HUB1 Antibody

Shipped with Ice Packs
In Stock

Description

Role in Pre-mRNA Splicing

Hub1 binds non-covalently to spliceosomal proteins like Snu66, modulating spliceosome activity. Key discoveries include:

  • Spliceosomal Interaction: Hub1 binds to the HIND (Hub1-INteracting Domain) motif in Snu66, altering spliceosome composition and enabling recognition of non-canonical splice sites .

  • Alternative Splicing: In S. cerevisiae, Hub1 mediates alternative splicing of SRC1 pre-mRNA by promoting exon skipping and intron retention .

  • Human Cell Studies: Depletion of Hub1 via siRNA causes severe splicing defects, nuclear mRNA retention, and apoptosis due to mitotic errors .

Cellular Consequences of Hub1 Depletion

  • Cell Cycle Arrest: Hub1-deficient human cells exhibit G2/M phase arrest and mitotic catastrophes (e.g., defective chromosome segregation) .

  • Apoptosis: Prolonged Hub1 depletion triggers caspase-mediated apoptosis, linked to splicing speckle disorganization and mRNA processing failures .

Clinical and Pathological Relevance

  • Autoantigenicity: Hub1 is an autoantigen in adult T-cell leukemia (ATL), with ~70% of ATL patients producing anti-Hub1 antibodies .

  • Disease Associations: Hub1 (UBL5) is implicated in mitochondrial stress responses, metabolic disorders, and cancer, though direct disease-causing mutations remain unreported .

Comparative Analysis Across Species

FeatureYeast Hub1Human Hub1 (UBL5)
EssentialityNon-essentialEssential for viability
Splicing RoleModulates specific alternative splicingRequired for critical intron/exon selection
LocalizationNuclear splicing specklesNuclear/cytoplasmic shuttling under stress
Binding PartnersSnu66, spliceosomal tri-snRNPSnu66, kinases, stress-response factors

Future Research Directions

  • Mechanistic Studies: Elucidate how Hub1 fine-tunes spliceosome activity for specific introns.

  • Therapeutic Targets: Explore Hub1’s role in cancers linked to splicing defects (e.g., leukemia) .

  • Stress Pathways: Investigate Hub1’s involvement in mitochondrial unfolded protein responses (UPR<sup>mt</sup>) and metabolic regulation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
HUB1 antibody; BRE1A antibody; RDO4 antibody; At2g44950/At2g44960 antibody; T13E15.24/T13E15.23 antibody; T14P1.25/T14P1.24 antibody; E3 ubiquitin-protein ligase BRE1-like 1 antibody; AtBRE1 antibody; EC 2.3.2.27 antibody; Protein HISTONE MONOUBIQUITINATION 1 antibody; AtHUB1 antibody; Protein REDUCED DORMANCY 4 antibody; RING-type E3 ubiquitin transferase BRE1-like 1 antibody
Target Names
HUB1
Uniprot No.

Target Background

Function
HUB1 (HISTONE MONOUBIQUITINATION 1) is an E3 ubiquitin-protein ligase that plays a crucial role in histone modification and gene regulation. It specifically monoubiquitinates histone H2B at lysine 143 (H2BK143ub1), marking a site for epigenetic transcriptional activation. This modification is essential for the subsequent methylation of histone H3 at lysine 4 (H3K4me) and potentially H3K79me, contributing significantly to the histone code and gene regulation. HUB1 forms a ubiquitin ligase complex in collaboration with the E2 enzyme UBC2/RAD6. This complex is involved in various biological processes, including flowering time regulation, defense against necrotrophic fungal pathogens, and the control of seed dormancy and germination.
Gene References Into Functions
  1. Two RING E3 ligases, HUB1 and HUB2 (HISTONE MONOUBIQUITINATION 2), are essential for the selective transcriptional activation of four cuticle biosynthesis genes in Arabidopsis thaliana. PMID: 24319075
  2. Studies indicate that alterations in HUB1 expression levels affect plant fitness parameters, highlighting the significant role of histone H2B monoubiquitination in plant development and overall fitness. PMID: 23073007
  3. Double mutant analysis between hub1-1 and elongata (elo) revealed an embryo-lethal phenotype, suggesting a synergistic genetic interaction between these genes. PMID: 22762858
  4. A critical role is proposed for chromatin remodeling in seed dormancy through H2B monoubiquitination mediated by HUB1 and HUB2. PMID: 17329563
  5. Research demonstrates that HUB1 is a functional homolog of the human and yeast BRE1 proteins, suggesting its involvement in gene activation and cell cycle regulation through chromatin modifications. PMID: 17329565
  6. HUB1 upregulates the expression of FLC (FLOWERING LOCUS C), and interacts with UBC1 and UBC2 to repress flowering. PMID: 18980658
  7. Studies describe the role of HUB1 in disease resistance in Arabidopsis thaliana, highlighting its importance in plant defense mechanisms. PMID: 19286969

