ASF1B Human

ASF1 Anti-Silencing Function 1 Homolog B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Cellular Functions

ASF1B regulates chromatin dynamics through histone deposition and exchange, enabling replication-dependent and independent nucleosome assembly .

Key Mechanisms

  • Nucleosome Assembly: Cooperates with chromatin assembly factor 1 (CAF-1) to facilitate replication-coupled histone deposition during DNA replication .

  • Histone H3.3 Synergy: Promotes cell cycle progression and β-cell proliferation by stabilizing histone H3.3 deposition at replication-independent sites .

  • Immune Modulation: Correlates with increased infiltration of immune cells (e.g., CD8+ T cells, neutrophils) but also enhances Treg cell activity and immune checkpoint expression (e.g., PD-1) .

Interactions

Partner ProteinFunction
TLK1/TLK2Phosphorylates ASF1B, regulating cell cycle progression
CHAF1A/CHAF1BCollaborates in replication-dependent chromatin assembly
CDK9Stabilizes CDK9 to promote cervical cancer progression

Clinical Implications

ASF1B is implicated in oncogenesis and serves as a prognostic biomarker.

Cancer-Associated Roles

Cancer TypePrognostic Impact
HCCHigh expression: HR = 1.71 (OS), HR = 1.72 (PFS)
Cervical CancerKnockdown reduces tumor growth in murine models
Breast/ProstateAssociated with aggressive disease progression

Tissue Expression and Localization

ASF1B exhibits tissue-specific nuclear expression, with prominent activity in reproductive and tumor tissues .

TissueExpression LevelLocalization
TestisHighNucleus
LiverModerateNucleus
Tumor TissuesElevatedNucleus

Key Studies

  1. HCC: ASF1B knockdown inhibits proliferation and colony formation in Hep3B/Huh7 cells, with enrichment in cell cycle pathways (e.g., DNA replication, mismatch repair) .

  2. β-Cell Proliferation: Overexpression increases S-phase entry and mitotic progression, dependent on histone-binding capacity .

  3. Immune Microenvironment: Positively correlates with Treg infiltration and immune checkpoints, suggesting potential as an immunotherapy target .

Therapeutic Strategies

  • Targeting ASF1B: Inhibition may suppress tumor growth in cancers like HCC and cervical cancer .

  • Combination Therapies: Pairing with anti-PD-1/PD-L1 therapies could enhance efficacy in high-ASF1B-expressing tumors .

Product Specs

Introduction
ASF1 Anti-Silencing Function 1 Homolog B (ASF1B), a member of the H3/H4 histone chaperone protein family, shares similarities with the yeast anti-silencing function-1 protein. Acting as a substrate for tousled-like kinase, a family of cell cycle-regulated kinases, ASF1B is believed to play a vital role in regulating chromatin's nucleosome structure. This is achieved by ensuring a consistent supply of histones to nucleosome assembly sites. In conjunction with CAF-1 (chromatin assembly factor 1), ASF1B facilitates replication-dependent chromatin assembly. Notably, ASF1B exhibits high expression levels in the testis and lower levels in the colon, small intestine, and thymus. Spermatogenesis is known to be dependent on ASF1B.
Description
Recombinantly produced in E.Coli, ASF1B Human Recombinant is a single, non-glycosylated polypeptide chain. It consists of 210 amino acids (with amino acids 1 to 202 present) and has a molecular weight of 23.4kDa. An 8 amino acid His-tag is fused to the C-terminus of ASF1B. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
The product appears as a sterile, colorless solution after filtration.
Formulation
The ASF1B protein solution is provided at a concentration of 1mg/ml. It is formulated in a buffer containing 20mM Tris-HCl (pH 8.0), 10% glycerol, 0.15M NaCl, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), the product should be stored at 4°C. For extended storage, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
SDS-PAGE analysis confirms a purity exceeding 85%.
Synonyms
Histone chaperone ASF1B, Anti-silencing function protein 1 homolog B, hAsf1, hAsf1b, CCG1-interacting factor A-II, CIA-II, hCIA-II, ASF1B.
Source
Escherichia Coli.
Amino Acid Sequence
MAKVSVLNVA VLENPSPFHS PFRFEISFEC SEALADDLEW KIIYVGSAES EEFDQILDSV LVGPVPAGRH MFVFQADAPN PSLIPETDAV GVTVVLITCT YHGQEFIRVG YYVNNEYLNP ELRENPPMKP DFSQLQRNIL ASNPRVTRFH INWDNNMDRL EAIETQDPSL GCGLPLNCTP
IKGLGLPGCI PGLLPENSMD CILEHHHHHH
.

Q&A

What is ASF1B and how does it differ from ASF1A in human cells?

ASF1B is one of two mammalian paralogs of the Anti-silencing function 1 (ASF1) histone chaperone family. While ASF1A and ASF1B share approximately 70% sequence identity, they exhibit significant functional differences. ASF1A participates in pathways not exclusive to S-phase (including DNA damage repair), whereas ASF1B is primarily involved in cell proliferation. Their expression patterns also differ substantially - ASF1A is ubiquitously expressed across cell types, while ASF1B expression is predominantly limited to proliferating tissues and is greatly reduced in terminally differentiated and quiescent cells .

Methodologically, when investigating ASF1 paralog functions, researchers should consider:

  • Tissue-specific expression analysis using qPCR or western blotting

  • Cell cycle phase-specific localization studies using synchronized cell populations

  • Paralog-specific antibodies for immunoprecipitation experiments

How does ASF1B interact with histone proteins in the human system?

ASF1B functions as a histone chaperone that primarily interacts with H3-H4 dimers. This interaction occurs through a specific binding interface, and the histone-binding capability is essential for ASF1B's biological functions. Research has demonstrated that a histone binding-deficient mutant of ASF1B (V94R) fails to induce β-cell proliferation, indicating that histone binding to ASF1B is required for its proliferation-inducing effects .

While ASF1B interacts with multiple histone H3 variants (H3.1, H3.2, and H3.3), it has different functional outcomes with each. When co-expressed with histone H3.3, ASF1B shows enhanced ability to promote β-cell proliferation. In contrast, overexpression of histones H3.1 and H3.2 does not significantly impact ASF1B-mediated induction of proliferation .

What mechanisms underlie ASF1B-induced human β-cell proliferation?

ASF1B promotes human β-cell proliferation through multiple coordinated mechanisms:

  • Transcriptional regulation: ASF1B overexpression creates distinct transcriptional signatures with approximately 460 differentially expressed genes. Of these, 286 are induced while 179 are repressed . The upregulated genes include numerous cell cycle regulators that facilitate G1/S transition.

  • Cell cycle checkpoint regulation: ASF1B significantly increases the population of cells in S-phase (from ~0.5% to ~13%) with a corresponding decrease in G1 phase population (from 97% to 77%) .

  • Histone variant cooperation: ASF1B works synergistically with histone H3.3A to promote proliferation, while suppression of endogenous H3.3 attenuates ASF1B's proliferative effect .

Methodologically, researchers can assess these mechanisms through:

  • RNA sequencing to identify ASF1B-responsive genes

  • Flow cytometry for cell cycle analysis

  • BrdU incorporation assays to measure proliferation rates

  • [³H]-thymidine incorporation into newly synthesized DNA

How can researchers effectively measure ASF1B-induced proliferation in human β-cells?

Multiple complementary techniques should be employed to comprehensively assess ASF1B-induced proliferation:

  • BrdU incorporation: Human islets transduced with adenoviral ASF1B (Ad-ASF1B) show a >4-fold increase in BrdU-positive β-cells compared to controls when pulsed with BrdU 24 hours post-transduction .

  • [³H]-thymidine incorporation: This technique measures DNA synthesis by quantifying radioactive thymidine uptake. ASF1B overexpression induces ~1.5-fold higher [³H]-thymidine incorporation (~120 CPM/μg protein) compared to controls (70 CPM/μg protein) .

  • Flow cytometry analysis of DNA content: This approach enables quantification of cell populations in different cell cycle phases. The table below summarizes typical results:

Cell Cycle PhaseControl IsletsASF1B-Overexpressing Islets
G0/G1~97%~77%
S-phase~0.5%~13%
G2/M~2.5%~10%
  • 2D FACS analysis: Using BrdU as a marker of DNA synthesis and propidium iodide (PI) for DNA content provides more detailed cell cycle progression data. This technique confirms significant increases in S-phase population (from ~1% to ~11%) .

What role does ASF1B play in DNA double-strand break (DSB) repair?

ASF1B participates in DNA double-strand break repair through its interaction with the 53BP1-RIF1 pathway:

  • Recruitment dynamics: Both ASF1A and ASF1B are recruited to sites of laser-induced DNA damage. While ASF1A recruitment is primarily dependent on 53BP1-RIF1, ASF1B shows only modestly reduced recruitment in 53BP1- or RIF1-null cells, suggesting additional recruitment mechanisms .

  • Spatial distribution: ASF1B colocalizes with 53BP1 and RIF1 at distal chromatin flanking DSBs. This spatial arrangement is influenced by BRCA1, which excludes 53BP1 (and consequently RIF1 and ASF1) from chromatin proximal to DSBs in the S/G2 phase .

  • Pathway involvement: ASF1 acts in the same non-homologous end joining (NHEJ) pathway as RIF1 but functions through a parallel pathway with the shieldin complex. Importantly, its histone chaperone activity is essential for this function, as demonstrated by the inability of the histone-binding-defective V94R mutant to rescue defects in DNA damage resistance .

Experimental approaches for investigating ASF1B in DSB repair include:

  • Laser microirradiation combined with live-cell imaging

  • Immunofluorescence microscopy to analyze protein distribution patterns

  • Epistasis analysis using knockouts of pathway components

  • DNA damage sensitivity assays with DSB-inducing agents like etoposide

How does the RIF1-ASF1 complex influence chromatin structure during DNA repair?

The RIF1-ASF1 complex plays a crucial role in modulating chromatin structure to facilitate DNA repair through several mechanisms:

  • Histone modifications: ASF1-bound non-nucleosomal H3-H4 heterodimers contain pre-modified H3K9me1, particularly under genotoxic conditions. This modification is enriched in the RIF1-ASF1 complex following DNA damage .

  • Chromatin compaction: ASF1 promotes compaction of adjacent chromatin through heterochromatinization, which protects broken DNA ends from BRCA1-mediated resection .

  • Interaction specificity: ASF1A interacts more strongly with RIF1 than ASF1B does. This interaction occurs through ASF1A's E36/D37 residues and RIF1's B-domain (R1217/R1218/Q1219) . Point mutations in these regions significantly reduce the interaction.

  • Competition with other pathways: ASF1's binding to RIF1 occurs in a manner similar to its interaction with CAF-1 and HIRA, suggesting these interactions are mutually exclusive and may represent regulatory checkpoints in the repair process .

For researching these mechanisms, appropriate methodologies include:

  • Co-immunoprecipitation to study protein-protein interactions

  • ChIP-seq to analyze histone modifications at damage sites

  • Proximity ligation assays to visualize protein interactions in situ

  • Mutagenesis studies targeting specific interaction domains

What are the most effective gene delivery systems for studying ASF1B function in primary human cells?

When studying ASF1B in primary human cells, particularly in β-cells, adenoviral vectors have proven highly effective:

  • Adenoviral transduction: Adenoviruses containing ASF1B (Ad-ASF1B) achieve significant overexpression in human islets within 48 hours. This approach enables efficient gene delivery to post-mitotic cells like human islet cells, which are otherwise difficult to transfect .

  • Transduction protocol optimization: For human islet studies, a 12-hour transduction period followed by a 24-hour post-transduction recovery yields optimal results for proliferation assays. This timing allows for protein expression while minimizing viral toxicity .

  • Co-expression strategies: To study functional interactions, co-transduction with multiple adenoviral constructs (e.g., ASF1B plus histone variants) can be employed. This approach revealed the synergistic effect of ASF1B and H3.3A on β-cell proliferation .

  • Controls and validation: Appropriate controls such as Ad-GFP or Ad-LacZ are essential. Verification of transgene expression should be performed using both immunohistochemistry and western blot analysis .

Alternative approaches for different experimental contexts include:

  • Lentiviral systems for stable expression and integration

  • CRISPR-Cas9 for genomic editing of endogenous ASF1B

  • Inducible expression systems for temporal control of ASF1B levels

How can researchers distinguish between ASF1B's direct effects on proliferation versus secondary effects on cellular function?

Distinguishing direct from indirect effects of ASF1B on cellular processes requires multi-faceted experimental approaches:

  • Temporal analysis: Monitoring gene expression changes and cellular phenotypes at multiple time points following ASF1B induction helps establish causality. Early transcriptional changes (within 24-48 hours) are more likely to represent direct effects .

  • Domain-specific mutants: Using ASF1B mutants with altered functionality provides insight into mechanism. For example, the histone binding-deficient V94R mutant demonstrates that histone interaction is essential for proliferation induction .

  • Functional assays: Complementing proliferation markers with functional assessments (e.g., glucose-stimulated insulin secretion for β-cells) helps determine whether ASF1B affects specialized cellular functions. Studies show that ASF1B overexpression induces proliferation without affecting insulin secretion or content within a 48-72 hour window .

  • Pathway inhibition: Selectively inhibiting downstream pathways can help delineate the mechanisms through which ASF1B operates. For example, suppressing histone H3.3 attenuates ASF1B's proliferative effect, indicating its dependence on this pathway .

  • Cell-type specificity: Analyzing the effects of ASF1B across different cell types within the same tissue provides insight into specificity. While ASF1B expression increases in both α-cells and β-cells following adenoviral transduction, the proliferative effect is specific to β-cells .

What is the potential of ASF1B as a therapeutic target for diabetes through β-cell regeneration?

ASF1B shows promise as a therapeutic target for diabetes through its ability to induce human β-cell proliferation. Several lines of evidence support this potential:

  • Disease relevance: The expression of ASF1B correlates with resistance to diabetes in mouse models. In a study comparing diabetes-resistant B6 and diabetes-susceptible BTBR mice, obesity induced the expression of ASF1B and other cell cycle regulatory genes in islets from B6 mice but not BTBR mice .

  • Human β-cell specificity: ASF1B overexpression induces proliferation specifically in human β-cells but not in other islet cell types, suggesting a targeted effect that could minimize off-target complications .

  • Functional preservation: ASF1B-induced proliferation does not negatively impact glucose-stimulated insulin secretion or insulin content in the short term, suggesting that newly proliferating cells may maintain functionality .

  • Long-term functionality: While ASF1B does not impair β-cell function within 48-72 hours, longer-term studies are needed to ensure that proliferating β-cells retain their differentiated phenotype and functionality.

  • Delivery methods: Developing β-cell-specific delivery methods for ASF1B modulators would be necessary to avoid potential off-target effects in other proliferative tissues.

  • Safety concerns: Given ASF1B's role in promoting proliferation, careful evaluation is needed to ensure that its therapeutic activation does not increase cancer risk.

How do ASF1B expression patterns correlate with human pathological conditions beyond diabetes?

ASF1B expression patterns have significant implications for various human pathological conditions:

  • Cancer: Given that ASF1B is predominantly expressed in proliferating tissues, its dysregulation has been implicated in cancer progression. ASF1B plays a major role in cellular proliferation across various cancer types .

  • DNA repair disorders: ASF1B's involvement in DNA damage response pathways suggests that its dysfunction could contribute to genomic instability syndromes. The RIF1-ASF1 complex promotes changes in high-order chromatin structure to stimulate the NHEJ pathway for DSB repair .

  • Cell cycle disorders: ASF1B's role in regulating the G1/S transition suggests that abnormal expression could contribute to disorders characterized by dysregulated cell proliferation.

For investigating these correlations, researchers should consider:

  • Analysis of ASF1B expression in tissue microarrays across multiple disease states

  • Correlation studies between ASF1B expression levels and disease progression or outcomes

  • Integration of ASF1B status with other molecular markers for improved disease stratification

  • Single-cell analysis to identify cell type-specific roles of ASF1B in heterogeneous tissues

What are the main challenges in studying ASF1B-histone interactions in primary human tissues?

Researching ASF1B-histone interactions in primary human tissues presents several technical challenges:

  • Limited tissue availability: Primary human tissues, especially pancreatic islets, are scarce and heterogeneous, complicating large-scale studies of ASF1B-histone interactions.

  • Post-mitotic state: Most primary human tissues exist in a post-mitotic state with very low replication rates, making it difficult to study the dynamics of ASF1B during normal cell cycle progression without artificial manipulation .

  • Complex histone variant discrimination: Distinguishing between different histone variants (H3.1, H3.2, H3.3) and their specific interactions with ASF1B requires highly specific antibodies or tagged proteins, which may alter native interactions .

  • Temporal dynamics: ASF1B-histone interactions are dynamic and may change during different phases of the cell cycle or in response to various stimuli, requiring sophisticated live-cell imaging approaches.

  • Context-dependent functionality: The effects of ASF1B may vary significantly depending on cell type and physiological context, necessitating carefully controlled comparative studies.

Innovative approaches to address these challenges include:

  • Development of organoid systems from primary tissues to expand material availability

  • In situ proximity ligation assays to visualize endogenous protein interactions

  • Advanced chromatin immunoprecipitation techniques with improved sensitivity

  • Single-cell approaches to account for cellular heterogeneity within tissues

How can contradictions in ASF1B functional data between different experimental systems be reconciled?

Researchers often encounter contradictory results when studying ASF1B across different experimental systems. These can be reconciled through:

  • Systematic comparison of experimental conditions:

    • Cell type differences: ASF1B functions differently in various cell types. For example, it specifically induces proliferation in β-cells but not in α-cells within the same islet preparation .

    • Species differences: Findings from yeast, mouse, and human systems may differ due to evolutionary divergence in ASF1 functions.

    • Cell cycle state: The initial proliferative state of cells may influence ASF1B's effects, particularly in quiescent versus actively dividing cells.

  • Analysis of experimental timeframes:

    • Short-term versus long-term effects: ASF1B may induce proliferation without affecting function in the short term (48-72 hours), but longer-term consequences could differ .

    • Adaptive responses: Cells may adapt to altered ASF1B levels over time, masking or changing initial effects.

  • Consideration of interacting partners:

    • Availability of histone variants: The relative abundance of H3.3 versus H3.1/H3.2 in different systems may affect ASF1B function .

    • Competition between binding partners: ASF1B interactions with RIF1, CAF-1, and HIRA appear mutually exclusive, suggesting that the predominant interaction may vary across experimental systems .

  • Technical standardization:

    • Expression level control: Standardizing the degree of ASF1B overexpression or knockdown across systems.

    • Functional readout consistency: Using multiple, consistent measures of proliferation (BrdU incorporation, [³H]-thymidine incorporation, FACS analysis) across different studies.

Product Science Overview

Introduction

ASF1B, or Anti-Silencing Function 1 Homolog B, is a member of the H3/H4 family of histone chaperone proteins. It is similar to the anti-silencing function-1 protein found in yeast. This protein plays a crucial role in the regulation of chromatin structure and function, which is essential for various cellular processes such as DNA replication, repair, and transcription.

Gene and Protein Information

The ASF1B gene is a protein-coding gene that is involved in chromatin regulation and acetylation pathways . The gene is located on chromosome 19 and is known to produce a histone chaperone protein that facilitates histone deposition, exchange, and removal during nucleosome assembly and disassembly . The ASF1B protein is also involved in the nuclear import of the histone H3-H4 dimer together with importin-4 (IPO4) .

Function

ASF1B cooperates with chromatin assembly factor 1 (CAF-1) to promote replication-dependent chromatin assembly . It specifically recognizes and binds newly synthesized histones with the monomethylation of H3 ‘Lys-9’ (H3K9me1) and diacetylation at ‘Lys-5’ and ‘Lys-12’ of H4 (H4K5K12ac) marks in the cytosol . Unlike ASF1A, ASF1B does not participate in replication-independent nucleosome deposition .

Biological Importance

ASF1B is required for gonad development and has been associated with various diseases, including Hermansky-Pudlak Syndrome 9 and Spastic Paraplegia 7, Autosomal Recessive . The protein’s role in ensuring a constant supply of histones at sites of nucleosome assembly is critical for maintaining chromatin structure and function .

Applications

ASF1B antibodies are used in various research applications, including Western Blotting (WB), ELISA, Immunohistochemistry (IHC), Immunocytochemistry (ICC), Flow Cytometry (FACS), and Neutralization (Neut) . These antibodies are essential tools for studying the protein’s function and its role in chromatin regulation.

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.