AOC3 is a copper-containing amine oxidase with a topaquinone cofactor that exists as both a membrane-bound protein and a soluble form in human serum. The soluble form is derived from a cleavage product of the membrane-bound protein . AOC3 displays dual functionality: it acts as an adhesion molecule facilitating leukocyte binding to endothelial cells and as an enzyme catalyzing the oxidative deamination of primary amines .
This protein has attracted significant research interest due to its involvement in various inflammatory processes and potential as a therapeutic target for conditions including atherosclerosis, non-alcoholic steatohepatitis (NASH), and other inflammatory diseases . The gene for human AOC3 is located on chromosome 11, and its mouse ortholog is well-studied in various disease models .
Human AOC3 is a Type II integral membrane protein spanning from Gly27 to Asn763 in its mature form . Researchers have determined the crystal structure of a soluble, proteolytically cleaved form of human AOC3 (sAOC3) extracted from human plasma at 2.6 Å and 2.95 Å resolutions .
The enzyme contains a topaquinone (TPQ) cofactor that can exist in both "on-copper" (inactive) and "off-copper" (active) conformations. In the 2.6 Å structure, an imidazole molecule is hydrogen-bonded to the TPQ cofactor in its inactive on-copper conformation, while in the 2.95 Å structure, an imidazole molecule is covalently bound to the active off-copper conformation of TPQ .
Several key amino acid residues have been identified that influence substrate specificity:
Residue | Function |
---|---|
Met211 | Key residue for substrate specificity |
Thr212 | Involved in imidazole binding in substrate channel |
Tyr394 | Involved in imidazole binding in substrate channel |
Leu469 | Key residue for substrate specificity |
A triple mutant (Met211Val/Tyr394Asn/Leu469Gly) was shown to change substrate preferences of human AOC3 toward those of human AOC2, another copper-containing monoamine oxidase .
AOC3 expression varies significantly across different tissues:
Tissue Type | AOC3 Expression Level |
---|---|
Lung Endothelium | High |
Heart Endothelium | High |
Intestinal Endothelium | High |
Brain | Low |
Spleen | Low |
Kidney | Low |
Liver | Low |
The highest expression is found in the endothelium of lung, heart, and intestine, while expression is low in tissues such as brain, spleen, kidney, and liver .
AOC3 is primarily expressed in:
Vascular smooth muscle cells (VSMCs)
Endothelial cells (to a lesser extent)
Adipocytes (highly expressed)
Interestingly, AOC3 is not expressed by macrophages . In vascular tissues, AOC3 is predominantly found in the media layer and is upregulated during vessel inflammation .
AOC3 vascular expression is regulated at sites of inflammation through its release from intracellular granules where the protein is stored . In mouse models, AOC3 protein expression gradually increases in the abdominal aorta with age, reaching statistical significance at 70 weeks .
AOC3 catalyzes the oxidative deamination of primary amines in a reaction that produces:
The enzyme is sensitive to inhibition by semicarbazide, which is why it is sometimes referred to as semicarbazide-sensitive amine oxidase (SSAO) .
AOC3 can accept a variety of primary amines with different chemical features:
Substrate | Relative Activity | Km Value Range |
---|---|---|
Methylamine | High | - |
Benzylamine | High | - |
Dopamine | High | - |
Cysteamine | High | - |
Branched-chain amines | Moderate | - |
Aliphatic amines | Moderate | - |
Aminoacetone | Variable | - |
The measured k<sub>cat</sub>/K<sub>m</sub> values for human AOC3 generally range between 10² and 10⁴ M⁻¹s⁻¹ . The K<sub>m</sub>(O₂) of AOC3 approximates the partial pressure of oxygen found in the interstitial space .
Comparison of purified murine to human enzyme indicates k<sub>cat</sub>/K<sub>m</sub> values that are within 3 to 4-fold, with the exception of methylamine and aminoacetone that are approximately 10-fold more active with human AOC3 . This suggests caution is needed when screening the efficacy of inhibitors designed against human enzymes in non-transgenic mouse models .
