AOC1 Human

Amine Oxidase Copper Containing 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Structure and Biochemical Properties

AOC1 is a homodimeric glycoprotein with isoforms varying in molecular mass due to glycosylation. The recombinant human AOC1 protein (expressed in HEK293 cells) has a molecular mass of 84.2 kDa and comprises 738 amino acids, including a C-terminal 6xHis tag . Key features include:

PropertyDetail
Gene LocationChromosome 7q34
SubstratesHistamine, putrescine, spermine, spermidine
InhibitorsAmiloride, aminoguanidine
Tissue ExpressionHigh: kidneys, placenta, intestines; Moderate: lungs, brain

The enzyme catalyzes oxidative deamination, producing hydrogen peroxide and aldehydes . Its inhibition by amiloride links it to epithelial sodium channel regulation .

Fibromyalgia and Histamine Intolerance

AOC1 deficiency is implicated in histamine intolerance, leading to symptoms like migraines, allergies, and gastrointestinal disorders . A 2023 study of 100 women with fibromyalgia identified four AOC1 gene variants (rs10156191, rs1049742, rs1049793, rs2052129) associated with symptom severity :

SNPMinor Allele FrequencyClinical Correlation
rs1015619131.5%Increased dry skin intensity and low stool consistency
rs104974210%Linked to higher Fibromyalgia Impact Questionnaire (FIQ) scores
rs104979332.5%Aggravated GI disorders and sleep disturbances
rs205212927%Correlation with migraine frequency

Patients with ≥5 risk alleles showed worse FIQ scores (p=0.020p = 0.020), suggesting cumulative genetic effects .

Cancer Progression

AOC1 overexpression promotes colorectal cancer (CRC) proliferation and metastasis. Key findings include:

Research Applications

Recombinant AOC1 (e.g., PRO-2657) is used to study histamine metabolism and enzyme dysfunction. Specifications include :

  • Purity: >90% (SDS-PAGE).

  • Formulation: 0.25 mg/mL in PBS (pH 7.4) with 10% glycerol.

  • Stability: Stable at 4°C for 2–4 weeks; long-term storage at -20°C with carrier protein (e.g., 0.1% HSA).

Therapeutic and Diagnostic Potential

  • Biomarker: AOC1 levels in blood or tissues may predict CRC prognosis and fibromyalgia severity .

  • Targeted Therapy: Inhibiting AOC1 could suppress tumor growth, while enzyme supplementation might alleviate histamine intolerance .

Future Directions

  • Genetic Studies: Larger cohorts to confirm AOC1 variant linkages and haplotype effects .

  • Mechanistic Insights: Role in EMT and AKT pathways warrants exploration for anti-cancer drug development .

Product Specs

Introduction

Amine Oxidase Copper Containing 1, also known as AOC1, is an enzyme classified under the copper-containing amine oxidase protein family. It plays a crucial role in the oxidation of various biogenic amines, encompassing neurotransmitters, xenobiotic amines, and histamine. Deficiencies in AOC1 have been linked to conditions such as dietary histamine intolerance and histaminosis. Moreover, this enzyme has been implicated in tumor progression by promoting AKT signaling and epithelial-mesenchymal transition (EMT) in stomach cancer.

Description

Recombinant human AOC1, expressed in HEK cells, is a single, glycosylated polypeptide chain with a molecular weight of 84.2 kDa. It encompasses amino acids 20 to 751, totaling 738 amino acids. The protein includes a 6-amino acid His-tag fused at the C-terminus to facilitate purification, which is achieved through proprietary chromatographic techniques.

Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation

The AOC1 solution is provided at a concentration of 0.25 mg/ml and is formulated in a buffer consisting of 10% glycerol and Phosphate-Buffered Saline (PBS) at a pH of 7.4.

Stability
For short-term storage (up to 4 weeks), the solution can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To further enhance stability during long-term storage, consider adding a carrier protein such as HSA or BSA to a final concentration of 0.1%. It is important to avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity

The purity of AOC1 is determined to be greater than 90% using SDS-PAGE analysis.

