Recombinant Mouse Histone H2A type 2-A (Hist2h2aa1)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Mouse Histone H2A type 2-A (Hist2h2aa1)

Recombinant Mouse Histone H2A type 2-A, also known as Hist2h2aa1, is a protein involved in the structure and function of chromatin in mice. Histones are crucial for DNA packaging into nucleosomes, which are the fundamental units of chromatin. This specific histone variant plays a role in maintaining chromatin structure and facilitating gene expression by influencing DNA accessibility to transcriptional machinery.

Structure and Function

Histones are alkaline proteins that form the core of nucleosomes, around which DNA wraps. The structure of histones includes a globular domain and a long N-terminal tail that protrudes from the nucleosome core. These tails are subject to various post-translational modifications, which can alter chromatin structure and affect gene expression.

Histone H2A type 2-A is part of the H2A family, which includes several variants with distinct functions. These variants can influence chromatin dynamics, DNA repair, and gene regulation. For example, the H2AX variant is involved in DNA damage response, while other variants like H2A.Z and macroH2A are associated with transcriptional regulation and chromatin remodeling.

Research Findings

Research on histone variants has shown their importance in various biological processes:

  • Expression Patterns: Studies on mouse histone H2A genes have demonstrated that these genes are expressed in a cell cycle-dependent manner, with replication-dependent genes showing peak expression during the S-phase of the cell cycle .

  • Chromatin Modifications: Histone modifications, such as acetylation and methylation, play critical roles in regulating gene expression. For instance, histone H3 K9 acetylation is associated with active gene expression .

  • Biological Roles: Histone variants are involved in maintaining genome stability, regulating gene expression, and responding to DNA damage. For example, H2AX is phosphorylated in response to DNA double-strand breaks, marking damaged regions for repair .

Recombinant Production

Recombinant histones are produced using bacterial or insect cell expression systems. This involves cloning the histone gene into an expression vector, followed by transformation into a host cell line. The recombinant protein is then purified for use in biochemical assays or structural studies.

Applications in Research

Recombinant histones are used in various research applications:

  • Chromatin Assembly: Recombinant histones can be used to assemble nucleosomes in vitro, allowing researchers to study chromatin structure and dynamics.

  • Epigenetic Studies: Histone modifications can be studied using recombinant histones, providing insights into epigenetic regulation of gene expression.

  • Protein Interactions: Recombinant histones can be used to investigate interactions with other chromatin-associated proteins.

Data Tables

While specific data tables for Recombinant Mouse Histone H2A type 2-A (Hist2h2aa1) are not readily available, general information on histone structure and function can be summarized as follows:

Histone TypeFunctionModification
H2AChromatin structure, DNA repairPhosphorylation, ubiquitination
H2BChromatin dynamics, transcriptionAcetylation, ubiquitination
H3Chromatin structure, gene regulationMethylation, acetylation
H4Chromatin compaction, gene expressionAcetylation

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 preparation.
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 collect 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%, provided as a reference for your consideration.
Shelf Life
Shelf life depends on storage conditions, buffer components, 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. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type is determined during manufacturing.
The tag type is determined during the production process. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Hist2h2aa1;; Hist2h2aa2Histone H2A type 2-A; H2a-614; H2a-615; Histone H2A.2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-130
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Hist2h2aa1;
Target Protein Sequence
SGRGKQGGK ARAKAKSRSS RAGLQFPVGR VHRLLRKGNY AERVGAGAPV YMAAVLEYLT AEILELAGNA ARDNKKTRII PRHLQLAIRN DEELNKLLGK VTIAQGGVLP NIQAVLLPKK TESHHKAKGK
Uniprot No.

Target Background

Function

Histone H2A type 2-A (Hist2h2aa1): Background and Function

A core component of the nucleosome, Histone H2A type 2-A plays a critical role in DNA packaging and regulation. Nucleosomes compact DNA into chromatin, influencing the accessibility of DNA to cellular machinery involved in transcription, repair, replication, and chromosomal stability. Histone modifications, collectively known as the histone code, and nucleosome remodeling dynamically regulate DNA accessibility.

