Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Molecules ; 26(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34833887

ABSTRACT

Pyropia haitanensis is an important laver species in China. Its quality traits are closely related to the content of glutamic acid. Glutamate dehydrogenase (GDH) is a crucial enzyme in the glutamic acid metabolism. In this study, two GDH genes from P. haitanensis, PhGDH1 and PhGDH2, were cloned and successfully expressed in Escherichia coli. The in vitro enzyme activity assay demonstrated that the catalytic activity of PhGDHs is mainly in the direction of ammonium assimilation. The measured Km values of PhGDH1 for NADH, (NH4)2SO4, and α-oxoglutarate were 0.12, 4.99, and 0.16 mM, respectively, while the corresponding Km values of PhGDH2 were 0.02, 3.98, and 0.104 mM, respectively. Site-directed mutagenesis results showed that Gly193 and Thr361 were important catalytic residues for PhGDH2. Moreover, expression levels of both PhGDHs were significantly increased under abiotic stresses. These results suggest that PhGDHs can convert α-oxoglutarate to glutamic acid, and enhance the flavor and stress resistance of P. haitanensis.


Subject(s)
Glutamate Dehydrogenase/metabolism , Glutamic Acid/metabolism , Rhodophyta/metabolism , Biochemical Phenomena , China , Glutamate Dehydrogenase/physiology , Mutagenesis, Site-Directed , Rhodophyta/genetics , Stress, Physiological/physiology
2.
J Neurochem ; 157(3): 802-815, 2021 05.
Article in English | MEDLINE | ID: mdl-33421122

ABSTRACT

INTRODUCTION: Mammalian glutamate dehydrogenase (hGDH1 in human cells) interconverts glutamate to α-ketoglutarate and ammonia while reducing NAD(P) to NAD(P)H. During primate evolution, humans and great apes have acquired hGDH2, an isoenzyme that underwent rapid evolutionary adaptation concomitantly with brain expansion, thereby acquiring unique catalytic and regulatory properties that permitted its function under conditions inhibitory to its ancestor hGDH1. Although the 3D-structures of GDHs, including hGDH1, have been determined, attempts to determine the hGDH2 structure were until recently unsuccessful. Comparison of the hGDH1/hGDH2 structures would enable a detailed understanding of their evolutionary differences. This work aimed at the determination of the hGDH2 crystal structure and the analysis of its functional implications. Recombinant hGDH2 was produced in the Spodoptera frugiperda ovarian cell line Sf21, using the Baculovirus expression system. Purification was achieved via a two-step chromatography procedure. hGDH2 was crystallized, X-ray diffraction data were collected using synchrotron radiation and the structure was determined by molecular replacement. The hGDH2 structure is reported at a resolution of 2.9 Å. The enzyme adopts a novel semi-closed conformation, which is an intermediate between known open and closed GDH1 conformations, differing from both. The structure enabled us to dissect previously reported biochemical findings and to structurally interpret the effects of evolutionary amino acid substitutions, including Arg470His, on ADP affinity. In conclusion, our data provide insights into the structural basis of hGDH2 properties, the functional evolution of hGDH isoenzymes, and open new prospects for drug design, especially for cancer therapeutics.


Subject(s)
Brain/enzymology , Brain/physiology , Glutamate Dehydrogenase/physiology , Neoplasms/enzymology , Neoplasms/physiopathology , Amino Acid Substitution , Animals , Cell Line , Crystallization , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/chemistry , Humans , Models, Molecular , Molecular Structure , Mutation , Protein Conformation , Recombinant Proteins , Spodoptera , X-Ray Diffraction
3.
PLoS One ; 15(2): e0229288, 2020.
Article in English | MEDLINE | ID: mdl-32078638

