GAD1 Human

Glutamate Decarboxylase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

GAD1 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 247 amino acids (1-224) and having a molecular mass of 27.7kDa.
GAD1 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Glutamate Decarboxylase 1 (GAD1) is one of the enzymes responsible for converting L-glutamic acid into gamma-aminobutyric acid (GABA). GAD1 has been identified as a potential autoimmune target in type II diabetes, with the enzyme being recognized by autoantibodies and autoreactive T cells in affected individuals. Additionally, GAD1 may play a role in stiff man syndrome. A deficiency in GAD1 can lead to pyridoxine-dependent seizures.
Description
Recombinant human GAD1, expressed in E. coli, is a single, non-glycosylated polypeptide chain. This protein consists of 247 amino acids (residues 1-224) with a molecular weight of 27.7 kDa. The N-terminus of GAD1 is fused to a 23 amino acid His-tag. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
The GAD1 solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4M Urea.
Stability
For optimal storage, the product should be kept at 4°C if it will be used within 2-4 weeks. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 85% as assessed by SDS-PAGE analysis.
Synonyms
Glutamate Decarboxylase 1 (Brain, 67kDa), GAD, Glutamate Decarboxylase 67 KDa Isoform, 67 KDa Glutamic Acid Decarboxylase, GAD-67, EC 4.1.1.15, CPSQ1, SCP, Glutamate Decarboxylase 1 (Brain, 67kD), Glutamate Decarboxylase 1, GAD67, EC 4.1.1, GAD1.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMASSTPS SSATSSNAGA DPNTTNLRPT TYDTWCGVAH GCTRKLGLKI CGFLQRTNSL EEKSRLVSAF KERQSSKNLL SCENSDRDAR FRRTETDFSN LFARDLLPAK NGEEQTVQFL LEVVDILLNY VRKTFDRSTK VLDFHHPHQL LEGMEGFNLE LSDHPESLEQ ILVDCRDTLK YGVRTGHPRF FNQLSTGLDI IGLAGEWLTS TANTNMPSDM RECWLLR.

Q&A

What is GAD1/GAD67 and what is its primary function in humans?

GAD1, also known as GAD67, is the enzyme responsible for the conversion of glutamic acid to gamma-aminobutyric acid (GABA), which serves as the major inhibitory neurotransmitter in higher brain regions. It also functions as a putative paracrine hormone in pancreatic islets . GAD occurs in two molecular forms - 65 kDa and 67 kDa - that are encoded by separate genes (GAD2 and GAD1, respectively) and share approximately 65% amino acid identity. The GAD1 protein is highly conserved across species, highlighting its evolutionary significance and essential biological role .

Where is GAD1 primarily expressed in the human body?

GAD1 demonstrates tissue-specific expression patterns in humans. It is primarily expressed in the central nervous system (CNS), particularly in GABAergic neurons. Beyond the brain, GAD1 is also found in pancreatic islet cells, where it may be involved in paracrine signaling. Additionally, GAD1 expression has been detected in reproductive tissues including the testis, oviduct, and ovary . Within the brain, expression patterns vary across different cell types, with the Human Protein Atlas providing detailed information on cell-type specificity through their tau specificity score, which ranges from 0 to 1 and indicates how specifically the gene is expressed across cells or tissues .

What transcript variants of GAD1 exist in humans?

Recent research using RNA sequencing and PCR technologies has significantly expanded our understanding of GAD1 transcript diversity. A study published in 2018 reported the discovery of 10 novel transcripts of GAD1 in the human brain, adding to previously known variants like the full-length GAD1 transcript (GAD67) and GAD25 . Interestingly, four novel GAD1 transcripts (designated as 8A, 8B, I80, and I86) demonstrate a lifespan trajectory expression pattern that is anticorrelated with the expression of the full-length GAD1 transcript, suggesting developmental regulation of alternative splicing . This transcript diversity likely contributes to the complex regulation of GABA synthesis in different brain regions and developmental stages.

How is GAD1 implicated in neuropsychiatric disorders?

