ADH1A Human

Alcohol Dehydrogenase 1A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

ADH1A is located on chromosome 4 (4q23) within a cluster of six other alcohol dehydrogenase genes (ADH1B, ADH1C, ADH4, ADH5, ADH6, and ADH7) . The gene spans 15 exons and encodes the alpha subunit of class I alcohol dehydrogenase, forming homo- or heterodimers with beta (ADH1B) and gamma (ADH1C) subunits .

Ethanol Metabolism

ADH1A catalyzes the rate-limiting oxidation of ethanol to acetaldehyde, with a K<sub>m</sub> of 4.0 mM and turnover rate of 30 min<sup>−1</sup> . Its activity is critical in early development, as it is predominantly expressed in fetal and infant livers .

Retinol and Bile Acid Metabolism

Beyond ethanol, ADH1A oxidizes retinol to retinaldehyde (a precursor of retinoic acid) and participates in bile acid synthesis by metabolizing 5β-cholestane-3α,7α,12α,26-tetrol .

Transcriptional Regulation

  • Bile Acids: Chenodeoxycholic acid (CDCA) and synthetic FXR agonists upregulate ADH1A expression in human hepatocytes, unlike rodent models .

  • Epigenetic Suppression: In hepatocellular carcinoma (HCC), mTOR/HDAC1 signaling suppresses ADH1A transcription, promoting tumor progression .

Cancer

  • Hepatocellular Carcinoma (HCC): Suppression of ADH1A and ALDH2 by mTOR/HDAC1 dysregulation leads to acetaldehyde accumulation, DNA damage, and oncogenesis .

  • Prognostic Value: Low ADH1A expression correlates with aggressive tumors and poor survival in HCC, lung adenocarcinoma, and gastric cancer .

Alcohol-Related Disorders

While ADH1A itself is not polymorphic, its interaction with polymorphic ADH1B/ADH1C isoforms influences ethanol metabolism rates and susceptibility to alcohol dependence .

Table 2: ADH1A in Disease Contexts

DiseaseMechanismClinical ImplicationSource
HCCEpigenetic suppression by HDAC1Biomarker for tumor progression
Fetal Alcohol SyndromeEthanol clearance in fetal liverDevelopmental toxicity risk

Research and Therapeutic Insights

  • Structural Studies: X-ray crystallography (e.g., PDB 1HSO) reveals substrate-binding pocket dynamics, aiding inhibitor design .

  • Therapeutic Targets: HDAC1 inhibitors and mTOR antagonists (e.g., rapamycin) restore ADH1A expression, showing promise in HCC treatment .

Product Specs

Introduction
Alcohol dehydrogenase 1A (ADH1A) is an enzyme that plays a crucial role in ethanol metabolism. It converts organic alcohols to ketones or aldehydes and vice versa. ADH1A is vital for eliminating ethanol produced by gut microorganisms. While most active in the fetus and infant liver, its activity decreases during gestation and remains low in adulthood.
Description
This recombinant ADH1A protein is produced in E. coli and lacks glycosylation. It consists of 375 amino acids (residues 1-375) and has a molecular weight of 42kDa. A 20 amino acid His-tag is fused to the N-terminus to facilitate purification. The protein is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The ADH1A solution is supplied at a concentration of 1mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0), 1mM DTT, 10% glycerol, and 0.1M NaCl.
Stability
For short-term storage (up to 4 weeks), keep at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for prolonged storage. Avoid repeated freezing and thawing.
Purity
Purity is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Alcohol dehydrogenase 1A, Alcohol dehydrogenase subunit alpha, ADH1A, ADH1.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSTAGKVIKC KAAVLWELKK PFSIEEVEVA PPKAHEVRIK MVAVGICGTD DHVVSGTMVT PLPVILGHEA AGIVESVGEG VTTVKPGDKV IPLAIPQCGK CRICKNPESN YCLKNDVSNP QGTLQDGTSR FTCRRKPIHH FLGISTFSQY TVVDENAVAK
IDAASPLEKV CLIGCGFSTG YGSAVNVAKV TPGSTCAVFG LGGVGLSAIM GCKAAGAARI IAVDINKDKF AKAKELGATE CINPQDYKKP IQEVLKEMTD GGVDFSFEVI GRLDTMMASL LCCHEACGTS VIVGVPPDSQ NLSMNPMLLL TGRTWKGAIL GGFKSKECVP KLVADFMAKK
FSLDALITHV LPFEKINEGF DLLHSGKSIR TILMF.