ABSTRACT

The GluD1 gene is associated with susceptibility for schizophrenia, autism, depression, and bipolar disorder. However, the function of GluD1 and how it is involved in these conditions remain elusive. In this study, we generated a Grid1 gene-knockout (GluD1-KO) mouse line with a pure C57BL/6N genetic background and performed several behavioral analyses. Compared to a control group, GluD1-KO mice showed no significant anxiety-related behavioral differences, evaluated using behavior in an open field, elevated plus maze, a light-dark transition test, the resident-intruder test of aggression and sensorimotor gating evaluated by the prepulse inhibition test. However, GluD1-KO mice showed (1) higher locomotor activity in the open field, (2) decreased sociability and social novelty preference in the three-chambered social interaction test, (3) impaired memory in contextual, but not cued fear conditioning tests, and (4) enhanced depressive-like behavior in a forced swim test. Pharmacological studies revealed that enhanced depressive-like behavior in GluD1-KO mice was restored by the serotonin reuptake inhibitors imipramine and fluoxetine, but not the norepinephrine transporter inhibitor desipramine. In addition, biochemical analysis revealed no significant difference in protein expression levels, such as other glutamate receptors in the synaptosome and postsynaptic densities prepared from the frontal cortex and the hippocampus. These results suggest that GluD1 plays critical roles in fear memory, sociability, and depressive-like behavior.


Subject(s)
Anxiety/pathology , Depression/pathology , Fear , Glutamate Dehydrogenase/physiology , Interpersonal Relations , Memory Disorders/pathology , Social Behavior Disorders/pathology , Animals , Anxiety/etiology , Behavior, Animal , Depression/etiology , Male , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Social Behavior Disorders/etiology
4.
J Neurochem ; 153(1): 80-102, 2020 04.
Article in English | MEDLINE | ID: mdl-31886885

ABSTRACT

Glutamate dehydrogenase (GDH) is essential for the brain function and highly regulated, according to its role in metabolism of the major excitatory neurotransmitter glutamate. Here we show a diurnal pattern of the GDH acetylation in rat brain, associated with specific regulation of GDH function. Mornings the acetylation levels of K84 (near the ADP site), K187 (near the active site), and K503 (GTP-binding) are highly correlated. Evenings the acetylation levels of K187 and K503 decrease, and the correlations disappear. These daily variations in the acetylation adjust the GDH responses to the enzyme regulators. The adjustment is changed when the acetylation of K187 and K503 shows no diurnal variations, as in the rats after a high dose of thiamine. The regulation of GDH function by acetylation is confirmed in a model system, where incubation of the rat brain GDH with acetyl-CoA changes the enzyme responses to GTP and ADP, decreasing the activity at subsaturating concentrations of substrates. Thus, the GDH acetylation may support cerebral homeostasis, stabilizing the enzyme function during diurnal oscillations of the brain metabolome. Daytime and thiamine interact upon the (de)acetylation of GDH in vitro. Evenings the acetylation of GDH from control animals increases both IC50GTP and EC50ADP . Mornings the acetylation of GDH from thiamine-treated animals increases the enzyme IC50GTP . Molecular mechanisms of the GDH regulation by acetylation of specific residues are proposed. For the first time, diurnal and thiamine-dependent changes in the allosteric regulation of the brain GDH due to the enzyme acetylation are shown.


Subject(s)
Brain/enzymology , Circadian Rhythm/physiology , Glutamate Dehydrogenase/physiology , Thiamine/pharmacology , Acetyl Coenzyme A/pharmacology , Acetylation , Allosteric Regulation/drug effects , Animals , Cerebral Cortex/enzymology , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/chemistry , Male , Mitochondria/enzymology , NAD/pharmacology , Rats , Rats, Wistar
5.
Mol Psychiatry ; 24(10): 1451-1460, 2019 10.
Article in English | MEDLINE | ID: mdl-30824864

ABSTRACT

Ionotropic glutamate delta receptors do not bind glutamate and do not generate ionic current, resulting in difficulty in studying the function and trafficking of these receptors. Here, we utilize chimeric constructs, in which the ligand-binding domain of GluD1 is replaced by that of GluK1, to examine its synaptic trafficking and plasticity. GluD1 trafficked to the synapse, but was incapable of expressing long-term potentiation (LTP). The C-terminal domain (CT) of GluD1 has a classic PDZ-binding motif, which is critical for the synaptic trafficking of other glutamate receptors, but we found that its binding to PSD-95 was very weak, and deleting the PDZ-binding motif failed to alter synaptic trafficking. However, deletion of the entire CT abolished synaptic trafficking, but not surface expression. We found that mutation of threonine (T) T923 to an alanine disrupted synaptic trafficking. Therefore, GluD1 receptors have strikingly different trafficking mechanisms compared with AMPARs. These results highlight the diversity of ionotropic glutamate receptor trafficking rules at a single type of synapse. Since this receptor is genetically associated with schizophrenia, our findings may provide an important clue to understand schizophrenia.


