GLUD1 Human

Glutamate Dehydrogenase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to GLUD1 Human

GLUD1 (glutamate dehydrogenase 1) is a mitochondrial matrix enzyme catalyzing the oxidative deamination of L-glutamate to α-ketoglutarate and ammonia. It serves as a critical regulator of nitrogen metabolism, energy homeostasis, and insulin secretion. Expressed predominantly in liver, pancreas, brain, and kidney, GLUD1 is ubiquitously involved in metabolic pathways but absent in muscle tissue . Mutations in GLUD1 are linked to hyperinsulinism-hyperammonemia syndrome (HI/HA), while its downregulation is associated with renal cancer progression .

Molecular Structure and Gene Organization

  • Location: Chromosome 10q23.2 (human), spans 13 exons.

  • Paralogs: GLUD2 (intronless, X-linked) originated via retrotransposition of GLUD1 .

  • Alternative Splicing: Generates 27 transcript variants, including non-coding forms .

Catalytic Activity

GLUD1 reversibly converts glutamate to α-ketoglutarate, with equilibrium favoring glutamate synthesis under standard conditions. Key reactions:

  • Oxidative deamination: Glutamate → α-ketoglutarate + NH₄⁺ (NAD⁺/NADP⁺-dependent).

  • Reductive amination: α-ketoglutarate → glutamate (reverse reaction) .

Allosteric Regulation

GLUD1 activity is modulated by energy-sensing molecules:

RegulatorEffectMechanism
ADPActivationDestabilizes abortive complexes, opens catalytic cleft .
GTPInhibitionStabilizes closed conformation, slows product release .
LeucineActivationBinds at subunit interface, enhances substrate affinity .
ATPDual role (inhibition/activation)Low [ATP]: inhibits via GTP site; high [ATP]: weak inhibition .

Hyperinsulinism-Hyperammonemia Syndrome (HI/HA)

  • Causes: Dominant GLUD1 mutations (e.g., R496H, H454Y) reduce GTP sensitivity, increasing insulin secretion and ammonia levels .

  • Symptoms: Neonatal hypoglycemia, hyperammonemia, intellectual disability .

Renal Cell Carcinoma (RCC)

Recent studies highlight GLUD1’s tumor-suppressive role in clear cell RCC (ccRCC):

FindingMechanismClinical Correlation
GLUD1 downregulationPromoter hypermethylationPoor prognosis, advanced tumor stages .
PI3K/Akt/mTOR inhibitionReduced cell proliferation/migrationEnhanced sensitivity to tyrosine kinase inhibitors (TKIs) .
Immune Microenvironment (TIME) suppressionLower immunosuppressive markersPotential biomarker for immunotherapy response .

Recombinant GLUD1 Production

  • Source: E. coli (His-tagged, 528 amino acids).

  • Formulation: 1 mg/ml in 20 mM Tris-HCl (pH 8.0), 0.4 M urea, 10% glycerol .

PropertyDetails
Molecular Weight58.4 kDa (monomer)
PuritySterile filtered, non-glycosylated
StabilityStable in urea/glycerol buffer

Tumor-Suppressive Potential in ccRCC

GLUD1 overexpression suppresses ccRCC progression via:

  1. Metabolic Reprogramming: Reduces glutamine dependency.

  2. Pathway Inhibition: Blocks PI3K/Akt/mTOR signaling.

  3. Epigenetic Modulation: Targets promoter methylation for therapeutic intervention .

Comparative Analysis: GLUD1 vs. GLUD2

ParameterGLUD1GLUD2
LocationMitochondrial matrixCytosol
ExpressionLiver, pancreas, brain, kidneyNervous system (neurons)
FunctionEnergy metabolism, ammonia detoxNeurotransmitter glutamate synthesis
RegulationADP, GTP, ATP, LeuNot energy-sensitive
Disease LinkHI/HA, diabetes, ccRCCNeurological disorders

