AP3M1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receiving them. Delivery times may vary depending on the shipping method and location. For specific delivery times, please contact your local distributor.
Synonyms
Adapter-related protein complex 3 mu-1 subunit antibody; AP-3 adapter complex mu3A subunit antibody; AP-3 complex subunit mu-1 antibody; AP3M1 antibody; AP3M1_HUMAN antibody; Clathrin adaptor complex AP3 mu 3A subunit antibody; Mu-adaptin 3A antibody; mu3A antibody; Mu3A-adaptin antibody
Target Names
AP3M1
Uniprot No.

Target Background

Function

AP3M1 Antibody is a component of the AP-3 complex, an adaptor-related complex that is not clathrin-associated. This complex is associated with the Golgi region as well as more peripheral structures. It facilitates the budding of vesicles from the Golgi membrane and is potentially involved in trafficking to lysosomes. In collaboration with the BLOC-1 complex, AP-3 plays a crucial role in directing cargo into vesicles assembled at cell bodies for delivery into neurites and nerve terminals.

Gene References Into Functions
  1. Structural basis for the recognition of tyrosine-based sorting signals by the mu3A subunit of the AP-3 adaptor complex PMID: 23404500
  2. Research findings demonstrate that the intact and stable AP-3 complex is essential for HIV-1 assembly and release. Furthermore, the involvement of the AP-3 complex in the late stages of the HIV-1 replication cycle is independent of clathrin-mediated endocytosis. PMID: 22875976
  3. Current findings suggest that AP3 complex genes may not play a significant role in the pathogenesis of schizophrenia. PMID: 19481122
Database Links

HGNC: 569

OMIM: 610366

KEGG: hsa:26985

STRING: 9606.ENSP00000347408

UniGene: Hs.500104

Protein Families
Adaptor complexes medium subunit family
Subcellular Location
Golgi apparatus. Cytoplasmic vesicle membrane; Peripheral membrane protein; Cytoplasmic side.

Q&A

What is AP3M1 and why is it an important research target?

AP3M1 (Adaptor-Related Protein Complex 3, mu 1 Subunit) is a medium adaptin subunit of the AP-3 complex, which is a heterotetramer involved in vesicle trafficking pathways. The AP-3 complex facilitates vesicle budding from the Golgi membrane and plays a direct role in trafficking to lysosomes. AP3M1 has also been shown to interact with the HIV-1 virulence protein Nef, making it an important target for research on intracellular trafficking mechanisms and viral pathogenesis . Understanding AP3M1 function requires specific antibodies for detecting its expression, localization, and interactions in experimental systems, which makes AP3M1 antibodies essential tools in cell biology and neuroscience research .

What criteria should be considered when selecting an AP3M1 antibody for research?

When selecting an AP3M1 antibody, researchers should consider multiple validation parameters to ensure experimental success. First, verify the antibody's reactivity with your species of interest; available AP3M1 antibodies show reactivity with human, mouse, and rat samples, with some also reacting with chicken, cow, dog, hamster, and horse samples . Second, confirm that the antibody has been validated for your specific application (WB, IHC, IF, ELISA) through published literature or manufacturer validation data . Third, consider the antibody's binding epitope - some target the C-terminal region (e.g., AA 340-390), while others target internal regions, which may affect protein detection under certain experimental conditions . Finally, evaluate the purification method (affinity chromatography via peptide column) and storage conditions (typically 4°C for three months or -20°C for up to one year) to ensure antibody stability and performance .

How do polyclonal and monoclonal AP3M1 antibodies differ in research applications?

