Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Hum Mol Genet ; 33(4): 355-373, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37944084

RESUMEN

GRID1 and GRID2 encode the enigmatic GluD1 and GluD2 proteins, which form tetrameric receptors that play important roles in synapse organization and development of the central nervous system. Variation in these genes has been implicated in neurodevelopmental phenotypes. We evaluated GRID1 and GRID2 human variants from the literature, ClinVar, and clinical laboratories and found that many of these variants reside in intolerant domains, including the amino terminal domain of both GRID1 and GRID2. Other conserved regions, such as the M3 transmembrane domain, show different intolerance between GRID1 and GRID2. We introduced these variants into GluD1 and GluD2 cDNA and performed electrophysiological and biochemical assays to investigate the mechanisms of dysfunction of GRID1/2 variants. One variant in the GRID1 distal amino terminal domain resides at a position predicted to interact with Cbln2/Cbln4, and the variant disrupts complex formation between GluD1 and Cbln2, which could perturb its role in synapse organization. We also discovered that, like the lurcher mutation (GluD2-A654T), other rare variants in the GRID2 M3 domain create constitutively active receptors that share similar pathogenic phenotypes. We also found that the SCHEMA schizophrenia M3 variant GluD1-A650T produced constitutively active receptors. We tested a variety of compounds for their ability to inhibit constitutive currents of GluD receptor variants and found that pentamidine potently inhibited GluD2-T649A constitutive channels (IC50 50 nM). These results identify regions of intolerance to variation in the GRID genes, illustrate the functional consequences of GRID1 and GRID2 variants, and suggest how these receptors function normally and in disease.


Asunto(s)
Sistema Nervioso Central , Receptores de Glutamato , Humanos , Sistema Nervioso Central/metabolismo , Mutación , Dominios Proteicos , Receptores de Glutamato/metabolismo
2.
Int J Mol Sci ; 25(10)2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38791334

RESUMEN

Human evolution is characterized by rapid brain enlargement and the emergence of unique cognitive abilities. Besides its distinctive cytoarchitectural organization and extensive inter-neuronal connectivity, the human brain is also defined by high rates of synaptic, mainly glutamatergic, transmission, and energy utilization. While these adaptations' origins remain elusive, evolutionary changes occurred in synaptic glutamate metabolism in the common ancestor of humans and apes via the emergence of GLUD2, a gene encoding the human glutamate dehydrogenase 2 (hGDH2) isoenzyme. Driven by positive selection, hGDH2 became adapted to function upon intense excitatory firing, a process central to the long-term strengthening of synaptic connections. It also gained expression in brain astrocytes and cortical pyramidal neurons, including the CA1-CA3 hippocampal cells, neurons crucial to cognition. In mice transgenic for GLUD2, theta-burst-evoked long-term potentiation (LTP) is markedly enhanced in hippocampal CA3-CA1 synapses, with patch-clamp recordings from CA1 pyramidal neurons revealing increased sNMDA receptor currents. D-lactate blocked LTP enhancement, implying that glutamate metabolism via hGDH2 potentiates L-lactate-dependent glia-neuron interaction, a process essential to memory consolidation. The transgenic (Tg) mice exhibited increased dendritic spine density/synaptogenesis in the hippocampus and improved complex cognitive functions. Hence, enhancement of neuron-glia communication, via GLUD2 evolution, likely contributed to human cognitive advancement by potentiating synaptic plasticity and inter-neuronal connectivity.


Asunto(s)
Cognición , Glutamato Deshidrogenasa , Ácido Glutámico , Plasticidad Neuronal , Animales , Humanos , Ácido Glutámico/metabolismo , Cognición/fisiología , Glutamato Deshidrogenasa/metabolismo , Glutamato Deshidrogenasa/genética , Ratones , Ácido Láctico/metabolismo , Potenciación a Largo Plazo , Ratones Transgénicos , Células Piramidales/metabolismo , Hipocampo/metabolismo , Evolución Molecular , Sinapsis/metabolismo
3.
Int J Mol Sci ; 25(8)2024 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-38673928

RESUMEN

There are two paralogs of glutamate dehydrogenase (GDH) in humans encoded by the GLUD1 and GLUD2 genes as a result of a recent retroposition during the evolution of primates. The two human GDHs possess significantly different regulation by allosteric ligands, which is not fully characterized at the structural level. Recent advances in identification of the GDH ligand binding sites provide a deeper perspective on the significance of the accumulated substitutions within the two GDH paralogs. In this review, we describe the evolution of GLUD1 and GLUD2 after the duplication event in primates using the accumulated sequencing and structural data. A new gibbon GLUD2 sequence questions the indispensability of ancestral R496S and G509A mutations for GLUD2 irresponsiveness to GTP, providing an alternative with potentially similar regulatory features. The data of both GLUD1 and GLUD2 evolution not only confirm substitutions enhancing GLUD2 mitochondrial targeting, but also reveal a conserved mutation in ape GLUD1 mitochondrial targeting sequence that likely reduces its transport to mitochondria. Moreover, the information of GDH interactors, posttranslational modification and subcellular localization are provided for better understanding of the GDH mutations. Medically significant point mutations causing deregulation of GDH are considered from the structural and regulatory point of view.