Product Specs

Introduction
Glutamate dehydrogenase 1, mitochondrial precursor (GLUD1) is a member of the Glu/Leu/Phe/Val dehydrogenases family. It is a mitochondrial enzyme that catalyzes the conversion of L-glutamate to alpha-ketoglutarate, playing a crucial role in nitrogen metabolism in both plants and animals. GLUD1 is ubiquitous in organisms and its catalytic activity is essential for regulating amino acid-induced insulin secretion. Mutations in the GLUD1 gene can lead to hyperinsulinism-hyperammonemia syndrome (HHS), a genetic disorder characterized by elevated levels of insulin and ammonia in the bloodstream. GLUD1 activity is allosterically activated by ADP and inhibited by GTP and ATP.
Description
Recombinant human GLUD1 protein expressed in E. coli. This protein is a single, non-glycosylated polypeptide chain consisting of 528 amino acids (residues 54-558) with a molecular weight of 58.4 kDa. The protein includes a 23 amino acid His-tag fused at the N-terminus.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The GLUD1 solution is provided at a concentration of 1 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.4 M Urea, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein such as HSA or BSA (0.1%) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity is determined to be greater than 80.0% by SDS-PAGE analysis.
Synonyms
Glutamate Dehydrogenase 1, GLUD, GDH 1, EC 1.4.1.3, GDH, GDH1, Glutamate Dehydrogenase (NAD(P)+), Glutamate Dehydrogenase 1 Mitochondrial, EC 1.4.1, GLUD1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSSEAVADR EDDPNFFKMV EGFFDRGASI VEDKLVEDLR TRESEEQKRN RVRGILRIIK PCNHVLSLSF PIRRDDGSWE VIEGYRAQHS QHRTPCKGGI RYSTDVSVDE VKALASLMTY KCAVVDVPFG GAKAGVKINP KNYTDNELEK ITRRFTMELA KKGFIGPGID VPAPDMSTGE REMSWIADTY ASTIGHYDIN AHACVTGKPI SQGGIHGRIS ATGRGVFHGI ENFINEASYM SILGMTPGFG DKTFVVQGFG NVGLHSMRYL HRFGAKCIAV GESDGSIWNP DGIDPKELED FKLQHGSILG FPKAKPYEGS ILEADCDILI PAASEKQLTK SNAPRVKAKI IAEGANGPTT PEADKIFLER NIMVIPDLYL NAGGVTVSYF EWLKNLNHVS YGRLTFKYER DSNYHLLMSV QESLERKFGK HGGTIPIVPT AEFQDRISGA SEKDIVHSGL AYTMERSARQ IMRTAMKYNL GLDLRTAAYV NAIEKVFKVY NEAGVTFT.

Q&A

What is GLUD1 and what are its primary functions in human cells?

GLUD1 (glutamate dehydrogenase 1) is a mitochondrial matrix enzyme that plays a crucial role in nitrogen and glutamate metabolism and energy homeostasis. In humans, it catalyzes the reversible conversion of glutamate to α-ketoglutarate and ammonia, using NAD+ or NADP+ as cofactors. This enzyme is highly expressed in the liver, brain, pancreas, and kidney, but shows notably low expression in muscle tissue . In pancreatic β-cells, GLUD1 participates in insulin secretion mechanisms, while in neural tissue it contributes to both the synthesis and catabolism of glutamate and potentially ammonia detoxification .

Methodologically, when studying GLUD1's basic functions, researchers should implement enzyme activity assays that measure the oxidative deamination of glutamate or the reductive amination of α-ketoglutarate under varying conditions of pH, temperature, and substrate concentration to establish baseline enzymatic parameters.

How is the GLUD1 gene organized in the human genome?

The human GLUD1 gene spans approximately 45 kilobases and is organized into 13 exons . The gene's promoter region contains potential binding sites for various regulatory factors, including Sp1, AP-1, and AP-2, suggesting complex transcriptional regulation . Genomic analyses have revealed that GLUD1 belongs to a gene family that includes several pseudogenes (GLUDP2, GLUDP3, GLUDP4, and GLUDP5) .