Synonyms

Amiloride-sensitive amine oxidase (copper-containing), DAO, Diamine oxidase, Amiloride-binding protein 1, Amine oxidase copper domain-containing protein 1, Histaminase, Kidney amine oxidase, KAO, ABP, ABP1, DAO1

Source

HEK293 Cells.

Amino Acid Sequence

EPSPGTLPRK AGVFSDLSNQ ELKAVHSFLW SKKELRLQPS STTTMAKNTV FLIEMLLPKK YHVLRFLDKG ERHPVREARA VIFFGDQEHP NVTEFAVGPL PGPCYMRALS PRPGYQSSWA SRPISTAEYA LLYHTLQEAT KPLHQFFLNT TGFSFQDCHD RCLAFTDVAP RGVASGQRRS WLIIQRYVEG YFLHPTGLEL LVDHGSTDAG HWAVEQVWYN GKFYGSPEEL ARKYADGEVD VVVLEDPLPG GKGHDSTEEP PLFSSHKPRG DFPSPIHVSG PRLVQPHGPR FRLEGNAVLY GGWSFAFRLR SSSGLQVLNV HFGGERIAYE VSVQEAVALY GGHTPAGMQT KYLDVGWGLG SVTHELAPGI DCPETATFLD TFHYYDADDP VHYPRALCLF EMPTGVPLRR HFNSNFKGGF NFYAGLKGQV LVLRTTSTVY NYDYIWDFIF YPNGVMEAKM HATGYVHATF YTPEGLRHGT RLHTHLIGNI HTHLVHYRVD LDVAGTKNSF QTLQMKLENI TNPWSPRHRV VQPTLEQTQY SWERQAAFRF KRKLPKYLLF TSPQENPWGH KRTYRLQIHS MADQVLPPGW QEEQAITWAR YPLAVTKYRE SELCSSSIYH QNDPWHPPVV FEQFLHNNEN IENEDLVAWV TVGFLHIPHS EDIPNTATPG NSVGFLLRPF NFFPEDPSLA SRDTVIVWPR DNGPNYVQRW IPEDRDCSMP PPFSYNGTYR PVHHHHHH

Q&A

What is the AOC1 gene and what is its role in human physiology?

AOC1 (Amine oxidase copper containing 1) encodes a metal-binding membrane glycoprotein that oxidatively deaminates putrescine, histamine, and related compounds. This enzyme plays a crucial role in the metabolism of biogenic amines, particularly histamine degradation. The gene is also known by several synonyms including ABP1, KAO, DAO1, DAO, and ABP .

The encoded protein is inhibited by amiloride, a diuretic that acts by closing epithelial sodium ion channels. Functionally, AOC1 has 3,202 documented associations with various biological entities spanning 7 categories, including molecular profiles, chemicals, diseases, phenotypes, functional terms, structural features, and cell types/tissues .

Methodologically, when studying AOC1's basic function, researchers typically employ enzyme activity assays with specific substrates like putrescine or histamine, coupled with spectrophotometric detection of hydrogen peroxide produced during the oxidative deamination process.

What are the common SNP variants of the AOC1 gene and how are they detected?

The most commonly studied single nucleotide polymorphisms (SNPs) of the AOC1 gene include:

SNP Variantrs NumberNucleotide Change
c.47C>Trs10156191C to T at position 47
c.995C>Trs1049742C to T at position 995
c.1990C>Grs10449793C to G at position 1990
c.691G>Trs2052129G to T at position 691

These variants are typically detected using multiplex PCR amplification of the regions of interest within the AOC1 gene. For instance, in clinical research, DNA can be obtained from oral mucosa samples using automated platforms like M2000SP (Abbott Molecular), followed by analysis with specialized kits such as Realtype® (Progenie Molecular SLU) .

For research purposes, genetic DAO enzyme deficiency is commonly defined as the presence of at least one allele variant of the AOC1 gene. Statistical analysis of these variants typically involves comparison with reference populations (such as European population samples) in Hardy-Weinberg equilibrium extracted from databases like the Allele Frequency Aggregator (ALFA) .