Asunto(s)
Evolución Molecular , Glutamato Deshidrogenasa , Procesamiento Proteico-Postraduccional , Animales , Humanos , Glutamato Deshidrogenasa/metabolismo , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/química , Ligandos , Mutación , Primates/genética
4.
J Struct Biol ; 211(2): 107546, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32512155

RESUMEN

GluD2 receptor belongs to the orphan delta family of glutamate receptor ion channels. These receptors play key roles in synaptogenesis and synaptic plasticity and are associated with multiple neuronal disorders like schizophrenia, autism spectrum disorder, cerebellar ataxia, intellectual disability, paraplegia, retinal dystrophy, etc. Despite the importance of these receptors in CNS, insights into full-length GluD2 receptor structure is missing till-date. Here we report cryo-electron microscopy structure of the rat GluD2 receptor in the presence of calcium ions and the ligand 7-chlorokynurenic acid, elucidating its 3D architecture. The structure reveals a non-swapped architecture at the extracellular amino-terminal (ATD), and ligand-binding domain (LBD) interface similar to that observed in GluD1; however, the organization and arrangement of the ATD and LBD domains in GluD2 are unique. While our results demonstrate that non-swapped architecture is conserved in the delta receptor family, they also highlight the differences that exist between the two member receptors; GluD1 and GluD2.


Asunto(s)
Microscopía por Crioelectrón , Neuronas/ultraestructura , Receptores de Glutamato/ultraestructura , Receptores Ionotrópicos de Glutamato/ultraestructura , Animales , Sistema Nervioso Central/patología , Sistema Nervioso Central/ultraestructura , Humanos , Ligandos , Plasticidad Neuronal/genética , Neuronas/metabolismo , Neuronas/patología , Dominios Proteicos/genética , Ratas , Receptores de Glutamato/genética , Receptores Ionotrópicos de Glutamato/genética
5.
Neurochem Res ; 44(1): 154-169, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29777493

RESUMEN

Human evolution is characterized by brain expansion and up-regulation of genes involved in energy metabolism and synaptic transmission, including the glutamate signaling pathway. Glutamate is the excitatory transmitter of neural circuits sub-serving cognitive functions, with glutamate-modulation of synaptic plasticity being central to learning and memory. GLUD2 is a novel positively-selected human gene involved in glutamatergic transmission and energy metabolism that underwent rapid evolutionary adaptation concomitantly with prefrontal cortex enlargement. Two evolutionary replacements (Gly456Ala and Arg443Ser) made hGDH2 resistant to GTP inhibition and allowed distinct regulation, enabling enhanced enzyme function under high glutamatergic system demands. GLUD2 adaptation may have contributed to unique human traits, but evidence for this is lacking. GLUD2 arose through retro-positioning of a processed GLUD1 mRNA to the X chromosome, a DNA replication mechanism that typically generates pseudogenes. However, by finding a suitable promoter, GLUD2 is thought to have gained expression in nerve and other tissues, where it adapted to their particular needs. Here we generated GLUD2 transgenic (Tg) mice by inserting in their genome a segment of the human X chromosome, containing the GLUD2 gene and its putative promoter. Double IF studies of Tg mouse brain revealed that the human gene is expressed in the host mouse brain in a pattern similar to that observed in human brain, thus providing credence to the above hypothesis. This expressional adaptation may have conferred novel role(s) on GLUD2 in human brain. Previous observations, also in GLUD2 Tg mice, generated and studied independently, showed that the non-redundant function of hGDH2 is markedly activated during early post-natal brain development, contributing to developmental changes in prefrontal cortex similar to those attributed to human divergence. Hence, GLUD2 adaptation may have influenced the evolutionary course taken by the human brain, but understanding the mechanism(s) involved remains challenging.


Asunto(s)
Adaptación Fisiológica/fisiología , Encéfalo/fisiología , Evolución Molecular , Glutamato Deshidrogenasa/biosíntesis , Heterocigoto , Animales , Expresión Génica , Glutamato Deshidrogenasa/química , Glutamato Deshidrogenasa/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Estructura Secundaria de Proteína , Cromosoma X/genética
6.
Proc Natl Acad Sci U S A ; 113(19): 5358-63, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27118840

RESUMEN

Whereas all mammals have one glutamate dehydrogenase gene (GLUD1), humans and apes carry an additional gene (GLUD2), which encodes an enzyme with distinct biochemical properties. We inserted a bacterial artificial chromosome containing the human GLUD2 gene into mice and analyzed the resulting changes in the transcriptome and metabolome during postnatal brain development. Effects were most pronounced early postnatally, and predominantly genes involved in neuronal development were affected. Remarkably, the effects in the transgenic mice partially parallel the transcriptome and metabolome differences seen between humans and macaques analyzed. Notably, the introduction of GLUD2 did not affect glutamate levels in mice, consistent with observations in the primates. Instead, the metabolic effects of GLUD2 center on the tricarboxylic acid cycle, suggesting that GLUD2 affects carbon flux during early brain development, possibly supporting lipid biosynthesis.