Q&A

What is the genomic organization of ADH1A and how does it relate to other ADH genes?

ADH1A is one of seven separate alcohol dehydrogenase genes that have been identified, characterized, and mapped to a gene cluster located on Chromosome 4 in humans. The ADH genes are classified into distinct classes based on enzymatic and DNA/protein sequence characteristics, with ADH1A belonging to the class I alcohol dehydrogenase isoenzymes .

The human ADH system has unique complexity due to a gene triplication and polymorphism that occurs among the class I isoenzymes, giving rise to ADH1A, ADH1B, and ADH1C genes. This triplication and extensive polymorphism appear to be unique to humans, as several animal species have been found to express only two class I isoenzymes .

How does ADH1A expression differ throughout human development?

In humans, the product of the ADH1A gene is the sole isoenzyme expressed in fetal liver, with the expression of the ADH1B and ADH1C genes rising just before and after birth, respectively . This sequential expression pattern suggests a programmed developmental regulation of these genes and potentially distinct metabolic roles during different developmental stages.

The unique developmental expression pattern of ADH1A raises important questions about why humans possess three distinct class I isoenzymes and why these isoenzymes show differences in expression during development. Understanding these patterns may provide insights into the specific substrates affected by the presence of ethanol during fetal development .

What are the evolutionary characteristics of ADH1A across species?

The ADH1A isoenzyme is found only in humans and certain primates, such as the rhesus monkey . This limited distribution across species suggests a relatively recent evolutionary origin for ADH1A, possibly related to primate-specific metabolic adaptations.

The evolutionary uniqueness of ADH1A may reflect adaptations to specific dietary or metabolic challenges faced by humans and close primate relatives. Researchers should consider this limited species distribution when designing studies and selecting appropriate animal models for ADH1A research.

What are the key structural features of the ADH1A protein?

The three-dimensional structure of ADH1A has been determined at 2.5 Å resolution, revealing several unique features compared to other class I alcohol dehydrogenases . The ADH1A structure shows a slightly more closed conformation when compared with other human class I structures, with the catalytic domain rotated approximately 1.5 degrees further toward the coenzyme-binding cleft .

Key crystallographic data for the ADH1A structure include:

  • Space group: P21

  • Cell dimensions: a=55.7 Å, b=100.2 Å, c=69.1 Å; α=90°, β=104.9°, γ=90°

  • Resolution range: 44-2.5 Å

  • Completeness: 92.9% (83.0% in the highest resolution shell)

  • Rmerge: 9.2% (29.4% in the highest resolution shell)

How do amino acid substitutions affect ADH1A function compared to other class I ADH isoenzymes?

Although ADH1A and ADH1B*1 isoenzymes differ by 23 amino acid substitutions, the unique structural and functional features of ADH1A can be reduced to two critical amino acid replacements: Arg 47 → Gly and Phe 93 → Ala .

The substitution of Gly for Arg at position 47 is particularly significant as Arg 47 typically interacts electrostatically with and donates hydrogen bonds to the adenosine phosphate of the bound cofactor NAD(H) in most class I isoenzymes . This substitution promotes a greater extent of domain closure in the ADH1A isoenzyme.

The substrate-binding pockets of each isoenzyme possess unique topologies that dictate their distinct but overlapping substrate preferences. While ADH1B1 has the most restrictive substrate-binding site near the catalytic zinc atom, both ADH1A and ADH1C2 possess amino acid substitutions that correlate with their better efficiency for the oxidation of secondary alcohols .

What experimental approaches can characterize ADH1A substrate specificity?

To characterize ADH1A substrate specificity, researchers should:

  • Perform steady-state kinetic analysis with various primary and secondary alcohols, measuring parameters such as kcat and KM to determine enzyme efficiency.

  • Conduct comparative analysis with other ADH isoenzymes under identical conditions to identify substrate preference patterns.

  • Use site-directed mutagenesis to investigate the role of specific amino acids (particularly positions 47 and 93) in determining substrate specificity.

  • Employ molecular docking and molecular dynamics simulations to predict interactions between ADH1A and potential substrates.

  • Analyze crystal structures of ADH1A in complex with various substrates to directly observe binding interactions.

These approaches can help understand how the unique structural features of ADH1A, particularly its more closed conformation and distinctive substrate-binding pocket, contribute to its specific catalytic properties .

What are the key polymorphisms in ADH1A and their functional consequences?