For researchers studying GLUD1 genomic organization, it is recommended to employ comprehensive genomic sequencing techniques covering not only exonic regions but also intronic and promoter regions to capture regulatory elements. Comparative genomic approaches can help distinguish functional GLUD1 from its pseudogenes.

What expression patterns does GLUD1 show across human tissues?

GLUD1 demonstrates distinctive tissue-specific expression patterns. It is expressed at high levels in metabolically active organs including the liver, brain, pancreas, and kidney, while showing minimal expression in muscle tissue . This distribution pattern correlates with tissues that have high energy demands or significant roles in ammonia metabolism.

When investigating tissue expression patterns, researchers should combine quantitative PCR, western blotting, and immunohistochemistry techniques. RNA-seq and proteomics approaches can provide more comprehensive profiles across multiple tissues simultaneously. Tissue microarrays can efficiently compare expression across numerous samples.

How do mutations in GLUD1 contribute to hyperinsulinism/hyperammonemia syndrome?

Mutations in GLUD1 are linked to hyperinsulinism/hyperammonemia syndrome (HA/HI), characterized by a combination of mild-to-moderate hyperammonemia and relatively mild, late-onset hypoglycemia . These mutations typically alter the enzyme's regulation, particularly its inhibition by GTP, leading to increased enzyme activity. The dysregulated GLUD1 activity in pancreatic β-cells results in excessive glutamate oxidation, generating more ATP, which subsequently triggers inappropriate insulin release and hypoglycemia.

Methodologically, researchers investigating this mechanism should:

  • Use site-directed mutagenesis to introduce clinically observed mutations into GLUD1 expression constructs

  • Perform enzyme kinetic analyses comparing wild-type and mutant GLUD1 under varying conditions of allosteric regulators

  • Utilize patient-derived induced pluripotent stem cells differentiated into pancreatic β-cells to study the effects in a physiologically relevant context

  • Develop islet-specific GLUD1 mutant mouse models to observe systemic effects

What experimental approaches best measure GLUD1 activity in human samples?

Accurate measurement of GLUD1 activity in human samples presents several methodological challenges. The following approaches are recommended:

  • Spectrophotometric assays measuring NAD(P)H production/consumption during glutamate oxidation or α-ketoglutarate reduction

  • Radiometric assays using 14C-labeled substrates for greater sensitivity

  • Polarographic oxygen consumption measurements for mitochondrial preparations

  • Mass spectrometry-based metabolomic approaches to track isotope-labeled glutamate metabolism

When working with clinical samples, researchers should be aware that:

  • Fresh tissue samples yield more reliable activity measurements than frozen samples

  • Mitochondrial isolation protocols must maintain membrane integrity

  • GLUD1 activity should be normalized to total protein or mitochondrial content

  • Potential confounding activities from other dehydrogenases should be controlled with specific inhibitors

How does transgenic GLUD1 expression affect neuronal function and vulnerability?

Transgenic expression of GLUD1 in neurons creates an in vivo model of moderate excess glutamate release in the CNS. Studies have shown that this leads to:

  • Increased in vivo release of glutamate after neuronal depolarization in the striatum

  • Enhanced frequency and amplitude of miniature EPSCs in the hippocampal CA1 region

  • Selective neuronal vulnerability in specific brain regions (e.g., CA1 but not CA3)

  • Age-dependent decreases in MAP2A labeling of dendrites and synaptophysin labeling of presynaptic terminals

  • Reduced long-term potentiation of synaptic activity

  • Decreased spine density in dendrites of CA1 neurons

Researchers studying GLUD1's role in neuronal function should employ a multi-method approach including:

  • Electrophysiological recordings to measure synaptic transmission parameters

  • Microdialysis to quantify neurotransmitter release in vivo

  • Immunohistochemistry to assess structural changes in neurons and synapses

  • Behavioral testing to correlate molecular/cellular changes with functional outcomes

  • Age-stratified analyses to capture progressive changes

What is the relationship between GLUD1 and cellular energy metabolism?

