AP3S1 Human

Assembly Protein Complex 3 Subunit-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Key Functions:

  1. Vesicle Budding and Trafficking: Facilitates vesicle formation from the Golgi membrane, directing cargoes to lysosomes and lysosome-related organelles (e.g., melanosomes, platelet dense granules) .

  2. Collaboration with BLOC-1 Complex: Synergizes with BLOC-1 to deliver cargoes to neurites and nerve terminals, critical for neuronal function and immune cell degranulation (e.g., NK cells, CD8+ T-cells) .

Tissue Expression Profile

AP3S1 exhibits broad tissue expression, with notable activity in:

  • Brain (neuronal structures)

  • Liver (hepatocytes)

  • Skeletal Muscle

  • Immune Cells (T-cells, NK cells) .

Data from the Human Protein Atlas indicates robust mRNA and protein expression in:

TissueExpression LevelSubcellular Localization
BrainHighGolgi apparatus, cytoplasm
LiverModerateGolgi, lysosomes
Lymph NodeHighImmune cell vesicles

Cancer Biology and Prognostic Significance

Recent pan-cancer analyses reveal AP3S1 as a potential oncogene with immunosuppressive roles:

Molecular Pathways:

AP3S1 is enriched in:

  • Cell Cycle Regulation

  • ER-to-Golgi Transport

  • Immune Response Pathways (e.g., Toll-like receptor signaling, neutrophil degranulation) .

Functional Partners and Interactions

AP3S1 interacts with adaptor complexes and regulatory proteins:

PartnerFunctionInteraction Score
AP3B1AP-3 complex assembly; lysosomal trafficking0.999
AP3D1Vesicle formation; neuronal cargo delivery0.999
BLOC-1 ComplexSynergistic targeting of cargoes to lysosomes and neuritesN/A
AP-2 ComplexEndocytic vesicle formation (clathrin-dependent)0.968

Genetic and Evolutionary Context

  • Gene ID: 1176 .

  • Aliases: CLAPS3, Sigma3A, Adapter-related protein complex 3 sigma-1 .

  • Orthologs: Conserved across mammals (e.g., mouse Ap3s1) .

Product Specs

Introduction
AP-3 complex subunit sigma-1 (AP3S1), a component of the non-clathrin-associated adapter-related complex AP-3, plays a crucial role in intracellular trafficking. This protein is found in the Golgi region and peripheral structures, where it participates in vesicle budding from the Golgi membrane and protein trafficking to lysosomes.
Description
Recombinant human AP3S1, expressed in E. coli, is a single polypeptide chain encompassing amino acids 1-193 of the protein sequence. This 23.8 kDa protein is fused to a 20 amino acid His-tag at the N-terminus and purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The AP3S1 solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 10% glycerol, and 1 mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for extended storage periods. Repeated freezing and thawing should be avoided.
Purity
The purity of the AP3S1 protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
CLAPS3, Sigma3A ,AP-3 complex subunit sigma-1, AP-3 complex subunit sigma-3A, Adapter-related protein complex 3 sigma-1 subunit, Clathrin-associated/assembly/adapter protein, small 3, Sigma-3A-adaptin, Sigma-adaptin 3a.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MIKAILIFNN HGKPRLSKFY QPYSEDTQQQ IIRETFHLVS RDENVCNFL EGGLLIGGSD NKLIYRHYAT LYFVFCVDSS ESELGILDLI QVFVETLDKC ENVCELDLI FHVDKVHNIL AEMVMGGMVL ETNMNEIVTQ IDAQNKLEKS EAGLAGAPAR AVSAVKNMNL PEIPRNINIG DISIKVPNLP SFK.

Q&A

What is AP3S1 and what is its primary cellular function?

AP3S1 (Adaptor-related protein complex 3 subunit sigma-1) encodes a subunit of the AP-3 complex, which is an adaptor-related complex not associated with clathrin. This complex is primarily localized to the Golgi region and more peripheral cellular structures. Functionally, AP3S1 facilitates the budding of vesicles from the Golgi membrane and is directly involved in trafficking to lysosomes . In concert with the BLOC-1 complex, AP3S1 is required to target cargo into vesicles assembled at cell bodies for delivery into neurites and nerve terminals .