Polyclonal and monoclonal AP3M1 antibodies present distinct advantages in research scenarios based on their inherent properties. Polyclonal AP3M1 antibodies, such as ABIN6991550 and A12628, recognize multiple epitopes on the AP3M1 protein, offering higher sensitivity for detecting low-abundance proteins in techniques like Western blotting (1:1000-1:8000 dilution) and immunohistochemistry (1:50-1:500 dilution) . These antibodies provide robust signal amplification but may show batch-to-batch variation. In contrast, monoclonal AP3M1 antibodies (e.g., R08-8D2) recognize a single epitope with high specificity, making them ideal for distinguishing between highly homologous proteins like AP3M1 and AP3M2 . The choice between polyclonal and monoclonal antibodies should be guided by experimental requirements: use polyclonals for maximum detection sensitivity and monoclonals when absolute specificity is required, particularly in co-immunoprecipitation experiments or when discriminating between closely related AP complex subunits .

What are the optimal dilution ranges for AP3M1 antibodies in different applications?

Optimal dilution ranges for AP3M1 antibodies vary significantly across different experimental applications and should be carefully optimized for each specific antibody. For Western blot analysis, the recommended dilution ranges from 1:1000 to 1:8000, with antibodies like 12114-1-AP showing successful detection at 1-2 μg/mL in human brain tissue lysates . For immunohistochemistry applications on paraffin-embedded sections, start with dilutions between 1:50 and 1:500, with 5 μg/mL being an effective concentration for mouse brain tissue . Immunofluorescence applications typically require higher antibody concentrations, with recommended starting dilutions of 1:50 to 1:500 or approximately 20 μg/mL for mouse brain tissue and HeLa cells . For ELISA applications, optimal dilutions need to be empirically determined for each system. It is essential to perform preliminary titration experiments with positive controls (HEK-293 cells, NIH/3T3 cells for WB; mouse cerebellum tissue for IHC) to establish the optimal working concentration for your specific experimental conditions and antibody lot .

What sample preparation techniques enhance AP3M1 detection in Western blotting?

Effective sample preparation is critical for successful AP3M1 detection in Western blotting due to its moderate abundance and calculated molecular weight of approximately 47 kDa. Begin by extracting proteins using a lysis buffer containing protease inhibitors to prevent degradation of AP3M1, which is particularly important when working with brain tissue samples where proteolytic activity is high . For optimal protein denaturation, heat samples at 95°C for 5 minutes in reducing sample buffer containing SDS and β-mercaptoethanol. When loading the gel, aim for 20-50 μg of total protein per lane, as AP3M1 has been successfully detected in human brain tissue lysate at antibody concentrations of 1-2 μg/mL . Use freshly prepared or properly stored samples, as repeated freeze-thaw cycles can degrade AP3M1. After transfer to membranes, block with 5% non-fat milk or BSA for at least 1 hour to reduce background. When probing for AP3M1, be aware that commercially available antibodies typically detect bands around the expected molecular weight of 47 kDa, though post-translational modifications may result in slight variations in observed molecular weight .

How should antigen retrieval be optimized for AP3M1 immunohistochemistry?

Antigen retrieval optimization is crucial for successful AP3M1 immunohistochemistry, particularly when working with formalin-fixed, paraffin-embedded tissues where epitope masking can significantly impact antibody binding. For AP3M1 detection, heat-induced epitope retrieval (HIER) methods have shown superior results compared to enzymatic retrieval. The primary recommended approach is to use TE buffer at pH 9.0, which has been validated with mouse cerebellum tissue . Alternatively, citrate buffer at pH 6.0 can be used, though this may provide variable results depending on the specific antibody and tissue type . When performing the retrieval, optimal conditions include heating the sections in the selected buffer to 95-100°C for 15-20 minutes, followed by allowing the sections to cool gradually to room temperature for approximately 20 minutes. For tissues with high levels of endogenous peroxidase activity, incorporate a peroxidase blocking step using 3% hydrogen peroxide for 10 minutes after antigen retrieval. Following optimization, application of AP3M1 antibody at a dilution of 1:50-1:500 has shown successful staining, with 5 μg/mL being an effective concentration for detecting AP3M1 in mouse brain tissue samples .

How can researchers distinguish between AP3M1 and AP3M2 cross-reactivity?