Several significant polymorphisms have been identified in ADH1A, particularly in the 5' regulatory region of the gene. Notable polymorphisms include:

  • rs1229966 (SNP3) at bp -1291, which could significantly alter the secondary structure of ADH1A mRNA according to computational predictions .

  • rs1826909 at bp -5601 (4.3 kb to SNP3)

  • rs4147531 at bp -55 (1.2 kb to SNP3)

  • rs2866151 at intron 8 (3.2 kb to SNP2)

  • rs13134764 at bp -31 (1.26 kb to SNP3)

  • rs904092 at bp -2022 (731 bp to SNP3)

These polymorphisms, particularly those in the regulatory regions, may affect ADH1A expression levels and thus influence alcohol metabolism and other physiological processes. The functional consequences of these variations have been investigated in relation to substance dependence and personality traits .

How should researchers design genotyping strategies for comprehensive ADH1A variant analysis?

For comprehensive ADH1A variant analysis, researchers should:

  • Prioritize SNPs with known functional consequences or strong associations with phenotypes, such as rs1229966, which could alter mRNA structure.

  • Include both coding and regulatory region polymorphisms, with particular attention to the 5' regulatory region where several significant associations have been reported.

  • Consider linkage disequilibrium patterns to select tag SNPs that efficiently capture genetic variation across the gene.

  • Use high-throughput genotyping methods for multiple SNPs (e.g., SNP arrays or next-generation sequencing) rather than single-SNP approaches.

  • Include flanking regions and other nearby ADH genes to capture potential regulatory elements and enable haplotype analysis.

  • Account for population differences in allele frequencies and linkage disequilibrium patterns when designing the genotyping panel.

  • Validate genotyping results with different methods or replicate samples to ensure accuracy.

This comprehensive approach will provide a more complete picture of genetic variation in ADH1A and its potential functional consequences .

What statistical approaches are most effective for analyzing ADH1A genetic associations with complex traits?

Based on previous successful studies, effective statistical approaches for analyzing ADH1A genetic associations include:

  • Multilevel genetic analysis: Examine associations at multiple genetic levels (diplotypes, haplotypes, genotypes, and alleles) to capture different aspects of genetic variation .

  • Stepwise multivariate analysis of covariance (MANCOVA): This approach allows comprehensive examination of relationships between ADH1A variants and multiple related phenotypes simultaneously .

  • Stepwise analysis of covariance (ANCOVA): For decomposing multivariate effects and analyzing relationships between ADH1A and individual traits or components .

  • Stepwise logistic regression analysis: Particularly useful for binary outcomes such as substance dependence diagnosis .

  • Consideration of admixture effects: Account for potential population stratification, especially in ethnically diverse samples .

  • Stratified analysis: Analyze associations separately by sex, age, ethnicity, and affected status to identify potential moderating effects of these factors .

  • Correction for multiple testing: Implement appropriate statistical corrections when testing multiple SNPs or phenotypes to minimize false positive results.

These approaches have been successfully applied in studies demonstrating associations between ADH1A variation and both personality traits and substance dependence .

What evidence supports the role of ADH1A in alcohol dependence risk?

Multiple studies have demonstrated significant associations between ADH1A variation and alcohol dependence (AD). Edenberg et al. (2006) genotyped 14 SNPs within or near ADH1A and found several SNPs distributed across the gene (from the upstream region through exon 8) were significantly associated with AD, including rs1826909, rs4147531, and rs2866151 .

Luo et al. (2006) reported positive associations between ADH1A diplotypes and AD, providing further evidence for ADH1A's role in AD risk . Similarly, Kuo et al. (2008) found that two SNPs in the upstream region of ADH1A (rs13134764 and rs904092) were significantly associated with AD in an Irish affected sibling pair sample, with rs904092 showing interaction with rs3762894 at the 5' end of ADH4 .

These consistent findings across multiple independent studies strongly support a role for ADH1A variation in modifying risk for alcohol dependence.

How does ADH1A contribute to both alcohol and drug dependence vulnerability?

Research suggests that ADH1A may contribute to shared genetic vulnerability for different types of substance dependence. Luo et al. (2007c) reported positive associations between ADH1A diplotypes and both alcohol dependence (AD) and drug dependence (DD) .

This shared genetic influence is consistent with findings for other genes in the ADH family. Many studies have shown that DD and AD share susceptibility genes including ADH4, ADH1A, ADH1B, ADH1C, ADH5, ADH6, and ADH7 .