The protein has a molecular weight of approximately 21 kDa and consists of 193 amino acids. It contains specific domains that enable its interaction with other components of the vesicular trafficking machinery. AP3S1 is evolutionarily conserved, highlighting its fundamental importance in cellular processes across species. Understanding these basic properties is essential for researchers designing experiments to investigate its roles in normal and pathological states.

What experimental methods are most reliable for detecting AP3S1 expression?

Several complementary approaches are recommended for robust AP3S1 detection:

RNA-level detection:

  • RT-qPCR using validated primers specific to AP3S1

  • RNA-sequencing data analysis, as employed in TCGA and GTEx databases

  • In situ hybridization for tissue-specific localization

Protein-level detection:

  • Western blotting using validated antibodies (e.g., ab113099) at concentrations between 1-2 μg/mL

  • Immunohistochemistry for tissue samples

  • Immunofluorescence for subcellular localization

Database resources for expression analysis:

  • TCGA and GTEx for transcriptomic data across cancer and normal tissues

  • UALCAN and Human Protein Atlas (HPA) for protein expression data

  • cBioPortal for methylation and copy number alteration data

When designing AP3S1 detection experiments, researchers should include appropriate positive controls such as mouse kidney tissue lysate, where the protein shows consistent expression . Validation using multiple detection methods is strongly recommended, especially when studying differential expression across tissue types or disease states.

How does AP3S1 expression vary across normal human tissues?

AP3S1 expression shows tissue-specific patterns in normal human tissues, which is important for establishing baselines when studying its dysregulation in disease states. Comprehensive analysis using data from the GTEx database reveals differential expression patterns across tissue types. Expression is notably maintained in tissues with high vesicular trafficking requirements, consistent with AP3S1's role in the vesicular transport system .

When studying AP3S1 expression in specific tissue contexts, researchers should always include matched normal tissues as controls. The Human Protein Atlas provides valuable immunohistochemistry data showing protein-level expression across normal tissues that should be consulted when designing tissue-specific studies . Understanding this baseline variation is critical for correctly interpreting expression changes in pathological conditions.

What evidence supports AP3S1 as a potential pan-cancer oncogene?

Multiple lines of evidence establish AP3S1 as a potential pan-cancer oncogene:

Overexpression patterns:

  • AP3S1 is significantly overexpressed in 16 of 33 cancer types analyzed through TCGA and GTEx data

  • Particularly high expression is observed in Adrenocortical carcinoma (ACC), Breast invasive carcinoma (BRCA), Cholangiocarcinoma (CHOL), and multiple other cancer types

  • Protein-level overexpression confirmed through UALCAN database and Human Protein Atlas in HNSC, LIHC, LUAD, and KIRC

Prognostic significance:

Functional implications:

  • GSEA and GSVA analyses demonstrate AP3S1's involvement in multiple malignant pathways

  • AP3S1 was identified as a tumor driver gene in a study analyzing 1,145 esophageal squamous cell carcinoma samples

These consistent findings across multiple cancer types and analytical approaches strongly support AP3S1's role as a pan-cancer oncogene, making it a valuable target for cancer research.

How does AP3S1 influence the tumor immune microenvironment?

