AOC1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to AOC1 Antibody

AOC1 antibodies are immunoreagents designed to bind specifically to the AOC1 protein, a membrane glycoprotein encoded by the AOC1 gene. This enzyme catalyzes the oxidative deamination of histamine, putrescine, and other polyamines, producing hydrogen peroxide and aldehydes. Dysregulation of AOC1 is linked to tumor progression, kidney development, and immune responses . Antibodies against AOC1 are widely used to study its expression, localization, and mechanistic roles in disease models.

Cancer Studies

AOC1 antibodies have been pivotal in uncovering dual roles for AOC1 in oncology:

  • Prostate Cancer: AOC1 acts as a tumor suppressor by promoting ferroptosis via ROS generation. Antibody-based assays confirmed its downregulation in aggressive tumors .

  • Colorectal/Gastric Cancer: AOC1 overexpression correlates with metastasis and poor prognosis. Knockdown studies using siRNA (validated by AOC1 antibodies) showed reduced proliferation and migration in vitro and in vivo .

  • Hepatocellular Carcinoma (HCC): High AOC1 expression drives metastasis via IL-6/JAK/STAT3 signaling, as demonstrated by immunohistochemistry .

Developmental Biology

AOC1 antibodies identified its regulation by the Wilms tumor protein (WT1) during kidney and gonad development, linking polyamine metabolism to organogenesis .

Validation and Quality Control

Boster Bio’s AOC1 antibody undergoes rigorous validation:

  • Specificity: Verified using recombinant AOC1 protein and blocking peptide experiments .

  • Reproducibility: Batch-to-blot consistency confirmed across WB (1:500–1:2000 dilution) and ICC/IF (1:50–1:200) .

  • Cross-Reactivity: No off-target binding observed in human, mouse, or rat tissues .

Diagnostic Potential

AOC1 antibodies are used to assess AOC1 levels in tumor biopsies. For example:

  • CRC Liver Metastases: AOC1 expression is significantly higher in metastatic lesions compared to primary tumors .

  • Prostate Cancer Staging: Low AOC1 levels correlate with higher Gleason scores and lymph node invasion .

Therapeutic Targeting

AOC1’s role in polyamine metabolism makes it a candidate for small-molecule inhibitors. Preclinical studies using AOC1 antibodies have identified ferroptosis induction as a strategy to combat therapy-resistant cancers .

Purchasing and Handling

  • Storage: Store at -20°C; avoid repeated freeze-thaw cycles .

  • Recommended Dilutions:

    • WB: 1:500–1:2000

    • ICC/IF: 1:50–1:200 .

Future Directions

  • Mechanistic Studies: Elucidate AOC1’s interaction with transcription factors like SOX15 or WT1 .

  • Clinical Trials: Explore AOC1-targeted therapies, such as antibody-drug conjugates (ADCs) or siRNA delivery systems .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks lead time (made-to-order)
Synonyms
AOC1 antibody; ERD12 antibody; At3g25760 antibody; K13N2.10 antibody; K13N2_8Allene oxide cyclase 1 antibody; chloroplastic antibody; EC 5.3.99.6 antibody; Early-responsive to dehydration 12 protein antibody
Target Names
AOC1
Uniprot No.

Target Background

Function
This antibody targets a protein involved in the biosynthesis of 12-oxo-phytodienoic acid (OPDA), a key precursor to jasmonic acid.
Database Links

KEGG: ath:AT3G25760

STRING: 3702.AT3G25760.1

UniGene: At.26518

Protein Families
Allene oxide cyclase family
Subcellular Location
Plastid, chloroplast.
Tissue Specificity
Highly expressed in fully developed leaves.

Q&A

What is AOC1 and why is it significant in cancer research?

AOC1 (Amine Oxidase Copper Containing 1) is a protein-coding gene that encodes a metal-binding membrane glycoprotein responsible for oxidatively deaminating putrescine, histamine, and related compounds . This protein has gained significant attention in cancer research due to its upregulation in several cancer types, particularly colorectal cancer and gastric cancer . Recent studies have established that AOC1 expression is significantly increased in human CRC tissues, especially in liver metastases, and this elevated expression correlates with worse prognosis in cancer patients . The significance of AOC1 in cancer research extends beyond its role as a biomarker, as functional studies have demonstrated that it actively promotes cancer progression by enhancing cellular proliferation, migration, and inhibiting apoptosis . The protein's involvement in multiple cellular pathways, including the AKT signaling pathway, makes it a promising therapeutic target for cancer treatment and an important molecule for understanding cancer biology .

What are the most commonly used applications for AOC1 antibodies in research?