Distinguishing between AP3M1 and AP3M2 cross-reactivity presents a significant challenge due to the high sequence homology between these adaptin subunits. Several polyclonal anti-AP3M1 antibodies, including ABIN6991550, explicitly note potential cross-reactivity with AP3M2 . To address this issue, researchers should implement multiple validation strategies: First, include known positive and negative control samples where AP3M1 or AP3M2 expression has been independently verified. Second, perform knockout/knockdown validation experiments using siRNA or CRISPR-Cas9 systems targeting AP3M1 specifically, which will reveal the degree of antibody specificity. Third, consider using monoclonal antibodies, such as R08-8D2, which may offer higher specificity for distinguishing between these closely related proteins . Fourth, conduct parallel Western blots with antibodies targeting different epitopes of AP3M1 to confirm consistent detection patterns. Finally, when cross-reactivity cannot be eliminated, complement antibody-based detection with mRNA expression analysis using AP3M1-specific primers to corroborate protein-level findings. For critical experiments where absolute specificity is required, researchers should consider the functional distinction that AP3M1 shows preferential binding to the YQRL motif from the cytosolic domain of TGN38 compared to AP3M2 .

What factors contribute to variable AP3M1 detection across different tissue types?

Variable AP3M1 detection across tissue types stems from multiple biological and technical factors that researchers must account for when interpreting results. Biologically, AP3M1 expression levels differ naturally among tissues, with notably higher expression in neuronal tissues, reflecting the protein's important role in neurotransmitter vesicle trafficking . Within tissues, cellular heterogeneity further complicates detection, as AP3M1 expression varies among cell types. The subcellular localization of AP3M1 also affects detection sensitivity—it associates with the Golgi region and peripheral structures, requiring appropriate sample preparation to access these compartments . Technically, tissue-specific matrix effects can interfere with antibody binding, while endogenous protein modifications (phosphorylation, ubiquitination) may mask epitopes or alter antibody recognition. Fixation methods significantly impact AP3M1 detection; formalin fixation can cross-link proteins and obscure epitopes, necessitating optimization of antigen retrieval methods (TE buffer pH 9.0 or citrate buffer pH 6.0) for each tissue type . Researchers should validate antibody performance across multiple tissues, optimize protocols for each tissue type (particularly fixation duration and antigen retrieval), and consider using multiple detection methods (e.g., combining IHC with IF or WB) to confirm results in tissues with challenging detection profiles.

What are the potential causes of non-specific bands in AP3M1 Western blots?

Non-specific bands in AP3M1 Western blots can arise from multiple sources that require systematic troubleshooting to resolve. First, antibody cross-reactivity with structurally similar proteins represents a major source of non-specificity, particularly with AP3M2 due to high sequence homology . Second, degradation products of AP3M1 may appear as lower molecular weight bands, especially in samples that have undergone multiple freeze-thaw cycles or inadequate protease inhibition during extraction. Third, post-translational modifications of AP3M1 can create bands at higher molecular weights than the calculated 47 kDa, with possible phosphorylation or ubiquitination events altering migration patterns . Fourth, detection system artifacts, particularly when using highly sensitive chemiluminescent substrates, can generate false-positive signals. To minimize non-specific bands: (1) optimize primary antibody concentration through careful titration experiments (1:1000-1:8000 dilution range for most AP3M1 antibodies) ; (2) increase blocking stringency using 5% BSA instead of milk for phosphorylated protein detection; (3) incorporate additional washing steps with higher detergent concentrations; (4) consider using monoclonal antibodies like R08-8D2 for higher specificity ; and (5) validate bands using positive control lysates from HEK-293 or NIH/3T3 cells where AP3M1 expression has been confirmed .

How can AP3M1 antibodies be utilized in co-immunoprecipitation studies of vesicular trafficking complexes?