AP3S1 plays a significant role in shaping the tumor immune microenvironment (TIME) through multiple mechanisms:

Immune cell infiltration:

  • AP3S1 expression positively correlates with immunosuppressive cells including tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and regulatory T cells (Tregs)

  • Negative correlation observed with cytotoxic immune cells including natural killer (NK) cells and CD8+ T cells

  • Influences stromal scores, immune scores, and ESTIMATE scores in most tumors analyzed

Immune-related pathways:

  • Gene set enrichment analysis (GSEA) reveals AP3S1's involvement in both adaptive and innate immune systems

  • Associated with neutrophil degranulation, cytokine signaling, antigen processing, and Toll-like receptor cascades

  • GSVA analysis shows positive association with IL6 JAK STAT3 signaling, IL2 STAT5 signaling, TGF-beta signaling, and interferon responses

Immune checkpoint regulation:

  • Positive correlation with key immunosuppressive genes including PD-1, PD-L1, CTLA4, LAG3, and TIGIT across most cancer types

  • Associated with immune checkpoint pathways and antigen processing mechanisms

Chemokine regulation:

  • Correlates with expression of chemokines involved in TAM recruitment (CCL2, CCR2, CXCR4, CCR5)

  • Associated with immunosuppressive chemokines (CCL3, CCL4, CCL5, CCL22) that mediate T cell suppression

These findings suggest AP3S1 contributes to an immunosuppressive microenvironment in tumors, potentially explaining its association with poor prognosis and highlighting its relevance for immunotherapy research.

What methodologies are recommended for studying AP3S1's role in cancer progression?

Researchers investigating AP3S1's role in cancer progression should employ a multi-faceted approach:

In silico analysis:

  • Utilize TCGA and GTEx data for expression analysis across tumor types

  • Perform survival analyses (Kaplan-Meier and Cox regression) to establish prognostic significance

  • Conduct gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) to identify associated pathways

  • Analyze tumor mutational burden (TMB) and microsatellite instability (MSI) correlations

In vitro experimental approaches:

  • Gene knockdown/overexpression studies in relevant cancer cell lines

  • Migration and invasion assays to assess metastatic potential

  • Co-culture systems with immune cells to evaluate effects on immune function

  • Western blotting with validated antibodies (e.g., ab113099) for protein detection

In vivo models:

  • Xenograft models with AP3S1 knockdown/overexpression

  • Syngeneic mouse models to study tumor-immune interactions

  • Patient-derived xenografts to evaluate clinical relevance

Clinical sample analysis:

  • Tissue microarray analysis of patient samples

  • Correlation of AP3S1 expression with clinical parameters and outcomes

  • Single-cell analysis to assess expression in specific cell populations within the tumor microenvironment

When designing these studies, researchers should include multiple cancer types to validate AP3S1's pan-cancer role and incorporate appropriate controls to account for tissue-specific expression patterns.

How can researchers effectively investigate the relationship between AP3S1 and immune checkpoint molecules?

To thoroughly investigate the relationship between AP3S1 and immune checkpoint molecules, researchers should implement the following methodological approaches:

Correlation analyses:

  • Perform comprehensive correlation analysis between AP3S1 expression and immune checkpoint genes (PD-1, PD-L1, CTLA4, LAG3, TIGIT) across multiple cancer datasets

  • Use both Pearson and Spearman correlation tests to account for different distribution patterns

  • Visualize correlations using heatmaps to identify cancer-specific patterns

Mechanistic investigations:

  • Conduct AP3S1 knockdown/overexpression experiments in cancer cell lines to assess direct effects on immune checkpoint expression

  • Employ ChIP-seq to identify potential direct regulatory relationships

  • Use luciferase reporter assays to examine transcriptional regulation

Functional immune assays:

  • Set up co-culture systems with T cells to assess functional consequences of AP3S1 modulation

  • Measure T cell activation markers, proliferation, and cytokine production

  • Employ immune killing assays to evaluate cytotoxic T cell function

In vivo validation:

  • Develop mouse models with modulated AP3S1 expression

  • Assess response to immune checkpoint inhibitor therapy

  • Perform immune cell phenotyping of tumor-infiltrating lymphocytes

Clinical correlations:

  • Analyze patient samples for co-expression patterns

  • Stratify patients based on both AP3S1 and immune checkpoint expression

  • Correlate with response to immunotherapy in treatment cohorts

This comprehensive approach will help elucidate whether AP3S1 directly regulates immune checkpoint molecules or whether they are co-regulated by shared upstream mechanisms, providing valuable insights for immunotherapy development.