How is AOC1 expression measured in different experimental contexts?

AOC1 expression can be measured at multiple levels using various techniques:

  • mRNA level measurement:

    • Quantitative real-time PCR (qRT-PCR) is the gold standard for measuring AOC1 mRNA expression

    • RNA sequencing (RNA-seq) for genome-wide expression analysis that includes AOC1

    • Microarray analysis for high-throughput expression profiling

  • Protein level measurement:

    • Western blotting for semi-quantitative protein detection

    • Immunohistochemistry (IHC) staining for tissue localization and expression assessment

    • ELISA for quantitative protein measurement in biological fluids

In research settings, as demonstrated in colorectal cancer studies, comprehensive analysis typically combines multiple approaches. For instance, researchers have examined AOC1 expression levels in paired colorectal cancer and peritumoral tissues, as well as distant liver metastatic tissues, using qRT-PCR, western blotting, and immunohistochemistry staining simultaneously .

The choice of methodology depends on the specific research question, available samples, and required sensitivity. For monitoring changes in cell culture experiments, qRT-PCR and western blotting are commonly employed, while patient-derived samples often benefit from IHC to preserve spatial information.

What methodologies are most effective for studying AOC1 knockdown effects in cancer cell lines?

When investigating AOC1 knockdown effects in cancer cell lines, researchers typically employ a multi-faceted approach combining genetic manipulation with functional assays:

Genetic Manipulation Strategies:

  • siRNA or shRNA-mediated knockdown for temporary or stable AOC1 suppression

  • CRISPR-Cas9 gene editing for complete knockout models

  • Inducible knockdown systems for temporal control of AOC1 expression

Functional Assessment Methods:
For proliferation assessment, the Cell Counting Kit-8 (CCK-8) assay has proven effective. The protocol typically involves:

  • Plating approximately 1,000 cells/well in 96-well plates with complete medium (DMEM with 10% FBS)

  • After 24h incubation, adding 10 μL/well of CCK-8 solution and incubating for 1.5h at 37°C in dark conditions

  • Measuring absorbance at 450 nm using a microplate reader

  • Repeating measurements for 5 consecutive days to establish growth curves

For colony formation capacity, researchers typically:

  • Plate approximately 1,000 cells in 6-well plates with DMEM containing 10% FBS

  • Culture for 2 weeks

  • Wash with PBS and fix with 4% paraformaldehyde for 30 min

  • Stain with 0.1% crystal violet for 30 min

  • Count colonies to quantify growth potential

Migration assessment typically employs:

  • Transwell migration assays for directional cell movement

  • Wound healing assays for cell motility in a monolayer context

To establish causality, comprehensive studies should include rescue experiments where AOC1 is re-expressed in knockdown cells to confirm phenotype reversal.

How do AOC1 genetic variants impact enzyme activity, and what are the implications for disease susceptibility?

AOC1 genetic variants have significant impacts on enzyme activity, with downstream effects on histamine metabolism and disease susceptibility:

Enzyme Activity Impact:
The four main SNP variants (rs10156191, rs1049742, rs10449793, and rs2052129) affect enzyme kinetics differently:

  • Some variants reduce substrate binding affinity

  • Others affect the catalytic rate of the enzyme

  • Certain variants may alter protein stability or folding

Disease Association Analysis:
Statistical approaches for studying these associations include:

  • Calculating variant prevalence in case-control studies using chi-square or Fisher's exact tests

  • Comparing genotype frequencies between patient populations and reference databases

  • Assessing Hardy-Weinberg equilibrium to validate genotyping accuracy and detect selection pressures

For genetic association studies, researchers typically establish genetic DAO enzyme deficiency as the presence of at least one allele variant of the AOC1 gene. This definition allows for stratification of populations in association studies with various conditions, particularly those involving histamine intolerance or inflammation .