Database Links
Protein Families
Histone H2A family
Subcellular Location
Nucleus. Chromosome.

Q&A

What is the genomic context of Hist2h2aa1 in mice?

Hist2h2aa1 is one of four H2a genes located in the Hist2 cluster on mouse chromosome 3. The mouse genome contains at least 18 replication-dependent histone H2a genes distributed across three main Hist gene clusters: 13 genes in the Hist1 cluster (chromosome 13), 4 genes in the Hist2 cluster (chromosome 3), and 1 gene in the Hist3 cluster (chromosome 11) . These clusters contain genes encoding all five histone protein families (H1, H2A, H2B, H3, and H4) with Hist2h2aa1 representing one of the H2A variants within the Hist2 cluster.

What is the expression pattern of Hist2h2aa1 during the cell cycle?

As a replication-dependent histone, Hist2h2aa1 follows a characteristic expression pattern throughout the cell cycle. Expression levels increase as cells progress from G1 to S phase, peak during the middle of S-phase (2-4 hours after the beginning of S-phase), and then decrease toward the end of S-phase (6 hours) . This pattern differs significantly from replication-independent histones like H2afz, which lacks a pronounced peak during S-phase, although interestingly, H2afz expression also decreases at the end of S-phase similarly to replication-dependent genes .

What epigenetic mechanisms regulate Hist2h2aa1 expression?

Chromatin immunoprecipitation (ChIP) followed by quantitative PCR (qPCR) analyses have revealed a strong correlation between histone H3 K9 acetylation levels in promoter regions and the expression of histone genes . Specifically, genes with higher expression levels (such as Hist3h2a and H2afz) show significantly higher H3 K9 acetylation in their promoters compared to low-expression genes like Hist1h2aa .

The following table demonstrates H3 K9 acetylation levels in promoter regions of different histone genes compared to Hist2h2aa1:

Promoter RegionCycle Threshold ValuesAcetylation Difference (A-B)
Hist1h2aa promoter (low expression)No antibody (A): 26.9, 26.95
H3K9ac antibody (B): 27.61, 27.06
-0.71, -0.11
Hist3h2a promoter (high expression)No antibody (A): 27.46, 27.15
H3K9ac antibody (B): 24.14, 23.59
3.32, 3.56
H2afz promoter (highest expression)No antibody (A): 29.45, 30.58
H3K9ac antibody (B): 26.47, 26.68
2.98, 3.9
γ-satellite (heterochromatin control)No antibody (A): 8.2, 8.17
H3K9ac antibody (B): 8.81, 8.57
-0.61, -0.4

Note: Higher positive values in the difference column indicate greater H3 K9 acetylation levels .

How can researchers design experiments to investigate functional differences between Hist2h2aa1 and other H2A variants?

When investigating functional differences between histone variants, a fractional factorial experimental design approach can be valuable. This methodology allows researchers to examine multiple factors with a minimum number of experimental runs to determine significant effects .

For Hist2h2aa1 research, key experimental factors to consider include:

  • Cell cycle phase (G1, S, G2/M)

  • Cellular differentiation state

  • Treatment conditions (e.g., DNA damage, transcriptional inhibitors)

  • Genetic background (wild-type vs. knockout models)

  • Post-translational modifications

A two-level fractional factorial design would allow researchers to test these factors at high (+1) and low (-1) levels to identify significant main effects and interactions. For example, when studying Hist2h2aa1 incorporation into chromatin under different conditions, researchers could compare synchronized cells at different cell cycle phases with various treatments to determine when and how this variant is preferentially incorporated .

What techniques can detect subtle functional differences between highly similar histone variants like Hist2h2aa1 and other H2A family members?

Detecting functional differences between histone variants with high sequence similarity requires sophisticated methodological approaches:

  • Variant-specific antibodies: Develop antibodies that recognize unique epitopes in Hist2h2aa1, focusing on regions with amino acid differences compared to other variants.