Subject(s)
Glutamate Dehydrogenase/metabolism , Receptors, Glutamate/metabolism , Animals , Carrier Proteins/genetics , Glutamate Dehydrogenase/physiology , Glutamic Acid/metabolism , Hippocampus/metabolism , Long-Term Potentiation , Membrane Proteins/metabolism , Mice , Neuronal Plasticity/physiology , Neurons/metabolism , Patch-Clamp Techniques , Protein Binding , Protein Transport/physiology , Receptors, AMPA/metabolism , Receptors, Glutamate/genetics , Receptors, Opioid, delta/metabolism , Synapses/metabolism
6.
Mol Psychiatry ; 23(3): 691-700, 2018 03.
Article in English | MEDLINE | ID: mdl-28696429

ABSTRACT

Human mutations of the GRID1 gene encoding the orphan delta1 glutamate receptor-channel (GluD1) are associated with schizophrenia but the explicit role of GluD1 in brain circuits is unknown. Based on the known function of its paralog GluD2 in cerebellum, we searched for a role of GluD1 in slow glutamatergic transmission mediated by metabotropic receptor mGlu1 in midbrain dopamine neurons, whose dysfunction is a hallmark of schizophrenia. We found that an mGlu1 agonist elicits a slow depolarizing current in HEK cells co-expressing mGlu1 and GluD1, but not in cells expressing mGlu1 or GluD1 alone. This current is abolished by additional co-expression of a dominant-negative GluD1 dead pore mutant. We then characterized mGlu1-dependent currents in dopamine neurons from midbrain slices. Both the agonist-evoked and the slow postsynaptic currents are abolished by expression of the dominant-negative GluD1 mutant, pointing to the involvement of native GluD1 channels in these currents. Likewise, both mGlu1-dependent currents are suppressed in GRID1 knockout mice, which reportedly display endophenotypes relevant for schizophrenia. It is known that mGlu1 activation triggers the transition from tonic to burst firing of dopamine neurons, which signals salient stimuli and encodes reward prediction. In vivo recordings of dopamine neurons showed that their spontaneous burst firing is abolished in GRID1 knockout mice or upon targeted expression of the dominant-negative GluD1 mutant in wild-type mice. Our results de-orphanize GluD1, unravel its key role in slow glutamatergic transmission and provide insights into how GRID1 gene alterations can lead to dopaminergic dysfunctions in schizophrenia.


Subject(s)
Dopaminergic Neurons/metabolism , Glutamate Dehydrogenase/genetics , Receptors, Glutamate/genetics , Animals , Cerebellum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/physiology , Glutamate Dehydrogenase/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glutamate/physiology , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/genetics , Single-Cell Analysis
7.
Shanghai Kou Qiang Yi Xue ; 26(4): 358-362, 2017 Aug.
Article in Chinese | MEDLINE | ID: mdl-29199326

ABSTRACT

PURPOSE: To investigate the effect of glutamate dehydrogenase 1 (GLUD1) on proliferation, osteogenic differentiation and mineralization of human dental pulp stem cells (hDPSCs). METHODS: hDPSCs were isolated by tissue-explant method in vitro, and shGLUD1 lentivirus was transfected to knock down the expression of GLUD1. RT-PCR and Western blot were performed to detect the expression of GLUD1. CCK8 assay was used to evaluate cell proliferation. After culture with osteogenic inducing medium for 14 days, alizarin red staining was used to detect the formation of mineralization nodules, and RT-PCR and immunofluorescence staining were performed to detect the expression of Runx2 and OCN, respectively. The data were analyzed with SPSS 20.0 software package. RESULTS: The expression of GLUD1 was significantly increased in hDPSCs after osteogenic induction compared with the control. After transfection with shGLUD1 lentivirus, GLUD1 expression was significantly decreased (P<0.05). Compared with the control group, mineralization nodule formation was significantly decreased in shGLUD1 group after osteogenic induction. The expression of OCN (late-staged markers for osteogenic differentiation) were significantly decreased both in mRNA and protein levels, while the expression of Runx2 (early-staged markers for osteoblast differentiation) was up-regulated. CONCLUSIONS: shGLUD1 inhibits the proliferation, mineralization and the late stage of osteogenic differentiation of hDPSCs in vitro. GLUD1 may play an important role in osteogenic differentiation of hDPSCs.