When conducting Mendelian Randomization studies to establish causal relationships between AOC1 variants and disease outcomes, robust statistical approaches include:

  • Selection of instrumental variables with p-value thresholds below 5 × 10^-8

  • Focus on cis-SNPs located within 1 megabase of the gene

  • Assessment of instrument strength through variance (R²) and F-statistics calculations

What is the relationship between AOC1 expression and cancer progression, particularly in colorectal cancer?

Research has established significant correlations between AOC1 expression and colorectal cancer (CRC) progression:

Expression Pattern in Progression:
AOC1 expression significantly increases in human CRC tissues compared to normal tissue, with even higher expression observed in liver metastases. This progressive increase correlates with worse clinical outcomes, suggesting AOC1 as a potential biomarker for disease progression .

Biological Mechanisms:
Functional analyses have revealed that AOC1 promotes aggressive CRC phenotypes through several mechanisms:

  • Enhanced proliferation (demonstrated through CCK-8 and colony formation assays)

  • Increased migration capacity (shown in Transwell and wound healing assays)

  • Induction of Epithelial-Mesenchymal Transition (EMT), a key process in metastasis formation

In vivo Confirmation:
Xenograft tumor formation experiments in nude mice have confirmed that AOC1 knockdown inhibits tumor growth in vivo, providing strong evidence for its role in tumor progression beyond cell culture systems .

The consistent findings across in vitro, in vivo, and patient-derived samples establish AOC1 as both a prognostic biomarker and potential therapeutic target in CRC.

What are the current methodological approaches for studying the functional associations of AOC1 with other biological entities?

AOC1 has been identified as having 3,202 functional associations with biological entities spanning various categories. Investigating these interactions requires sophisticated methodological approaches:

Expression Correlation Analysis:

  • Tissue-specific expression profiling using databases like Allen Brain Atlas and BioGPS

  • Co-expression network analysis to identify genes with similar expression patterns

  • Cell type-specific expression analysis to identify tissue-specific functions

Protein-Protein Interaction Studies:

  • Co-immunoprecipitation followed by mass spectrometry to identify binding partners

  • Proximity labeling techniques (BioID, APEX) to capture transient interactions

  • Yeast two-hybrid screening for binary interaction mapping

Functional Genomics Approaches:

  • CRISPR-Cas9 screens to identify synthetic lethal interactions

  • RNA-seq after AOC1 manipulation to detect downstream transcriptional effects

  • Metabolomics profiling to identify changes in histamine and related compounds

Computational Prediction Methods:

  • Network-based approaches using existing protein interaction databases

  • Machine learning algorithms to predict functional associations based on protein features

  • Systems biology modeling to integrate multi-omics data

When analyzing these complex datasets, researchers typically employ:

  • Standardized value scoring to rank associations by strength

  • Ontology enrichment analysis to identify biological processes overrepresented in AOC1 networks

  • Pathway analysis to contextualize AOC1 within established biological systems

How can researchers effectively design experiments to study the potential of AOC1 as a therapeutic target?

Designing experiments to evaluate AOC1 as a therapeutic target requires systematic approaches spanning multiple research phases:

Target Validation Phase:

  • Genetic Manipulation Studies:

    • Compare the effects of transient knockdown (siRNA) versus stable knockdown (shRNA)

    • Utilize inducible systems to assess the temporal requirements of AOC1 inhibition

    • Employ CRISPR-Cas9 for complete gene knockout to determine maximum potential effect

  • Pharmacological Inhibition:

    • Screen existing DAO inhibitors, including amiloride derivatives

    • Assess dose-response relationships in multiple cell lines

    • Compare genetic versus pharmacological inhibition to identify potential off-target effects

Biomarker Development:

  • Identify patient subgroups likely to benefit from AOC1 targeting through:

    • Analysis of AOC1 expression across patient cohorts

    • Correlation with specific genetic backgrounds

    • Association with treatment response in existing datasets

Preclinical Efficacy Assessment:

  • Utilize multiple model systems including:

    • Cell line panels representing disease heterogeneity

    • Patient-derived organoids to maintain tumor complexity

    • Xenograft models in nude mice with various treatment schedules

Combination Strategies:

  • Investigate synergistic potential with:

    • Standard-of-care chemotherapeutics

    • Targeted therapies addressing complementary pathways

    • Immunotherapeutic approaches

Resistance Mechanism Prediction:

  • Employ long-term selection experiments with sublethal AOC1 inhibition

  • Perform whole-genome or targeted sequencing to identify resistance mutations

  • Develop strategies to overcome or prevent resistance development

What statistical approaches are most appropriate for analyzing AOC1 genetic variant data?

When analyzing AOC1 genetic variant data, researchers should employ specific statistical approaches tailored to genetic association studies:

For Population-Based Studies:

  • Chi-square tests or Fisher's exact tests for comparing variant frequencies between groups

  • Hardy-Weinberg equilibrium testing to validate genotyping quality and detect potential selection bias

  • Calculation of variance (R²) and F-statistics to assess the strength of instrumental variables in Mendelian Randomization studies

For Genotype-Phenotype Correlations:

  • Logistic regression for binary outcomes (e.g., disease presence/absence)

  • Linear regression for continuous traits (e.g., enzyme activity levels)

  • Cox proportional hazard models for time-to-event data (e.g., survival analysis)

For Complex Genetic Models:

  • Calculation of genotype-specific relative risks or odds ratios

  • Adjustment for covariates including age, sex, and ethnicity

  • Correction for multiple testing using Bonferroni or False Discovery Rate methods

Software Recommendations:

  • R statistical software (v4.0.0 or newer) for comprehensive analysis

  • PLINK for genome-wide association analysis

  • SNPTEST for detailed variant association testing

  • Haploview for linkage disequilibrium analysis and haplotype reconstruction

The specific formula for calculating variance explained by genetic variants is:
R² = [2 × β² × EAF × (1 − EAF)]/[2 × β² × EAF × (1 − EAF) + 2 × SE² × N × EAF × (1 − EAF)]

Where:

  • β = effect size of the SNP on the phenotype

  • N = sample size

  • EAF = effect allele frequency

  • SE = standard error associated with the β estimate

When assessing instrument strength in Mendelian Randomization, an F-value above 10 is typically considered strong evidence for reducing weak instrument bias .

How should researchers design cell-based assays to study AOC1 function and regulation?

Designing robust cell-based assays for AOC1 research requires careful consideration of multiple factors:

Cell Line Selection:

  • Choose physiologically relevant cell types that naturally express AOC1

  • For CRC studies, use established lines like HCT116, SW480, or HT29

  • Include multiple cell lines to account for genetic background variation

  • Consider patient-derived primary cells or organoids for increased relevance

Expression Modulation Strategies:

  • For knockdown studies: optimize transfection conditions for each cell type

  • For overexpression: use inducible systems to control expression levels

  • Include appropriate controls (scrambled siRNA, empty vectors)

  • Verify modulation efficiency by qRT-PCR and western blotting

Functional Assay Design:
For proliferation assessment using Cell Counting Kit-8:

  • Optimize cell density (typically ~1,000 cells/well for CRC lines)

  • Include at least five technical replicates

  • Measure at consistent timepoints (24, 48, 72, 96, and 120 hours)

  • Normalize to initial timepoint (0h) values

For colony formation assays:

  • Determine optimal seeding density through preliminary experiments

  • Culture for sufficient duration (typically 2 weeks for CRC lines)

  • Use automated colony counting software for objective quantification

  • Report both colony number and size distribution

For migration analysis:

  • Standardize cell density and starvation conditions

  • Capture images at consistent timepoints

  • Use automated image analysis for wound closure quantification

  • Include positive and negative controls for assay validation

Quality Control Measures:

  • Perform mycoplasma testing regularly

  • Verify cell line identity through STR profiling

  • Use low-passage cells to minimize genetic drift

  • Include technical and biological replicates (minimum n=3)

Data Analysis:

  • Apply appropriate statistical tests (typically ANOVA or two-tailed Student's t-test)

  • Present comprehensive data including means, standard deviations, and individual data points

  • Include power calculations to justify sample sizes

  • Report exact p-values rather than significance thresholds

What are the considerations for selecting appropriate animal models in AOC1 research?