  • CRISPR/Cas9-mediated tagging: Insert small epitope tags or fluorescent proteins to track specific variants without disrupting function.

  • Mass spectrometry analysis: Employ quantitative proteomic approaches with high resolution mass spectrometry to distinguish variants that differ by only a few amino acids.

  • ChIP-seq with variant-specific antibodies: Map genomic distribution patterns to identify preferential incorporation sites.

  • Protein interaction studies: Use proximity labeling techniques (BioID, APEX) to identify differential protein interactions with specific variants.

  • Single-molecule imaging: Track incorporation and dynamics of individual histone molecules in living cells to detect variant-specific behaviors.

These approaches have revealed functional differences between similar variants like H2A.Z-1 and H2A.Z-2, which differ by only 3-6 amino acids yet show distinct functional properties .

What are the critical considerations for the expression and purification of recombinant Hist2h2aa1?

Successful expression and purification of recombinant Hist2h2aa1 requires attention to several methodological details:

  • Expression system selection: While bacterial expression systems (especially E. coli) are commonly used for histone production, they lack post-translational modification machinery. Depending on your research question, consider insect cell or mammalian expression systems if post-translational modifications are crucial.

  • Purification strategy: Histones are typically purified under denaturing conditions using ion exchange chromatography followed by size exclusion chromatography. Specific considerations for Hist2h2aa1 include:

    • Initial extraction with high salt buffers (2M NaCl) to dissociate histones from DNA

    • Sequential chromatography steps using cation exchange (SP Sepharose) followed by gel filtration

    • Refolding protocols if expressed as inclusion bodies in bacterial systems

  • Quality control assessments:

    • SDS-PAGE and Western blotting to confirm purity and identity

    • Mass spectrometry to verify intact mass and detect post-translational modifications

    • Circular dichroism spectroscopy to confirm proper secondary structure

    • Functional assays such as nucleosome assembly tests to verify biological activity

  • Storage conditions: Purified Hist2h2aa1 should be stored in buffer containing 10-20% glycerol at -80°C to maintain stability and prevent aggregation.

How can researchers effectively analyze the incorporation of Hist2h2aa1 into nucleosomes?

Analyzing Hist2h2aa1 incorporation into nucleosomes requires multiple complementary approaches:

  • In vitro nucleosome reconstitution:

    • Combine purified Hist2h2aa1 with other core histones (H2B, H3, H4) in equimolar ratios

    • Add DNA with known nucleosome positioning sequences

    • Use salt dialysis method (starting from 2M NaCl and gradually reducing to physiological concentrations)

    • Verify assembly by native PAGE and sucrose gradient ultracentrifugation

  • Micrococcal nuclease (MNase) digestion assays:

    • Treat chromatin with MNase to digest linker DNA while preserving nucleosomal DNA

    • Extract and analyze DNA fragments by gel electrophoresis

    • Compare protection patterns between nucleosomes containing canonical H2A versus Hist2h2aa1

  • Biophysical characterization:

    • Use analytical ultracentrifugation to assess nucleosome stability

    • Apply FRET (Förster Resonance Energy Transfer) to measure conformational dynamics

    • Employ hydrogen-deuterium exchange mass spectrometry to identify structural differences

  • Genome-wide mapping:

    • Perform ChIP-seq with Hist2h2aa1-specific antibodies

    • Analyze enrichment patterns relative to genomic features (promoters, enhancers, etc.)

    • Compare with other H2A variants to identify unique distribution patterns

What controls should be included when studying the effects of Hist2h2aa1 on gene expression?