What statistical approaches should be employed when analyzing AP3S1 expression in relation to patient outcomes?

Robust statistical analysis is crucial when evaluating AP3S1's clinical significance:

Survival analysis techniques:

Expression analysis:

  • Compare means using t-tests (paired for tumor/normal from same patient)

  • ANOVA for comparing multiple groups (e.g., tumor stages)

  • Non-parametric alternatives (Mann-Whitney, Kruskal-Wallis) for non-normally distributed data

  • Present data as mean ± standard deviation (SD) as recommended in published AP3S1 studies

Correlation methods:

  • Use Spearman's correlation for ordinal data or non-linear relationships

  • Apply Pearson's correlation for continuous, normally distributed variables

  • Implement multiple testing correction (e.g., Benjamini-Hochberg) to control false discovery rate

Cut-point determination:

  • Employ ROC curve analysis to determine optimal expression thresholds

  • Use median split if no clear biological threshold exists

  • Consider quartile analysis to examine dose-dependent effects

Integrated analysis:

  • Develop nomograms incorporating AP3S1 with clinical parameters

  • Perform decision curve analysis to assess clinical utility

  • Calculate C-index to evaluate predictive accuracy

Statistical significance should be consistently defined as p < 0.05 across analyses, in accordance with standard practice in AP3S1 research . Power calculations should be performed to ensure adequate sample sizes for detecting clinically meaningful differences.

How can researchers reconcile conflicting findings regarding AP3S1 expression across different cancer types?

Addressing contradictory findings requires systematic investigation through several approaches:

Methodological standardization:

  • Compare detection methods (RNA-seq, microarray, qPCR, IHC, Western blot) used across studies

  • Standardize analytical pipelines for expression quantification

  • Verify antibody specificity and RNA probe design across studies

  • Ensure comparable normalization methods when analyzing expression data

Biological context considerations:

  • Analyze tissue-specific regulatory mechanisms that may influence AP3S1 function

  • Examine cancer subtype heterogeneity within broadly defined cancer types

  • Consider tumor microenvironment differences that may modulate AP3S1's role

  • Evaluate genetic background variations that might affect AP3S1 function

Integration of multiple data types:

  • Compare gene expression with protein levels to identify post-transcriptional effects

  • Analyze epigenetic modifications, including DNA methylation data from cBioPortal

  • Assess copy number alterations that might explain expression differences

  • Consider alternate splicing events that could produce functionally distinct isoforms

Meta-analysis approach:

  • Perform systematic review and meta-analysis of published studies

  • Weight findings based on sample size, methodological rigor, and validation approaches

  • Test for publication bias that might skew reported associations

  • Use random-effects models to account for between-study heterogeneity

When analyzing AP3S1 expression, researchers should note that while AP3S1 is overexpressed in most cancers, including BRCA, CHOL, HNSC, KIRC, KIRP, LIHC, and LUAD, it shows lower expression in PRAD and KICH compared to adjacent normal tissues . These tissue-specific differences highlight the importance of context-dependent analysis rather than assuming universal patterns.

What experimental controls are essential when studying AP3S1 in cancer models?

Rigorous experimental design for AP3S1 research requires comprehensive controls:

Expression analysis controls:

  • Matched normal-tumor tissue pairs from the same patient when possible

  • Non-malignant cell lines of the same tissue origin

  • Reference genes with stable expression across experimental conditions

  • Positive control tissues known to express AP3S1 (e.g., mouse kidney tissue for Western blotting)

Functional studies controls:

  • Empty vector controls for overexpression studies

  • Scrambled/non-targeting siRNA/shRNA for knockdown experiments

  • Isotype controls for antibody-based experiments

  • Wild-type unmodified cells alongside genetically modified lines

Specificity controls:

  • Secondary antibody-only controls for immunostaining

  • Multiple siRNA/shRNA constructs targeting different regions of AP3S1

  • Rescue experiments with siRNA-resistant constructs

  • Validation of antibody specificity through knockout/knockdown samples

Biological replicates:

  • Minimum of three biological replicates for all experiments

  • Use of multiple cell lines representing the same cancer type

  • Patient-derived samples from diverse demographic backgrounds

  • Independent validation in separate cohorts

Technical considerations:

  • Include loading controls (e.g., GAPDH, β-actin) for Western blotting

  • Concentration gradient testing for antibodies (1-2 μg/mL range recommended)

  • Multiple primer pairs for qPCR validation

  • Both positive and negative controls for all assays

Implementing these controls ensures that observed effects are specifically attributable to AP3S1 rather than experimental artifacts or non-specific effects, enhancing the reproducibility and reliability of research findings.

What cell line models are most appropriate for AP3S1 functional studies?

Selection of appropriate cell line models is critical for meaningful AP3S1 research:

Cancer types with strong AP3S1 associations:

  • Based on expression data, prioritize cell lines from cancers with established AP3S1 overexpression :

    • Breast cancer (e.g., MCF-7, MDA-MB-231)

    • Liver cancer (e.g., HepG2, Huh7)

    • Lung adenocarcinoma (e.g., A549, H1299)

    • Head and neck squamous cell carcinoma (e.g., SCC-9, FaDu)

    • Kidney renal clear cell carcinoma (e.g., 786-O, A498)

Expression verification:

  • Verify baseline AP3S1 expression levels before selecting specific cell lines

  • Include both high and low AP3S1-expressing lines for comparative studies

  • Consider cell lines with naturally occurring AP3S1 mutations or amplifications

Functional considerations:

  • For vesicular trafficking studies, use cell lines with well-characterized Golgi and lysosomal markers

  • For immune interaction studies, select lines that respond to immune modulators

  • For metastasis research, use cell lines with established metastatic potential

Technical attributes:

  • Prioritize cell lines with good transfection efficiency for genetic manipulation

  • Consider growth characteristics that facilitate your experimental design

  • Use authenticated cell lines with stable genotypes across passages

Complementary models:

  • Pair immortalized cell lines with primary cell cultures

  • Consider 3D organoid models for more physiologically relevant contexts

  • For immune interaction studies, include co-culture systems with immune cells

Researchers should verify AP3S1 expression in their chosen cell lines using validated antibodies such as ab113099 at appropriate concentrations (1-2 μg/mL) before proceeding with functional studies, as expression levels may vary even within cell lines of the same cancer type.

How should researchers design experiments to investigate AP3S1's dual role in vesicular trafficking and cancer immunity?

Investigating AP3S1's dual functionality requires carefully designed experimental approaches:

Vesicular trafficking investigations:

  • Live cell imaging with fluorescently tagged AP3S1 to track vesicular movement

  • Co-localization studies with markers for Golgi (GM130), lysosomes (LAMP1), and endosomes (EEA1)

  • Electron microscopy to visualize ultrastructural vesicular changes upon AP3S1 modulation

  • Cargo trafficking assays measuring transport kinetics of known lysosomal proteins

Immune function investigations:

  • Analysis of cytokine secretion profiles following AP3S1 manipulation

  • Flow cytometry to assess immune checkpoint protein surface expression

  • Co-culture systems with various immune cell populations (T cells, macrophages, NK cells)

  • Chemotaxis assays to evaluate immune cell recruitment capacity

Integrative approaches:

  • Investigate how AP3S1-mediated vesicular trafficking affects immune protein localization

  • Study exosome composition and production following AP3S1 modulation

  • Examine antigen presentation efficiency in AP3S1-manipulated cancer cells

  • Analyze trafficking of immune receptors and ligands to cell surface

Molecular mechanistic studies:

  • Identify AP3S1 protein interaction partners using co-immunoprecipitation or proximity labeling

  • Map phosphorylation sites that might regulate AP3S1's different functions

  • Perform domain mapping to identify regions responsible for specific functions

  • Use CRISPR-Cas9 to generate domain-specific mutations rather than complete knockouts

Translational investigations:

  • Correlate vesicular trafficking markers with immune infiltration in patient samples

  • Develop dual-targeting approaches addressing both functions simultaneously

  • Investigate how existing cancer therapies affect AP3S1's dual functionality

This comprehensive approach will help elucidate how AP3S1's canonical role in vesicular trafficking connects to its newly discovered functions in cancer immunity, potentially revealing novel therapeutic opportunities.