GAD1 has been extensively studied in relation to various neuropsychiatric disorders, with schizophrenia receiving particular attention. Multiple autopsy studies have found that both mRNA and protein levels of GAD1 are lower in multiple cortical areas and hippocampus of schizophrenia patients . Furthermore, methylation levels of CpG loci within the putative GAD1 promoter have been significantly associated with schizophrenia-risk SNPs (such as rs3749034) and with the expression of certain GAD1 transcripts like GAD25 in the dorsolateral prefrontal cortex (DLPFC) . Additional findings indicate that schizophrenia patients who had completed suicide and/or were positive for nicotine exposure had significantly higher full-length GAD1 expression in the DLPFC, suggesting complex interactions between GAD1 expression, external factors, and disease manifestation .

How do GAD1 expression levels change during human brain development and aging?

The expression pattern of GAD1 throughout the human lifespan shows remarkable complexity. The full-length GAD1 transcript (GAD67) follows a distinct developmental trajectory that differs from several of its splice variants. Research has revealed that four novel GAD1 transcripts (8A, 8B, I80, and I86) demonstrate a lifespan trajectory expression pattern that is anticorrelated with the expression of the full-length GAD1 transcript . This inverse relationship suggests a developmental switch in GAD1 transcript usage that may reflect changing requirements for GABA synthesis during brain maturation.

These expression patterns vary across different brain regions, with particularly notable changes in the prefrontal cortex and hippocampus. The developmental regulation of GAD1 involves not only changes in total expression but also shifts in the relative abundance of different transcript variants, pointing to the importance of alternative splicing in GAD1 regulation throughout life. For researchers studying developmental aspects of GAD1, it is crucial to consider these temporal patterns and employ age-matched controls in experimental designs to account for the natural variability in GAD1 expression across the lifespan.

What are the challenges in interpreting genetic association studies of GAD1 in schizophrenia?

Interpreting genetic association studies of GAD1 in schizophrenia presents numerous methodological and conceptual challenges. Schizophrenia is clinically heterogeneous, and different patient populations may have different underlying genetic architectures. Studies with Chinese, Japanese, and other populations have shown inconsistent results, suggesting population-specific effects .

Statistical power represents another major challenge. Smaller studies may detect associations that aren't replicated in larger genome-wide association studies (GWAS). As noted in one study, "inconsistencies between genetic studies in smaller samples and in large GWAS are often observed... This may be a result of the etiological and phenotypic heterogeneity of the illness" .

The selection of single nucleotide polymorphisms (SNPs) further complicates interpretation. Different studies examine different SNPs across the GAD1 gene, making direct comparisons difficult. Some research has utilized comprehensive tagging approaches covering the entire gene region, while others focused on specific SNPs of interest . Additionally, phenotypic definition varies between studies, with some focusing on paranoid subtypes, others on early-onset cases, etc. These phenotypic differences may account for some of the inconsistencies observed across studies.

What epigenetic mechanisms regulate GAD1 expression in the human brain?

Several epigenetic mechanisms contribute to the regulation of GAD1 expression in the human brain. DNA methylation plays a significant role, with methylation levels of CpG loci within the putative GAD1 promoter having been significantly associated with schizophrenia-risk SNPs and with the expression of certain GAD1 transcripts in the dorsolateral prefrontal cortex (DLPFC) . This suggests that DNA methylation is a key regulator of GAD1 expression patterns.

Histone modifications represent another important epigenetic mechanism regulating GAD1. Alterations in histone acetylation and methylation at the GAD1 locus have been observed in psychiatric disorders, suggesting chromatin-based regulation of gene expression. The accessibility of the GAD1 gene to transcription machinery is influenced by these modifications, affecting its expression levels in different brain regions and cell types.

Additionally, alternative splicing of GAD1 appears to be epigenetically regulated, as evidenced by the differential expression of splice variants in relation to DNA methylation states . The complex interplay between genetic variation, epigenetic modifications, and environmental factors contributes to the developmental regulation of GAD1 and may explain some of the dysregulation observed in neuropsychiatric disorders.

How do single-cell analysis techniques reveal cell-type-specific expression patterns of GAD1?

Single-cell analysis techniques have revolutionized our understanding of cell-type-specific expression patterns of GAD1 in the human brain. Single-cell RNA sequencing (scRNA-seq) allows for the characterization of GAD1 expression at single-cell resolution, revealing distinct expression patterns across different neuronal and non-neuronal cell types . This technology has demonstrated that GAD1 expression is not uniform across all GABAergic neurons but shows considerable heterogeneity even within this broad cell class.

