ASB8 Human

Ankyrin Repeat And SOCS Box Containing 8 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Function and Mechanism

ASB8 functions as a substrate-recognition component of SCF-like ECS (Elongin-Cullin-SOCS-box protein) E3 ubiquitin ligase complexes, mediating ubiquitination and proteasomal degradation of target proteins . Key roles include:

  • Targeted degradation: Facilitates K48-linked ubiquitination of TBK1/IKKi, reducing IRF3 phosphorylation and immune signaling .

  • Cancer association: A truncated mutant lacking the SOCS box suppresses lung adenocarcinoma cell growth, suggesting a role in oncogenesis .

Functional partners (STRING database) :

ProteinInteraction ScoreRole
Elongin-C (ELOC)0.878Transcription elongation factor
Elongin-B (ELOB)0.838Regulates RNA polymerase II activity
Cullin-5 (CUL5)0.807Scaffold for ECS E3 ligase complexes
NEDD80.660Modifies cullins for ligase activation

Tissue Expression and Subcellular Localization

ASB8 exhibits tissue-specific expression:

  • Highest expression: Skeletal muscle .

  • Moderate expression: Heart, brain, placenta, liver, kidney, pancreas .

  • Cancer cells: Detected in lung carcinoma cell lines (SPC-A1, A549, NCI-H446) .

Subcellularly, ASB8 localizes to the cytoplasm, as shown in hepatocellular carcinoma cells (BEL-7402) .

Research Applications and Experimental Relevance

  • Interactions: Binds Elongin B/C complexes in vitro, enabling substrate targeting .

  • Cancer research: Truncated mutants lacking the SOCS box inhibit lung adenocarcinoma cell proliferation, linking ASB8 to tumor suppression .

Genomic and Evolutionary Context

  • Genomic location: Chromosome 12q13 .

  • Orthologs: High sequence identity (96%) with mouse ASB8 .

  • Phylogenetic conservation: Predicted roles in signal transduction and ubiquitination across species (e.g., zebrafish) .

Product Specs

Introduction
ASB8, part of the SCF-like ECS (Elongin-Cullin-SOCS-box protein) E3 ubiquitin-protein ligase complex, acts as a substrate-recognition component. It enables the ubiquitination and subsequent proteasomal degradation of specific target proteins.
Description
Produced in E.coli, Recombinant Human ASB8 is a single, non-glycosylated polypeptide chain. It consists of 311 amino acids (1-288) and has a molecular weight of 34.0kDa. The protein is fused to a 23 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear solution, sterile and filtered.
Formulation
The ASB8 solution (0.25mg/ml) is prepared with 20mM Tris-HCl buffer (pH 8.0), 0.15M NaCl, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), store the vial at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 90.0% via SDS-PAGE analysis.
Synonyms
Ankyrin Repeat And SOCS Box Containing 8, ASB-8.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMSSSMWY IMQSIQSKYS LSERLIRTIA AIRSFPHDNV EDLIRGGADV NCTHGTLKPL HCACMVSDAD CVELLLEKGA EVNALDGYNR TALHYAAEKD EACVEVLLEY GANPNALDGN RDTPLHWAAF KNNAECVRAL LESGASVNAL DYNNDTPLSW AAMKGNLESV SILLDYGAEV RVINLIGQTP ISRLVALLVR GLGTEKEDSC FELLHRAVGH FELRKNGTMP REVARDPQLC EKLTVLCSAP GTLKTLARYA VRRSLGLQYL PDAVKGLPLP ASLKEYLLLL E

Q&A

What is the basic structure and function of human ASB8?

Human ASB8 is a protein-coding gene that belongs to the ankyrin repeat and SOCS box-containing (ASB) protein family. Structurally, ASB8 contains multiple domains including ankyrin repeats and a SOCS box domain. The protein features a specific domain architecture with ankyrin repeat-containing domains and SOCS box-like domains that form part of the protein's functional regions .