Subject(s)
Cell Differentiation , Dental Pulp , Glutamate Dehydrogenase , Osteogenesis , Cell Proliferation , Cells, Cultured , Dental Pulp/cytology , Glutamate Dehydrogenase/physiology , Humans , Stem Cells
8.
J Hepatol ; 64(4): 860-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26639393

ABSTRACT

BACKGROUND & AIMS: Recently, spatial-temporal/metabolic mathematical models have been established that allow the simulation of metabolic processes in tissues. We applied these models to decipher ammonia detoxification mechanisms in the liver. METHODS: An integrated metabolic-spatial-temporal model was used to generate hypotheses of ammonia metabolism. Predicted mechanisms were validated using time-resolved analyses of nitrogen metabolism, activity analyses, immunostaining and gene expression after induction of liver damage in mice. Moreover, blood from the portal vein, liver vein and mixed venous blood was analyzed in a time dependent manner. RESULTS: Modeling revealed an underestimation of ammonia consumption after liver damage when only the currently established mechanisms of ammonia detoxification were simulated. By iterative cycles of modeling and experiments, the reductive amidation of alpha-ketoglutarate (α-KG) via glutamate dehydrogenase (GDH) was identified as the lacking component. GDH is released from damaged hepatocytes into the blood where it consumes ammonia to generate glutamate, thereby providing systemic protection against hyperammonemia. This mechanism was exploited therapeutically in a mouse model of hyperammonemia by injecting GDH together with optimized doses of cofactors. Intravenous injection of GDH (720 U/kg), α-KG (280 mg/kg) and NADPH (180 mg/kg) reduced the elevated blood ammonia concentrations (>200 µM) to levels close to normal within only 15 min. CONCLUSION: If successfully translated to patients the GDH-based therapy might provide a less aggressive therapeutic alternative for patients with severe hyperammonemia.


Subject(s)
Hyperammonemia/drug therapy , Liver Diseases/drug therapy , Animals , Glutamate Dehydrogenase/physiology , Ketoglutaric Acids/therapeutic use , Male , Mice , Mice, Inbred C57BL
9.
EMBO J ; 34(13): 1786-800, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25953831

ABSTRACT

Coupling cell cycle with nutrient availability is a crucial process for all living cells. But how bacteria control cell division according to metabolic supplies remains poorly understood. Here, we describe a molecular mechanism that coordinates central metabolism with cell division in the α-proteobacterium Caulobacter crescentus. This mechanism involves the NAD-dependent glutamate dehydrogenase GdhZ and the oxidoreductase-like KidO. While enzymatically active GdhZ directly interferes with FtsZ polymerization by stimulating its GTPase activity, KidO bound to NADH destabilizes lateral interactions between FtsZ protofilaments. Both GdhZ and KidO share the same regulatory network to concomitantly stimulate the rapid disassembly of the Z-ring, necessary for the subsequent release of progeny cells. Thus, this mechanism illustrates how proteins initially dedicated to metabolism coordinate cell cycle progression with nutrient availability.


Subject(s)
Caulobacter crescentus/cytology , Caulobacter crescentus/enzymology , Caulobacter crescentus/metabolism , Cell Division/genetics , Glutamate Dehydrogenase/physiology , Bacterial Proteins/metabolism , Caulobacter crescentus/genetics , Cytoskeletal Proteins/metabolism , Gene Deletion , Glutamate Dehydrogenase/metabolism , NAD/metabolism , Organisms, Genetically Modified , Protein Binding
11.
Mol Biol Cell ; 23(19): 3851-62, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22875990