When selecting animal models for AOC1 research, researchers must consider several critical factors to ensure translational relevance:

Model Selection Criteria:

  • Expression Pattern Similarity:

    • Verify AOC1 expression patterns in the model organism compared to humans

    • Confirm conservation of regulatory elements and promoter regions

    • Assess tissue distribution similarity, particularly in target organs

  • Functional Conservation:

    • Evaluate sequence homology at protein level

    • Confirm enzymatic activity similarities

    • Assess substrate specificity conservation

  • Model-Specific Advantages:

    • Mouse models: genetic manipulation capabilities, physiological similarity

    • Zebrafish: rapid development, cost-effective drug screening

    • Rat models: larger size for surgical interventions, certain metabolic similarities

Genetic Modification Approaches:

  • Conventional knockout for complete AOC1 ablation

  • Conditional knockout for tissue-specific or temporal control

  • Knock-in models for studying specific variants

  • Humanized models expressing human AOC1 variants

Experimental Design Considerations:
For xenograft tumor studies:

  • Use immunocompromised strains (e.g., nude mice) to prevent rejection

  • Calculate appropriate sample sizes based on expected effect size

  • Establish clear endpoints based on tumor volume or animal welfare

  • Include both male and female animals to account for sex differences

  • Randomize animals to treatment groups to minimize bias

Ethical and Regulatory Compliance:

  • Adhere to the 3Rs principle (Replacement, Reduction, Refinement)

  • Obtain appropriate institutional review board approval

  • Follow species-specific welfare guidelines

  • Ensure proper training of personnel

Analysis and Reporting Standards:

  • Pre-register study protocols to prevent reporting bias

  • Report according to ARRIVE guidelines

  • Include detailed methodology for reproducibility

  • Present individual animal data points alongside group statistics

The choice of animal model should ultimately be guided by the specific research question, with careful consideration of how the model's biology may affect interpretation of results related to AOC1 function.

How is AOC1 research contributing to personalized medicine approaches?

AOC1 research is increasingly informing personalized medicine strategies through several key pathways:

Biomarker Development:
The identification of AOC1 as a prognostic biomarker in colorectal cancer represents a significant advancement. High expression of AOC1 correlates with worse clinical outcomes and is an independent risk factor for poor prognosis. This enables potential patient stratification for treatment intensity based on AOC1 expression levels .

Pharmacogenomic Applications:
The characterized SNP variants of AOC1 (rs10156191, rs1049742, rs10449793, and rs2052129) present opportunities for personalized therapeutic approaches:

  • Patients with certain variants may require adjusted medication dosages

  • Variant profiles could predict response to histamine-targeting therapies

  • Genetic screening could identify individuals at risk for adverse drug reactions

Therapeutic Target Identification:
Functional analysis demonstrating that AOC1 knockdown inhibits the proliferation and migration of cancer cells provides a foundation for targeted therapy development. The validation of these effects in xenograft models further strengthens the case for AOC1 as a druggable target .

Integration with Multi-Omics Data:
The extensive functional associations of AOC1 (3,202 associations across 7 biological categories) enable integration with:

  • Transcriptomic profiles for pathway analysis

  • Proteomic data for interaction networks

  • Metabolomic signatures for functional impact assessment

This multi-dimensional approach allows for the development of comprehensive patient profiles that can inform treatment decisions based on individual molecular landscapes.

What are the emerging technologies that may advance AOC1 research?