Show More

Hide All

Database Links

KEGG: ath:AT2G44950

STRING: 3702.AT2G44950.1

UniGene: At.43266

Protein Families
BRE1 family
Subcellular Location
Nucleus.
Tissue Specificity
Ubiquitously expressed.

Q&A

What is HUB1 and what are its primary functions?

HUB1 (HTLV-I U5RE Binding Protein 1) is a protein that binds to a possible repressor element of the long terminal repeat of the human T lymphotropic virus type I (HTLV-I) . Unlike typical ubiquitin-like proteins, HUB1 binds proteins non-covalently and independently of ATP, with its YY motif shown to be nonessential for this binding activity . This protein has been associated with diverse physiological functions, including:

  • Cell cycle progression and polarized growth

  • Mitochondrial unfolded protein response

  • mRNA splicing, particularly through interaction with spliceosomal components

Most significantly, HUB1 has been identified as modifying the spliceosome in a way that enables it to tolerate and use certain non-canonical 5' splice sites, representing a novel mechanism for splice site utilization guided by non-covalent modification .

How does HUB1 interact with the spliceosome?

HUB1 interacts with the spliceosome primarily through binding to Snu66, a component of the (U4/U6.U5) tri-small nuclear ribonucleoprotein particle (snRNP) . This interaction has been verified through various experimental approaches:

  • Co-immunoprecipitation assays with antibodies specific for yeast HUB1 and Snu66 confirm their interaction in vivo

  • The binding occurs through a specific element called HIND (HUB1 interaction domain) in Snu66

  • Examination of Snu66 revealed two highly similar elements (72% identity) arranged in tandem that mediate this interaction

  • HUB1 can also co-immunoprecipitate with Prp8, a central spliceosomal protein, in a reaction mediated by Snu66

In Hub1-deficient cells, researchers observed an overrepresentation of certain proteins from U1 and U2 snRNPs, but not of the tri-snRNP, indicating that while HUB1 modification affects spliceosomal composition, the basic makeup of snRNPs is preserved .

What is the significance of HUB1 as an autoantigen?

HUB1 has emerged as a significant autoantigen that frequently elicits humoral immune responses, particularly in patients with adult T cell leukemia (ATL) . When researchers examined the reactivity of IgG serum samples against purified HUB1 protein through Western blot analysis, they documented the following pattern:

Subject GroupPositive HUB1 Antibody ReactivityPercentageSample Size
ATL Patients2170.0%30
Asymptomatic HTLV-I Carriers1041.7%24
Healthy Donors937.5%24

This significantly higher rate of antibody production against HUB1 in ATL patients compared to carriers and healthy individuals suggests a potential relationship between this autoimmune response and ATL development . The autoantigenicity of HUB1 makes antibodies against it valuable potential biomarkers for disease progression in HTLV-I-related pathologies.

What techniques are commonly used to detect HUB1 antibodies in research settings?

Several methodological approaches have been employed to detect and study HUB1 antibodies in research:

  • SEREX Method (Serological Identification of Antigens by Recombinant Expression Cloning): This technique has been used for immunological screening of cDNA phage expression libraries with IgG antibodies from patient serum to identify HUB1 as an autoantigen .