AOC1 antibodies are versatile tools in cancer research with several established applications for investigating its biological functions and clinical relevance. Immunohistochemistry (IHC) stands as one of the most frequently used techniques, allowing researchers to detect endogenous levels of AOC1 protein in tissue microarrays (TMAs) and paraffin-embedded tissue sections . This application has proven particularly valuable for evaluating AOC1 expression in paired tumor and peritumoral tissues, as well as in metastatic samples . Western blotting represents another crucial application, enabling quantitative assessment of AOC1 protein expression following experimental manipulations such as gene knockdown or overexpression . Additionally, AOC1 antibodies are employed in immunoprecipitation assays to study protein-protein interactions and in flow cytometry to analyze AOC1 expression in different cell populations . For cancer progression studies, these antibodies are often used in conjunction with proliferation, migration, and apoptosis assays to establish connections between AOC1 expression and cellular phenotypes .

How should AOC1 antibodies be stored and handled to maintain optimal activity?

Proper storage and handling of AOC1 antibodies are critical for maintaining their specificity and sensitivity in experimental applications. According to manufacturer protocols, AOC1 antibodies should be stored at -20°C for long-term preservation of antibody integrity and activity . The antibody is typically formulated in a solution containing PBS (pH 7.4), glycerol (40%), and sodium azide (0.05%) to maintain stability during storage . When handling the antibody, it's essential to avoid repeated freeze-thaw cycles, which can lead to protein denaturation and loss of activity; instead, prepare small aliquots for single use upon receiving the antibody . During experimental procedures, maintain the antibody on ice when in use and return it to the appropriate storage temperature promptly to prevent degradation . For immunohistochemistry applications, optimization of antibody dilution is necessary, with a recommended starting dilution of 1:500 as used in published studies . Prior to application in critical experiments, researchers should validate each new lot of antibody using positive control tissues known to express AOC1, such as colorectal or gastric cancer tissues, to ensure consistent performance .

What are the optimal protocols for AOC1 antibody-based immunohistochemistry in cancer tissues?

For optimal AOC1 antibody-based immunohistochemistry in cancer tissues, researchers should follow a detailed protocol that includes several critical steps to ensure specific and reproducible staining. Begin with tissue preparation by deparaffinizing sections and performing antigen retrieval using citrate buffer (pH 6.0), a crucial step for unmasking epitopes that may have been altered during fixation . Following this, block endogenous peroxidases using 3% hydrogen peroxide, and prevent non-specific binding by applying 5% goat serum as a blocking agent . For primary antibody incubation, apply the AOC1 antibody at a 1:500 dilution and incubate overnight at 4°C to ensure thorough binding to the target antigen . After washing with PBS three times for 5 minutes each, apply an appropriate secondary antibody for 1 hour at room temperature, followed by visualization using diaminobenzidine chromogen and counterstaining with hematoxylin . For scoring and interpretation, researchers have established a detailed system based on staining intensity, with scores of 0 (negative), 1 (weak), 2 (moderate), and 3 (strong), which can be used to categorize samples into low and high expression groups . Include positive controls (known AOC1-expressing tissues) and negative controls (primary antibody omitted) in each staining batch to validate results, and consider using this approach to analyze tissue microarrays for high-throughput evaluation of AOC1 expression across multiple samples .

How can researchers effectively validate AOC1 antibody specificity?

Validating AOC1 antibody specificity is essential to ensure reliable and reproducible experimental results when studying this protein in cancer research contexts. A comprehensive validation approach should employ multiple complementary techniques, beginning with Western blot analysis to confirm that the antibody detects a single band of the expected molecular weight (approximately 85-90 kDa) in positive control samples . Performing parallel experiments with experimental knockdown models, such as siRNA-mediated AOC1 silencing, provides further validation by demonstrating reduced or absent antibody binding in AOC1-depleted samples compared to controls . For immunohistochemistry applications, researchers should compare staining patterns between tissues known to have high AOC1 expression (e.g., colorectal cancer tissues) and those with low or absent expression, while also conducting blocking peptide competition assays where pre-incubation of the antibody with its immunogen peptide should eliminate specific staining . Cross-reactivity testing against related family members, particularly other copper-containing amine oxidases, would further establish specificity . Additionally, comparing staining patterns obtained with different antibodies targeting distinct epitopes of AOC1 can provide confirmation of specificity, as consistent patterns across different antibodies strongly suggest true target detection rather than cross-reactivity .

What are the recommended protocols for AOC1 knockdown experiments in cancer cell lines?