What approaches show promise for targeting AP3S1 in cancer therapy research?

Several strategies warrant investigation for therapeutic targeting of AP3S1:

Direct targeting approaches:

  • Small molecule inhibitors targeting AP3S1 protein-protein interactions

  • Antisense oligonucleotides or siRNA for transcript reduction

  • PROTAC-based degradation of AP3S1 protein

  • Peptide inhibitors disrupting AP3S1 complex assembly

Pathway-based strategies:

  • Targeting upstream regulators of AP3S1 expression

  • Inhibiting downstream effectors in AP3S1-associated pathways

  • Combination approaches targeting multiple components of AP3S1-related signaling

Immune-focused interventions:

  • Combining AP3S1 inhibition with immune checkpoint blockade

  • Developing strategies to reverse AP3S1-mediated immune suppression

  • Targeting AP3S1-dependent recruitment of immunosuppressive cells

Vesicular trafficking modulation:

  • Disrupting AP3S1's role in cancer-specific cargo trafficking

  • Targeting cancer-specific AP3S1 complex components

  • Developing lysosome-targeting therapies in AP3S1-overexpressing cancers

Cancer type-specific approaches:

  • Prioritizing intervention in cancers where AP3S1 shows strongest prognostic association (BRCA, GBM, HNSC, KIRP, LIHC, LUAD, MESO, PAAD, and UVM)

  • Developing context-dependent targeting strategies based on cancer-specific AP3S1 functions

Research should focus on therapeutic window considerations, as AP3S1 has important physiological functions in normal tissues. Conditional or cancer-specific targeting approaches may help minimize potential toxicities while maximizing anti-tumor effects.

What are the key technical challenges in developing AP3S1 as a prognostic biomarker?

Developing AP3S1 as a clinically useful biomarker faces several technical hurdles:

Standardization challenges:

  • Establishing standardized detection methods across laboratories

  • Defining clinically relevant expression thresholds for different cancer types

  • Ensuring reproducibility of results across different technical platforms

  • Validating antibody specificity for immunohistochemistry applications

Sample considerations:

  • Addressing tumor heterogeneity through multi-region sampling

  • Standardizing sample collection, processing, and storage protocols

  • Developing methods for detection in liquid biopsies

  • Accounting for normal tissue contamination in tumor samples

Analytical complexities:

  • Integrating AP3S1 with existing prognostic markers into unified models

  • Determining if mRNA or protein assessment provides superior prognostic value

  • Accounting for potential post-translational modifications

  • Developing cancer-specific vs. pan-cancer analytical approaches

Clinical validation requirements:

  • Conducting large, prospective validation studies across diverse patient populations

  • Demonstrating added value beyond existing prognostic factors

  • Establishing predictive value for specific treatment responses

  • Determining optimal timing for biomarker assessment (diagnosis, recurrence, etc.)

Implementation barriers:

  • Developing cost-effective testing methods suitable for routine clinical use

  • Creating standardized reporting systems for AP3S1 status

  • Training pathologists in consistent scoring methods

  • Establishing quality control systems for biomarker testing

Addressing these challenges requires collaborative efforts between academic researchers, industry partners, and regulatory agencies to move AP3S1 from a research biomarker to a clinically validated prognostic tool.

What future research directions are most promising for understanding AP3S1's role in cancer biology?