The mechanism for this shared vulnerability remains to be fully elucidated, but may involve:

  • Shared neurobiological pathways affected by ADH1A function or expression

  • Potential roles of ADH1A in metabolizing endogenous substrates that affect reward or stress systems

  • Developmental effects of ADH1A expression patterns that influence neural systems relevant to addiction vulnerability

  • Possible effects of ADH1A on the oxidation of neurotransmitters such as dopamine, serotonin, and norepinephrine

What methodological challenges arise when investigating gene-environment interactions involving ADH1A?

When investigating gene-environment interactions involving ADH1A, researchers face several methodological challenges:

  • Sample size requirements: Detecting gene-environment interactions typically requires substantially larger sample sizes than main genetic effects alone.

  • Environmental assessment precision: Accurate measurement of relevant environmental exposures (e.g., alcohol consumption patterns, stress exposure) is critical but often difficult to standardize.

  • Developmental timing: The sequential expression pattern of ADH genes during development suggests critical periods when environmental factors may have particularly strong effects.

  • Potential confounding by population stratification: ADH1A allele frequencies may differ between populations, potentially confounding associations if not properly accounted for .

  • Defining appropriate phenotypes: Substance use disorders are heterogeneous, and defining phenotypes that capture biologically meaningful subtypes is challenging.

  • Accounting for other genetic factors: Interactions between ADH1A and other genes (e.g., ADH4 as demonstrated by Kuo et al. 2008) add complexity to models .

  • Longitudinal assessment: Understanding how gene-environment interactions evolve over time requires resource-intensive longitudinal study designs.

Addressing these challenges requires careful study design, precise measurement techniques, appropriate statistical methods, and often, large collaborative studies.

What specific personality traits show association with ADH1A variation?

Research has identified significant associations between ADH1A genetic variants and specific personality traits across different population groups:

  • Agreeableness and Conscientiousness: These traits were associated with ADH1A diplotypes, haplotypes, genotypes, and/or alleles in three of four phenotype groups studied: European-American substance dependence (SD) subjects, healthy subjects, and African-American SD subjects (p-values ranging from 1.7×10^-4 to 0.055) .

  • Neuroticism: This trait was specifically associated with ADH1A diplotype, haplotypes, and genotypes in African-American SD subjects (p-values ranging from 0.001 to 0.031) .

Interestingly, these associations were not detected in college students, suggesting that the relationship between ADH1A and personality may be influenced by factors such as age, development stage, or environmental exposures .

How do demographic factors moderate ADH1A-personality associations?

Previous studies have demonstrated that gene effects on personality traits can differ substantially based on demographic factors. For ADH1A specifically, the research suggests:

  • Ethnicity effects: Associations between ADH1A and personality traits showed some differences between European-Americans and African-Americans. For example, Neuroticism was associated with ADH1A variants in African-American SD subjects but not in other groups .

  • Age/developmental effects: The absence of significant associations in college students compared to older adults suggests age or developmental stage may moderate ADH1A-personality relationships .

  • Affection status effects: The pattern of associations differed somewhat between substance-dependent subjects and healthy individuals, suggesting that disease status interacts with genetic effects .

  • Sex differences: While specific sex differences in ADH1A effects weren't detailed in the search results, previous research on related genes suggests sex can be an important moderator of genetic effects on personality .

These moderating effects highlight the importance of considering demographic factors in study design and analysis when investigating ADH1A-personality associations.

What neurobiological mechanisms might explain the link between ADH1A and personality traits?

Several potential neurobiological mechanisms could explain the observed associations between ADH1A variation and personality traits:

  • Neurotransmitter metabolism: The ADH1A enzyme (αα ADH) may be involved in the oxidation of neurotransmitters such as dopamine, serotonin, and norepinephrine, which play critical roles in personality and behavior regulation .

  • Developmental effects: As the sole class I ADH isoenzyme expressed during fetal development, ADH1A may influence early neurodevelopmental processes that establish the neurobiological substrates of personality .

  • Interaction with alcohol metabolism: Variation in alcohol metabolism efficiency could affect sensitivity to alcohol's effects on personality-relevant neural circuits, particularly in individuals with alcohol exposure.

  • Shared genetic architecture: ADH1A may be in linkage disequilibrium with other genes that more directly influence personality traits.

  • Endogenous substrate metabolism: Beyond alcohol, ADH1A may metabolize currently unidentified endogenous substrates that influence neural function relevant to personality traits.

