amino Acids/metabolism , Hippocampus/metabolism , Organic Anion Transporters, Sodium-Dependent/physiology , Animals , Astrocytes/drug effects , D-Aspartic Acid/metabolism , D-Aspartic Acid/pharmacology , Glutamate Plasma Membrane Transport Proteins/physiology , Hippocampus/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Wistar
19.
Plant J ; 52(6): 1169-80, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18005230

ABSTRACT

In plants, malate is a central metabolite and fulfills a large number of functions. Vacuolar malate may reach very high concentrations and fluctuate rapidly, whereas cytosolic malate is kept at a constant level allowing optimal metabolism. Recently, a vacuolar malate transporter (Arabidopsis thaliana tonoplast dicarboxylate transporter, AttDT) was identified that did not correspond to the well-characterized vacuolar malate channel. We therefore hypothesized that a member of the aluminum-activated malate transporter (ALMT) gene family could code for a vacuolar malate channel. Using GFP fusion constructs, we could show that AtALMT9 (A. thaliana ALMT9) is targeted to the vacuole. Promoter-GUS fusion constructs demonstrated that this gene is expressed in all organs, but is cell-type specific as GUS activity in leaves was detected nearly exclusively in mesophyll cells. Patch-clamp analysis of an Atalmt9 T-DNA insertion mutant exhibited strongly reduced vacuolar malate channel activity. In order to functionally characterize AtALMT9 as a malate channel, we heterologously expressed this gene in tobacco and in oocytes. Overexpression of AtALMT9-GFP in Nicotiana benthamiana leaves strongly enhanced the malate current densities across the mesophyll tonoplasts. Functional expression of AtALMT9 in Xenopus oocytes induced anion currents, which were clearly distinguishable from endogenous oocyte currents. Our results demonstrate that AtALMT9 is a vacuolar malate channel. Deletion mutants for AtALMT9 exhibit only slightly reduced malate content in mesophyll protoplasts and no visible phenotype, indicating that AttDT and the residual malate channel activity are sufficient to sustain the transport activity necessary to regulate the cytosolic malate homeostasis.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Malates/metabolism , Organic Anion Transporters/metabolism , Vacuoles/metabolism , Aluminum/metabolism , Aluminum/pharmacology , Animals , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/physiology , Biological Transport/drug effects , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acid Transporters/metabolism , Dicarboxylic Acid Transporters/physiology , Gene Expression Regulation, Plant , Glucuronidase/genetics , Glucuronidase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Membrane Potentials/physiology , Mutation , Oocytes/metabolism , Oocytes/physiology , Organic Anion Transporters/genetics , Organic Anion Transporters/physiology , Patch-Clamp Techniques , Plants, Genetically Modified , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Nicotiana/genetics , Nicotiana/metabolism , Xenopus
20.
PLoS Genet ; 3(6): e95, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17571923

ABSTRACT

To investigate whether alterations in mitochondrial metabolism affect longevity in Drosophila melanogaster, we studied lifespan in various single gene mutants, using inbred and outbred genetic backgrounds. As positive controls we included the two most intensively studied mutants of Indy, which encodes a Drosophila Krebs cycle intermediate transporter. It has been reported that flies heterozygous for these Indy mutations, which lie outside the coding region, show almost a doubling of lifespan. We report that only one of the two mutants lowers mRNA levels, implying that the lifespan extension observed is not attributable to the Indy mutations themselves. Moreover, neither Indy mutation extended lifespan in female flies in any genetic background tested. In the original genetic background, only the Indy mutation associated with altered RNA expression extended lifespan in male flies. However, this effect was abolished by backcrossing into standard outbred genetic backgrounds, and was associated with an unidentified locus on the X chromosome. The original Indy line with long-lived males is infected by the cytoplasmic symbiont Wolbachia, and the longevity of Indy males disappeared after tetracycline clearance of this endosymbiont. These findings underscore the critical importance of standardisation of genetic background and of cytoplasm in genetic studies of lifespan, and show that the lifespan extension previously claimed for Indy mutants was entirely attributable to confounding variation from these two sources. In addition, we saw no effects on lifespan of expression knockdown of the Indy orthologues nac-2 and nac-3 in the nematode Caenorhabditis elegans.


Subject(s)
Cytoplasm/physiology , Dicarboxylic Acid Transporters/physiology , Drosophila Proteins/physiology , Drosophila/genetics , Longevity/genetics , Symporters/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Cytoplasm/genetics , DNA, Mitochondrial/physiology , Dicarboxylic Acid Transporters/genetics , Drosophila/growth & development , Drosophila/physiology , Drosophila Proteins/genetics , Female , Longevity/physiology , Male , Mitochondria/genetics , Mitochondria/physiology , Molecular Sequence Data , Mutation , Symporters/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...