Utilizing AP3M1 antibodies in co-immunoprecipitation (co-IP) studies requires careful optimization to preserve native protein-protein interactions within vesicular trafficking complexes. Begin by selecting antibodies validated for immunoprecipitation applications, ensuring they target epitopes that remain accessible in the native AP3M1 conformation and do not interfere with protein-protein interaction domains. When preparing lysates, use gentle non-denaturing buffers (typically containing 1% NP-40 or 0.5% Triton X-100) to maintain the integrity of AP-3 complex associations with its binding partners. Pre-clear lysates with appropriate control IgG and protein A/G beads to reduce non-specific binding. For the immunoprecipitation, cross-link the AP3M1 antibody to protein A/G beads to prevent antibody co-elution with the target protein. After incubation (typically 4°C overnight), perform stringent washing steps while maintaining buffer conditions that preserve complex integrity. When analyzing co-IP results, probe not only for AP3M1 (47 kDa) but also for known AP-3 complex components including AP3D1, AP3B1/B2, and AP3S1/S2 to confirm successful complex isolation . For studying AP3M1's interaction with the HIV-1 Nef protein or TGN38 cytosolic domain, include appropriate controls to verify specific binding to the YQRL motif that distinguishes AP3M1 from AP3M2 function . Advanced applications may incorporate proximity labeling techniques such as BioID or APEX2 fused to AP3M1 to identify novel interaction partners in the vesicular trafficking pathway.

What methodological approaches can differentiate between total and active AP3M1 in cellular systems?

Differentiating between total and active AP3M1 protein pools requires sophisticated methodological approaches that target specific conformational or biochemical states of the protein. To assess total AP3M1 levels, conventional Western blotting with antibodies targeting conserved epitopes (such as those within amino acids 340-390) provides baseline expression data across different cellular conditions . For active AP3M1 analysis, researchers should implement multiple complementary techniques: First, subcellular fractionation combined with immunofluorescence microscopy can identify the Golgi-associated pool of AP3M1, which represents functionally engaged protein in vesicle trafficking . Density gradient fractionation followed by Western blot analysis allows quantification of membrane-associated (active) versus cytosolic (inactive) AP3M1 pools. Second, co-immunoprecipitation studies focused on AP3M1 interactions with cargo proteins containing sorting signals (particularly those with the YQRL motif) can selectively isolate active AP3M1 complexes . Third, phosphorylation-specific antibodies may help distinguish active from inactive AP3M1, though these are not yet widely available commercially. Finally, fluorescence recovery after photobleaching (FRAP) or fluorescence resonance energy transfer (FRET) microscopy using tagged AP3M1 constructs can provide dynamic information about protein activation states in living cells. Researchers should validate these approaches using positive controls where AP3M1 activity is modulated, such as through serum starvation or treatment with trafficking inhibitors.

How can super-resolution microscopy enhance AP3M1 trafficking studies using specific antibodies?

Super-resolution microscopy techniques offer unprecedented opportunities to visualize AP3M1-mediated trafficking events beyond the diffraction limit of conventional microscopy. When implementing these advanced imaging approaches, researchers should optimize immunofluorescence protocols starting with dilutions between 1:50-1:500 and consider several methodological refinements . For Structured Illumination Microscopy (SIM), use highly cross-adsorbed secondary antibodies with minimal spectral overlap to achieve resolution of ~100 nm, enabling visualization of AP3M1 distribution within Golgi subdomains and budding vesicles. For Stimulated Emission Depletion (STED) microscopy, carefully titrate both primary and secondary antibody concentrations to minimize background while maintaining signal intensity, achieving resolutions of ~30-50 nm that can distinguish individual AP3M1-coated vesicles. DNA-PAINT and STORM techniques require specialized buffer systems and photoswitchable fluorophore-conjugated secondary antibodies to reach ~10-20 nm resolution, revealing precise nanoscale organization of AP3M1 within trafficking complexes. For all super-resolution applications, sample preparation becomes critical—use thin sections (≤10 μm), optimize fixation to preserve cellular ultrastructure (4% PFA for 10-15 minutes), and employ enhanced antigen retrieval methods. Multi-color imaging combining AP3M1 with cargo proteins or other adaptor complex subunits can reveal functional interactions at unprecedented resolution. Researchers should validate super-resolution findings with complementary techniques such as electron microscopy immunogold labeling to confirm the specificity of observed structures.