  • Recombinant Protein Expression: HUB1 cDNA has been expressed as a fusion protein with glutathione S-transferase, followed by thrombin treatment to isolate purified HUB1 protein for antibody studies .

  • Western Blot Analysis: This technique has been used to examine the reactivity of IgG serum samples from patients, carriers, and healthy donors against purified HUB1 protein .

  • Co-immunoprecipitation Assays: Antibodies specific for HUB1 and Snu66 have been employed in these assays to verify their interactions in vivo .

  • Yeast Two-hybrid (Y2H) Assays: These have been used to confirm HUB1's interaction with Snu66 and identify the specific binding domains involved .

What is the relationship between HUB1 and HTLV-I?

HUB1 (HTLV-I U5RE binding protein 1) is named for its ability to bind to a possible repressor element of the long terminal repeat (LTR) of the human T lymphotropic virus type I (HTLV-I) . This relationship extends beyond nomenclature into functional relevance in several ways:

  • HUB1 potentially regulates HTLV-I viral gene expression through its interaction with the viral LTR

  • ATL, a malignancy associated with HTLV-I infection, shows significantly higher prevalence of anti-HUB1 antibodies (70%) compared to asymptomatic HTLV-I carriers (41.7%)

  • The increased production of antibodies against HUB1 in ATL patients suggests its potential role in disease progression from HTLV-I infection to ATL development

  • HUB1's identification through immunological screening of a cDNA library from an ATL cell line (ST1) further strengthens its association with HTLV-I-related pathology

How can HUB1 antibodies be used to study spliceosomal modifications?

HUB1 antibodies provide valuable tools for investigating the complex mechanisms of spliceosomal modifications through several sophisticated methodological approaches:

  • Co-immunoprecipitation coupled with mass spectrometry: HUB1 antibodies can isolate HUB1-associated spliceosomal complexes, allowing identification of the protein composition modifications induced by HUB1. This approach has revealed that in Hub1-deficient cells, certain proteins from U1 and U2 snRNPs are overrepresented, indicating HUB1's role in spliceosomal composition regulation .

  • Comparative immunoprecipitation: By comparing spliceosomal complexes isolated from wild-type versus Hub1-deficient cells, researchers can identify specific spliceosomal alterations dependent on HUB1's presence. This comparison has shown that while HUB1 deficiency affects U1 and U2 snRNPs representation, the basic makeup of snRNPs remains preserved .

  • Sequential co-immunoprecipitation: This technique can reveal multi-protein interactions, such as how HUB1 antibodies can co-immunoprecipitate Prp8 (a central spliceosomal protein) in a reaction mediated by Snu66, demonstrating HUB1's integration into established spliceosomal networks .

  • Structural studies of HUB1-modified spliceosomes: Antibodies against HUB1 can help isolate spliceosomes for structural analysis using techniques like cryo-electron microscopy, potentially revealing how HUB1 binding physically alters spliceosome conformation to accommodate non-canonical splice sites.

  • Splice-site selection assays: Using reporter constructs with various splice site configurations, researchers can determine how HUB1 antibody depletion affects the spliceosome's ability to recognize non-canonical splice sites.

What experimental approaches can differentiate between free HUB1 and spliceosome-bound HUB1?

Differentiating between free and spliceosome-bound HUB1 requires specialized experimental techniques:

  • Differential centrifugation and glycerol gradient fractionation: These techniques can separate free HUB1 from larger spliceosomal complexes, followed by Western blotting with HUB1 antibodies to identify the protein's distribution.

  • Size exclusion chromatography: This approach separates protein complexes based on size, allowing researchers to distinguish between free HUB1 and HUB1 incorporated into the much larger spliceosomal complexes.

  • Conformation-specific antibodies: Development of antibodies that specifically recognize HUB1 in its free form versus when bound to Snu66 through the HIND domain can provide direct differentiation capabilities.

  • Proximity ligation assays (PLA): This technique can detect HUB1-Snu66 interactions in situ, visualizing only HUB1 that is in close proximity to spliceosomal proteins rather than the free protein.