Asunto(s)
Encéfalo/metabolismo , Ciclo del Ácido Cítrico/fisiología , Glutamato Deshidrogenasa/metabolismo , Ácido Glutámico/metabolismo , Metaboloma/fisiología , Transcriptoma/fisiología , Animales , Glutamato Deshidrogenasa/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Proteínas Recombinantes/metabolismo , Especificidad de la Especie
7.
Int J Mol Sci ; 20(7)2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30979012

RESUMEN

Staufen2 (Stau2) is an RNA-binding protein that is involved in dendritic spine morphogenesis and function. Several studies have recently investigated the role of Stau2 in the regulation of its neuronal target mRNAs, with particular focus on the hippocampus. Here, we provide evidence for Stau2 expression and function in cerebellar Purkinje cells. We show that Stau2 downregulation (Stau2GT) led to an increase of glutamate receptor ionotropic delta subunit 2 (GluD2) in Purkinje cells when animals performed physical activity by voluntary wheel running compared with the age-matched wildtype (WT) mice (C57Bl/6J). Furthermore, Stau2GT mice showed lower performance in motor coordination assays but enhanced motor learning abilities than did WT mice, concomitantly with an increase in dendritic GluD2 expression. Together, our results suggest the novel role of Stau2 in Purkinje cell synaptogenesis in the mouse cerebellum.


Asunto(s)
Envejecimiento , Encéfalo/fisiología , Regulación de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Células de Purkinje/metabolismo , Proteínas de Unión al ARN/genética , Receptores de Glutamato/genética , Animales , Cerebelo/citología , Cerebelo/fisiología , Femenino , Eliminación de Gen , Masculino , Ratones Endogámicos C57BL , Actividad Motora , Células de Purkinje/citología , ARN Mensajero/genética , Receptores de Glutamato/análisis
8.
J Neurosci ; 36(48): 12129-12143, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27903723

RESUMEN

Shank2 is a multidomain scaffolding protein implicated in the structural and functional coordination of multiprotein complexes at excitatory postsynaptic sites as well as in psychiatric disorders, including autism spectrum disorders. While Shank2 is strongly expressed in the cerebellum, whether Shank2 regulates cerebellar excitatory synapses, or contributes to the behavioral abnormalities observed in Shank2-/- mice, remains unexplored. Here we show that Shank2-/- mice show reduced excitatory synapse density in cerebellar Purkinje cells in association with reduced levels of excitatory postsynaptic proteins, including GluD2 and PSD-93, and impaired motor coordination in the Erasmus test. Shank2 deletion restricted to Purkinje cells (Pcp2-Cre;Shank2fl/fl mice) leads to similar reductions in excitatory synapse density, synaptic protein levels, and motor coordination. Pcp2-Cre;Shank2fl/fl mice do not recapitulate autistic-like behaviors observed in Shank2-/- mice, such as social interaction deficits, altered ultrasonic vocalizations, repetitive behaviors, and hyperactivity. However, Pcp2-Cre;Shank2fl/fl mice display enhanced repetitive behavior in the hole-board test and anxiety-like behavior in the light-dark test, which are not observed in Shank2-/- mice. These results implicate Shank2 in the regulation of cerebellar excitatory synapse density, motor coordination, and specific repetitive and anxiety-like behaviors. SIGNIFICANCE STATEMENT: The postsynaptic side of excitatory synapses contains multiprotein complexes, termed the postsynaptic density, which contains receptors, scaffolding/adaptor proteins, and signaling molecules. Shank2 is an excitatory postsynaptic scaffolding protein implicated in the formation and functional coordination of the postsynaptic density and has been linked to autism spectrum disorders. Using Shank2-null mice and Shank2-conditional knock-out mice with a gene deletion restricted to cerebellar Purkinje cells, we explored functions of Shank2 in the cerebellum. We found that Shank2 regulates excitatory synapse density, motor coordination, and specific repetitive and anxiety-like behaviors, but is not associated with autistic-like social deficits or repetitive behaviors.