AOC3 plays a crucial role in the adherence of certain lymphocyte subtypes to inflamed endothelial tissues . Through real-time imaging, it has been shown that AOC3 mediates slow rolling, firm adhesion, and transmigration of leukocytes in vessels at inflammatory sites and lymphoid tissues .
The adhesive function of AOC3 is involved in the process of leukocyte extravasation, a critical feature of inflammatory responses. The role of AOC3 amine oxidase activity in this process is not fully defined, but it appears to be carbohydrate-dependent .
The relationship between the enzymatic activity of AOC3 and its adhesion properties remains an area of active investigation. Studies have shown that the soluble form of AOC3 is derived mainly from endothelial cells during inflammation, and its inhibition diminishes leukocyte migration and inflammation in various pathologies .
Research on the role of AOC3 in atherosclerosis has yielded contradictory findings:
Study Finding | Impact on Atherosclerosis |
---|---|
AOC3 knockout in ApoE⁻/⁻ mice | Increased plaque size at early stages (15 weeks) |
AOC3 knockout in ApoE⁻/⁻ mice | Increased CD3+ T cell infiltration in plaques |
AOC3 knockout in ApoE⁻/⁻ mice | Increased MCP-1 expression |
AOC3 inhibition (some studies) | Reduction in atheromatous plaques |
AOC3 inhibition (other studies) | Increase in atheromatous plaques |
In 15-week-old ApoE⁻/⁻AOC3⁻/⁻ mice, the absence of AOC3 was associated with increased lesion size, α-smooth muscle actin (α-SMA), and CD3 staining in the plaque, independently of cholesterol modification . At 25 weeks, advanced plaques were larger with equivalent staining for α-SMA, while CD3+ cells increased in the media from ApoE⁻/⁻AOC3⁻/⁻ mice .
The mechanism appears to involve vascular smooth muscle cell dedifferentiation associated with higher T cell recruitment in plaques, explained by MCP-1 (Monocyte Chemoattractant Protein-1) augmentation .
AOC3 has emerged as a potential therapeutic target for NASH:
A Phase IIa, double-blind, placebo-controlled, multicenter trial investigated the AOC3 inhibitor BI 1467335 in adults with NASH
The trial showed that BI 1467335 dose-dependently inhibited AOC3 activity
Doses ≥3 mg achieved >80% inhibition of AOC3 activity at Week 4
At Week 12, doses ≥3 mg dose-dependently reduced liver injury biomarkers alanine aminotransferase (ALT) and caspase-cleaved cytokeratin 18 (CK-18 caspase)
AOC3 levels are elevated in:
Diabetes
Serum of ApoE⁻/⁻ mice (increased 20-fold in 70-week-old mice compared to 11-week-old or 70-week-old wild-type mice)
AOC3 is considered an independent marker of atherosclerosis and can extend to coronary artery disease .