For post-mortem human brain tissue, where intact cell isolation is challenging, single-nucleus RNA sequencing (snRNA-seq) provides an alternative approach to assess nuclear transcripts at single-nucleus resolution. This technique has been particularly valuable for studying GAD1 expression in archived human brain samples.

The Human Protein Atlas employs the Tau specificity score as a numerical indicator of gene expression specificity across cells or tissues, with values ranging from 0 to 1 . This metric helps researchers quantify the cell-type specificity of GAD1 expression and identify the particular cell populations where GAD1 is most highly expressed or most significantly altered in disease states.

What are the optimal methods for detecting GAD1/GAD67 in human brain tissue samples?

The detection of GAD1/GAD67 in human brain tissue samples can be accomplished through several complementary techniques, each with specific advantages. For protein detection, immunohistochemistry (IHC) using specific antibodies like the Human GAD1/GAD67 Antibody (MAB2086) allows visualization of GAD1 expression patterns in tissue sections . This approach is particularly useful for examining the cellular and regional distribution of GAD1.

Western blotting provides a quantitative method for measuring total GAD1 protein levels, while ELISA offers a highly sensitive approach for measuring GAD1 in tissue homogenates and biological fluids, with detection limits as low as 0.56ng/mL . The Human Glutamate Decarboxylase 1 (Brain GAD1) ELISA Kit allows for the accurate detection of GAD1 levels in human serum, plasma, and cell culture supernatants .

For transcript analysis, RT-qPCR remains the gold standard for measuring mRNA expression levels of different GAD1 transcript variants. RNA sequencing provides the most comprehensive characterization of all transcript variants, including novel isoforms like the recently discovered variants (8A, 8B, I80, and I86) . The optimal detection method should be selected based on the specific research question, sample availability, and whether protein or transcript levels are of primary interest.

What are the best approaches for studying GAD1 splice variants in human brain samples?

Studying GAD1 splice variants in human brain samples requires a strategic combination of techniques. For identification of splice variants, RNA sequencing is the most comprehensive approach. Long-read RNA-seq technologies like PacBio or Oxford Nanopore provide full-length transcript information, facilitating the identification of novel splice variants. This approach led to the discovery of 10 novel transcripts of GAD1 in the human brain .

For quantification of specific splice variants, RT-qPCR with primers designed to target unique exon-exon junctions can provide accurate measurements of relative abundance. When designing such experiments, it's important to consider that expression of certain GAD1 variants (8A, 8B, I80, and I86) shows an inverse correlation with the full-length transcript across the lifespan .

Spatial distribution analysis of GAD1 splice variants can be achieved through in situ hybridization techniques using probes specific to unique regions of each variant. This approach provides valuable information about the regional and cellular localization of different GAD1 transcripts within the brain.

For functional characterization, expression vectors containing specific splice variants can be used to study their properties in cell models. Additionally, protein analysis using isoform-specific antibodies (when available) or mass spectrometry can identify peptides unique to specific protein isoforms resulting from alternative splicing.

What statistical approaches are recommended for analyzing GAD1 genetic association data in complex disorders?

Analyzing GAD1 genetic association data in complex disorders like schizophrenia requires robust statistical approaches to handle potential confounding factors and multiple testing issues. For single SNP association tests, allelic tests comparing allele frequencies between cases and controls (e.g., Fisher's exact test, chi-square test) provide a starting point. Genotypic tests comparing genotype distributions (e.g., Cochran-Armitage trend test) can also be informative .

Haplotype analysis is particularly valuable when studying the GAD1 gene. Sliding window approaches analyzing haplotypes across the GAD1 gene region and haplotype block identification using methods like Gabriel's algorithm can reveal associations not detected by single SNP analyses .

Multiple testing correction is essential when analyzing multiple SNPs or haplotypes. Options include the conservative Bonferroni correction, False Discovery Rate (FDR) control, and permutation-based methods that generate empirical p-values through permutation of case-control status .

Population stratification must be controlled using approaches such as Principal Component Analysis (PCA), including top principal components as covariates, or mixed models that account for cryptic relatedness and population structure . These statistical approaches are crucial for generating reliable genetic association data, especially given the previous inconsistent findings in GAD1 genetic studies.

How can epigenetic modifications of the GAD1 gene be accurately measured in human brain tissue?