Asunto(s)
Ansiedad/fisiopatología , Cerebelo/fisiopatología , Trastornos de Traumas Acumulados/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Desempeño Psicomotor/fisiología , Sinapsis/patología , Animales , Conducta Animal/fisiología , Recuento de Células , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Sinapsis/fisiología
9.
J Neurosci ; 36(17): 4846-58, 2016 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-27122040

RESUMEN

UNLABELLED: Although injured axons usually do not regenerate in the adult CNS, parallel fibers (PFs) regenerate synaptic connections onto cerebellar Purkinje cells (PCs). In this study, we investigated the role of GluD2 in this regenerative process after PF transection using GluD2-knock-out (KO) mice. All dendritic spines on distal dendrites were innervated by PFs in sham-operated wild-type controls, whereas one-third were devoid of innervation in GluD2-KO mice. In both genotypes, a steep drop in the number of PF synapses occurred with a reciprocal surge in the number of free spines on postlesion day 1, when the PF territory aberrantly expanded toward the proximal dendrites. In wild-type mice, the territory and number of PF synapses were nearly fully restored to normal on postlesion day 7, although PF density remained low. Moreover, presynaptic and postsynaptic elements were markedly enlarged, and the PF terminal-to-PC spine contact ratio increased from 1:1 to 1:2 at most synapses. On postlesion day 30, the size and contact ratio of PF synapses returned to sham-operated control values and PF density recovered through the sprouting and elongation of PF collaterals. However, GluD2-KO mice showed neither a hypertrophic response nor territorial restoration 7 d postlesion, nor the recovery of PF axons or synapses on postlesion day 30. This suggests that PF wiring regenerates initially by inducing hypertrophic responses in surviving synaptic elements (hypertrophic phase), followed by collateral formation by PF axons and retraction of PF synapses (remodeling phase). Without GluD2, no transition to these regenerative phases occurs. SIGNIFICANCE STATEMENT: The glutamate receptor GluD2 expressed at parallel fiber (PF)-Purkinje cell (PC) synapses regulates the formation and maintenance of the synapses. To investigate the role of GluD2 in their extraordinarily high regenerative capacity, the process after surgical transection of PFs was compared between wild-type and GluD2-knock-out mice. We discovered that, in wild-type mice, PF synapses regenerate initially by inducing hypertrophic responses in surviving synaptic elements, and then by sprouting and elongation of PF collaterals. Subsequently, hypertrophied PF synapses remodel into compact synapses. In GluD2-knock-out mice, PF wiring remains in the degenerative phase, showing neither a hypertrophic response nor recovery of PF axons or synapses. Our finding thus highlights that synaptic connection in the adult brain can regenerate with aid of GluD2.


Asunto(s)
Cerebelo/citología , Fibras Nerviosas/fisiología , Plasticidad Neuronal , Células de Purkinje/fisiología , Receptores de Glutamato , Sinapsis/fisiología , Animales , Axones/metabolismo , Axones/ultraestructura , Dendritas/metabolismo , Dendritas/ultraestructura , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Células de Purkinje/ultraestructura , Sinapsis/ultraestructura
10.
BMC Med Genet ; 18(1): 144, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29207948

RESUMEN

BACKGROUND: Spinocerebellar ataxias comprise a large and heterogeneous group of disorders that may present with isolated ataxia, or ataxia in combination with other neurologic or non-neurologic symptoms. Monoallelic or biallelic GRID2 mutations were recently reported in rare cases with cerebellar syndrome and variable degree of ataxia, ocular symptoms, hypotonia and developmental delay. CASE PRESENTATION: We report on a consanguineous family with autosomal recessive childhood onset of slowly progressive cerebellar ataxia and delayed psychomotor development in three siblings. MRI of an adult and affected family member revealed slightly widened cerebral and cerebellar sulci, suggesting generalized brain atrophy, and mild cerebellar atrophy. Using whole exome sequencing we identified a novel homozygous missense variant [c.2128C > T, p.(Arg710Trp)] in GRID2 that segregates with the disease. The missense variant is located in a conserved region encoding the extracellular serine-binding domain of the GluD2 protein and predicts a change in conformation of the protein. CONCLUSION: The widespread supratentorial brain abnormalities, absence of oculomotor symptoms, increased peripheral muscle tone and the novel missense mutation add to the clinical and genetic variability in GRID2 associated cerebellar syndrome. The neuroradiological findings in our family indicate a generalized neurodegenerative process to be taken into account in other families segregating complex clinical features and GRID2 mutations.