ABSTRACT

In pancreatic ß-cells, glutamate dehydrogenase (GDH) modulates insulin secretion, although its function regarding specific secretagogues is unclear. This study investigated the role of GDH using a ß-cell-specific GDH knockout mouse model, called ßGlud1(-/-). The absence of GDH in islets isolated from ßGlud1(-/-) mice resulted in abrogation of insulin release evoked by glutamine combined with 2-aminobicyclo[2.2.1]heptane-2-carboxylic acid or l-leucine. Reintroduction of GDH in ßGlud1(-/-) islets fully restored the secretory response. Regarding glucose stimulation, insulin secretion in islets isolated from ßGlud1(-/-) mice exhibited half of the response measured in control islets. The amplifying pathway, tested at stimulatory glucose concentrations in the presence of KCl and diazoxide, was markedly inhibited in ßGlud1(-/-) islets. On glucose stimulation, net synthesis of glutamate from α-ketoglutarate was impaired in GDH-deficient islets. Accordingly, glucose-induced elevation of glutamate levels observed in control islets was absent in ßGlud1(-/-) islets. Parallel biochemical pathways, namely alanine and aspartate aminotransferases, could not compensate for the lack of GDH. However, the secretory response to glucose was fully restored by the provision of cellular glutamate when ßGlud1(-/-) islets were exposed to dimethyl glutamate. This shows that permissive levels of glutamate are required for the full development of glucose-stimulated insulin secretion and that GDH plays an indispensable role in this process.


Subject(s)
Glutamate Dehydrogenase/physiology , Glutamic Acid/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Aspartic Acid/biosynthesis , Calcium Signaling , Cells, Cultured , Female , Glucose/physiology , Glutamate Dehydrogenase/deficiency , Glutamate Dehydrogenase/genetics , Glutamic Acid/biosynthesis , Glutamic Acid/metabolism , Glutamine/physiology , Insulin Secretion , Insulin-Secreting Cells/enzymology , Leucine/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout
12.
J Endocrinol ; 212(3): 239-42, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22232141

ABSTRACT

In this issue of Journal of Endocrinology, Dr Han and colleagues report a protective effect of the glutamate dehydrogenase activator 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) under diabetes-like conditions that impair ß-cell function in both a pancreatic ß-cell line and db/db mice. Based on these observations, the authors suggest that BCH could serve as a novel treatment modality in type 2 diabetes. The present commentary discusses the importance of the findings. Some additional questions are raised, which may be addressed in future investigations, as there is some concern regarding the BCH treatment of ß-cell failure.


Subject(s)
Amino Acids, Cyclic/pharmacology , Diabetes Mellitus/physiopathology , Glutamate Dehydrogenase/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Animals
13.
J Endocrinol ; 212(3): 307-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22131441

ABSTRACT

2-Aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH) is an activator of glutamate dehydrogenase (GDH), which is a mitochondrial enzyme with an important role in insulin secretion. We investigated the effect of BCH on the high-glucose (HG)-induced reduction in glucose-stimulated insulin secretion (GSIS), the HG/palmitate (PA)-induced reduction in insulin gene expression, and HG/PA-induced ß-cell death. We also studied whether long-term treatment with BCH lowers blood glucose and improves ß-cell integrity in db/db mice. We evaluated GSIS, insulin gene expression, and DNA fragmentation in INS-1 cells exposed to HG or HG/PA in the presence or absence of BCH. An in vivo study was performed in which 7-week-old diabetic db/db mice were treated with BCH (0.7  g/kg, n = 10) and placebo (n = 10) every other day for 6 weeks. After treatment, an intraperitoneal glucose tolerance test and immunohistological examinations were performed. Treatment with BCH blocked HG-induced GSIS inhibition and the HG/PA-induced reduction in insulin gene expression in INS-1 cells. In addition, BCH significantly reduced HG/PA-induced INS-1 cell death and phospho-JNK level. BCH treatment improved glucose tolerance and insulin secretion in db/db mice. BCH treatment also increased the ratio of insulin-positive ß-cells to total islet area (P < 0.05) and reduced the percentage of ß-cells expressing cleaved caspase 3 (P < 0.05). In conclusion, the GDH activator BCH improved glycemic control in db/db mice. This anti-diabetic effect may be associated with improved insulin secretion, preserved islet architecture, and reduced ß-cell apoptosis.


