Recombinant Xenopus laevis Histone chaperone asf1a-B (asf1ab)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized fulfillment.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to settle the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50% and can serve as a guideline.
Shelf Life
Shelf life depends on several factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its inclusion.
Synonyms
asf1ab; asf1aHistone chaperone asf1a-B; Anti-silencing function protein 1 homolog A-B
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-200
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
asf1ab
Target Protein Sequence
MAKVQILNMV VLDNPCPFHN PFQFEITFEC IEDLPDDLEW KIIYVGSAES EEYDQTLDSV LVGPVPAGRH MFVFQADAPN CSLIPESDAV GVTVVLITCT YRGQEFIRVG YYVNNEYSDP ELRENPPLKA HFGQLQRNIL ASNPRVTRFH INWECTSEAK MEDIENVDPA SNTMLPPNCA PSKGLAAALN TLPENSMDCM
Uniprot No.

Target Background

Function
Histone chaperone facilitating histone deposition, exchange, and removal during nucleosome assembly and disassembly.
Database Links

KEGG: xla:380002

UniGene: Xl.57985

Protein Families
ASF1 family
Subcellular Location
Nucleus.

Q&A

What is ASF1a-B in Xenopus laevis and how does it differ from mammalian ASF1 proteins?

ASF1a-B (also called asf1ab) in Xenopus laevis is a histone chaperone that belongs to the anti-silencing function 1 (ASF1) family. In most vertebrates, including Xenopus, there are two paralogous genes of ASF1: ASF1a and ASF1b, which share approximately 71% sequence homology but differ mainly in their C-terminal sequences . While yeast has only a single ASF1 protein, the evolutionary duplication event at the ancestor of jawed vertebrates led to the divergence of ASF1a and ASF1b in vertebrates .

In Xenopus laevis specifically, ASF1a-B functions primarily in histone dynamics during embryonic development, DNA replication, transcription, and DNA repair. The protein facilitates the deposition of H3-H4 histone dimers onto chromatin in a DNA synthesis-independent manner .

What are the known functional domains of Xenopus ASF1a-B and their significance?

The ASF1 protein contains several key functional domains:

  • N-terminal core domain: Highly conserved domain that forms a β-sandwich structure responsible for binding histone H3-H4 dimers .

  • Hydrophobic groove: Located on the surface of ASF1, this region interacts with B-domains of partner proteins like HIRA and CAF-1 p60 .

  • C-terminal region: More divergent between ASF1a and ASF1b, this region plays a crucial role in specificity for different binding partners and contributes to their distinct functions .

For Xenopus ASF1a-B specifically, the protein contains regions that interact with:

  • Histone H3-H4 dimers

  • HIRA complex components

  • CAF-1 complex proteins

  • Other histone chaperones involved in chromatin assembly

These interactions allow ASF1a-B to orchestrate proper histone deposition during embryonic development in Xenopus .

What are the recommended methods for expressing and purifying recombinant Xenopus ASF1a-B?

Expression System and Purification Protocol:

  • Expression System Selection: E. coli is the preferred system for recombinant ASF1a-B expression, similar to the approach used for other Xenopus histone proteins .

  • Vector Construction:

    • Clone the Xenopus ASF1a-B coding sequence into a bacterial expression vector (pET or pGEX systems)

    • Include an affinity tag (His6 or GST) to facilitate purification

    • Consider using a TEV protease cleavage site for tag removal

  • Expression Conditions:

    • Transform into BL21(DE3) or Rosetta E. coli strains

    • Induce expression with 0.5-1 mM IPTG

    • Express at 18°C overnight to enhance solubility

  • Purification Protocol:

    • Lyse cells in buffer containing 50 mM Tris-HCl pH 7.5, 500 mM NaCl, 5% glycerol, 1 mM DTT

    • Apply to appropriate affinity column (Ni-NTA for His-tagged or Glutathione Sepharose for GST-tagged)

    • Include a size exclusion chromatography step using FPLC for >98% purity

  • Quality Control:

    • Verify purity by SDS-PAGE (expected MW ~22 kDa)

    • Confirm identity using Western blot with ASF1-specific antibodies

    • Validate functionality through histone binding assays

What functional assays are most effective for studying ASF1a-B activity in Xenopus embryonic systems?

Key Functional Assays:

  • In vitro Nucleosome Assembly Assays:

    • Mix purified recombinant ASF1a-B with H3-H4 tetramers and DNA

    • Monitor nucleosome formation using gel mobility shift assays

    • Quantify assembly efficiency using micrococcal nuclease digestion patterns

  • Chromatin Assembly in Xenopus Egg Extracts:

    • Add recombinant ASF1a-B to Xenopus egg extracts with demembranated sperm chromatin

    • Analyze histone deposition kinetics using immunofluorescence

    • Measure nucleosome density through micrococcal nuclease digestion

  • Morpholino-mediated Knockdown:

    • Microinject ASF1a-B-specific morpholino oligonucleotides into Xenopus embryos

    • Examine effects on embryonic development, particularly gastrulation and neural development

    • Monitor histone deposition using H3.3 reporter constructs

  • Histone Variant Deposition Assays:

    • Express tagged histone variants (H3.3-GFP) in embryos

    • Isolate chromatin and immunoprecipitate ASF1a-B

    • Analyze associated histones by Western blot or mass spectrometry

  • DNA Synthesis-Independent Assembly (DSIA) Assay:

    • Use aphidicolin-treated Xenopus egg extracts to block DNA synthesis

    • Add labeled histones and monitor their incorporation into sperm chromatin

    • Measure the contribution of ASF1a-B to histone deposition by immunodepletion or addition of blocking antibodies

How does ASF1a-B function during early embryonic development in Xenopus laevis?

ASF1a-B plays critical roles during early Xenopus development, particularly in the regulation of chromatin structure and embryonic gene expression:

  • Zygotic Genome Activation (ZGA):

    • ASF1a-B facilitates the transition from maternal to zygotic control of development

    • It contributes to the chromatin remodeling events preceding the mid-blastula transition (MBT)

    • ASF1a-B helps establish permissive chromatin states for early zygotic transcription

  • Gastrulation and Germ Layer Formation:

    • ASF1a-B is required for proper mesoderm formation and morphogenetic movements

    • Depletion of ASF1a-B leads to gastrulation defects and shortened trunks in Xenopus embryos

    • It regulates the expression of key developmental genes involved in axis specification

  • Neural Development:

    • ASF1a-B is essential for proper neural plate formation and neural tube closure

    • It interacts with developmental transcription factors to regulate lineage-specific gene expression

    • Loss of ASF1a-B function results in neural developmental abnormalities

  • Paternal Genome Reprogramming:

    • Unlike in Drosophila where ASF1 doesn't localize to decondensing sperm nuclei, in Xenopus (and other mammals), ASF1a-B is detected in both maternal and paternal pronuclei

    • ASF1a, but not ASF1b, is required for histone H3.3 loading into the paternal genome after fertilization

What is the differential role of ASF1a versus ASF1b in Xenopus embryonic development?

Despite their similarity, ASF1a and ASF1b exhibit distinct functions during Xenopus embryonic development:

FeatureASF1aASF1b
H3.3 DepositionRequired for H3.3 loading into paternal pronucleiNot essential for paternal H3.3 loading
Embryonic LethalityKnockdown causes severe developmental defectsKnockdown causes less severe defects
Histone ModificationsRegulates H3K56 acetylationMinimal effect on H3K56ac
Transcription FactorsInteracts with Oct4 and regulates pluripotency gene expressionLess involved in pluripotency regulation
Cell ProliferationMinimal effect on cell proliferation in early embryosCritical for cell proliferation, affects PCNA nuclear accumulation
Binding PartnersPreferentially interacts with HIRA complexPreferentially interacts with CAF-1 complex
Developmental TimingEssential throughout embryonic developmentParticularly important during rapid cleavage stages

Research has shown that in Xenopus and other vertebrates:

  • ASF1a regulates H3K56 acetylation levels and pluripotency factor expression (such as Oct4)

  • ASF1b primarily safeguards pre-implantation embryo development by regulating cell proliferation

  • ASF1a, but not ASF1b, is necessary for histone H3.3 assembly in paternal pronuclei after fertilization

  • ASF1b is more highly expressed in rapidly proliferating cells and tissues, while ASF1a expression is more constitutive

How do ASF1a and ASF1b achieve functional specificity despite high sequence similarity?

The functional specificity between ASF1a and ASF1b, despite their high sequence homology, is achieved through multiple mechanisms:

What molecular mechanisms explain the interaction of Xenopus ASF1a-B with H3-H4 dimers during chromatin assembly?

The interaction between Xenopus ASF1a-B and H3-H4 dimers involves several well-characterized molecular mechanisms:

  • Physical Binding Interface:

    • ASF1 binds a H3-H4 dimer through a conserved hydrophobic pocket that recognizes the C-terminal helix of histone H3

    • Key residues in the ASF1 binding pocket include V94, which is critical for histone binding (mutation V94R abolishes histone binding)

    • The interaction prevents H3-H4 tetramer formation, as ASF1 binds at the H3-H3' interface that would normally mediate tetramerization

  • Histone Variant Selectivity:

    • ASF1a-B can interact with both canonical H3.1/H3.2 and variant H3.3 in complex with H4

    • Studies indicate that H3.3 may have preferential interactions with ASF1a-B during certain developmental processes

    • Co-expression of histone H3.3 enhances ASF1B-mediated effects on cell proliferation, while H3.1 and H3.2 do not show the same effect

  • Handoff Mechanisms:

    • ASF1a-B functions as a "histone escort" that transfers H3-H4 dimers to downstream deposition factors

    • For DNA synthesis-independent deposition, ASF1a hands H3.3-H4 to the HIRA complex

    • For replication-coupled assembly, ASF1b transfers H3.1-H4 to the CAF-1 complex

    • These handoff events are regulated by additional protein interactions and possibly post-translational modifications

  • Chaperone Network Integration:

    • In Xenopus embryos, ASF1a-B functions within a broader network of histone chaperones

    • It works alongside other chaperones like NAP1, which primarily handles H2A-H2B dimers

    • The coordinated action of these chaperones ensures proper nucleosome assembly during embryonic development

What are the current technical challenges in studying ASF1a-B function in Xenopus embryonic systems?

Current Technical Challenges:

  • Paralogue-Specific Manipulation:

    • Difficulty in selectively targeting ASF1a versus ASF1b due to high sequence similarity

    • The need for highly specific antibodies or genetic tools to distinguish the paralogs

    • Challenges in creating paralog-specific knockouts without compensatory effects

  • Temporal Control of Depletion:

    • Complete knockout of both ASF1a and ASF1b is likely lethal

    • Current approaches using morpholinos have limitations in targeting specific developmental windows

    • Need for inducible or tissue-specific depletion systems in Xenopus

  • Monitoring Chromatin Dynamics:

    • Technical challenges in visualizing histone deposition events in live embryos

    • Limited tools for genome-wide assessment of nucleosome positioning in early embryos

    • Difficulty in distinguishing direct versus indirect effects of ASF1a-B depletion

  • Structural Studies:

    • The C-terminal regions of ASF1a and ASF1b remain unstructured in available structures

    • Challenges in obtaining co-crystal structures with full-length interaction partners

    • Limited structural information specific to Xenopus ASF1a-B

  • Embryonic Material Limitations:

    • Restricted amounts of material from early embryonic stages

    • Challenges in biochemical purification from embryos without contamination

    • Need for improved methods to isolate stage-specific chromatin complexes

How do discrepancies in ASF1a-B function between different model organisms impact our understanding of its role in Xenopus?

Research on ASF1 across different model organisms has revealed important similarities and discrepancies that affect our understanding of its function in Xenopus:

  • Species-Specific Divergence:

    • In Drosophila, ASF1 does not reside in decondensing sperm nuclei, while in Xenopus and mammals, ASF1a-B is detected in both maternal and paternal pronuclei

    • This suggests different mechanisms of paternal genome reprogramming across species

  • Developmental Role Variation:

    • In mice, ASF1a knockout causes embryonic lethality at midgestation, while ASF1b is dispensable for development

    • In Xenopus, both ASF1a and ASF1b appear necessary for proper embryonic development

    • This suggests potential differences in paralog subfunctionalization across vertebrates

  • Contextual Function Differences:

    • In yeast (with single ASF1), the protein interacts equally with HIRA and CAF-1

    • In mammals and Xenopus, ASF1a preferentially interacts with HIRA while ASF1b favors CAF-1

    • This indicates that paralog-specific functions evolved after gene duplication in vertebrates

  • Cell Type-Specific Requirements:

    • In mammalian cells, ASF1b is strongly associated with proliferation and elevated in cancer cells

    • Similar proliferation-associated expression has not been thoroughly confirmed in Xenopus

    • Understanding whether these patterns are conserved requires further research

  • Research Gap Analysis:

    • Discrepancies observed between in vitro (recombinant proteins) and in vivo studies

    • Need for integrated approaches that bridge biochemical, structural, and developmental studies

    • Importance of species-specific validation rather than assuming functional conservation

What emerging research directions might advance our understanding of ASF1a-B function in Xenopus development?

Promising Future Research Directions:

  • Genome Editing Approaches:

    • CRISPR/Cas9-mediated generation of ASF1a and ASF1b mutant Xenopus lines

    • Creation of tagged endogenous ASF1a-B for live imaging studies

    • Paralog-specific domain swapping to identify functional determinants

  • Single-Cell Multi-Omics:

    • Single-cell RNA-seq combined with ATAC-seq to map ASF1a-B effects on chromatin accessibility and transcription

    • Cell type-specific profiling of ASF1a-B occupancy during development

    • Spatial transcriptomics to visualize ASF1a-B-dependent gene expression patterns

  • Histone Variant Dynamics:

    • Investigation of ASF1a-B's role in deposition of specific histone variants during Xenopus development

    • Tracking the fate of maternal versus newly synthesized histones during early development

    • Determining how ASF1a-B contributes to the establishment of bivalent chromatin domains

  • Interactome Studies:

    • Comprehensive mapping of stage-specific ASF1a-B interactomes during Xenopus development

    • Identification of developmental transcription factors that interact with ASF1a-B

    • Characterization of potential Xenopus-specific ASF1a-B binding partners

  • Disease Models:

    • Investigation of ASF1a-B in Xenopus models of developmental disorders

    • Examination of potential links to congenital disorders of chromatin regulation

    • Exploration of ASF1a-B in regenerative processes in Xenopus

  • Comparative Evolutionary Studies:

    • Detailed comparison of ASF1a and ASF1b functions across vertebrate species

    • Examination of selection pressures on ASF1 genes during evolution

    • Reconstruction of the ancestral ASF1 function and evolutionary trajectory

How can recombinant Xenopus ASF1a-B be used to study nucleosome assembly in vitro?

Recombinant Xenopus ASF1a-B serves as a valuable tool for studying nucleosome assembly through several methodological approaches:

  • Reconstituted Nucleosome Assembly Systems:

    • Combine purified ASF1a-B with recombinant Xenopus histones H3-H4

    • Add DNA templates of interest and additional assembly factors (e.g., NAP1 for H2A-H2B deposition)

    • Monitor nucleosome formation using gel mobility shift assays, MNase digestion, or electron microscopy

  • Sequential Histone Deposition Assays:

    • Use ASF1a-B to deliver H3-H4 dimers to downstream chaperones (HIRA or CAF-1)

    • Study the handoff mechanism using fluorescently labeled histones and FRET analysis

    • Investigate how different histone variants (H3.1 vs. H3.3) are processed through this pathway

  • Histone Modification Analysis:

    • Examine how pre-existing histone modifications affect ASF1a-B binding and nucleosome assembly

    • Study the role of ASF1a-B in facilitating specific histone modifications (e.g., H3K56 acetylation)

    • Use modified recombinant histones to assess their impact on assembly kinetics

  • Chromatin Remodeling Studies:

    • Investigate how ASF1a-B-assembled nucleosomes interact with chromatin remodeling complexes

    • Compare the stability and positioning of nucleosomes assembled via ASF1a versus ASF1b

    • Assess how nucleosomes assembled with ASF1a-B respond to transcription factors and RNA polymerase

  • In Vitro Transcription Systems:

    • Use ASF1a-B to assemble nucleosomes on promoter templates

    • Study how ASF1a-B-mediated chromatin assembly affects transcriptional regulation

    • Compare with other assembly pathways to understand differential effects on gene expression

What controls and validation steps are essential when using recombinant ASF1a-B in experimental systems?

Critical Controls and Validation Steps:

  • Protein Quality Validation:

    • Verify purity (>95%) by SDS-PAGE and size exclusion chromatography

    • Confirm proper folding using circular dichroism spectroscopy

    • Validate histone binding activity through pull-down assays with recombinant H3-H4

    • Ensure the absence of contaminating nucleases or proteases

  • Activity Controls:

    • Include histone-binding deficient mutant (V94R) as a negative control

    • Compare wild-type ASF1a with ASF1b to distinguish paralog-specific effects

    • Use heat-inactivated protein as an additional negative control

    • Include recombinant yeast ASF1 as a reference for conserved functions

  • Physiological Relevance Assessment:

    • Verify that recombinant protein concentrations fall within physiological ranges in Xenopus embryos

    • Compare results from recombinant systems with immunodepletion-reconstitution experiments in egg extracts

    • Validate key findings using alternative approaches (e.g., morpholinos, dominant-negative constructs)

  • Specificity Controls:

    • Test for non-specific DNA binding using empty vector DNA or irrelevant sequences

    • Assess binding to other histone types (H2A-H2B) to confirm specificity for H3-H4

    • Include competition assays with unlabeled components to verify binding specificity

  • System-Specific Controls:

    • When using Xenopus egg extracts, include mock-depleted extracts as controls

    • For embryo injections, include appropriate injection controls (buffer, control morpholinos)

    • In cell culture experiments, compare effects in different cell types to assess context-dependence

How do recent discoveries about ASF1's role in DNA damage repair relate to its functions in Xenopus embryonic development?

Recent research has revealed important connections between ASF1's DNA damage repair functions and its developmental roles in Xenopus:

  • DNA Damage Response in Rapid Cell Cycles:

    • Xenopus embryos undergo extremely rapid cell divisions before the mid-blastula transition

    • ASF1a-B likely suppresses aberrant checkpoint activation during these rapid cycles through its interaction with histone variant H2A.X.3 (H2A.X-F in Xenopus)

    • This function may be crucial for maintaining genomic integrity during the synchronous cleavage divisions

  • RIF1-ASF1 Pathway in DNA Repair:

    • Recent studies identified a 53BP1-RIF1-ASF1 pathway in DNA damage repair

    • ASF1a is recruited to DNA damage sites by interacting with RIF1 through residues E36 and D37

    • ASF1a and ASF1b show different recruitment patterns, with ASF1a strongly dependent on 53BP1-RIF1 and ASF1b showing additional recruitment mechanisms

  • Chromatin Restoration After Repair:

    • In Xenopus egg extracts, ASF1a-B likely facilitates chromatin restoration after DNA repair

    • This function would be particularly important during early development when maintaining genomic integrity is critical

    • The HIRA-ASF1a pathway has been implicated in nucleosome assembly at sites of DNA damage

  • Developmental Timing of Repair Mechanisms:

    • DNA damage response mechanisms change dramatically at the mid-blastula transition

    • The role of ASF1a-B in DNA repair may differ between pre-MBT and post-MBT embryonic stages

    • Understanding these stage-specific functions remains an active area of investigation

  • Connection to Developmental Disorders:

    • Defects in histone chaperone function during development could lead to accumulated DNA damage

    • This might contribute to developmental abnormalities when ASF1a-B function is compromised

    • The connection between ASF1's repair functions and developmental roles requires further investigation

What controversies exist regarding the redundant versus specific functions of ASF1a and ASF1b in vertebrate development?

Several unresolved controversies and debates persist regarding ASF1a and ASF1b functions in vertebrate development:

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.