Several AOC3 inhibitors have been developed and studied:
Inhibitor | Target | Effect |
---|---|---|
BI 1467335 | AOC3 | Dose-dependent inhibition of AOC3 activity; reduction in liver injury biomarkers in NASH patients |
LJP1586 | AOC3 | Used to measure differentiation markers in human vascular smooth muscle cells |
Imidazole | AOC3 | Inhibitory role at high concentrations used in crystallization |
Semicarbazide | AOC3 | General inhibitor of amine oxidase activity |
A Phase IIa trial of BI 1467335 in NASH patients revealed:
Mean AOC3 activities relative to baseline at Week 12 were: 90.4% (placebo), 26.5% (1 mg), 10.4% (3 mg), 5.0% (6 mg), 3.3% (10 mg)
All tested BI 1467335 doses were well tolerated with no clinically relevant treatment-emergent safety signals
The trial was registered with ClinicalTrials.gov (NCT03166735) and the European Union Drug Regulating Authorities Clinical Trials Database (EudraCT 2016-000499-83)
Despite promising results in some conditions, caution is warranted when targeting AOC3:
The inhibition of AOC3 in limiting atherosclerosis shows contradictory results
The dual role of AOC3 impacts therapeutic strategies using pharmacological regulators of SSAO activity
AOC3 may have an important role in atherosclerosis independent of its canonical inflammatory effect
Several aspects of AOC3 biology require further investigation:
The in vivo substrate(s) of AOC3 remain unknown but could provide valuable clues to the enzyme's function
The precise relationship between AOC3's enzymatic and adhesion functions needs clarification
The contradictory findings regarding AOC3's role in atherosclerosis warrant resolution
The availability of purified human AOC3 provides valuable research opportunities:
Expression of untagged, soluble human AOC3 in insect cells provides a relatively simple means of obtaining pure enzyme for research
Differentiated murine 3T3-L1 adipocytes show a uniform distribution of AOC3 on the cell surface and whole cell Km values that are reasonably close to values measured using purified enzymes
These studies support the relevance of kinetic parameters measured with isolated AOC3 variants to adipocyte function
Future research on AOC3 should focus on:
Identifying physiological substrates of AOC3
Resolving the dual functional roles of AOC3 in different tissues
Developing specific inhibitors that could target pathological functions while preserving beneficial ones
Further clinical trials to evaluate the efficacy and safety of AOC3 inhibitors in various conditions
VAP-1, AOC3, HPAO, VAP1, Membrane primary amine oxidase, Copper amine oxidase, HPAO, Semicarbazidesensitive amine oxidase, SSAO, Vascular adhesion protein 1.
ADPGRGGDGG EPSQLPHCPS VSPSAQPWTH PGQSQLFADL SREELTAVMR FLTQRLGPGL VDAAQARPSD NCVFSVELQL PPKAAALAHL DRGSPPPARE ALAIVFFGRQ PQPNVSELVV GPLPHPSYMR DVTVERHGGP LPYHRRPVLF QEYLDIDQMI FNRELPQASG LLHHCCFYKH RGRNLVTMTT APRGLQSGDR ATWFGLYYNI SGAGFFLHHV GLELLVNHKA LDPARWTIQK VFYQGRYYDS LAQLEAQFEA GLVNVVLIPD NGTGGSWSLK SPVPPGPAPP LQFYPQGPRF SVQGSRVASS LWTFSFGLGA FSGPRIFDVR FQGERLVYEI SLQEALAIYG GNSPAAMTTR YVDGGFGMGK YTTPLTRGVD CPYLATYVDW HFLLESQAPK TIRDAFCVFE QNQGLPLRRH HSDLYSHYFG GLAETVLVVR SMSTLLNYDY VWDTVFHPSG AIEIRFYATG YISSAFLFGA TGKYGNQVSE HTLGTVHTHS AHFKVDLDVA GLENWVWAED MVFVPMAVPW SPEHQLQRLQ VTRKLLEMEE QAAFLVGSAT PRYLYLASNH SNKWGHPRGY RIQMLSFAGE PLPQNSSMAR GFSWERYQLA VTQRKEEEPS SSSVFNQNDP WAPTVDFSDF INNETIAGKD LVAWVTAGFL HIPHAEDIPN TVTVGNGVGF FLRPYNFFDE DPSFYSADSI YFRGDQDAGA CEVNPLACLP QAAACAPDLP AFSHGGFSHN HHHHHH
Human AOC3 is a Type II integral membrane protein with a topaquinone cofactor. It spans from Gly27 to Asn763 in its mature form (accession number Q16853) and shares approximately 83% amino acid sequence identity with mouse AOC3 . AOC3 functions both as an adhesion molecule mediating leukocyte-endothelial interactions and as an enzyme catalyzing oxidative deamination of primary amines. Its structure includes a transmembrane domain anchoring it to cell membranes, though a soluble form also circulates in human serum .