Subject(s)
Amino Acids, Cyclic/pharmacology , Diabetes Mellitus/physiopathology , Glutamate Dehydrogenase/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Animals , Apoptosis/drug effects , Blood Glucose/analysis , Cell Line, Tumor , Diabetes Mellitus/drug therapy , Drug Synergism , Enzyme Activation/drug effects , Gene Expression/drug effects , Glucose/pharmacology , Insulin/blood , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulinoma , Mice , Pancreatic Neoplasms , Rats
14.
Neurochem Int ; 59(4): 510-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21600947

ABSTRACT

Glutamate dehydrogenase (GDH) is a mitochondrial enzyme linking the Krebs cycle to the multifunctional amino acid glutamate. Thereby, GDH plays a pivotal role between carbohydrate and protein metabolisms, controlling production and consumption of the messenger molecule glutamate in neuroendocrine cells. GDH activity is under the control of several regulators, conferring to this enzyme energy-sensor property. Indeed, GDH directly depends on the provision of the co-factor NADH/NAD(+), rendering the enzyme sensitive to the redox status of the cell. Moreover, GDH is allosterically regulated by GTP and ADP. GDH is also regulated by ADP-ribosylation, mediated by a member of the energy-sensor family sirtuins, namely SIRT4. In the brain, GDH ensures the cycling of the neurotransmitter glutamate between neurons and astrocytes. GDH also controls ammonia metabolism and detoxification, mainly in the liver and kidney. In pancreatic ß-cells, the importance of GDH as a key enzyme in the regulation of insulin secretion is now well established. Inhibition of GDH activity decreases insulin release, while activating mutations are associated with a hyperinsulinism syndrome. Although GDH enzyme catalyzes the same reaction in every tissue, its function regarding metabolic homeostasis varies greatly according to specific organs. In this review, we will discuss specificities of GDH regulation in neuroendocrine cells, in particular pancreatic islets and central nervous system.


Subject(s)
Central Nervous System/enzymology , Energy Metabolism/physiology , Glutamate Dehydrogenase/metabolism , Homeostasis/physiology , Islets of Langerhans/enzymology , Animals , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/physiology , Humans , Mitochondria/metabolism
15.
Neurochem Int ; 59(4): 482-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21397649

ABSTRACT

It has been previously demonstrated that ammonia exposure of neurons and astrocytes in co-culture leads to net synthesis not only of glutamine but also of alanine. The latter process involves the concerted action of glutamate dehydrogenase (GDH) and alanine aminotransferase (ALAT). In the present study it was investigated if the glutamine synthetase (GS) inhibitor methionine sulfoximine (MSO) would enhance alanine synthesis by blocking the GS-dependent ammonia scavenging process. Hence, co-cultures of neurons and astrocytes were incubated for 2.5h with [U-(13)C]glucose to monitor de novo synthesis of alanine and glutamine in the absence and presence of 5.0 mM NH(4)Cl and 10 mM MSO. Ammonia exposure led to increased incorporation of label but not to a significant increase in the amount of these amino acids. However, in the presence of MSO, glutamine synthesis was blocked and synthesis of alanine increased leading to an elevated content intra- as well as extracellularly of this amino acid. Treatment with MSO led to a dramatic decrease in glutamine content and increased the intracellular contents of glutamate and aspartate. The large increase in alanine during exposure to MSO underlines the importance of the GDH and ALAT biosynthetic pathway for ammonia fixation, and it points to the use of a GS inhibitor to ameliorate the brain toxicity and edema induced by hyperammonemia, events likely related to glutamine synthesis.


Subject(s)
Alanine/metabolism , Ammonia/metabolism , Astrocytes/metabolism , Glutamate Dehydrogenase/physiology , Glutamine/antagonists & inhibitors , Glutamine/biosynthesis , Neurons/metabolism , Alanine Transaminase/metabolism , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Glutamate-Ammonia Ligase/antagonists & inhibitors , Glutamate-Ammonia Ligase/metabolism , Hyperammonemia/chemically induced , Hyperammonemia/metabolism , Methionine Sulfoximine/pharmacology , Mice , Mice, Inbred Strains , Neurons/drug effects , Neurons/enzymology
16.
Neurochem Int ; 59(4): 473-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21397652