What considerations are important when developing phospho-specific AP3M1 antibodies for signaling research?

Developing phospho-specific AP3M1 antibodies for signaling research requires careful consideration of multiple factors to ensure specificity and utility. First, researchers must identify physiologically relevant phosphorylation sites through phosphoproteomic analysis or literature review, focusing on sites that regulate AP3M1 function or are modulated in response to cellular signaling. Once candidate sites are identified, synthetic phosphopeptides should be designed containing the phosphorylated residue of interest with 7-15 flanking amino acids to provide context for antibody recognition. These phosphopeptides must be carefully purified and their phosphorylation status verified by mass spectrometry before immunization. Host selection is critical—rabbits are commonly used for polyclonal antibody development due to their robust immune response to phosphoepitopes . During antibody production, dual-purification strategies should be employed: first affinity purification against the phosphopeptide, followed by negative selection against the corresponding non-phosphorylated peptide to remove antibodies that recognize the unmodified sequence. Validation of phospho-specific AP3M1 antibodies must include: (1) ELISA testing against phosphorylated and non-phosphorylated peptides; (2) Western blot analysis comparing lysates from control cells, phosphatase-treated samples, and cells treated with kinase activators/inhibitors; (3) immunofluorescence studies with parallel phosphatase treatment controls; and (4) confirmation using mass spectrometry or mutational analysis of the target phosphorylation site. Researchers should also determine the specific kinases and phosphatases regulating these sites to develop comprehensive models of AP3M1 regulation in vesicular trafficking.

How can multiplexed immunoassays be developed using AP3M1 antibodies for systems biology approaches?

Developing multiplexed immunoassays incorporating AP3M1 antibodies enables systems biology investigations of vesicular trafficking networks with unprecedented detail. When designing such assays, researchers should consider several methodological approaches and optimization strategies. For multiplex immunofluorescence imaging, implement tyramide signal amplification or sequential antibody labeling and stripping to detect AP3M1 alongside 5-10 additional proteins within the same sample. Start with validated AP3M1 antibody dilutions (1:50-1:500) and adjust based on signal-to-noise ratios in the multiplexed context . For protein microarray applications, immobilize AP3M1 antibodies alongside antibodies against other trafficking proteins on functionalized glass slides, optimizing antibody concentration and spotting buffer composition to maintain native conformations. When developing multiplex flow cytometry assays for vesicle analysis, conjugate AP3M1 antibodies to distinguishable fluorophores or metal isotopes (for mass cytometry) that permit simultaneous detection with other markers. For Luminex-based suspension assays, couple purified AP3M1 antibodies to spectrally distinct beads, determining optimal coupling conditions through titration experiments. During data analysis, employ computational approaches such as principal component analysis, clustering algorithms, or Bayesian network modeling to identify relationships between AP3M1 and other components of trafficking pathways. Validate multiplexed findings using orthogonal techniques including co-immunoprecipitation or proximity ligation assays. When interpreting results, be aware of potential cross-reactivity with AP3M2 and implement appropriate controls to distinguish signals . These approaches enable quantitative assessment of AP3M1 dynamics in relation to multiple components of vesicular trafficking systems simultaneously.

What controls are essential for validating AP3M1 antibody specificity in research applications?