For effective AOC1 knockdown experiments in cancer cell lines, researchers should implement a comprehensive protocol that ensures efficient gene silencing and proper assessment of functional outcomes. Begin by selecting appropriate cancer cell lines that express detectable levels of AOC1, such as colorectal cancer cell lines (SW480, HCT116) or gastric cancer lines (AGS, MKN45), based on the cancer type being studied . For transient knockdown, design multiple siRNAs targeting different regions of the AOC1 mRNA to ensure specificity and robust silencing, with transfection performed using established reagents like Lipofectamine 2000 at a final siRNA concentration of 50 nM . For stable knockdown, utilize lentiviral shRNA vectors followed by puromycin selection to establish cell lines with sustained AOC1 depletion, which is particularly valuable for long-term assays and in vivo experiments . Validation of knockdown efficiency is crucial and should be performed using both qRT-PCR to assess mRNA levels and Western blotting to confirm protein reduction, with measurements taken at 24-48 hours post-transfection for siRNA approaches . Following successful knockdown validation, conduct functional assays including proliferation (CCK-8 and colony formation), migration (Transwell and wound healing), and apoptosis studies (flow cytometry with Annexin V/PI staining) to comprehensively assess the impact of AOC1 depletion on cancer cell phenotypes .

What signaling pathways are affected by AOC1 expression in cancer cells?

AOC1 exerts its pro-oncogenic effects through modulation of several key signaling pathways that collectively promote cancer cell survival, proliferation, and metastatic potential. The AKT signaling pathway has been identified as a primary mechanism through which AOC1 influences cancer progression, with studies in gastric cancer cells demonstrating that AOC1 knockdown significantly reduces AKT phosphorylation, thereby inhibiting this pro-survival pathway . Additionally, AOC1 interfaces with epithelial-to-mesenchymal transition (EMT) signaling, as evidenced by functional analyses showing that AOC1 knockdown in colorectal cancer cells inhibits EMT, a critical process for cancer cell invasion and metastasis . The mitochondrial apoptosis pathway is also regulated by AOC1, with silencing experiments revealing that AOC1 knockdown increases the expression of pro-apoptotic factors (Bax, Caspase-9, Caspase-3) while decreasing anti-apoptotic protein Bcl2, thereby promoting cancer cell death . Furthermore, AOC1's enzymatic function in deaminating polyamines potentially impacts polyamine metabolism pathways, which are known regulators of cell proliferation, migration, and apoptosis . The extensive crosstalk between these signaling networks creates a complex regulatory environment wherein AOC1 acts as a multifaceted oncogenic driver, positioning it as both a biomarker and a potential therapeutic target in cancer management .

What are the emerging therapeutic applications targeting AOC1 in cancer treatment?

Emerging therapeutic applications targeting AOC1 represent a promising frontier in cancer treatment strategy development, particularly for gastrointestinal malignancies where AOC1 overexpression correlates with poor clinical outcomes. Preclinical evidence from xenograft tumor formation studies in nude mice has demonstrated that knockdown of AOC1 significantly inhibits tumor growth in vivo, providing proof-of-concept for AOC1-targeted therapeutic approaches . Several potential intervention strategies are being explored, including the development of small molecule inhibitors designed to block AOC1's enzymatic activity, thereby interfering with polyamine metabolism and downstream signaling events that promote cancer progression . RNA interference-based therapeutics (siRNA, shRNA) delivered via nanoparticles represent another promising approach, as demonstrated by the effectiveness of AOC1 knockdown in reducing cancer cell proliferation, migration, and survival in experimental models . Antibody-drug conjugates targeting AOC1 could potentially deliver cytotoxic agents specifically to cancer cells with high AOC1 expression, minimizing off-target effects . Additionally, combination therapies that pair AOC1 inhibition with established treatments may prove particularly effective, especially given AOC1's role in regulating the AKT pathway, which is implicated in treatment resistance mechanisms . As research advances, clinical trials evaluating these approaches will be crucial for translating the mounting preclinical evidence for AOC1's therapeutic potential into effective treatment options for cancer patients .

What are common challenges in AOC1 immunohistochemistry and how can they be addressed?

Researchers performing AOC1 immunohistochemistry may encounter several technical challenges that can impact staining quality and interpretation reliability. Background staining represents a frequent issue that can obscure specific AOC1 signals, often arising from insufficient blocking or non-specific antibody binding; this can be addressed by optimizing blocking conditions (extending blocking time to 1 hour with 5% goat serum) and carefully titrating primary antibody concentration based on preliminary experiments . Antigen retrieval challenges are also common, as suboptimal retrieval can reduce AOC1 detection in formalin-fixed, paraffin-embedded tissues; researchers should evaluate multiple retrieval methods (heat-induced epitope retrieval with citrate buffer at pH 6.0 has proven effective) and durations to maximize signal while preserving tissue integrity . Variability in staining intensity between batches can compromise comparative analyses, necessitating the inclusion of positive control tissues in each staining run and potentially implementing automated staining platforms to enhance consistency . Quantification difficulties arise when translating visual assessments into meaningful data; establishing a standardized scoring system based on staining intensity (0-3 scale) and extent (percentage of positive cells) as described in published protocols can improve reproducibility and inter-observer agreement . For tissue microarray applications, researchers should address heterogeneity concerns by including multiple cores per case and validating findings on whole-tissue sections when possible .