GLUD1 occupies a critical position at the intersection of amino acid and carbohydrate metabolism. By catalyzing the reversible conversion of glutamate to α-ketoglutarate, GLUD1 can:

  • Feed the TCA cycle during times of high energy demand

  • Participate in glutamate synthesis when ammonia levels are high

  • Contribute to the regulation of cellular redox status through NAD+/NADH balance

Research approaches to study GLUD1's role in energy metabolism should include:

  • Metabolic flux analysis using isotope-labeled substrates

  • Seahorse XF analyzer measurements of oxygen consumption rates under various substrate conditions

  • Integration of transcriptomic and metabolomic data to map GLUD1's position in metabolic networks

  • Comparative studies across tissues with different metabolic profiles

How does GLUD1 function differ between neural and non-neural tissues?

GLUD1 exhibits tissue-specific functional variations that are particularly important when comparing neural and non-neural tissues. In the brain, where glutamate serves as both a metabolic intermediate and a neurotransmitter, GLUD1 appears to function in both glutamate synthesis and catabolism . This dual role makes GLUD1 particularly important for neurotransmitter recycling and potentially for ammonia detoxification in neural tissue.

Researchers investigating tissue-specific functions should:

  • Compare enzyme kinetics and regulatory properties of GLUD1 isolated from different tissues

  • Perform tissue-specific gene knockdown/knockout experiments

  • Use tissue-specific promoters for transgenic models

  • Analyze tissue-specific protein interaction networks through co-immunoprecipitation and mass spectrometry

What methodological considerations are important when studying GLUD1 pseudogenes?

The human genome contains several GLUD1 pseudogenes (GLUDP2, GLUDP3, GLUDP4, and GLUDP5) that complicate genetic and expression analyses . These pseudogenes show varying degrees of similarity to the functional GLUD1 gene, with some representing truncated versions of GLUD1.

When studying GLUD1 in the context of its pseudogene family, researchers should:

  • Design primers and probes that specifically target unique regions of GLUD1 not shared with pseudogenes

  • Use long-read sequencing technologies to resolve complex genomic regions

  • Employ careful sequence alignment and phylogenetic analysis to understand the evolutionary relationships between GLUD1 and its pseudogenes

  • Consider the possibility of pseudogene transcription and potential regulatory roles of pseudogene transcripts

  • Validate antibody specificity when studying protein expression to ensure they do not cross-react with potential pseudogene products

How can GLUD1 mutations be effectively screened in hyperinsulinism patients?

For clinical researchers investigating GLUD1 mutations in hyperinsulinism patients, a systematic screening approach is recommended:

  • Initial clinical assessment focusing on:

    • Presence of mild-to-moderate hyperammonemia

    • Relatively mild, late-onset hypoglycemia

    • Protein-sensitive hypoglycemia

  • Genetic screening methodology:

    • Targeted sequencing of GLUD1 exons and exon-intron boundaries

    • Focus on hotspot regions in exons 6, 7, 11, and 12 where most pathogenic mutations cluster

    • Consider multiplex ligation-dependent probe amplification (MLPA) to detect larger deletions/duplications

    • Whole exome sequencing when targeted approaches are negative but clinical suspicion remains high

  • Functional validation of novel variants:

    • In vitro enzyme activity assays with recombinant proteins

    • Structural modeling to predict effects on protein conformation and allosteric regulation

    • Cell-based assays measuring insulin secretion in response to amino acids

What are the contradictions in current GLUD1 research that need resolution?

Several areas of GLUD1 research present contradictory findings that require methodological approaches to resolve:

  • Tissue-specific roles of GLUD1 vs. GLUD2 (the hominoid-specific paralog):

    • Systematic comparison of expression patterns using tissue-specific RNA-seq

    • Paralog-specific antibodies for immunohistochemistry

    • CRISPR-based selective targeting of each paralog

  • Direction of GLUD1 activity in vivo (glutamate oxidation vs. synthesis):

    • Metabolic flux analysis with stable isotopes

    • Measurement of reaction directionality under physiological concentrations of substrates and products

    • In vivo microdialysis combined with mass spectrometry

  • Regulatory mechanisms controlling GLUD1 activity:

    • Comprehensive analysis of post-translational modifications

    • Identification of tissue-specific protein interaction partners

    • Investigation of transcriptional and post-transcriptional regulation

How should researchers design transgenic models to study GLUD1 function?