  • Cross-linking followed by immunoprecipitation: Chemical cross-linking can fix HUB1-spliceosome interactions prior to immunoprecipitation with spliceosomal protein antibodies, allowing specific analysis of the bound fraction.

  • Native gel electrophoresis: This technique preserves protein-protein interactions and can separate free HUB1 from spliceosome-bound HUB1 based on significant differences in complex size and charge.

What are the differences in HUB1 antibody reactivity between ATL patients, HTLV-I carriers, and healthy donors?

Analysis of HUB1 antibody reactivity reveals significant differences across populations that may have important diagnostic and prognostic implications:

Subject GroupPositive HUB1 Antibody ReactivityPercentageSample SizeStatistical Significance
ATL Patients21/3070.0%30p < 0.05 compared to both other groups
HTLV-I Carriers10/2441.7%24p > 0.05 compared to healthy donors
Healthy Donors9/2437.5%24Reference group

These findings indicate that while anti-HUB1 antibodies can be detected even in healthy individuals, there is a significant elevation in antibody prevalence among ATL patients (70.0%) compared to asymptomatic HTLV-I carriers (41.7%) and healthy donors (37.5%) .

Several hypotheses might explain this pattern:

  • Increased exposure of HUB1 antigens during ATL development, possibly due to cellular damage or altered subcellular localization

  • Post-translational modifications of HUB1 in ATL cells that increase immunogenicity

  • Disruption of immunological tolerance in ATL patients leading to broader autoantibody responses

  • Potential cross-reactivity between viral proteins and HUB1 epitopes

The differential antibody reactivity suggests that HUB1 antibody testing might serve as a valuable adjunct diagnostic tool in distinguishing ATL from asymptomatic HTLV-I infection.

How does the HUB1-Snu66 interaction affect experimental outcomes in splicing assays?

The HUB1-Snu66 interaction significantly influences experimental outcomes in splicing assays through several mechanisms:

  • Modification of spliceosome composition: In Hub1-deficient cells, researchers have observed an overrepresentation of certain proteins from U1 and U2 snRNPs, but not of the tri-snRNP . This alteration in spliceosomal protein composition can affect experimental outcomes in splicing efficiency assays.

  • Regulation of non-canonical splice site recognition: HUB1 binding to Snu66 through the HIND element appears critical for enabling the spliceosome to tolerate and utilize certain non-canonical 5' splice sites . This has profound implications for experiments examining alternative splicing.

  • Temperature-dependent effects: Conditional mutants of HUB1 show moderate RNA splicing defects, particularly at high temperatures . This temperature sensitivity should be carefully controlled in experimental designs involving HUB1 or Snu66 manipulations.

  • Differential effects with overexpression versus depletion: While HUB1-deficient cells show splicing impairments and spliceosomal alterations, overexpression studies have shown no significant changes in spliceosome composition . This asymmetrical response requires consideration when designing gain-of-function versus loss-of-function experiments.

  • Integration of Prp8 into the spliceosome: HUB1 can co-immunoprecipitate with Prp8, a central spliceosomal protein, through its interaction with Snu66 . This integration into the core spliceosomal machinery suggests that HUB1-Snu66 interactions may affect fundamental aspects of the splicing reaction.

What methodological approaches can resolve contradictory data in HUB1 antibody studies?

Resolving contradictory data in HUB1 antibody studies requires systematic methodological approaches:

  • Comprehensive antibody validation: Establish detailed validation protocols including Western blotting against recombinant HUB1, immunoprecipitation efficiency testing, and specificity verification against HUB1-knockout controls. This helps identify antibodies with appropriate specificity and sensitivity.

  • Epitope mapping: Different antibodies targeting distinct domains of HUB1 may yield varying results depending on protein conformation or interaction status. Precise characterization of recognition epitopes allows more accurate interpretation of seemingly contradictory results.

  • Standardized sample preparation: Variations in sample processing (protein extraction methods, buffer compositions, detergents used) can significantly affect HUB1 detection. Implementing consistent protocols across laboratories minimizes these technical variables.