Several cutting-edge technologies are poised to significantly advance AOC1 research:

Single-Cell Technologies:

  • Single-cell RNA sequencing to map AOC1 expression at unprecedented resolution

  • Single-cell proteomics to detect cell-specific AOC1 protein levels

  • Spatial transcriptomics to visualize AOC1 expression in tissue context

Advanced Genetic Engineering:

  • Base editing for precise modification of AOC1 variants

  • Prime editing for scarless gene correction

  • CRISPR screening with improved specificity for AOC1 pathway analysis

Organoid and Tissue Models:

  • Patient-derived organoids for personalized drug screening

  • Organ-on-chip technology for studying AOC1 in tissue microenvironments

  • 3D bioprinting for complex tissue architecture recapitulation

Computational Approaches:

  • AI-driven protein structure prediction for improved understanding of AOC1 variants

  • Network-based drug repurposing to identify existing drugs targeting AOC1

  • Integrative multi-omics analysis to contextualize AOC1 within cellular systems

Advanced Imaging:

  • Live-cell imaging with fluorescent AOC1 reporters

  • Super-resolution microscopy for subcellular localization

  • Intravital microscopy for in vivo dynamics

High-Throughput Screening:

  • CRISPR activation/inhibition screens to identify AOC1 regulators

  • Small molecule libraries for AOC1 modulator discovery

  • Massively parallel reporter assays for regulatory element mapping

These technologies will enable researchers to address current limitations in AOC1 research, potentially accelerating translation to clinical applications for conditions including colorectal cancer and histamine-related disorders.

How can researchers address the current challenges in understanding AOC1's role in disease pathways?

Researchers face several challenges in fully elucidating AOC1's role in disease pathways, which can be addressed through specific methodological approaches:

Challenge: Tissue Heterogeneity
Solution Approaches:

  • Employ single-cell technologies to resolve cell type-specific expression

  • Use laser capture microdissection to isolate specific tissue compartments

  • Develop computational deconvolution methods for bulk tissue data

  • Utilize spatial transcriptomics to preserve contextual information

Challenge: Functional Redundancy
Solution Approaches:

  • Conduct simultaneous knockdown of AOC1 and related amine oxidases

  • Apply systems biology approaches to map compensatory mechanisms

  • Utilize temporal inhibition strategies to identify adaptive responses

  • Develop specific inhibitors that distinguish between related enzymes

Challenge: Translating In Vitro Findings
Solution Approaches:

  • Validate findings across multiple cell types and experimental systems

  • Employ patient-derived organoids that better recapitulate in vivo conditions

  • Conduct rigorous in vivo studies with appropriate animal models

  • Design clinical correlation studies to validate mechanistic hypotheses

Challenge: Complex Regulatory Networks
Solution Approaches:

  • Apply network analysis to identify key nodes interacting with AOC1

  • Utilize epigenetic profiling to map regulatory elements controlling AOC1

  • Implement machine learning to predict context-dependent regulation

  • Develop mathematical models to simulate pathway dynamics

Challenge: Clinical Heterogeneity
Solution Approaches:

  • Stratify patient cohorts based on AOC1 expression and variant profiles

  • Conduct large-scale genetic association studies across diverse populations

  • Integrate clinical metadata with molecular profiles for comprehensive analysis

  • Employ longitudinal sampling to capture disease evolution

Challenge: Methodological Standardization
Solution Approaches:

  • Establish consensus protocols for AOC1 detection and activity measurement

  • Develop reference materials for assay calibration

  • Create publicly available datasets as benchmarks

  • Form collaborative networks to validate findings across laboratories

Addressing these challenges requires interdisciplinary approaches combining molecular biology, genetics, computational biology, and clinical research. By implementing these methodological solutions, researchers can develop a more comprehensive understanding of AOC1's role in disease pathways and accelerate translation to clinical applications.

What are the most significant recent advances in AOC1 research?

The field of AOC1 research has experienced several significant advances in recent years:

  • Establishment of AOC1 as a Cancer Biomarker:
    Research has definitively established that AOC1 expression significantly increases in human colorectal cancer tissues, especially in liver metastases, and correlates with worse prognosis. This positions AOC1 as both a prognostic biomarker and potential therapeutic target .