Asunto(s)
Encéfalo/patología , Mutación Missense , Receptores de Glutamato/genética , Adulto , Anciano , Secuencia de Aminoácidos , Atrofia , Secuencia de Bases , Sitios de Unión , Encéfalo/diagnóstico por imagen , Ataxia Cerebelosa/genética , Cerebelo/diagnóstico por imagen , Cerebelo/patología , Consanguinidad , Discapacidades del Desarrollo/genética , Femenino , Homocigoto , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Modelos Moleculares , Linaje , Conformación Proteica , Dominios Proteicos , Receptores de Glutamato/química , Receptores de Glutamato/metabolismo , Serina/metabolismo , Secuenciación del Exoma
11.
Neurochem Res ; 42(1): 92-107, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27422263

RESUMEN

Mammalian glutamate dehydrogenase1 (GDH1) (E.C. 1.4.1.3) is a mitochondrial enzyme that catalyzes the reversible oxidative deamination of glutamate to α-ketoglutarate and ammonia while reducing NAD+ and/or NADP+ to NADH and/or NADPH. It links amino acid with carbohydrate metabolism, contributing to Krebs cycle anaplerosis, energy production, ammonia handling and redox homeostasis. Although GDH1 was one of the first major metabolic enzymes to be studied decades ago, its role in cell biology is still incompletely understood. There is however growing interest in a novel GDH2 isoenzyme that emerged via duplication in primates and underwent rapid evolutionary selection concomitant with prefrontal human cortex expansion. Also, the anaplerotic function of GDH1 and GDH2 is currently under sharp focus as this relates to the biology of glial tumors and other neoplasias. Here we used antibodies specific for human GDH1 (hGDH1) and human GDH2 (hGDH2) to study the expression of these isoenzymes in human tissues. Results revealed that both hGDH1 and hGDH2 are expressed in human brain, kidney, testis and steroidogenic organs. However, distinct hGDH1 and hGDH2 expression patterns emerged. Thus, while the Sertoli cells of human testis were strongly positive for hGDH2, they were negative for hGDH1. Conversely, hGDH1 showed very high levels of expression in human liver, but hepatocytes were virtually devoid of hGDH2. In human adrenals, both hGDHs were densely expressed in steroid-producing cells, with hGDH2 expression pattern matching that of the cholesterol side chain cleavage system involved in steroid synthesis. Similarly in human ovaries and placenta, both hGDH1 and hGDH2 were densely expressed in estrogen producing cells. In addition, hGDH1, being a housekeeping enzyme, was also expressed in cells that lack endocrine function. Regarding human brain, study of cortical sections using immunofluorescence (IF) with confocal microscopy revealed that hGDH1 and hGDH2 were both expressed in the cytoplasm of gray and white matter astrocytes within coarse structures resembling mitochondria. Additionally, hGDH1 localized to the nuclear membrane of a subpopulation of astrocytes and of the vast majority of oligodendrocytes and their precursors. Remarkably, hGDH2-specific staining was detected in human cortical neurons, with different expression patterns having emerged. One pattern, observed in large cortical neurons (some with pyramidal morphology), was a hGDH2-specific labeling of cytoplasmic structures resembling mitochondria. These were distributed either in the cell body-axon or on the cell surface in close proximity to astrocytic end-feet that encircle glutamatergic synapses. Another pattern was observed in small cortical neurons with round dense nuclei in which the hGDH2-specific staining was found in the nuclear membrane. A detailed description of these observations and their functional implications, suggesting that the GDH flux is used by different cells to serve some of their unique functions, is presented below.


Asunto(s)
Cuerpo Celular/enzimología , Regulación Enzimológica de la Expresión Génica , Glutamato Deshidrogenasa/biosíntesis , Espacio Intracelular/enzimología , Secuencia de Aminoácidos , Encéfalo/enzimología , Cuerpo Celular/genética , Glutamato Deshidrogenasa/genética , Humanos , Espacio Intracelular/genética , Riñón/enzimología , Hígado/enzimología , Masculino , Testículo/enzimología
12.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 4): 587-94, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23519667

RESUMEN

PSD-93 (chapsyn-110, DLG2) is a member of the family of membrane-associated guanylate kinase (MAGUK) proteins. The MAGUK proteins are involved in receptor localization and signalling pathways. The best characterized MAGUK protein, PSD-95, is known to be involved in NMDA receptor signalling via its PDZ domains. The PDZ domains of PSD-95 and PSD-93 are structurally very similar, but relatively little is known about the function of PSD-93. PSD-93 has been suggested to interact with GluD2 from the family of ionotropic glutamate receptors. Here, the interactions of four residues (GTSI) representing the extreme C-terminus of GluD2 with PSD-93 PDZ1 have been investigated in the crystalline phase. Two different binding modes of these residues were observed, suggesting that the peptide is not tightly bound to PSD-93 PDZ1. In accordance, the two N-terminal PSD-93 PDZ domains show no appreciable binding affinity for a GluD2-derived C-terminal octapeptide, whereas micromolar affinity was observed for a GluN2B-derived C-terminal octapeptide. This indicates that if present, the interactions between GluD2 and PSD-93 involve more than the extreme terminus of the receptor. In contrast, the tumour-suppressor protein SCRIB PDZ3 shows low micromolar affinity towards the GluD2-derived octapeptide, which is in agreement with previous findings using high-throughput assays.