ABSTRACT

Glutamate dehydrogenase 1 (GLUD1) is a mitochondrial enzyme expressed in all tissues, including brain. Although this enzyme is expressed in glutamatergic pathways, its function as a regulator of glutamate neurotransmitter levels is still not well defined. In order to gain an understanding of the role of GLUD1 in the control of glutamate levels and synaptic release in mammalian brain, we generated transgenic (Tg) mice that over-express this enzyme in neurons of the central nervous system. The Tg mice have increased activity of GLUD, as well as elevated levels and increased synaptic and depolarization-induced release of glutamate. These mice suffer age-associated losses of dendritic spines, nerve terminals, and neurons. The neuronal losses and dendrite structural changes occur in select regions of the brain. At the transcriptional level in the hippocampus, cells respond by increasing the expression of genes related to neurite growth and synapse formation, indications of adaptive or compensatory responses to the effects of increases in the release and action of glutamate at synapses. Because these Tg mice live to a relatively old age they are a good model of the effects of a "hyperglutamatergic" state on the aging process in the nervous system. The mice are also useful in defining the molecular pathways affected by the over-activation of GLUD in glutamatergic neurons of the brain and spinal cord.


Subject(s)
Adaptation, Physiological , Disease Models, Animal , Glutamate Dehydrogenase/biosynthesis , Glutamic Acid/biosynthesis , Glutamic Acid/metabolism , Mice, Transgenic , Neurons/enzymology , Synaptic Transmission/physiology , Adaptation, Physiological/genetics , Animals , Brain/enzymology , Cell Polarity/genetics , Cell Polarity/physiology , Dendrites/enzymology , Dendrites/pathology , Genome, Human/genetics , Genome, Human/physiology , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/physiology , Glutamic Acid/physiology , Humans , Mice , Mice, Inbred C57BL , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/pathology , Spinal Cord/enzymology , Synaptic Transmission/genetics , Up-Regulation/genetics , Up-Regulation/physiology
17.
Biochem Soc Trans ; 39(2): 425-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21428913

ABSTRACT

The guiding principle of the IAS Medal Lecture and of the research it covered was that searching mathematical analysis, depending on good measurements, must underpin sound biochemical conclusions. This was illustrated through various experiences with the amino acid dehydrogenases. Topics covered in the present article include: (i) the place of kinetic measurement in assessing the metabolic role of GDH (glutamate dehydrogenase); (ii) the discovery of complex regulatory behaviour in mammalian GDH, involving negative co-operativity in coenzyme binding; (iii) an X-ray structure solution for a bacterial GDH providing insight into catalysis; (iv) almost total positive co-operativity in glutamate binding to clostridial GDH; (v) unexpected outcomes with mutations at the catalytic aspartate site in GDH; (vi) reactive cysteine as a counting tool in the construction of hybrid oligomers to probe the basis of allosteric interaction; (vii) tryptophan-to-phenylalanine mutations in analysis of allosteric conformational change; (viii) site-directed mutagenesis to alter substrate specificity in GDH and PheDH (phenylalanine dehydrogenase); and (ix) varying strengths of binding of the 'wrong' enantiomer in engineered mutant enzymes and implications for resolution of racemates.


Subject(s)
Amino Acid Oxidoreductases/physiology , Biochemistry/trends , Allosteric Regulation/genetics , Allosteric Regulation/physiology , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Biochemistry/methods , Catalytic Domain/genetics , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Glutamate Dehydrogenase/physiology , Humans , Life , Mutagenesis, Site-Directed , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases/physiology , Protein Multimerization/genetics , Protein Subunits/chemistry , Protein Subunits/genetics , Tryptophan/chemistry , Tryptophan/genetics , Tryptophan/physiology
18.
Neurochem Int ; 59(4): 445-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21070828

ABSTRACT

Glutamate dehydrogenase (GDH) has been extensively studied for more than 50 years. Of particular interest is the fact that, while considered by most to be a 'housekeeping' enzyme, the animal form of GDH is heavily regulated by a wide array of allosteric effectors and exhibits extensive inter-subunit communication. While the chemical mechanism for GDH has remained unchanged through epochs of evolution, it was not clear how or why animals needed to evolve such a finely tuned form of this enzyme. As reviewed here, recent studies have begun to elucidate these issues. Allosteric regulation first appears in the Ciliates and may have arisen to accommodate evolutionary changes in organelle function. The occurrence of allosteric regulation appears to be coincident with the formation of an 'antenna' like feature rising off the tops of the subunits that may be necessary to facilitate regulation. In animals, this regulation further evolved as GDH became integrated into a number of other regulatory pathways. In particular, mutations in GDH that abrogate GTP inhibition result in dangerously high serum levels of insulin and ammonium. Therefore, allosteric regulation of GDH plays an important role in insulin homeostasis. Finally, several compounds have been identified that block GDH-mediated insulin secretion that may be to not only find use in treating these insulin disorders but to kill tumors that require glutamine metabolism for cellular energy.