Future research combining genetic association studies with functional neuroimaging, metabolomics, and experimental models could help elucidate these potential mechanisms .

What crystallization conditions are optimal for obtaining high-resolution ADH1A structures?

Based on the successful crystallization of ADH1A that yielded a 2.5 Å resolution structure, researchers should consider the following parameters for crystallization:

  • Space group: P21, which was used successfully for ADH1A crystallization

  • Cell dimensions: Approximately a=55.7 Å, b=100.2 Å, c=69.1 Å; α=90°, β=104.9°, γ=90°

  • Protein concentration: Should be optimized based on protein stability and solubility

  • Buffer conditions: Should maintain protein stability while promoting crystal formation

  • Precipitants: Common precipitants for ADH crystallization include polyethylene glycols and ammonium sulfate

  • Additives: Consider including cofactors (NAD+/NADH) to stabilize the protein structure

  • Temperature: Temperature can significantly affect crystallization kinetics and should be optimized

For comparative structural studies, researchers should note that ADH1A shows a slightly more closed conformation compared to other class I ADH structures, which may affect crystallization behavior .

How can researchers effectively analyze substrate specificity differences between ADH1A and other ADH isoenzymes?

To effectively analyze substrate specificity differences between ADH1A and other ADH isoenzymes, researchers should implement a systematic approach:

  • Steady-state kinetics: Determine kinetic parameters (kcat, KM, and catalytic efficiency kcat/KM) for a diverse panel of substrates including primary alcohols, secondary alcohols, and steroids across different ADH isoenzymes under identical conditions.

  • Structure-activity relationship analysis: Correlate substrate structural features with kinetic parameters to identify patterns of substrate preference.

  • Site-directed mutagenesis: Create targeted mutations at positions 47 and 93 (the key distinguishing residues of ADH1A) in various ADH backgrounds to isolate their effects on substrate specificity .

  • Product analysis: Use chromatographic and mass spectrometric techniques to identify and quantify reaction products with complex or chiral substrates.

  • Computational docking studies: Use the available crystal structures to perform in silico docking of different substrates and correlate binding energies with experimental data.

This comprehensive approach can reveal how the unique structural features of ADH1A, particularly its more closed conformation and the substitutions at positions 47 and 93, contribute to its distinct substrate preferences .

What techniques can effectively measure ADH1A expression patterns in different tissues and developmental stages?

To effectively measure ADH1A expression patterns across tissues and developmental stages, researchers should consider:

  • Quantitative PCR (qPCR): Design isoenzyme-specific primers to selectively amplify ADH1A transcripts, enabling precise quantification of gene expression levels.

  • RNA sequencing (RNA-seq): Perform transcriptome-wide analysis to measure ADH1A expression while simultaneously assessing global gene expression patterns.

  • In situ hybridization: Use ADH1A-specific probes to visualize expression patterns in tissue sections, providing spatial information about expression.

  • Immunohistochemistry: Develop and validate ADH1A-specific antibodies to detect protein expression in tissue sections.

  • Western blotting: Use isoenzyme-specific antibodies to quantify ADH1A protein levels in tissue extracts.

  • Single-cell RNA sequencing: Apply this technique to identify cell-specific expression patterns, particularly valuable in heterogeneous tissues.

  • Reporter gene assays: Create reporter constructs with the ADH1A promoter to investigate regulatory mechanisms controlling expression patterns.

These approaches are particularly important given the unique developmental expression pattern of ADH1A, which is the sole class I ADH isoenzyme expressed in fetal liver, with expression of ADH1B and ADH1C rising just before and after birth, respectively .

How might integrated multi-omics approaches advance our understanding of ADH1A function?

Integrated multi-omics approaches can significantly advance our understanding of ADH1A function through:

  • Genomics + Transcriptomics: Combining ADH1A genotyping with RNA-seq can reveal how genetic variants affect gene expression and splicing patterns across tissues and developmental stages.

  • Proteomics + Structural Biology: Mass spectrometry-based proteomics can identify ADH1A post-translational modifications and protein-protein interactions, while structural studies can reveal how these modifications affect enzyme function.

  • Metabolomics: Untargeted metabolomics in samples with different ADH1A genotypes or expression levels can identify potential endogenous substrates and metabolic pathways influenced by ADH1A activity.

  • Epigenomics: Assessing DNA methylation, histone modifications, and chromatin accessibility can elucidate regulatory mechanisms controlling the developmental expression pattern of ADH1A.