When designing transgenic models to study GLUD1 function, researchers should consider:

  • Choice of promoter:

    • Neuron-specific enolase promoter for neuronal expression

    • Tissue-specific promoters for targeted expression in liver, pancreas, or other tissues

    • Inducible promoter systems for temporal control of expression

  • Type of genetic modification:

    • Overexpression models to study effects of increased GLUD1 activity

    • Knockin models of specific mutations found in hyperinsulinism/hyperammonemia

    • Conditional knockout models to study tissue-specific requirements

    • CRISPR-engineered point mutations to study specific regulatory mechanisms

  • Validation approaches:

    • Quantification of GLUD1 mRNA and protein expression

    • Measurement of enzymatic activity in isolated mitochondria

    • Metabolomic profiling to assess broader metabolic impacts

    • Tissue-specific phenotyping (e.g., electrophysiology for neuronal models, glucose tolerance tests for pancreatic models)

The transgenic mouse model using neuron-specific enolase promoter to drive GLUD1 expression has successfully demonstrated the consequences of lifelong, moderate excess glutamate release in the CNS, revealing selective neuronal vulnerability and synaptic plasticity alterations .

What analytical techniques provide the most comprehensive understanding of GLUD1 function?

A multi-omics approach provides the most comprehensive understanding of GLUD1 function:

  • Genomics:

    • Whole genome sequencing to identify regulatory regions

    • ChIP-seq to map transcription factor binding sites

    • ATAC-seq to assess chromatin accessibility

  • Transcriptomics:

    • RNA-seq to measure expression levels and splice variants

    • Single-cell RNA-seq to capture cellular heterogeneity

    • Ribosome profiling to assess translation efficiency

  • Proteomics:

    • Mass spectrometry-based quantitative proteomics

    • Phosphoproteomics to map regulatory modifications

    • Interactome analysis through proximity labeling techniques

  • Metabolomics:

    • Targeted analysis of TCA cycle intermediates

    • Untargeted metabolic profiling

    • Flux analysis using stable isotope tracers

  • Functional assays:

    • Real-time measurements of mitochondrial respiration

    • Calcium imaging in response to glutamate

    • Neurotransmitter release quantification

Integration of these multi-omics data requires sophisticated computational approaches including network analysis, machine learning algorithms, and systems biology modeling.

Product Science Overview

Structure and Function

GLUD1 catalyzes the oxidative deamination of L-glutamate to 2-oxoglutarate and ammonia, using NAD+ or NADP+ as a cofactor . This reaction is reversible and is central to the metabolism of glutamate, an important neurotransmitter in the brain . The enzyme is composed of six identical subunits and is regulated by various allosteric effectors, including ADP (activator) and GTP/ATP (inhibitors) .

Expression and Regulation

GLUD1 is expressed at high levels in the liver, brain, pancreas, and kidney, but not in muscle . In the nervous tissue, it functions in both the synthesis and catabolism of glutamate and may play a role in ammonia detoxification . The regulation of GLUD1 is complex and involves multiple substrate and regulatory binding sites .

Clinical Significance

Mutations in the GLUD1 gene can lead to hyperinsulinism-hyperammonemia syndrome, a condition characterized by excessive insulin and ammonia levels in the blood . Additionally, GLUD1 has been implicated in various cancers, including breast cancer, where it plays a role in ammonia assimilation and tumor proliferation .

Recombinant GLUD1

Recombinant human GLUD1 is often produced in E. coli and is used in various research applications, including studies on enzyme kinetics, regulation, and structure . The recombinant protein typically includes an N-terminal His-tag for purification purposes and is used in assays such as Western Blotting and imaging .

Evolutionary Perspective

There are two paralogs of glutamate dehydrogenase in humans, GLUD1 and GLUD2, which arose from a recent retroposition event during primate evolution . These paralogs have different regulatory properties and are not fully characterized at the structural level .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.