How can researchers resolve inconsistent results between AOC1 mRNA and protein expression analyses?

Discrepancies between AOC1 mRNA and protein expression measurements represent a significant challenge in research settings that can arise from multiple biological and technical factors. Post-transcriptional regulation mechanisms, including microRNA-mediated inhibition of translation or alterations in mRNA stability, may lead to situations where mRNA levels do not proportionally correlate with protein expression; researchers should consider examining relevant regulatory factors, such as microRNAs predicted to target AOC1, when encountering such discrepancies . Differences in detection sensitivity between techniques can also contribute to inconsistencies, as qRT-PCR typically offers higher sensitivity for transcript detection compared to Western blotting for proteins; optimizing protein extraction protocols specifically for membrane-bound proteins like AOC1 can help address this disparity . Temporal differences in sampling may impact concordance, as protein expression changes often lag behind mRNA alterations; conducting time-course experiments with matched samples for both analyses can help characterize this relationship . Technical variability in normalization approaches presents another challenge, particularly when different reference genes or proteins are used across methods; employing multiple, validated reference controls for both mRNA (e.g., GAPDH, β-actin) and protein (e.g., β-actin, GAPDH) analyses can improve reliability . When persistent discrepancies occur, researchers should consider alternative techniques such as in situ hybridization paired with immunohistochemistry to examine both mRNA and protein expression within the same cellular context, providing spatial information that may reveal tissue- or cell-specific regulatory patterns .

What are promising areas for future investigation of AOC1 in cancer biology?

Several promising avenues for future AOC1 research in cancer biology present opportunities to deepen our understanding of its mechanisms and therapeutic potential. Single-cell transcriptomics and proteomics approaches represent a frontier for investigating cell-specific AOC1 expression patterns within the heterogeneous tumor microenvironment, potentially revealing distinct cellular subpopulations where AOC1 expression is particularly relevant to disease progression or treatment resistance . The establishment of AOC1-targeted drug discovery programs focused on developing specific inhibitors would advance translational applications, requiring detailed structural studies and high-throughput screening approaches to identify compounds that selectively modulate AOC1 activity with minimal off-target effects . Investigation of AOC1's role in cancer stem cell biology presents another intriguing direction, as preliminary evidence suggests connections between polyamine metabolism and stem cell maintenance in various cancer types, potentially linking AOC1 to tumor initiation and recurrence . Exploration of AOC1's involvement in immune response modulation within the tumor microenvironment could uncover interactions with immune cells that influence cancer progression and response to immunotherapies, particularly given the established roles of histamine (an AOC1 substrate) in immune regulation . Development of AOC1-based liquid biopsy approaches for non-invasive cancer detection and monitoring represents a clinically relevant research direction, requiring studies correlating circulating AOC1 levels with tissue expression and disease status across large patient cohorts .

How might novel AOC1 antibodies and detection methods advance cancer research?

The development of novel AOC1 antibodies and detection methods holds significant promise for advancing cancer research through enhanced sensitivity, specificity, and application versatility. Next-generation recombinant antibodies designed against specific epitopes of AOC1 could offer improved batch-to-batch consistency compared to traditional polyclonal antibodies, addressing a major challenge in longitudinal studies and multi-center research collaborations . Antibodies specifically targeting post-translational modifications of AOC1 would enable investigation of regulatory mechanisms affecting AOC1 function, as phosphorylation, glycosylation, and other modifications likely influence its enzymatic activity and subcellular localization in cancer cells . Development of fluorescently-labeled AOC1 antibodies compatible with multiplexed immunofluorescence and imaging mass cytometry would facilitate comprehensive spatial profiling of AOC1 in relation to other cancer biomarkers and microenvironmental features within the same tissue section, providing insights into cellular interactions and heterogeneity . Advanced in vivo imaging approaches utilizing radiolabeled or near-infrared labeled AOC1 antibodies could enable non-invasive monitoring of AOC1 expression in preclinical models and potentially patients, supporting personalized medicine approaches based on AOC1 status . Implementation of highly sensitive detection methods such as proximity ligation assays or digital ELISA platforms could improve quantification of AOC1 in limited biological samples, including liquid biopsies, potentially enabling earlier detection of AOC1-expressing cancers and more precise monitoring of treatment response .

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.