  • Single-cell Multi-omics: Applying single-cell technologies can reveal cell-specific effects of ADH1A variation on gene expression and metabolic processes.

  • Systems Biology Integration: Computational integration of multi-omics data can generate comprehensive models of ADH1A's role in cellular networks and physiological processes.

These approaches would be particularly valuable for understanding ADH1A's role during fetal development, when it is the sole class I ADH isoenzyme expressed .

What are the most promising therapeutic applications of ADH1A research?

Promising therapeutic applications emerging from ADH1A research include:

  • Personalized approaches to alcohol use disorders: ADH1A genetic variation could help identify individuals most likely to benefit from specific pharmacotherapies for alcohol dependence, based on metabolic differences .

  • Novel treatments for substance dependence: Understanding ADH1A's role in both alcohol and drug dependence could lead to treatments targeting shared vulnerability pathways .

  • Fetal alcohol spectrum disorder (FASD) prevention: As the primary ADH isoenzyme in fetal development, ADH1A-focused interventions might help mitigate alcohol exposure effects during pregnancy .

  • Personality disorder treatments: The association between ADH1A and personality traits suggests potential applications in personalizing treatments for personality disorders .

  • Cancer therapeutics: Given the role of other ADH enzymes in retinoid metabolism (important in cell differentiation), ADH1A modulators might have applications in cancer treatment.

  • Medication metabolism prediction: ADH1A genotyping could help predict metabolism of medications that are substrates for this enzyme, enabling personalized dosing.

These applications would build on the established associations between ADH1A variation and substance dependence, as well as its unique developmental expression pattern and substrate specificity profile .

How can researchers resolve contradictions in published ADH1A association studies?

To resolve contradictions in published ADH1A association studies, researchers should:

  • Conduct well-powered replication studies: Ensure adequate sample sizes to detect modest genetic effects and reduce the likelihood of both false positives and false negatives.

  • Perform meta-analyses: Systematically combine data from multiple studies to increase statistical power and identify sources of heterogeneity.

  • Standardize phenotype definitions: Use consistent diagnostic criteria and assessment instruments across studies to reduce phenotypic heterogeneity.

  • Account for population stratification: Use appropriate statistical methods and ancestry informative markers to control for population stratification effects .

  • Consider demographic moderators: Analyze how factors such as sex, age, ethnicity, and affection status might moderate genetic associations .

  • Examine multiple genetic levels: Analyze associations at the diplotype, haplotype, genotype, and allele levels to capture different aspects of genetic variation .

  • Include functional validation: Complement association studies with functional experiments to establish biological plausibility for genetic findings.

  • Investigate gene-environment interactions: Explore how environmental factors might modify genetic associations and contribute to contradictory findings.

By implementing these approaches, researchers can more effectively reconcile contradictory findings and build a more coherent understanding of ADH1A's role in health and disease .

Product Science Overview

Physiological Role

ADH1A is predominantly active in the liver, especially during fetal and infant stages. Its primary physiological function is the elimination of ethanol produced by microorganisms in the intestinal tract . As individuals age, the activity of ADH1A decreases, becoming only weakly active during adulthood .

Structure and Production

The human recombinant form of ADH1A is produced in E. coli and is a single, non-glycosylated polypeptide chain containing 395 amino acids. It has a molecular mass of approximately 42 kDa . The recombinant enzyme is often fused to a 20 amino acid His-tag at the N-terminus to facilitate purification through chromatographic techniques .

Stability and Storage

For optimal stability, the ADH1A solution should be stored at 4°C if it will be used within 2-4 weeks. For longer storage periods, it is recommended to freeze the solution at -20°C, preferably with a carrier protein such as 0.1% HSA (Human Serum Albumin) or BSA (Bovine Serum Albumin) to prevent degradation . It is important to avoid multiple freeze-thaw cycles to maintain the enzyme’s activity.

Applications

ADH1A is widely used in laboratory research, particularly in studies related to ethanol metabolism and its effects on the human body. It is also utilized in various biochemical assays to understand the enzyme’s kinetics and its interaction with different substrates and inhibitors .

Genetic Information

The ADH1A gene is located on the long arm of chromosome 4 and is part of a cluster that includes six additional alcohol dehydrogenase genes. These genes encode different subunits of the enzyme, including alpha, beta, and gamma subunits, which can form various homo- and heterodimers . Mutations in the ADH1A gene have been associated with variations in certain personality traits and substance dependence .

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.