The protein is primarily involved in:

  • Intracellular signal transduction pathways

  • Protein ubiquitination processes

  • Regulation of protein turnover through the ubiquitin-proteasome system

Its cellular localization is predominantly cytoplasmic, where it participates in protein-protein interactions through its ankyrin repeat domains while the SOCS box mediates interactions with elongin B/C complexes to form E3 ubiquitin ligase complexes.

How should researchers detect and quantify ASB8 expression in human tissues?

When studying ASB8 expression in human tissues, researchers should employ multiple complementary techniques to ensure robust detection and quantification:

Recommended methodological approach:

  • RNA-level detection:

    • RT-qPCR using validated primers specific to ASB8 transcript variants

    • RNA-Seq for comprehensive transcriptomic profiling

    • Northern blotting for validation of transcript size

  • Protein-level detection:

    • Western blotting with validated antibodies (confirm specificity with knockout controls)

    • Immunohistochemistry/Immunofluorescence for tissue localization

    • ELISA for quantitative measurement in tissue lysates

  • Cross-validation:

    • Always compare protein and mRNA levels as post-transcriptional regulation may affect correlation

    • Use multiple antibodies targeting different epitopes to confirm specificity

    • Include appropriate positive and negative controls in each experiment

Methodological consideration: When selecting antibodies, prioritize those validated for the specific application (WB, IHC, IP) and confirm specificity through knockout/knockdown validation experiments to avoid cross-reactivity with other ASB family members.

What are the established model systems for studying ASB8 function?

Researchers investigating ASB8 function should consider these established model systems, each with specific advantages:

Model SystemApplicationsAdvantagesLimitations
Human cell lines (HEK293, HeLa)Protein interaction studies, localization, functional assaysEasy to manipulate, well-characterizedMay not reflect tissue-specific regulation
Primary human cellsPhysiological studies, disease modelingMore physiologically relevantLimited availability, donor variability
CRISPR/Cas9 knockout modelsLoss-of-function studiesPrecise gene targetingPotential for off-target effects
Inducible expression systemsTemporal control of expressionAllows study of acute vs. chronic effectsLeaky expression can be problematic
Zebrafish modelsDevelopmental studies, in vivo functionOrthologous asb8 gene present, transparent embryosSome functional divergence from human

When selecting a model system, researchers should consider: (1) the specific research question, (2) available resources and expertise, (3) required physiological relevance, and (4) time constraints. For molecular interaction studies, cell lines are appropriate, while organismal functions may require animal models with the orthologous gene.

What experimental designs are optimal for investigating ASB8 substrate specificity?

Investigating ASB8 substrate specificity requires strategic experimental design incorporating multiple approaches:

Recommended methodological workflow:

  • Protein interaction identification:

    • Proximity labeling (BioID, APEX) to identify proteins in proximity to ASB8

    • Co-immunoprecipitation followed by mass spectrometry (MS)

    • Yeast two-hybrid screening for initial candidate identification

  • Validation of direct interactions:

    • In vitro binding assays with recombinant proteins

    • Surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) for binding kinetics

    • Domain mapping through truncation/mutation analysis

  • Ubiquitination target confirmation:

    • In vitro ubiquitination assays with purified components

    • Ubiquitin remnant profiling by MS in cells with ASB8 overexpression/knockout

    • Protein stability assays in the presence/absence of ASB8

  • Specificity determination:

    • Structural analysis of ASB8-substrate complexes

    • Competition assays with related ASB family members

    • Mutational analysis of substrate recognition motifs

The experimental approach should progress from identification to validation to functional confirmation, with appropriate controls at each stage. Researchers should be particularly mindful of the potential for false positives in interaction screens and include appropriate controls such as substrate-binding deficient mutants.

How should researchers address contradictory data regarding ASB8 signaling pathway involvement?

Contradictory findings regarding ASB8's role in signaling pathways require systematic investigation:

Methodological resolution approach:

  • Critical assessment of experimental conditions:

    • Evaluate cell type-specific differences (primary cells vs. immortalized lines)

    • Compare acute vs. chronic manipulations of ASB8 levels

    • Assess expression levels (physiological vs. overexpression)

    • Document passage number and culture conditions

  • Validation with multiple methodological approaches:

    • Use both loss-of-function (siRNA, CRISPR) and gain-of-function (overexpression) approaches

    • Apply both genetic and pharmacological interventions when possible

    • Utilize multiple independent reagents (different siRNAs, antibodies)

  • Pathway-specific validation:

    • Monitor multiple nodes within the pathway, not just end-points

    • Employ pathway-specific reporter assays

    • Perform epistasis experiments to position ASB8 within the pathway

  • Context-dependent regulation assessment:

    • Test pathway activity under different cell states (proliferation, differentiation)

    • Examine stimulus-dependent effects (growth factors, stress conditions)

    • Investigate post-translational modifications of ASB8 under different conditions

When publishing, researchers should explicitly address contradictions in the literature, detailing methodological differences that might explain discrepancies and providing comprehensive documentation of experimental conditions to allow proper interpretation.

What are the appropriate single-subject research designs for studying ASB8 in rare disease contexts?

When investigating ASB8 in rare disease contexts where large sample sizes are unavailable, single-subject research designs provide robust alternatives:

Methodological implementation:

  • A-B-A-B design application:

    • Baseline measurement of ASB8-related parameters (A1)

    • Introduction of therapeutic intervention targeting ASB8 pathway (B1)

    • Withdrawal of intervention to confirm return to baseline (A2)

    • Reintroduction of intervention to demonstrate reproducibility (B2)

  • Multiple baseline design for rare disease cohorts:

    • Staggered introduction of intervention across a small number of patients

    • Each patient serves as their own control

    • Different baseline lengths strengthen causal inference

    • Analysis focuses on intra-individual changes with intervention timing as the key variable

  • Single-cell analysis approach:

    • Perform deep molecular profiling of limited patient samples

    • Compare ASB8 pathway activity in affected vs. unaffected cells from the same patient

    • Use longitudinal sampling where possible to track disease progression

    • Integrate multi-omics data to compensate for limited sample numbers

  • Patient-derived model systems:

    • Generate iPSCs from patient samples for disease modeling

    • Create isogenic controls using gene editing

    • Perform functional rescue experiments with wild-type ASB8

    • Use organoid models to recapitulate tissue-specific phenotypes

What technical considerations are crucial when performing domain-function analysis of ASB8?

Domain-function analysis of ASB8 requires precise technical execution:

Critical methodological considerations:

  • Domain boundary definition:

    • Use multiple bioinformatic prediction tools rather than a single algorithm

    • Validate domain boundaries with limited proteolysis

    • Consider structural information from related proteins

    • Test multiple boundary options when creating truncation constructs

  • Expression construct design:

    • Carefully position epitope/fusion tags to avoid interference with domain function

    • Create both N- and C-terminal tagged versions to compare functionality

    • Include flexible linkers between domains and tags

    • Design domain-swapping experiments with related ASB family members

  • Functional validation approaches:

    • Test isolated domains and combinations for activity

    • Perform alanine-scanning mutagenesis of key residues

    • Use targeted missense mutations rather than large deletions

    • Employ rescue experiments with domain-specific mutants

  • Structural integrity verification:

    • Assess protein folding with circular dichroism or thermal shift assays

    • Confirm subcellular localization is maintained for mutant constructs

    • Evaluate protein stability and expression levels

    • Consider in silico molecular dynamics simulations to predict effects of mutations

Domain analysis should progress from computational prediction to experimental validation to functional testing. Researchers should be particularly vigilant about potential artifacts from improper domain truncation, which can lead to misfolded proteins and misleading results. When designing experiments, consider both loss-of-function and gain-of-function approaches to comprehensively assess domain contributions.

How should researchers integrate multi-omics approaches in ASB8 functional studies?

Multi-omics integration provides comprehensive insights into ASB8 function beyond single-method approaches:

Strategic implementation framework:

  • Experimental design for multi-omics integration:

    • Design experiments with matched samples across platforms

    • Include appropriate time points to capture dynamic changes

    • Incorporate perturbation conditions (ASB8 knockout, overexpression)

    • Consider single-cell approaches for heterogeneous samples

  • Recommended omics combination for ASB8:

    • Transcriptomics: RNA-seq to identify gene expression changes

    • Proteomics: Quantitative MS to detect protein abundance changes

    • Ubiquitylomics: Ubiquitin remnant profiling to identify substrates

    • Interactomics: Proximity labeling or IP-MS to map interaction networks

  • Analytical pipeline for integration:

    • Apply dimension reduction techniques for visualization

    • Perform pathway enrichment across multiple data types

    • Use network analysis to identify functional modules

    • Employ machine learning for pattern recognition

  • Validation of multi-omics findings:

    • Select key nodes for targeted validation

    • Confirm causal relationships with functional assays

    • Develop predictive models and test with new perturbations

    • Compare findings with publicly available datasets

A particularly effective approach is to examine the correlation between transcriptomic and proteomic changes following ASB8 perturbation, as discordance can reveal post-transcriptional regulation mechanisms potentially mediated by ASB8's role in protein ubiquitination.

What are the established protocols for studying ASB8 protein-protein interactions in physiologically relevant contexts?

Investigating ASB8 protein-protein interactions requires methods that preserve physiological relevance:

Methodological recommendations:

  • Endogenous interaction detection:

    • Co-immunoprecipitation with validated antibodies against endogenous proteins

    • Proximity ligation assay (PLA) for visualizing interactions in intact cells

    • FRET/BRET approaches with minimally tagged proteins at endogenous levels

    • Crosslinking mass spectrometry to capture transient interactions

  • Context-specific interaction mapping:

    • Perform interaction studies under relevant physiological stimuli

    • Compare interactions in different cell types where ASB8 is expressed

    • Examine cell cycle-dependent or differentiation-stage specific interactions

    • Investigate stress-induced changes in the ASB8 interactome

  • E3 ligase complex analysis:

    • Two-step purification strategies to isolate intact complexes

    • Activity-based probes to capture functionally active complexes

    • In situ labeling of ubiquitination substrates

    • Reconstitution of minimal functional complexes in vitro

  • Domain-specific interaction mapping:

    • Mutation of key residues in the ankyrin repeat versus SOCS box domains

    • Competition assays with domain-specific peptides

    • Structural analysis of co-crystalized interaction interfaces

    • Hydrogen-deuterium exchange mass spectrometry for interface mapping

When interpreting results, researchers should consider that different methods have inherent biases in detecting certain types of interactions. Cross-validation with multiple techniques strengthens confidence in identified interaction partners. Special attention should be paid to distinguishing direct from indirect interactions and quantifying interaction dynamics under different conditions.

How can researchers overcome technical challenges in distinguishing ASB8 from other ASB family members?

Distinguishing ASB8 from other ASB family members requires specialized approaches:

Technical solutions:

  • Antibody validation and selection:

    • Test antibodies against recombinant ASB family proteins for cross-reactivity

    • Validate antibody specificity using CRISPR knockout controls

    • Use epitopes from unique regions outside conserved domains

    • Consider monoclonal antibodies targeting unique peptide sequences

  • Nucleic acid detection specificity:

    • Design primers targeting unique regions with limited sequence homology

    • Include melt curve analysis to confirm amplicon specificity

    • Validate RNA-seq mapping parameters to avoid multi-mapped reads

    • Consider isoform-specific detection methods

  • Functional discrimination approaches:

    • Develop ASB8-specific substrate ubiquitination assays

    • Characterize unique interactors that distinguish from other family members

    • Identify cell type-specific expression patterns

    • Utilize rescue experiments with family members to test functional redundancy

  • Computational analysis strategies:

    • Apply stringent parameters in sequence alignment

    • Implement peptide uniqueness filters in proteomic analyses

    • Develop machine learning classifiers for distinguishing family members

    • Create ASB8-specific signature based on downstream effects

These approaches should be implemented as complementary strategies, with multiple methods providing convergent evidence for ASB8-specific effects. Researchers should explicitly address potential family member cross-reactivity in methods sections and include appropriate controls in experimental designs.

What statistical approaches are most appropriate for analyzing ASB8 expression data across diverse human tissues?

Analysis of ASB8 expression across tissues requires specialized statistical considerations:

Statistical methodology recommendations:

  • Data normalization approaches:

    • Apply tissue-specific normalization to account for compositional differences

    • Use multiple reference genes specific to each tissue type

    • Consider spike-in standards for absolute quantification

    • Apply quantile normalization only within similar tissue types

  • Differential expression analysis:

    • Use linear mixed models to account for within-subject correlations

    • Apply Bayesian approaches for small sample sizes

    • Consider non-parametric methods for non-normally distributed data

    • Adjust for tissue-specific confounding variables

  • Correlation analysis with clinical parameters:

    • Calculate tissue-specific correlation coefficients

    • Apply multivariate regression to identify independent associations

    • Use principal component analysis to reduce dimensionality

    • Implement mediation analysis to explore causal relationships

  • Visualization and interpretation:

    • Create tissue-specific expression heatmaps

    • Generate tissue-resolved network analyses

    • Plot expression against tissue-specific markers

    • Develop interactive visualizations for multi-dimensional exploration

When implementing these approaches, researchers should be particularly attentive to potential batch effects, which can be mistaken for biological differences. Multiple statistical tests should be accompanied by appropriate multiple testing corrections, and results should be interpreted in the context of biological significance beyond statistical significance.

What emerging technologies show promise for advancing ASB8 research?

Several cutting-edge technologies offer significant potential for advancing ASB8 research:

Emerging methodological approaches:

  • CRISPR-based functional genomics:

    • CRISPR activation/interference for endogenous gene modulation

    • Base editing for introducing precise point mutations

    • CRISPR screens targeting ASB8 pathway components

    • In vivo CRISPR delivery for tissue-specific manipulation

  • Advanced imaging technologies:

    • Super-resolution microscopy for subcellular localization

    • Live-cell imaging with split fluorescent proteins for interaction dynamics

    • Expansion microscopy for enhanced spatial resolution

    • Correlative light and electron microscopy for ultrastructural context

  • Single-cell multi-omics:

    • Integrated single-cell RNA and protein profiling

    • Spatial transcriptomics for tissue context

    • Single-cell interactome analysis

    • Lineage tracing with molecular recording

  • Structural biology advances:

    • Cryo-EM for complex structure determination

    • Integrative structural modeling combining multiple data types

    • AlphaFold2 prediction with experimental validation

    • Time-resolved structural methods for capturing conformational changes

Each of these technologies addresses specific limitations in current ASB8 research and offers opportunities for novel discoveries. Researchers should consider collaborative approaches to leverage these specialized technologies and focus on how they can answer previously intractable questions about ASB8 function, regulation, and therapeutic targeting.

How should researchers design longitudinal studies to investigate ASB8's role in disease progression?

Longitudinal studies examining ASB8 in disease contexts require careful methodological planning:

Design considerations:

  • Cohort selection and characterization:

    • Enroll patients at well-defined disease stages

    • Include pre-symptomatic individuals when possible

    • Collect comprehensive baseline data including ASB8 pathway components

    • Consider genetic stratification based on ASB8 variants

  • Sampling strategy:

    • Define optimal sampling frequency based on disease progression rate

    • Include event-triggered sampling at disease milestones

    • Establish protocols for consistent sample processing

    • Implement biobanking with future analysis options

  • Analytical approach:

    • Apply longitudinal statistical methods (mixed effects models, GEE)

    • Use trajectory analysis to identify patient subgroups

    • Implement change-point detection algorithms

    • Incorporate time-varying covariates in models

  • Integration with interventional studies:

    • Design intervention timing based on longitudinal biomarker changes

    • Use adaptive trial designs responsive to ASB8 pathway alterations

    • Collect samples pre- and post-intervention

    • Apply pharmacodynamic modeling to capture ASB8-related responses

These longitudinal approaches are particularly valuable for connecting ASB8 molecular alterations to disease phenotypes that develop over time. Researchers should plan for sufficient duration of follow-up based on the disease natural history and incorporate methods to minimize attrition bias in long-term studies.

How can researchers effectively position ASB8 findings within the broader ubiquitin-proteasome system literature?

Contextualizing ASB8 research within the ubiquitin-proteasome system requires strategic integration:

Integration framework:

  • Comparative analysis with other E3 ligases:

    • Systematically compare substrate specificity with related SOCS-box proteins

    • Position ASB8 within E3 ligase evolutionary hierarchies

    • Analyze shared vs. unique regulatory mechanisms

    • Evaluate functional redundancy and compensatory mechanisms

  • Pathway-level integration:

    • Map ASB8 substrates to biological pathways

    • Analyze interaction networks involving ASB8 and other UPS components

    • Consider tissue-specific UPS network architectures

    • Examine ASB8's role in UPS-related disease mechanisms

  • Methodological standardization:

    • Adopt standard assays used in the broader UPS field

    • Incorporate consensus reporting guidelines for E3 ligase studies

    • Use established substrate validation hierarchies

    • Implement compatible experimental conditions for cross-study comparisons

  • Knowledge synthesis approaches:

    • Contribute to UPS-focused databases with standardized data

    • Develop integrative computational models of ASB8 in UPS networks

    • Participate in consortium efforts studying E3 ligase biology

    • Apply systems biology approaches to position ASB8 in regulatory networks

By integrating ASB8 research into the broader UPS context, researchers can leverage existing knowledge frameworks while contributing novel insights about this specific E3 ligase component and its unique functional properties.

Product Science Overview

Structure and Function

The ASB8 protein is characterized by the presence of ankyrin repeats and a SOCS box. Ankyrin repeats are known for their role in protein-protein interactions, while the SOCS box is involved in protein degradation. The SOCS box proteins, including ASB8, act as a bridge between substrate proteins and E3 ubiquitin-protein ligases, facilitating the ubiquitination and subsequent proteasomal degradation of target proteins .

Expression and Localization

ASB8 is expressed in various tissues, with the highest expression observed in skeletal muscle. Other tissues with notable expression levels include the heart, brain, placenta, liver, kidney, and pancreas. The protein is predominantly localized in the cytoplasm .

Biological Pathways

ASB8 is involved in several biological pathways, including:

  • Class I MHC mediated antigen processing and presentation
  • Metabolism of proteins

These pathways highlight the importance of ASB8 in immune response and protein metabolism .

Research and Clinical Significance

Research has shown that ASB8 may be a substrate-recognition component of a SCF-like ECS (Elongin-Cullin-SOCS-box protein) E3 ubiquitin-protein ligase complex. This complex mediates the ubiquitination and degradation of target proteins, which is crucial for maintaining cellular homeostasis .

The study of ASB8 and its recombinant forms can provide insights into its role in various diseases and potential therapeutic applications. For instance, understanding the mechanisms of ASB8-mediated protein degradation can aid in the development of treatments for conditions associated with protein aggregation and degradation.

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.