  • Characterization of Functional Mechanisms:
    Functional analysis has demonstrated that AOC1 knockdown inhibits the proliferation and migration of cancer cells by modulating epithelial-mesenchymal transition (EMT) both in vitro and in vivo. This mechanistic insight provides a foundation for therapeutic development .

  • Comprehensive Genetic Variant Profiling:
    The detailed characterization of AOC1 SNP variants (rs10156191, rs1049742, rs10449793, and rs2052129) and their prevalence has enhanced our understanding of genetic DAO enzyme deficiency. This has implications for histamine metabolism and related disorders .

  • Development of Methodological Standards:
    Advances in experimental protocols for AOC1 research, including standardized cell proliferation assays (CCK-8), colony formation assays, and migration assessment techniques, have improved research reproducibility and reliability .

  • Expanded Functional Association Mapping:
    The identification of 3,202 functional associations between AOC1 and various biological entities across 7 categories has significantly expanded our understanding of this gene's role in multiple biological processes and potential disease pathways .

What are the key unanswered questions that should guide future AOC1 research?

Despite significant progress, several critical questions remain unanswered in AOC1 research:

  • Mechanism of Oncogenic Function:

    • How does AOC1 specifically promote cancer progression at the molecular level?

    • Which downstream pathways mediate its effects on proliferation and migration?

    • Are there context-dependent functions in different cancer types or subtypes?

  • Therapeutic Targeting Potential:

    • Can AOC1 inhibition effectively reduce tumor growth in clinical settings?

    • What are the potential side effects of AOC1 targeting given its role in histamine metabolism?

    • Are there specific patient populations that would benefit most from AOC1-targeted therapies?

  • Regulatory Networks:

    • What factors control AOC1 expression in normal and disease states?

    • How do epigenetic modifications influence AOC1 function?

    • Are there feedback mechanisms that modulate AOC1 activity?

  • Structure-Function Relationships:

    • How do specific genetic variants alter AOC1 protein structure and function?

    • What are the critical domains for substrate specificity and catalytic activity?

    • Can structural knowledge inform the design of specific inhibitors?

  • Clinical Translation:

    • How can AOC1 expression or genetic variants be effectively used for patient stratification?

    • What is the optimal methodology for AOC1 assessment in clinical samples?

    • Do circulating AOC1 levels correlate with tissue expression and have diagnostic value?

Product Science Overview

Structure and Function

AOC1 is characterized by its requirement for a copper ion per subunit and the cofactor topaquinone for its enzymatic activity . The enzyme catalyzes the oxidation of primary amines to aldehydes, with the subsequent release of ammonia and hydrogen peroxide. The general reaction can be summarized as follows:

RCH2NH2+H2O+O2RCHO+NH3+H2O2\text{RCH}_2\text{NH}_2 + \text{H}_2\text{O} + \text{O}_2 \rightarrow \text{RCHO} + \text{NH}_3 + \text{H}_2\text{O}_2

This reaction is crucial for the metabolism of amino groups and is involved in several metabolic pathways, including the urea cycle, histidine metabolism, and phenylalanine metabolism .

Physiological Role

AOC1 plays a significant role in the degradation of histamine, a biogenic amine involved in local immune responses, regulation of stomach acid, and functioning as a neurotransmitter . Deficiencies in AOC1 can lead to conditions such as dietary histamine intolerance and histaminosis .

Clinical Relevance

The enzyme’s ability to degrade histamine makes it a target for drug design, particularly in the treatment of inflammatory diseases and conditions related to histamine intolerance . Additionally, the structural information available for AOC1 has facilitated the development of computer-aided inhibitor design, which aims to create specific inhibitors that can modulate the enzyme’s activity without causing adverse effects .

Research and Development

Recent studies have focused on understanding the species-specific binding properties of AOC1 and its inhibitors. This research is essential for preclinical testing and the development of effective therapeutic agents . The structural bioinformatics and structural biology approaches applied in these studies provide valuable insights into the enzyme’s function and its interactions with potential inhibitors .

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 2025 TheBiotek. All Rights Reserved.