Accurately measuring epigenetic modifications of the GAD1 gene in human brain tissue requires specialized techniques targeting different types of modifications. For DNA methylation analysis, bisulfite sequencing remains the gold standard, providing single-nucleotide resolution of methylation status. This is particularly relevant as methylation levels of CpG loci within the putative GAD1 promoter have been significantly associated with schizophrenia-risk SNPs and with the expression of GAD1 transcripts .

Pyrosequencing offers a quantitative approach for analyzing methylation at specific CpG sites, while methylation arrays like the Illumina EPIC/450K arrays cover multiple CpG sites in GAD1 and allow comparison with genome-wide methylation patterns. For distinguishing between different cytosine modifications, oxidative bisulfite sequencing (oxBS-seq) can differentiate 5-methylcytosine (5mC) from 5-hydroxymethylcytosine (5hmC), which is particularly important in brain tissue where 5hmC is abundant.

For histone modification analysis, chromatin immunoprecipitation (ChIP) using antibodies against specific histone marks can be followed by qPCR for GAD1-specific regions or sequencing for genome-wide analysis. When working with human brain tissue, researchers must account for post-mortem changes, cell heterogeneity, and include appropriate reference standards to ensure accurate and interpretable results.

What are the optimal protein extraction methods for preserving GAD1/GAD67 enzymatic activity?

Preserving GAD1/GAD67 enzymatic activity during protein extraction requires specific methodological considerations. The buffer composition is crucial and should include a base buffer like 50 mM Tris-HCl (pH 7.4) or potassium phosphate buffer (pH 7.0-7.2). The cofactor pyridoxal 5'-phosphate (PLP, 0.2-1 mM) is essential as GAD1 is PLP-dependent . A reducing agent such as DTT or 2-mercaptoethanol helps maintain sulfhydryl groups, while protease inhibitors prevent degradation.

Tissue handling protocols are equally important. Samples should be maintained at 4°C throughout processing, and freeze-thaw cycles should be minimized. For brain tissue, rapid dissection on ice and immediate processing or flash-freezing is recommended. Gentle homogenization methods like Dounce homogenization preserve activity better than harsher methods such as sonication, which can generate heat and denature the enzyme.

For storage, adding 10-20% glycerol to the buffer helps stabilize the enzyme, and aliquots should be stored at -80°C for long-term preservation. When planning activity assays, include positive controls (e.g., commercial recombinant GAD1) and measure specific activity (units/mg protein) to normalize for extraction efficiency across samples.

How can researchers effectively design primers for studying GAD1 transcript variants?

Designing effective primers for studying GAD1 transcript variants requires targeting unique regions specific to each variant. For the novel transcripts discovered in human brain (8A, 8B, I80, and I86), primers should span their unique splice junctions to ensure specificity . When targeting shared regions, these forward primers should be combined with reverse primers specific to each transcript variant.

Standard primer design parameters apply: primers should typically be 18-25 nucleotides in length with a GC content of 40-60%. Primer pairs should have similar melting temperatures (within 5°C of each other), and secondary structures like hairpins and dimers should be avoided. Including 1-2 G/C bases at the 3' end provides stability for the extension step.

Several validation strategies should be employed. In silico validation using tools like Primer-BLAST confirms specificity, while experimental validation through gel electrophoresis verifies amplicon size. Sequencing verification of amplicons and inclusion of appropriate negative controls are essential quality checks.

Special considerations for GAD1 include awareness of the high sequence similarity between GAD1 and GAD2, designing primers that span introns when possible, and careful planning to distinguish between full-length GAD1 (GAD67) and shorter variants like GAD25 . These strategies ensure specific and accurate detection of the complex repertoire of GAD1 transcripts.

How is GAD1 research contributing to our understanding of schizophrenia pathophysiology?

GAD1 research has provided significant insights into schizophrenia pathophysiology, particularly regarding GABAergic neurotransmission dysfunction. Studies have consistently found reduced GAD1 mRNA and protein levels in multiple cortical areas and the hippocampus of schizophrenia patients . This reduction in GAD1 expression likely contributes to diminished GABA synthesis, disrupting the excitatory/inhibitory balance that is critical for normal brain function.

Epigenetic research has revealed that methylation levels of CpG loci within the putative GAD1 promoter are significantly associated with schizophrenia-risk SNPs and with the expression of specific GAD1 transcripts like GAD25 in the dorsolateral prefrontal cortex . These findings suggest that epigenetic dysregulation of GAD1 may be a molecular mechanism underlying some aspects of schizophrenia pathophysiology.