Subject(s)
Energy Metabolism/physiology , Evolution, Molecular , Glutamate Dehydrogenase/chemistry , Glutamate Dehydrogenase/physiology , Insulin/physiology , Allosteric Regulation/physiology , Animals , Humans , Structure-Activity Relationship
19.
Eur J Neurosci ; 32(2): 191-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20646056

ABSTRACT

Several C1q family members, especially the Cbln and C1q-like subfamilies, are highly and predominantly expressed in the central nervous system. Cbln1, a member of the Cbln subfamily, plays two unique roles at parallel fiber (PF)-Purkinje cell synapses in the cerebellum: the formation and stabilization of synaptic contact, and the control of functional synaptic plasticity by regulating the postsynaptic endocytotic pathway. The delta2 glutamate receptor (GluD2), which is predominantly expressed in Purkinje cells, plays similar critical roles in the cerebellum. In addition, viral expression of GluD2 or the application of recombinant Cbln1 induces PF-Purkinje cell synaptogenesis in vitro and in vivo. Antigen-unmasking methods were necessary to reveal the immunoreactivities for endogenous Cbln1 and GluD2 at the synaptic junction of PF synapses. We propose that Cbln1 and GluD2 are located at the synaptic cleft, where various proteins undergo intricate molecular interactions with each other, and serve as a bidirectional synaptic organizer.


Subject(s)
Complement C1q/physiology , Signal Transduction/physiology , Synapses/physiology , Animals , Cerebellum/physiology , Glutamate Dehydrogenase/physiology , Humans , Nerve Tissue Proteins/physiology , Protein Precursors/physiology
20.
PLoS Genet ; 4(8): e1000150, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18688271

ABSTRACT

Many new gene copies emerged by gene duplication in hominoids, but little is known with respect to their functional evolution. Glutamate dehydrogenase (GLUD) is an enzyme central to the glutamate and energy metabolism of the cell. In addition to the single, GLUD-encoding gene present in all mammals (GLUD1), humans and apes acquired a second GLUD gene (GLUD2) through retroduplication of GLUD1, which codes for an enzyme with unique, potentially brain-adapted properties. Here we show that whereas the GLUD1 parental protein localizes to mitochondria and the cytoplasm, GLUD2 is specifically targeted to mitochondria. Using evolutionary analysis and resurrected ancestral protein variants, we demonstrate that the enhanced mitochondrial targeting specificity of GLUD2 is due to a single positively selected glutamic acid-to-lysine substitution, which was fixed in the N-terminal mitochondrial targeting sequence (MTS) of GLUD2 soon after the duplication event in the hominoid ancestor approximately 18-25 million years ago. This MTS substitution arose in parallel with two crucial adaptive amino acid changes in the enzyme and likely contributed to the functional adaptation of GLUD2 to the glutamate metabolism of the hominoid brain and other tissues. We suggest that rapid, selectively driven subcellular adaptation, as exemplified by GLUD2, represents a common route underlying the emergence of new gene functions.


Subject(s)
Arabidopsis Proteins/metabolism , Glutamate Dehydrogenase (NADP+)/metabolism , Glutamate Dehydrogenase/physiology , Hominidae/genetics , Mitochondria/enzymology , Selection, Genetic , Amino Acid Sequence , Amino Acid Substitution , Animals , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Cell Line , Chlorocebus aethiops , Evolution, Molecular , Gene Duplication , Glutamate Dehydrogenase/chemistry , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase (NADP+)/chemistry , Glutamate Dehydrogenase (NADP+)/genetics , Hominidae/metabolism , Humans , Hylobates , Mice , Mitochondria/chemistry , Mitochondria/genetics , Molecular Sequence Data , Phylogeny , Protein Sorting Signals , Protein Transport , Sequence Alignment , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...