AOC3 catalyzes the oxidative deamination of small primary amines such as methylamine, benzylamine, and aminoacetone. This reaction produces aldehydes, ammonia, and hydrogen peroxide (H₂O₂) . The enzyme is characterized by its sensitivity to inhibition by semicarbazide, which is why it's also known as semicarbazide-sensitive amine oxidase (SSAO). The enzymatic activity contributes to inflammation modulation, vascular function, and cell signaling through the production of reactive products, particularly H₂O₂.
AOC3 plays a critical role in leukocyte extravasation—the process where immune cells migrate from blood vessels into tissues. It functions as an adhesion molecule on endothelial cells, facilitating the attachment and subsequent migration of leukocytes, particularly during inflammatory responses . Research demonstrates that AOC3 knockdown reduces CD4+ T-cell attachment to cells and decreases transendothelial migration in vitro, as well as reducing CD4+ T-cell trafficking to the lung in vivo . This adhesion function appears particularly important in inflammatory conditions and may contribute to pathological processes in various diseases.
AOC3 expression is highest in the endothelium of lung, heart, and intestine, with relatively lower expression in tissues such as brain, spleen, kidney, and liver . Significant expression has also been demonstrated in vascular smooth muscle cells (VSMCs), where it appears to influence differentiation and extracellular matrix organization . Within the vascular system, AOC3 displays cell-type specific expression patterns, with studies showing co-localization primarily with VSMCs in the media and atherosclerotic plaques, and some expression in endothelial cells .
AOC3 vascular expression is dynamically regulated during inflammation through its release from intracellular granules where the protein is stored under normal conditions . Upon inflammatory stimulation, these granules release AOC3, increasing its presence on the cell surface where it can participate in leukocyte adhesion. This regulated expression allows for rapid response to inflammatory signals and controlled immune cell recruitment. Additionally, the soluble form of AOC3 increases in serum during inflammatory conditions, potentially serving as a biomarker for vascular inflammation .
AOC3 expression shows disease-specific alterations:
Diabetic retinopathy: Significantly elevated in retinal vasculature, retinal parenchyma, and choroidal vasculature of patients with non-proliferative diabetic retinopathy compared to diabetic donors without retinopathy and non-diabetic controls
Lung cancer: Markedly decreased in tumor tissue compared to normal lung tissue at both mRNA and protein levels, with lower expression correlating with poorer survival probability
Atherosclerosis: Increased in vascular tissues with atherosclerotic progression, with significant age-dependent increases observed in ApoE−/− mouse models
These contrasting patterns suggest context-dependent regulation and functions of AOC3 across different pathological processes.
Researchers can employ several validated techniques for AOC3 detection and measurement:
For protein detection and localization:
Immunohistochemistry (IHC) using specific antibodies to visualize distribution in tissue sections
Western blotting for quantitative protein analysis, typically using validated antibodies such as MAB3957 (clone #393112)
Confocal microscopy with co-localization studies using cell-specific markers (e.g., α-SMA for VSMCs, VWF for endothelial cells)
For serum measurement:
Quantikine Human VAP-1 Immunoassay (R&D Systems) provides reliable quantification of soluble AOC3 in serum samples
For gene expression analysis:
RNA sequencing for comprehensive transcriptomic analysis
For enzymatic activity:
Assays measuring hydrogen peroxide production as an indicator of AOC3 activity
Inhibition studies using semicarbazide or more specific inhibitors like LJP1586
Robust AOC3 research requires appropriate controls:
Negative controls:
Positive controls:
Specificity controls:
Several established approaches allow manipulation of AOC3:
Genetic approaches:
RNA interference using AOC3-targeted shRNA or siRNA plasmids (e.g., pLKO_005 as control plasmid and AOC3-shRNA for knockdown)
CRISPR-Cas9 gene editing for knockout generation
Overexpression vectors for gain-of-function studies
Pharmacological approaches:
Semicarbazide as a traditional but non-specific inhibitor
Animal models:
AOC3−/− knockout mice
Disease-specific models such as ApoE−/−AOC3−/− double knockout mice for atherosclerosis studies
Conditional knockouts for tissue-specific investigations
Studies have demonstrated elevated AOC3 expression in human eyes with non-proliferative diabetic retinopathy (NPDR). Research by Borta et al. examined human donor eyes with clinically documented NPDR features including microaneurysms, hemorrhages, intraretinal microvascular abnormalities, venous beadings, macula edema, and hard exudates . They found increased AOC3 immunoreactivity in retinal vessels, retinal parenchyma, and choroidal vasculature compared to diabetic donors without retinopathy and non-diabetic controls .