Comprehensive validation of AP3M1 antibody specificity requires implementing multiple complementary control strategies across research applications. For Western blot specificity controls, researchers should include: (1) positive control lysates from cell lines with confirmed AP3M1 expression (HEK-293, NIH/3T3) ; (2) negative controls using AP3M1 knockout/knockdown samples generated via CRISPR-Cas9 or siRNA; (3) peptide competition assays where pre-incubating the antibody with excess immunogenic peptide should abolish specific signals; and (4) molecular weight verification, confirming detection at the expected 47 kDa . For immunohistochemistry and immunofluorescence applications, essential controls include: (1) omission of primary antibody to assess secondary antibody non-specificity; (2) tissue-specific positive controls (mouse cerebellum has been validated) ; (3) comparison of staining patterns across multiple antibodies targeting different AP3M1 epitopes; and (4) correlation with mRNA expression data from the same tissues. To address potential cross-reactivity with AP3M2, researchers should perform parallel detection in systems with differential expression of these homologs or use knockout models of each protein individually . For co-localization studies, quantitative analysis using Pearson's correlation coefficient or Manders' overlap coefficient with established markers of the Golgi apparatus or lysosomal compartments provides functional validation. Researchers should document all validation steps and clearly report the catalog number (e.g., ABIN6991550, A12628, 12114-1-AP) and lot number of antibodies used, as performance may vary between lots .

How should experiments be designed to study AP3M1's role in lysosomal trafficking pathways?

Designing experiments to elucidate AP3M1's role in lysosomal trafficking requires a multifaceted approach combining cellular, biochemical, and imaging methodologies. Begin with loss-of-function studies using siRNA knockdown or CRISPR-Cas9 knockout of AP3M1, verifying knockdown efficiency via Western blot (1:1000-1:8000 dilution of validated antibodies) . Then assess the functional consequences on lysosomal morphology and distribution using confocal microscopy with AP3M1 antibodies (1:50-1:500 dilution) co-stained with established lysosomal markers (LAMP1, LAMP2) . For quantitative analysis of trafficking dynamics, implement pulse-chase assays with lysosomal hydrolase precursors or fluorescently-labeled endocytic cargo, comparing their trafficking kinetics between control and AP3M1-depleted cells. To examine direct protein interactions, perform co-immunoprecipitation experiments using AP3M1 antibodies to isolate associated proteins, followed by mass spectrometry to identify novel binding partners . For high-resolution visualization of AP3M1's subcellular localization during trafficking events, combine immuno-electron microscopy with AP3M1 antibodies and proximity ligation assays to capture transient interactions with cargo proteins. To establish physiological relevance, design rescue experiments where wild-type and mutant (particularly in cargo-binding domains) AP3M1 constructs are expressed in knockout backgrounds. Throughout these studies, include appropriate controls: positive controls using cells with known AP3M1 expression patterns (HEK-293, NIH/3T3) , negative controls with primary antibody omission, and comparisons with other adaptor protein complexes to establish pathway specificity.

What experimental design considerations are important when using AP3M1 antibodies in brain tissue analysis?

Analysis of AP3M1 in brain tissue presents unique challenges requiring specialized experimental design considerations. When planning immunohistochemistry or immunofluorescence studies, researchers should implement specific methodological approaches: First, optimize fixation conditions—4% paraformaldehyde perfusion fixation for rodent models balances antigen preservation with structural integrity. Second, employ validated antigen retrieval methods specifically for neural tissues; TE buffer at pH 9.0 has shown superior results for AP3M1 detection in mouse cerebellum, with citrate buffer pH 6.0 as an alternative . Third, titrate antibody concentrations starting from established ranges (1:50-1:500 for IHC; 20 μg/mL for IF) but expect to optimize further due to the high lipid content of brain tissue . Fourth, implement comprehensive controls including AP3M1 knockout/knockdown brain sections, regional negative controls (areas with minimal AP3M1 expression), and developmental stage comparisons (as expression levels may vary). For co-localization studies, combine AP3M1 detection with neuron-specific markers (NeuN, MAP2), glia-specific markers (GFAP, Iba1), and synaptic markers (synaptophysin, PSD95) to characterize cell type-specific expression patterns. When analyzing AP3M1 expression in neurological disease models, pair IHC/IF with biochemical fractionation to distinguish changes in expression versus localization. For human post-mortem brain tissues, account for extended post-mortem intervals by adjusting fixation and antigen retrieval times. Finally, consider regional heterogeneity in protocol optimization, as antibody penetration and background can vary significantly between cortical, hippocampal, and cerebellar regions.

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.