Interestingly, environmental factors appear to influence GAD1 expression in schizophrenia. Research has shown that schizophrenia patients who had completed suicide and/or were positive for nicotine exposure had significantly higher full-length GAD1 expression in the DLPFC . This highlights the complex interplay between genetic vulnerability, environmental factors, and gene expression in schizophrenia.

What role does GAD1 play in human brain development and neuroplasticity?

GAD1 plays a crucial role in human brain development and neuroplasticity through its regulation of GABA synthesis. During development, GABA transitions from an excitatory to an inhibitory neurotransmitter, a shift that is fundamental to proper circuit formation. The discovery of novel GAD1 transcripts with expression patterns that are anticorrelated with the full-length transcript across the lifespan suggests that alternative splicing of GAD1 is developmentally regulated .

This developmental regulation of GAD1 transcript usage likely allows for precise control of GABA synthesis during critical periods of brain development. During these periods, GABAergic signaling influences neuronal migration, differentiation, and synapse formation. The temporal specificity of different GAD1 variants may contribute to the varying roles of GABA throughout development.

In terms of neuroplasticity, GAD1-mediated GABA synthesis is crucial for maintaining the excitatory/inhibitory balance that permits healthy neural adaptations. Alterations in GAD1 expression have been linked to changes in neuroplasticity in various contexts, including learning, memory formation, and adaptation to environmental challenges. The epigenetic regulation of GAD1, including DNA methylation patterns associated with specific SNPs, provides a mechanism for experience-dependent modulation of GABAergic function .

How do novel GAD1 transcripts influence GABA synthesis in different brain regions?

The discovery of 10 novel transcripts of GAD1 in the human brain has expanded our understanding of GABA synthesis regulation . These different transcripts likely contribute to region-specific and cell-type-specific control of GABA production. Four novel GAD1 transcripts (8A, 8B, I80, and I86) show a lifespan trajectory expression pattern that is anticorrelated with the expression of the full-length GAD1 transcript, suggesting they may have distinct roles in GABA synthesis regulation at different developmental stages .

The regional distribution of these novel transcripts may contribute to the known differences in GABAergic function across brain regions. For example, the prefrontal cortex and hippocampus, areas often implicated in schizophrenia, show specific patterns of GAD1 expression and regulation. The alternative transcripts may produce protein variants with different enzymatic properties, subcellular localization, or regulatory features.

Additionally, the expression of these novel transcripts appears to be subject to epigenetic regulation, as methylation levels of CpG loci in the GAD1 promoter are associated with expression of specific transcripts . This suggests that environmental factors and cellular states can influence which GAD1 transcript variants are expressed, providing a mechanism for context-specific regulation of GABA synthesis.

What advances in GAD1 antibody development have improved research capabilities?

Recent advances in GAD1 antibody development have significantly enhanced research capabilities for studying this important enzyme. The development of monoclonal antibodies with high specificity, such as the Human GAD1/GAD67 Antibody (MAB2086), has improved the reliability of immunodetection methods . These antibodies are generated against specific epitopes of the human GAD1 protein, allowing for precise targeting of the full-length protein or specific regions of interest.

Antibodies validated for multiple applications provide versatility in research approaches. For instance, antibodies suitable for both western blotting and immunohistochemistry allow correlation between quantitative protein measurements and spatial distribution analyses. The availability of antibodies recognizing epitopes that remain accessible in fixed tissue has enhanced the study of GAD1 in post-mortem human brain samples.

Additionally, the development of antibodies specific to different GAD1 isoforms resulting from alternative splicing has opened new avenues for investigating the functional diversity of GAD1 variants. These isoform-specific antibodies can help determine the subcellular localization and relative abundance of different GAD1 proteins in various brain regions and cell types.

How can GAD1 biomarkers be utilized in neuropsychiatric disorder research?

GAD1 biomarkers offer promising applications in neuropsychiatric disorder research, particularly for conditions involving GABAergic dysfunction. For schizophrenia research, measurements of GAD1 expression levels, methylation patterns, and genetic variants have provided insights into disease mechanisms . The methylation status of specific CpG sites in the GAD1 promoter region, especially those associated with schizophrenia-risk SNPs like rs3749034, may serve as epigenetic biomarkers .