Several promising research avenues could significantly advance understanding of AP3S1 in cancer:

Single-cell analysis:

  • Investigate cell type-specific expression patterns within the tumor microenvironment

  • Examine AP3S1's role in rare cell populations like cancer stem cells

  • Track evolutionary trajectories of AP3S1-expressing clones during cancer progression

  • Map spatial relationships between AP3S1-expressing cells and immune populations

Systems biology approaches:

  • Develop comprehensive protein interaction networks centered on AP3S1

  • Apply multi-omics integration to understand AP3S1 regulation

  • Use computational modeling to predict functional consequences of AP3S1 alterations

  • Identify synthetic lethal interactions for AP3S1-overexpressing cancers

Mechanistic investigations:

  • Elucidate how vesicular trafficking functions connect to immune regulation

  • Identify cancer-specific cargoes trafficked by AP3S1-containing complexes

  • Characterize structural biology of AP3S1 interactions for drug development

  • Determine if AP3S1 has non-canonical functions beyond vesicular trafficking

Translational research:

  • Develop AP3S1-targeted therapeutic approaches

  • Identify patient subgroups most likely to benefit from AP3S1-directed therapies

  • Investigate AP3S1 as a biomarker for immunotherapy response

  • Explore combinations of AP3S1 inhibition with standard cancer therapies

Evolutionary considerations:

  • Study AP3S1's role in therapy resistance development

  • Examine how AP3S1 influences metastatic progression

  • Investigate AP3S1 in the context of tumor dormancy and recurrence

  • Analyze AP3S1 expression changes during cancer evolution under treatment pressure

These research directions would address significant knowledge gaps while also advancing translational applications of AP3S1 biology in cancer diagnosis, prognosis, and treatment. Particular priority should be given to understanding the mechanistic basis of AP3S1's apparent dual role in vesicular trafficking and immune regulation, as this connection remains poorly understood despite its potential therapeutic significance.

Product Science Overview

Introduction

Assembly Protein Complex 3 Subunit-1 (APC3) is a crucial component of the anaphase-promoting complex/cyclosome (APC/C), a multi-subunit E3 ubiquitin ligase that regulates progression through the cell cycle by targeting specific proteins for degradation. The human recombinant form of APC3 is produced using recombinant DNA technology, which allows for the expression of the protein in a host organism, typically bacteria or yeast, to facilitate its study and use in various applications.

Structure and Function

APC3, also known as CDC27, is one of the core subunits of the APC/C complex. The APC/C complex is composed of at least 14 different subunits, and APC3 plays a pivotal role in the assembly and function of this complex. The primary function of APC/C is to ubiquitinate target proteins, marking them for degradation by the proteasome. This process is essential for the regulation of the cell cycle, particularly during the transition from metaphase to anaphase and the exit from mitosis.

APC3 contains multiple tetratricopeptide repeat (TPR) motifs, which are involved in protein-protein interactions. These motifs facilitate the binding of APC3 to other subunits of the APC/C complex as well as to its substrates and regulatory proteins. The proper assembly and function of the APC/C complex are critical for maintaining genomic stability and preventing uncontrolled cell proliferation.

Recombinant Production

The production of human recombinant APC3 involves the insertion of the gene encoding APC3 into an expression vector, which is then introduced into a host organism. Commonly used host organisms include Escherichia coli (E. coli) and Saccharomyces cerevisiae (yeast). The host cells are cultured under conditions that promote the expression of the recombinant protein, which is subsequently purified using various chromatographic techniques.

Recombinant APC3 is used in research to study the structure and function of the APC/C complex, as well as its role in cell cycle regulation and cancer. The availability of recombinant APC3 allows for detailed biochemical and biophysical analyses, which can provide insights into the mechanisms of APC/C-mediated ubiquitination and its regulation.

Applications in Research and Medicine

The study of APC3 and the APC/C complex has significant implications for understanding the molecular mechanisms underlying cell cycle regulation and its dysregulation in diseases such as cancer. By elucidating the structure and function of APC3, researchers can identify potential therapeutic targets for the development of anti-cancer drugs.

In addition to its role in basic research, recombinant APC3 can be used in high-throughput screening assays to identify small molecules that modulate the activity of the APC/C complex. These molecules could serve as lead compounds for the development of novel cancer therapies.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.