Asunto(s)
Guanilato-Quinasas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteínas Supresoras de Tumor/metabolismo , Comunicación Celular/fisiología , Cristalización , Cristalografía por Rayos X , Polarización de Fluorescencia , Guanilato-Quinasas/biosíntesis , Guanilato-Quinasas/química , Humanos , Microscopía de Fluorescencia por Excitación Multifotónica , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Estructura Terciaria de Proteína , Espectrometría de Fluorescencia , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/química
13.
Front Cell Dev Biol ; 11: 1281487, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38020911

RESUMEN

Glutamate dehydrogenases are enzymes that take part in both amino acid and energy metabolism. Their role is clear in many biological processes, from neuronal function to cancer development. The putative testis-specific Drosophila glutamate dehydrogenase, Bb8, is required for male fertility and the development of mitochondrial derivatives in spermatids. Testis-specific genes are less conserved and could gain new functions, thus raising a question whether Bb8 has retained its original enzymatic activity. We show that while Bb8 displays glutamate dehydrogenase activity, there are significant functional differences between the housekeeping Gdh and the testis-specific Bb8. Both human GLUD1 and GLUD2 can rescue the bb8 ms mutant phenotype, with superior performance by GLUD2. We also tested the role of three conserved amino acids observed in both Bb8 and GLUD2 in Gdh mutants, which showed their importance in the glutamate dehydrogenase function. The findings of our study indicate that Drosophila Bb8 and human GLUD2 could be novel examples of convergent molecular evolution. Furthermore, we investigated the importance of glutamate levels in mitochondrial homeostasis during spermatogenesis by ectopic expression of the mitochondrial glutamate transporter Aralar1, which caused mitochondrial abnormalities in fly spermatids. The data presented in our study offer evidence supporting the significant involvement of glutamate metabolism in sperm development.

14.
Neuron ; 109(4): 629-644.e8, 2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33352118

RESUMEN

The synaptotrophic hypothesis posits that synapse formation stabilizes dendritic branches, but this hypothesis has not been causally tested in vivo in the mammalian brain. The presynaptic ligand cerebellin-1 (Cbln1) and postsynaptic receptor GluD2 mediate synaptogenesis between granule cells and Purkinje cells in the molecular layer of the cerebellar cortex. Here we show that sparse but not global knockout of GluD2 causes under-elaboration of Purkinje cell dendrites in the deep molecular layer and overelaboration in the superficial molecular layer. Developmental, overexpression, structure-function, and genetic epistasis analyses indicate that these dendrite morphogenesis defects result from a deficit in Cbln1/GluD2-dependent competitive interactions. A generative model of dendrite growth based on competitive synaptogenesis largely recapitulates GluD2 sparse and global knockout phenotypes. Our results support the synaptotrophic hypothesis at initial stages of dendrite development, suggest a second mode in which cumulative synapse formation inhibits further dendrite growth, and highlight the importance of competition in dendrite morphogenesis.


Asunto(s)
Cerebelo/citología , Cerebelo/metabolismo , Dendritas/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Precursores de Proteínas/deficiencia , Células de Purkinje/metabolismo , Receptores de Glutamato/deficiencia , Animales , Dendritas/genética , Femenino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Embarazo , Unión Proteica/fisiología , Precursores de Proteínas/genética , Receptores de Glutamato/genética
15.
J Comp Neurol ; 528(6): 1003-1027, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31625608

RESUMEN

In the cerebellum, GluD2 is exclusively expressed in Purkinje cells, where it regulates synapse formation and regeneration, synaptic plasticity, and motor learning. Delayed cognitive development in humans with GluD2 gene mutations suggests extracerebellar functions of GluD2. However, extracerebellar expression of GluD2 and its relationship with that of GluD1 are poorly understood. GluD2 mRNA and protein were widely detected, with relatively high levels observed in the olfactory glomerular layer, medial prefrontal cortex, cingulate cortex, retrosplenial granular cortex, olfactory tubercle, subiculum, striatum, lateral septum, anterodorsal thalamic nucleus, and arcuate hypothalamic nucleus. These regions were also enriched for GluD1, and many individual neurons coexpressed the two GluDs. In the retrosplenial granular cortex, GluD1 and GluD2 were selectively expressed at PSD-95-expressing glutamatergic synapses, and their coexpression on the same synapses was shown by SDS-digested freeze-fracture replica labeling. Biochemically, GluD1 and GluD2 formed coimmunoprecipitable complex formation in HEK293T cells and in the cerebral cortex and hippocampus. We further estimated the relative protein amount by quantitative immunoblotting using GluA2/GluD2 and GluA2/GluD1 chimeric proteins as standards for titration of GluD1 and GluD2 antibodies. Intriguingly, the relative amount of GluD2 was almost comparable to that of GluD1 in the postsynaptic density fraction prepared from the cerebral cortex and hippocampus. In contrast, GluD2 was overwhelmingly predominant in the cerebellum. Thus, we have determined the relative extracerebellar expression of GluD1 and GluD2 at regional, neuronal, and synaptic levels. These data provide a molecular-anatomical basis for possible competitive and cooperative interactions of GluD family members at synapses in various brain regions.