In clinical studies, peripheral measurements of GAD1 or related markers may serve as accessible indicators of central GABAergic function. The availability of sensitive ELISA kits capable of detecting GAD1 in human serum, plasma, and cell lysates provides tools for such investigations . With detection sensitivity as low as 0.56ng/mL and a range of 1.56-100ng/mL, these assays can capture variations in GAD1 levels that might correlate with disease states or treatment responses .

Longitudinal studies examining GAD1 biomarkers in relation to disease progression, treatment response, or developmental trajectories can help identify at-risk individuals or predict outcomes. Furthermore, integrating GAD1 biomarkers with other measurements, such as neuroimaging findings or cognitive assessments, may provide a more comprehensive understanding of how GABAergic dysfunction contributes to neuropsychiatric disorders.

What emerging technologies will advance GAD1 research in coming years?

Several emerging technologies are poised to significantly advance GAD1 research in the coming years. Spatial transcriptomics technologies that preserve the spatial context of cells while providing transcriptome-wide data will revolutionize our understanding of GAD1 expression patterns within specific brain regions and circuits. These approaches will allow researchers to map the distribution of different GAD1 transcript variants with unprecedented spatial resolution.

CRISPR-based technologies for precise genome editing will enable more sophisticated manipulation of GAD1 expression and splicing in cellular and animal models. This will facilitate the investigation of causative relationships between specific GAD1 variants and neuronal function or disease phenotypes. Additionally, CRISPR-based epigenome editing tools will allow targeted modification of the epigenetic state at the GAD1 locus, providing insights into the regulatory mechanisms controlling its expression.

Single-cell multi-omics approaches that simultaneously profile the transcriptome, epigenome, and proteome of individual cells will provide integrated views of GAD1 regulation. These technologies will help unravel the complex relationships between genetic variation, epigenetic modifications, transcript diversity, and protein function in specific cell types relevant to neuropsychiatric disorders.

How might targeting GAD1 lead to novel therapeutic approaches for neuropsychiatric disorders?

Targeting GAD1 offers promising avenues for developing novel therapeutic approaches for neuropsychiatric disorders characterized by GABAergic dysfunction. Given the evidence of reduced GAD1 expression in schizophrenia , strategies aimed at enhancing GAD1 expression or activity could potentially address the underlying GABAergic deficit. Epigenetic modifiers, such as DNA methyltransferase inhibitors or histone deacetylase inhibitors, might be employed to counteract the hypermethylation observed at specific CpG sites in the GAD1 promoter region .

RNA-based therapeutics targeting specific GAD1 splice variants represent another promising approach. By modulating the balance between different GAD1 transcripts, it may be possible to normalize GABA synthesis in affected brain regions. This approach could be particularly valuable given the discovery of novel transcripts with expression patterns that differ from the full-length GAD1 during development .

Cell-based therapies involving the transplantation of GABAergic precursors or engineered cells with optimized GAD1 expression could potentially restore inhibitory circuit function in localized brain regions. Additionally, small molecule modulators that enhance GAD1 enzymatic activity or stability might provide a more traditional pharmacological approach to addressing GABAergic deficits in neuropsychiatric disorders.

Product Science Overview

Introduction

Glutamate Decarboxylase 1 (GAD1) is an enzyme that plays a crucial role in the central nervous system. It catalyzes the decarboxylation of glutamic acid to produce gamma-aminobutyric acid (GABA), a major inhibitory neurotransmitter in the brain. The recombinant form of this enzyme, produced through genetic engineering techniques, is used extensively in research and therapeutic applications.

Structure and Function

GAD1 is a pyridoxal 5’-phosphate (PLP)-dependent enzyme, meaning it requires PLP as a cofactor to function. The enzyme is encoded by the GAD1 gene and is primarily found in the brain, where it is involved in the synthesis of GABA. GABA is essential for regulating neuronal excitability and maintaining the balance between excitation and inhibition in the brain .

Recombinant Production

The recombinant form of GAD1 is produced by cloning the GAD1 gene into a suitable expression vector, which is then introduced into a host organism, typically Escherichia coli. The host organism expresses the GAD1 protein, which can be purified and used for various applications. This method allows for the production of large quantities of the enzyme, which is essential for research and therapeutic purposes .

Applications

Recombinant GAD1 is used in various research studies to understand its role in the central nervous system and its involvement in neurological disorders. It is also used in the development of therapeutic strategies for conditions such as epilepsy, anxiety, and depression, where GABAergic signaling is disrupted .

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.