The upregulated AOC3 likely contributes to inflammatory processes in diabetic retinopathy by facilitating leukocyte extravasation from the microvasculature, particularly neutrophil recruitment. This aligns with evidence that inflammatory processes contribute significantly to diabetic retinopathy development and that anti-inflammatory therapies can inhibit disease progression in animal models .
AOC3 appears to function as a tumor suppressor in lung cancer. Research demonstrates:
Downregulation in tumors: AOC3 is significantly decreased in lung cancer tissues compared to normal lung at both mRNA and protein levels
Correlation with survival: Lower AOC3 expression correlates with poorer survival probability across different patient cohorts
Epigenetic regulation: MicroRNA-3691-5p silences AOC3 expression in lung cancer, promoting tumor progression
EMT regulation: AOC3 downregulation increases migration and epithelial-mesenchymal transition (EMT) in cancer cells
Immune effects: Reduced AOC3 expression decreases CD4+ T-cell attachment to lung cancer cells and reduces immune cell trafficking to tumors, potentially enabling immune evasion
These findings suggest restoring AOC3 expression could represent a potential therapeutic strategy for lung cancer by both inhibiting EMT and enhancing anti-tumor immunity.
AOC3's role in atherosclerosis appears complex and somewhat contradictory. Studies using ApoE−/−AOC3−/− double knockout mice revealed:
Increased plaque formation: Absence of AOC3 associated with larger atherosclerotic lesions at both 15 and 25 weeks of age
Altered immune cell composition: Enhanced CD3+ T-cell infiltration in plaques and media, without changes in macrophage content
VSMC phenotype changes: Decreased expression of contractile markers in VSMCs, suggesting dedifferentiation
Inflammatory signaling: Elevated MCP-1 expression in younger (15-week-old) knockout mice
This contradicts some previous research suggesting AOC3 inhibition might reduce atherosclerotic plaques. The discrepancies might reflect:
Different methodologies (total knockout vs. inhibition)
Cell-specific effects (endothelial vs. VSMC roles)
Temporal factors in disease progression
Off-target effects of inhibitors (e.g., semicarbazide affects lysyl oxidase)
AOC3 expression in human coronary arteries shows co-localization primarily with VSMCs and some endothelial cells, suggesting cell type-specific functions within atherosclerotic environments .
When confronting contradictory results about AOC3, researchers should:
Analyze methodological differences systematically:
Compare knockout models vs. pharmacological inhibition approaches
Evaluate acute vs. chronic interventions
Consider species and strain differences
Examine disease models (genetic vs. induced) and severity
Investigate cell type-specific effects:
Design studies to isolate endothelial-specific vs. VSMC-specific functions
Use conditional knockout models targeting specific cell populations
Perform co-culture experiments to examine cellular interactions
Consider temporal dynamics:
Examine potential compensatory mechanisms:
Assess changes in related adhesion molecules or enzymes
Investigate alternative inflammatory pathways activated in AOC3 deficiency
Consider developmental adaptations in genetic models
AOC3 demonstrates distinct functions across different cell types:
In endothelial cells:
Primary function as an adhesion molecule facilitating leukocyte binding
Stored in intracellular granules and mobilized during inflammation
In vascular smooth muscle cells:
Associated with differentiation status and contractile phenotype
Expression increases during terminal differentiation
Influences extracellular matrix organization
In the tumor microenvironment:
Regulates immune cell recruitment and attachment
Influences cancer cell migration and EMT
These diverse functions may be explained by:
Cell-specific binding partners and signaling pathways
Different subcellular localization patterns
Varying enzymatic substrates available in different cellular contexts
Distinct regulatory mechanisms controlling expression
Modern molecular profiling approaches could resolve key knowledge gaps:
Single-cell RNA sequencing:
Map cell-specific expression patterns across tissues
Identify co-expressed genes suggesting functional networks
Track dynamic changes during disease progression
Proteomics approaches:
Identify AOC3 binding partners in different cell types
Characterize post-translational modifications affecting function
Quantify changes in the "AOC3 interactome" during disease states
Epigenetic profiling:
Map regulatory elements controlling AOC3 expression
Identify disease-associated epigenetic modifications
Characterize microRNA networks regulating AOC3
Metabolomics:
Identify physiological substrates for AOC3 enzymatic activity
Measure downstream metabolites produced by AOC3 function
Evaluate metabolic consequences of AOC3 inhibition
Several promising research directions warrant investigation:
Cell-specific functions:
Conditional knockout studies targeting endothelial cells vs. VSMCs
Investigation of AOC3's role in specific immune cell populations
Analysis of tissue-specific substrate preferences and enzymatic activities
Mechanistic studies:
Signaling pathways connecting AOC3 to VSMC differentiation
Molecular mechanisms underlying its dual adhesion/enzymatic functions
Interaction with extracellular matrix components in vascular remodeling
Therapeutic development:
Cell-specific targeting approaches for atherosclerosis
AOC3 restoration strategies for lung cancer
Novel selective inhibitors with improved specificity profiles
Biomarkers to predict response to AOC3-targeted interventions
Translational research:
Validation of findings in diverse human populations
Correlation of soluble AOC3 with disease progression and outcomes
Integration with other inflammatory and vascular biomarkers
Despite significant advances, several crucial questions remain:
Why does AOC3 show opposing expression patterns in different diseases?
Upregulated in diabetic retinopathy and atherosclerosis
Downregulated in lung cancer
What tissue-specific factors drive these divergent patterns?
How do the enzymatic and adhesion functions interact mechanistically?
Is the enzymatic activity required for adhesion function?
Do the adhesion properties influence substrate accessibility?
Can these functions be selectively targeted?
What explains the contradictory effects of AOC3 deficiency in atherosclerosis?
Why does AOC3 deletion sometimes increase and sometimes decrease plaque burden?
What determines whether inhibition will be beneficial or harmful?
How do compensatory mechanisms affect outcomes?
What are the key epigenetic regulators of AOC3 across different cell types?
Beyond miR-3691-5p in lung cancer, what other microRNAs regulate AOC3?
What transcription factors control cell-specific expression?
How does inflammation modify the epigenetic landscape around AOC3?
Amine Oxidase Copper Containing 3 (AOC3), also known as semicarbazide-sensitive amine oxidase (SSAO) and vascular adhesion protein 1 (VAP-1), is an enzyme that plays a crucial role in various physiological and pathological processes. This enzyme is part of the copper-containing amine oxidase family, which catalyzes the oxidative deamination of primary amines to aldehydes, ammonia, and hydrogen peroxide .
AOC3 is a membrane-bound enzyme that is predominantly expressed in the endothelial cells of blood vessels. It contains a copper ion at its active site, which is essential for its enzymatic activity. The enzyme facilitates the adhesion of leukocytes to the endothelium, a critical step in the inflammatory response. By oxidizing primary amines, AOC3 generates reactive aldehydes and hydrogen peroxide, which can modulate vascular function and leukocyte behavior .
AOC3 is involved in several physiological processes, including:
AOC3 has been associated with various pathological conditions, including:
Human recombinant AOC3 is produced using recombinant DNA technology, which involves inserting the gene encoding AOC3 into a suitable expression system, such as bacteria or mammalian cells. This allows for the large-scale production of the enzyme for research and therapeutic purposes. Recombinant AOC3 retains the enzymatic activity and functional properties of the native protein, making it a valuable tool for studying the enzyme’s role in health and disease.