Asunto(s)
Encéfalo/metabolismo , Glutamato Deshidrogenasa/metabolismo , Receptores de Glutamato/metabolismo , Animales , Perfilación de la Expresión Génica/métodos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL
16.
Metabolism ; 100: 153958, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31400387

RESUMEN

Glutamate dehydrogenase 1 (GDH1) contributes to glucose-stimulated insulin secretion in murine ß-cells, but not to basic insulin release. The implications of these findings for human biology are unclear as humans have two GDH-specific enzymes: hGDH1 (GLUD1-encoded) and hGDH2 (GLUD2-encoded), a novel enzyme that is highly activated by ADP and L-leucine. Here we studied in vivo glucose homeostasis in transgenic (Tg) mice generated by inserting the GLUD2 gene and its putative regulatory elements into their genome. Using specific antibodies, we observed that hGDH2 was co-expressed with the endogenous murine GDH1 in pancreatic ß-cells of Tg mice. Fasting blood glucose (FBG) levels were lower and of a narrower range in Tg (95% CI: 90.6-96.8 mg/dl; N = 26) than in Wt mice (95% CI: 136.2-151.4 mg/dl; N = 23; p < 0.0001), closely resembling those of healthy humans. GLUD2 also protected the host mouse from developing diabetes with advancing age. Tg animals maintained 2.6-fold higher fasting serum insulin levels (mean ±â€¯SD: 1.63 ±â€¯0.15 ng/ml; N = 12) than Wt mice (0.63 ±â€¯0.05 ng/ml; N = 12; p < 0.0001). Glucose loading (1 mg/g, given i.p.) induced comparable serum insulin increases in Tg and Wt mice, suggesting no significant GLUD2 effect on glucose-stimulated insulin release. L-leucine (0.25 mg/g given orally) induced a 2-fold increase in the serum insulin of the Wt mice, implying significant activation of the endogenous GDH1. However, L-leucine had little effect on the high insulin levels of the Tg mice, suggesting that, under the high ADP levels that prevail in ß-cells in the fasting state, glutamate flux through hGDH2 is close to maximal. Hence, the present data, showing that GLUD2 expression in Tg mice improves in vivo glucose homeostasis by boosting fasting serum insulin levels, suggest that evolutionary adaptation of hGDH2 has enabled humans to achieve narrow-range euglycemia by regulating glutamate-mediated basal insulin secretion.


Asunto(s)
Glucosa/metabolismo , Glutamato Deshidrogenasa/genética , Homeostasis , Secreción de Insulina , Animales , Glucosa 1-Deshidrogenasa/metabolismo , Humanos , Ratones , Ratones Transgénicos
17.
EBioMedicine ; 37: 56-67, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30314897

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most frequent and malignant primary brain tumor in adults and despite the progress in surgical procedures and therapy options, the overall survival remains very poor. Glutamate and α-KG are fundamental elements necessary to support the growth and proliferation of GBM cells. Glutamate oxidative deamination, catalyzed by GLUD2, is the predominant pathway for the production of α-KG. METHODS: GLUD2 emerged from the RNA-seq analysis of 13 GBM patients, performed in our laboratory and a microarray analysis of 77 high-grade gliomas available on the Geo database. Thereafter, we investigated GLUD2 relevance in cancer cell behavior by GLUD2 overexpression and silencing in two different human GBM cell lines. Finally, we overexpressed GLUD2 in-vivo by using zebrafish embryos and monitored the developing central nervous system. FINDINGS: GLUD2 expression was found associated to the histopathological classification, prognosis and survival of GBM patients. Moreover, through in-vitro functional studies, we showed that differences in GLUD2 expression level affected cell proliferation, migration, invasion, colony formation abilities, cell cycle phases, mitochondrial function and ROS production. In support of these findings, we also demonstrated, with in-vivo studies, that GLUD2 overexpression affects glial cell proliferation without affecting neuronal development in zebrafish embryos. INTERPRETATION: We concluded that GLUD2 overexpression inhibited GBM cell growth suggesting a novel potential drug target for control of GBM progression. The possibility to enhance GLUD2 activity in GBM could result in a blocked/reduced proliferation of GBM cells without affecting the survival of the surrounding neurons.