  • Multi-methodological verification: Cross-validate findings using complementary techniques such as ELISA, Western blotting, immunohistochemistry, and flow cytometry to distinguish true biological variations from method-specific artifacts.

  • Genetic approaches: Utilize CRISPR-Cas9 HUB1 knockout or knockdown cells as definitive negative controls to confirm antibody specificity and validate experimental findings.

  • Post-translational modification assessment: Evaluate whether contradictory results might be explained by differential detection of HUB1 with various post-translational modifications by different antibodies.

  • Inter-laboratory validation studies: Establish collaborative testing of the same samples across multiple laboratories using standardized protocols to identify reproducible findings versus lab-specific anomalies.

  • Detailed methodology reporting: Implement comprehensive documentation of experimental conditions, including buffer compositions, incubation times, and sample processing methods to enable precise comparison across studies.

What are the optimal experimental designs for studying HUB1's role in non-canonical splice site recognition?

Optimal experimental designs for investigating HUB1's role in non-canonical splice site recognition include:

  • Minigene splicing reporters: Construct reporter systems containing various non-canonical 5' splice sites coupled with downstream fluorescent or luminescent markers. These can be transfected into cells with normal or manipulated HUB1 levels to assess splicing efficiency.

  • CRISPR-Cas9 HUB1 knockout/knock-in systems: Generate cell lines with complete HUB1 deletion or specific mutations affecting the Snu66 interaction interface to determine the precise contribution of HUB1 to non-canonical splice site recognition.

  • Transcriptome-wide splicing analysis: Perform RNA-seq in cells with normal, depleted, or mutated HUB1 to identify all splice junctions affected by HUB1 status, with particular focus on non-canonical sites that might be especially sensitive to HUB1 depletion.

  • In vitro splicing assays: Develop cell-free splicing systems using nuclear extracts from cells with various HUB1 statuses, allowing precise biochemical analysis of splicing intermediates and kinetics.

  • Structure-function analysis: Create a series of HUB1 mutants with altered binding to Snu66 and assess their ability to rescue splicing defects in HUB1-deficient cells, correlating functional outcomes with structural changes.

  • Differential temperature experiments: Given the temperature sensitivity of conditional HUB1 mutants, design experiments that compare splicing outcomes across temperature ranges to identify conditions where HUB1's role becomes critical.

  • Co-depletion studies: Simultaneously manipulate HUB1 and other spliceosomal components to identify genetic interactions and potential compensatory mechanisms that might obscure HUB1's function in single-gene studies.

How can researchers develop highly specific antibodies against different functional domains of HUB1?

Developing highly specific antibodies against different functional domains of HUB1 requires strategic approaches:

  • Domain-specific peptide immunogens: Design synthetic peptides corresponding to distinct functional regions of HUB1, particularly:

    • The region that interacts with the HIND element of Snu66

    • The interface involved in non-covalent binding to other proteins

    • The YY motif region, despite its non-essentiality for binding

  • Recombinant fragment immunization: Express and purify discrete structural domains of HUB1 as immunogens, potentially increasing the likelihood of generating antibodies that recognize native protein conformations.

  • Phage display technology: Utilize phage display libraries to select antibodies with high specificity and affinity for particular HUB1 domains, followed by affinity maturation to enhance binding characteristics.

  • Conformation-specific antibody selection: Develop screening methods that select for antibodies recognizing specific conformational states of HUB1, such as free versus Snu66-bound forms.

  • Cross-reactivity elimination: Screen candidate antibodies against related ubiquitin-like proteins to eliminate those showing unwanted cross-reactivity.

  • Epitope mapping validation: Conduct comprehensive epitope mapping using techniques such as hydrogen-deuterium exchange mass spectrometry or X-ray crystallography of antibody-antigen complexes to confirm domain-specific binding.

  • Functional validation: Test antibodies for their ability to inhibit specific HUB1 functions, such as Snu66 binding or spliceosomal modification, providing confirmation of domain-specific recognition.

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.