When investigating how Hist2h2aa1 influences gene expression, several critical controls should be incorporated:

  • Variant controls:

    • Include other H2A variants, particularly those within the same cluster (Hist2h2ab, Hist2h2ac)

    • Use canonical H2A as a baseline reference

    • Include distantly related variants (like H2A.Z) to highlight functional distinctions

  • Expression level controls:

    • Monitor endogenous expression levels of all H2A variants throughout experiments

    • Validate overexpression or depletion efficiency using qRT-PCR and Western blotting

    • Ensure that expression manipulations are within physiologically relevant ranges

  • Cell cycle synchronization:

    • Given the cell-cycle dependent expression of Hist2h2aa1, synchronize cells to specific phases

    • Include time-course analyses to account for expression pattern changes

    • Use flow cytometry to confirm synchronization efficiency

  • Complementation experiments:

    • In knockdown/knockout studies, perform rescue experiments with wild-type Hist2h2aa1

    • Include rescue with other H2A variants to test functional redundancy

    • Use chimeric constructs to identify crucial regions for specific functions

  • Technical controls:

    • Include mock transfections/transductions

    • Use scrambled siRNA/shRNA for knockdown experiments

    • Perform biological and technical replicates with appropriate statistical analyses

How does Hist2h2aa1 compare functionally to other histone H2A variants?

The functional comparison between Hist2h2aa1 and other H2A variants reveals distinct roles despite high sequence similarities:

What research questions remain unresolved regarding Hist2h2aa1 function?

Despite advances in histone variant research, several critical questions about Hist2h2aa1 remain unanswered:

  • Genomic distribution patterns:

    • Does Hist2h2aa1 associate with specific genomic regions?

    • How does its distribution change throughout the cell cycle?

    • Is it enriched at particular chromatin features (promoters, enhancers, replication origins)?

  • Protein interaction networks:

    • Which chaperones specifically recognize and deposit Hist2h2aa1?

    • Does it interact with distinct chromatin remodeling complexes?

    • Are there Hist2h2aa1-specific readers that recognize this variant?

  • Post-translational modification landscape:

    • Which modifications are specific to Hist2h2aa1 compared to other H2A variants?

    • How do these modifications influence its function?

    • Which enzymes specifically target Hist2h2aa1 for modification?

  • Functional redundancy:

    • To what extent can other H2A variants compensate for Hist2h2aa1 depletion?

    • Are there cellular contexts where Hist2h2aa1 is essential?

    • What phenotypes arise from specific Hist2h2aa1 knockout/knockdown?

  • Role in development and disease:

    • Does Hist2h2aa1 expression or function change during developmental processes?

    • Is Hist2h2aa1 dysregulation associated with specific pathological conditions?

    • Could targeting Hist2h2aa1 function have therapeutic potential?

How can advanced technologies enhance our understanding of Hist2h2aa1 function?

Emerging technologies offer new opportunities to investigate Hist2h2aa1 functions:

  • Single-cell technologies:

    • Single-cell RNA-seq can reveal cell-type-specific expression patterns of Hist2h2aa1

    • Single-cell ATAC-seq combined with Hist2h2aa1 ChIP-seq can identify associations with chromatin accessibility

    • Single-cell proteomics may detect cell-specific post-translational modifications

  • Genome editing technologies:

    • CRISPR/Cas9-mediated precise editing to introduce specific mutations or tags

    • CUT&RUN or CUT&Tag methods for more sensitive chromatin profiling

    • CRISPR activation/interference to modulate Hist2h2aa1 expression

  • Structural biology approaches:

    • Cryo-EM to visualize Hist2h2aa1-containing nucleosomes at near-atomic resolution

    • Hydrogen-deuterium exchange mass spectrometry to map dynamic regions

    • X-ray crystallography to determine atomic-level structural differences

  • In vivo imaging:

    • Live-cell imaging of fluorescently tagged Hist2h2aa1 to track dynamics

    • Super-resolution microscopy to visualize chromatin organization

    • FRAP (Fluorescence Recovery After Photobleaching) to measure exchange kinetics

  • Systems biology integration:

    • Multi-omics approaches combining genomics, transcriptomics, and proteomics

    • Network analysis to position Hist2h2aa1 in broader cellular pathways

    • Mathematical modeling to predict functional outcomes of Hist2h2aa1 perturbations

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.