Asunto(s)
Glioblastoma/metabolismo , Glutamato Deshidrogenasa/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Glioblastoma/genética , Glioblastoma/patología , Glutamato Deshidrogenasa/genética , Humanos , Proteínas Mitocondriales/genética , Proteínas de Neoplasias/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
18.
Neuropharmacology ; 115: 92-99, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27276689

RESUMEN

The orphan Glutamate receptor Delta2 (GluD2) intrinsic ion channel activity is indirectly triggered by glutamate through stimulation of the metabotropic glutamate receptor 1 (mGlu1), in cerebellar Purkinje cells. However, the mechanisms of GluD2 ion channel opening are entirely unknown. In this work, we investigated the signaling pathways underlying the mGlu1-induced GluD2 current, performing whole-cell voltage-clamp recordings from mGlu1 and GluD2 transfected HEK293 cells. We show that the activation of GluD2 channels via DHPG-induced mGlu1 stimulation is Gαq-dependent. Moreover, inhibition of the downstream components of the mGlu1 canonical signaling pathway PLC and PKC with U73122 and GF109203X, respectively, strongly reduced the DHPG-induced GluD2 current. These results were further confirmed on endogenous receptors at the Parallel Fiber - Purkinje Cell cerebellar synapse, indicating that the opening of the GluD2 channel by mGlu1 receptor mobilizes the canonical Gq-PLC-PKC pathway. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.


Asunto(s)
Receptores de Glutamato/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Transducción de Señal/fisiología , Animales , Cerebelo/efectos de los fármacos , Cerebelo/fisiología , Estrenos/farmacología , Células HEK293 , Humanos , Indoles/farmacología , Maleimidas/farmacología , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Pirrolidinonas/farmacología , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
19.
Biol Trace Elem Res ; 173(2): 465-74, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27033232

RESUMEN

Iodine is a significant micronutrient. Iodine deficiency (ID)-induced hypothyroxinemia and hypothyroidism during developmental period can cause cerebellar dysfunction. However, mechanisms are still unclear. Therefore, the present research aims to study effects of developmental hypothyroxinemia caused by mild ID and hypothyroidism caused by severe ID or methimazole (MMZ) on parallel fiber-Purkinje cell (PF-PC) synapses in filial cerebellum. Maternal hypothyroxinemia and hypothyroidism models were established in Wistar rats using ID diet and deionized water supplemented with different concentrations of potassium iodide or MMZ water. Birth weight and cerebellum weight were measured. We also examined PF-PC synapses using immunofluorescence, and western blot analysis was conducted to investigate the activity of Neurexin1/cerebellin1 (Cbln1)/glutamate receptor d2 (GluD2) tripartite complex. Our results showed that hypothyroxinemia and hypothyroidism decreased birth weight and cerebellum weight and reduced the PF-PC synapses on postnatal day (PN) 14 and PN21. Accordingly, the mean intensity of vesicular glutamate transporter (VGluT1) and Calbindin immunofluorescence was reduced in mild ID, severe ID, and MMZ groups. Moreover, maternal hypothyroxinemia and hypothyroidism reduced expression of Neurexin1/Cbln1/GluD2 tripartite complex. Our study supports the hypothesis that developmental hypothyroxinemia and hypothyroidism reduce PF-PC synapses, which may be attributed to the downregulation of Neurexin1/Cbln1/GluD2 tripartite complex.


Asunto(s)
Regulación hacia Abajo , Glutamato Deshidrogenasa/biosíntesis , Hipotiroidismo/metabolismo , Yodo/deficiencia , Complejos Multiproteicos/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Precursores de Proteínas/biosíntesis , Células de Purkinje/metabolismo , Receptores de Superficie Celular/biosíntesis , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Femenino , Hipotiroidismo/patología , Células de Purkinje/patología , Ratas , Ratas Wistar , Sinapsis/patología
20.
Commun Integr Biol ; 6(6): e26466, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24563706

RESUMEN

Long-term changes in synaptic transmission in the central nervous system, such as long-term potentiation and long-term depression (LTD), are believed to underlie learning and memory in vivo. Despite intensive research, the precise molecular mechanisms underlying these phenomena have remained unclear. LTD is most commonly caused by the endocytosis of postsynaptic AMPA-type glutamate receptors, triggered by activity-induced serine phosphorylation of the GluA2 subunit. Interestingly, cerebellar LTD, which occurs at synapses between parallel fibers (PFs; axons of granule cells) and Purkinje cells, is unique in requiring an additional type of glutamate receptor, the δ2 receptor (GluD2). Cbln1 was recently identified as a GluD2 ligand that regulates PF synapse formation and maintenance. However, how GluD2 induces downstream signaling in Purkinje cells to regulate LTD induction is unknown. We here present evidence that GluD2 reduces the tyrosine phosphorylation level of the GluA2 subunit via PTPMEG, a protein tyrosine phosphatase that binds to GluD2's C-terminus. We also found that the serine phosphorylation of GluA2, a crucial step for AMPA-receptor endocytosis, requires prior tyrosine dephosphorylation. Thus, GluD2 may serve as a gatekeeper for LTD induction by coordinating interactions between GluA2's 2 phosphorylation sites.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda