ZNHIT1 Human

Zinc Finger HIT-Type Containing 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

ZNHIT1 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 177 amino acids (1-154) and having a molecular mass of 19.9kDa. ZNHIT1 is fused to a 23 amino acid His-tag at N-terminus.

Product Specs

Introduction
Zinc Finger HIT-Type Containing 1 (ZNHIT1), a member of the ZNHIT1 family, possesses a single HIT-type zinc finger. This protein, induced by DNA damage, appears to play a role in p53-mediated apoptosis induction. ZNHIT1 interacts with MAPK11 and MAPK14 and is a component of the chromatin-remodeling SRCAP complex, which consists of at least SRCAP, DMAP1, RUVBL1, RUVBL2, ACTL6A, YEATS4, ACTR6, and ZNHIT1. Additionally, ZNHIT1 binds to NR1D2, negating its inhibitory effect on APOC3 transcription without affecting its DNA-binding activity.
Description
Recombinant human ZNHIT1, produced in E. coli, is a single, non-glycosylated polypeptide chain composed of 177 amino acids (residues 1-154). It has a molecular weight of 19.9 kDa and includes a 23 amino acid His-tag at the N-terminus.
Physical Appearance
The product is a sterile, colorless solution.
Formulation
The ZNHIT1 solution is provided at a concentration of 1 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.0), 10% glycerol, and 0.4 M Urea.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles.
Purity
The purity of the product is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
Zinc Finger HIT-Type Containing 1, ZNHIT1, ZNFN4A1, Zinc Finger Protein, Subfamily 4A (HIT Domain Containing) Member 1, Zinc Finger HIT Domain Containing 1, Putative Cyclin G1 Interacting Protein, Cyclin-G1-Binding Protein 1, Zinc Finger Protein Subfamily 4A Member 1, P18 Hamlet, CG1I, H_DJ0747G18.14, p18Hamlet, Zinc Finger HIT Domain-Containing Protein 1, Zinc Finger HIT Type 1, CGBP1.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMVEKKTS VRSQDPGQRR VLDRAARQRR INRQLEALEN DNFQDDPHAG LPQLGKRLPQ FDDDADTGKK KKKTRGDHFK LRFRKNFQAL LEEQNLSVAE GPNYLTACAG PPSRPQRPFC AVCGFPSPYT CVSCGARYCT VRCLGTHQET RCLKWTV.

Q&A

What is ZNHIT1 and what is its primary function in human cells?

ZNHIT1 functions as a critical component of the SRCAP chromatin remodeling complex. Its primary role involves facilitating the incorporation of histone variant H2A.Z into specific genomic regions, particularly at transcription start sites (TSS) of genes involved in stem cell fate determination . This process is essential for transcriptional regulation of stemness-related genes. Mechanistically, ZNHIT1 promotes the interaction between H2A.Z and its chaperone YL1 by controlling YL1 phosphorylation, enabling precise targeting of H2A.Z incorporation . This epigenetic regulation is fundamental to maintaining both intestinal and hematopoietic stem cell populations.

Which stem cell populations are most directly influenced by ZNHIT1 activity?

Research has conclusively demonstrated that ZNHIT1 plays essential roles in at least two major stem cell populations:

  • Intestinal Stem Cells (ISCs): ZNHIT1 deletion in intestinal epithelium depletes Lgr5+ stem cells and disrupts intestinal homeostasis . It incorporates H2A.Z into the TSS regions of genes involved in Lgr5+ stem cell fate determination, including Lgr5, Tgfb1, and Tgfbr2 .

  • Hematopoietic Stem Cells (HSCs): ZNHIT1 restricts HSCs in a quiescent state, thereby preserving their function . It determines chromatin accessibility at distal enhancers of HSC quiescence genes, including Pten, Fstl1, and Klf4, for sustained transcription and consequent PI3K-Akt signaling inhibition .

These findings suggest that ZNHIT1 may play broader roles in adult stem cell maintenance across multiple tissues through its epigenetic regulatory functions.

How does ZNHIT1 contribute to cellular signaling pathways?

ZNHIT1 influences several key signaling pathways essential for stem cell maintenance:

  • TGF-β signaling: In intestinal stem cells, ZNHIT1 regulates the expression of TGF-β pathway components (Tgfb1 and Tgfbr2) through H2A.Z incorporation at their promoter regions . This regulation is crucial for Lgr5+ stem cell fate determination.

  • PI3K-Akt signaling: In hematopoietic stem cells, ZNHIT1 sustains the transcription of Pten, a negative regulator of the PI3K-Akt pathway . This inhibition of PI3K-Akt signaling is essential for maintaining HSC quiescence and preventing premature exhaustion.

  • Wnt signaling: ZNHIT1 affects the Lgr5+ intestinal stem cell population, which is highly dependent on Wnt signaling . Though indirect, this relationship suggests ZNHIT1 may influence the Wnt pathway in stem cell maintenance.

These pathways collectively establish the molecular framework through which ZNHIT1 contributes to stem cell homeostasis.

What methodological approaches are most effective for studying ZNHIT1-mediated chromatin remodeling?

Research into ZNHIT1's chromatin remodeling functions requires sophisticated methodological approaches:

ApproachApplicationTechnical Considerations
Conditional knockout modelsTissue-specific ZNHIT1 deletionGeneration of Znhit1 floxed alleles with tissue-specific Cre expression
ChIP-seq analysisMapping H2A.Z incorporation patternsHigh-quality antibodies against H2A.Z and ZNHIT1
ATAC-seqAssessing chromatin accessibility changesSample preparation optimization for rare stem cell populations
RNA-seqIdentifying transcriptional consequencesIntegration with epigenomic data for mechanistic insights
Co-immunoprecipitationCharacterizing protein interactionsOptimized conditions to capture transient interactions
Phosphorylation assaysStudying YL1 modificationPhospho-specific antibodies and mass spectrometry
Organoid culture systemsFunctional validation in 3D modelsAdaptation for specific stem cell populations

These approaches should be integrated to provide comprehensive insights into ZNHIT1's molecular functions in specific cellular contexts.

How can researchers distinguish between ZNHIT1-dependent and ZNHIT1-independent H2A.Z incorporation?

Distinguishing ZNHIT1-dependent from ZNHIT1-independent H2A.Z incorporation requires multiple complementary approaches:

  • Comparative genomics: Perform H2A.Z ChIP-seq in wild-type and ZNHIT1-knockout cells to identify genomic regions that specifically lose H2A.Z enrichment upon ZNHIT1 deletion . These regions represent ZNHIT1-dependent incorporation sites.

  • Sequential ChIP (Re-ChIP): This technique can detect co-occupancy of ZNHIT1 and H2A.Z at specific genomic loci, confirming direct involvement of ZNHIT1 in H2A.Z deposition.

  • Genomic context analysis: ZNHIT1-dependent H2A.Z incorporation shows distinct patterns, often occurring at promoters of stemness-related genes . Computational analysis of these patterns can help identify signature features.

  • Functional validation: Site-directed mutagenesis of putative ZNHIT1-dependent regions followed by reporter assays can confirm the functional relevance of specific incorporation sites.

  • Analysis of parallel H2A.Z incorporation pathways: Compare with regions dependent on other H2A.Z deposition mechanisms, such as those mediated by the NuA4 HAT complex components like KAT5 .

These approaches collectively enable precise mapping of ZNHIT1's contribution to the genomic H2A.Z landscape.

What is the molecular mechanism by which ZNHIT1 controls YL1 phosphorylation?

The molecular mechanism of ZNHIT1-mediated control of YL1 phosphorylation remains an active area of investigation, but current evidence suggests a multi-step process:

  • Direct interaction: ZNHIT1 physically interacts with YL1 (H2A.Z chaperone) within the SRCAP complex .

  • Regulation of kinase/phosphatase recruitment: ZNHIT1 likely recruits specific kinases or prevents phosphatase access to YL1, though the exact enzymes involved require further characterization.

  • Conformational changes: ZNHIT1 binding may induce conformational changes in YL1 that alter its susceptibility to phosphorylation at specific residues.

  • Functional consequences: This phosphorylation modification enhances YL1's affinity for H2A.Z, promoting efficient histone variant deposition at target loci .

  • Context-specific regulation: The phosphorylation status of YL1 may be differentially regulated in distinct cell types, contributing to tissue-specific functions of ZNHIT1.

Elucidating this mechanism requires advanced biochemical approaches including phospho-specific antibodies, mass spectrometry-based phosphoproteomic analysis, and structural studies of the ZNHIT1-YL1 interaction interface.

What are the most effective genetic tools for manipulating ZNHIT1 expression in human experimental systems?

For manipulating ZNHIT1 expression in human experimental systems, researchers should consider these approaches:

ApproachAdvantagesLimitationsBest Applications
CRISPR-Cas9 knockoutComplete loss-of-functionPotential compensatory mechanismsMechanistic studies requiring full ZNHIT1 elimination
Conditional CRISPR systemsTemporal control of gene deletionTechnical complexityStudying dynamic processes and avoiding developmental effects
Domain-specific mutationsTargeting specific functions while preserving othersRequires detailed structural knowledgeDissecting multifunctional roles of ZNHIT1
shRNA knockdownTunable reduction in expressionIncomplete silencing, off-targetsDose-dependent studies, situations where complete loss is lethal
Overexpression systemsGain-of-function analysisNon-physiological levelsRescue experiments, structure-function studies
Tagged ZNHIT1 variantsFacilitates tracking and purificationTag may interfere with functionInteraction studies, chromatin occupancy analysis

The optimal approach depends on the specific research question, with CRISPR-based methods generally providing the most definitive results for loss-of-function studies. For studying ZNHIT1 in stem cell contexts, inducible systems are particularly valuable due to the potential developmental consequences of constitutive deletion.

How should researchers design experiments to study ZNHIT1's role in stress response?

Designing experiments to study ZNHIT1's role in stress response requires careful consideration of multiple variables:

  • Stress model selection:

    • Replicative stress: Serial transplantation of HSCs, extended passaging

    • Oxidative stress: H₂O₂ treatment, hypoxia/reoxygenation

    • Inflammatory stress: Cytokine treatment, LPS exposure

    • DNA damage: Radiation, chemotherapeutic agents

  • Experimental controls:

    • Matched wild-type vs. ZNHIT1-knockout cells

    • Rescue conditions with ZNHIT1 re-expression

    • Dose-response and time-course analyses

  • Readouts:

    • Chromatin accessibility (ATAC-seq)

    • H2A.Z incorporation patterns (ChIP-seq)

    • Transcriptional changes (RNA-seq)

    • Functional assays (viability, proliferation, differentiation)

    • Stem cell maintenance markers

  • Single-cell approaches:

    • scRNA-seq to capture heterogeneous responses

    • Lineage tracing to follow cell fate decisions

  • In vivo validation:

    • Tissue-specific conditional knockout models

    • Stress challenge protocols (irradiation, inflammation)

    • Regeneration assays

These experimental designs should account for both acute and chronic stress responses, as ZNHIT1's role may differ between immediate adaptation and long-term resilience mechanisms, particularly in stem cell populations .

What technical challenges exist in analyzing ZNHIT1-associated chromatin complexes?

Analyzing ZNHIT1-associated chromatin complexes presents several technical challenges:

  • Complex isolation difficulties:

    • Chromatin remodeling complexes are often large and dynamic

    • Maintaining complex integrity during purification requires careful optimization

    • Low abundance in certain cell types limits yield

  • Interaction characterization challenges:

    • Transient or context-dependent interactions may be missed by standard techniques

    • Distinguishing direct from indirect interactions requires specialized approaches

    • Post-translational modifications can alter interaction profiles

  • Functional reconstitution limitations:

    • In vitro assembly of complete SRCAP complex is technically demanding

    • Reconstituting H2A.Z deposition activity requires multiple components

    • Measuring exchange activity needs specialized nucleosome substrates

  • Structural analysis constraints:

    • Large, flexible complexes are challenging for crystallography

    • Cryo-EM requires optimization for chromatin-associated complexes

    • Dynamic conformational changes may be difficult to capture

  • Technological approaches to overcome challenges:

    • Chemical crosslinking coupled with mass spectrometry (XL-MS)

    • Proximity labeling methods (BioID, APEX) for in vivo interaction mapping

    • Native mass spectrometry for intact complex analysis

    • Single-molecule approaches to capture dynamic events

Researchers must typically combine multiple complementary techniques to fully characterize ZNHIT1's interactions and functions within chromatin remodeling complexes .

How does ZNHIT1 maintain the balance between quiescence and activation in stem cells?

ZNHIT1 regulates the balance between quiescence and activation in stem cells through multiple mechanisms:

  • Epigenetic regulation of quiescence genes:

    • In HSCs, ZNHIT1 maintains chromatin accessibility at distal enhancers of quiescence genes including Pten, Fstl1, and Klf4

    • This accessibility enables sustained transcription of these key regulators

  • Signaling pathway modulation:

    • ZNHIT1 inhibits PI3K-Akt signaling through Pten regulation

    • This inhibition is crucial for maintaining HSC quiescence

    • Loss of ZNHIT1 leads to increased PI3K-Akt activity and premature stem cell activation

  • Selective H2A.Z incorporation:

    • ZNHIT1 directs H2A.Z incorporation at stemness gene promoters

    • This creates a permissive chromatin environment for expression of quiescence factors

    • The balance is maintained through precise control of H2A.Z deposition at target loci

  • Tissue-specific mechanisms:

    • In intestinal stem cells, ZNHIT1 regulates TGF-β signaling components important for self-renewal vs. differentiation decisions

    • In HSCs, ZNHIT1 prevents stem cell exhaustion by restricting cell cycle entry

Loss of ZNHIT1 disrupts this balance, leading to stem cell depletion through inappropriate activation and subsequent exhaustion, as demonstrated in both intestinal and hematopoietic systems .

What is the relationship between ZNHIT1 and other chromatin remodelers in stem cell maintenance?

ZNHIT1 functions within a network of chromatin remodelers that collectively establish and maintain stem cell identity:

  • Functional interactions within complexes:

    • ZNHIT1 is a component of the SRCAP chromatin remodeling complex

    • It functionally cooperates with other subunits including ACTL6A, DMAP1, VPS72, and YEATS4

    • These interactions are essential for targeted H2A.Z incorporation

  • Parallel H2A.Z incorporation pathways:

    • The NuA4 HAT complex (including KAT5) represents an alternative H2A.Z incorporation mechanism

    • ZNHIT1 and KAT5 may target different genomic regions for H2A.Z deposition

    • Both pathways contribute to stem cell-specific gene expression programs

  • Coordinated activity with other epigenetic regulators:

    • ZNHIT1-mediated H2A.Z incorporation likely coordinates with histone modifications

    • Potential cross-talk with DNA methylation machinery

    • Sequential or cooperative action with other remodeling complexes

  • Stem cell-specific interactions:

    • The relative importance of ZNHIT1 versus other remodelers varies by stem cell type

    • In intestinal stem cells, ZNHIT1 is essential for Lgr5+ cell maintenance

    • In HSCs, ZNHIT1 plays a critical role in preserving quiescence and preventing exhaustion

Understanding these relationships requires integrated analysis of multiple chromatin remodeling pathways in specific stem cell contexts .

How do cellular stress conditions affect ZNHIT1 function in different stem cell populations?

Cellular stress conditions modulate ZNHIT1 function in stem cell populations through several mechanisms:

  • Replicative stress effects:

    • Evidence suggests ZNHIT1, like other chromatin remodelers such as EZH2, helps maintain chromatin organization during replicative stress

    • ZNHIT1 appears especially critical for preserving HSC function during stress conditions

    • Its role in preventing chromatin instability becomes more pronounced under stress

  • Oxidative stress response:

    • While direct evidence specific to ZNHIT1 is emerging, other chromatin regulators in related pathways protect stem cells from oxidative damage

    • ZNHIT1 likely contributes to maintaining proper gene expression programs during oxidative stress

  • Inflammatory conditions:

    • In intestinal stem cells, ZNHIT1's regulation of TGF-β signaling may be particularly important during inflammatory stress

    • Its role in maintaining intestinal epithelium homeostasis suggests involvement in regenerative responses

  • Molecular adaptations under stress:

    • Stress may alter ZNHIT1's interactions with the SRCAP complex

    • Target specificity of ZNHIT1-mediated H2A.Z incorporation could shift under stress

    • Post-translational modifications of ZNHIT1 might change in response to stress signals

  • Differential requirements across stem cell types:

    • HSCs show particular dependence on ZNHIT1 for maintaining quiescence during stress

    • Intestinal stem cells require ZNHIT1 for proper regenerative responses

These stress-responsive functions highlight ZNHIT1's importance in stem cell resilience and tissue homeostasis under challenging conditions .

What single-cell approaches are most informative for studying ZNHIT1 function in rare stem populations?

Single-cell approaches offer powerful tools for studying ZNHIT1 in rare stem populations:

ApproachApplicationsTechnical ConsiderationsInsights into ZNHIT1 Function
scRNA-seqTranscriptional heterogeneity, lineage trajectoriesCell isolation protocols, low RNA contentIdentifies cell subpopulations with differential ZNHIT1 dependency
scATAC-seqChromatin accessibility at single-cell resolutionNuclear isolation, sparse data challengesMaps ZNHIT1-dependent accessible regions in rare stem cells
CUT&Tag/CUT&RUNHistone modifications and TF bindingAntibody specificity, low cell input protocolsProfiles H2A.Z incorporation patterns in limited stem cell numbers
Lineage tracing with genetic reportersIn vivo fate mappingRecombinase efficiency, reporter sensitivityTracks ZNHIT1-dependent fate decisions in stem cell progeny
Live cell imaging with tagged proteinsDynamic protein localizationTag interference, photodamageVisualizes ZNHIT1 dynamics during stem cell division/differentiation
Single-cell proteomicsProtein expression patternsSensitivity limitations, method developmentCorrelates ZNHIT1 levels with stem cell protein signatures
Single-cell multi-omicsIntegrated molecular profilingTechnical complexity, computational integrationLinks ZNHIT1-dependent chromatin states to gene expression

These approaches are particularly valuable for studying ZNHIT1 in intestinal and hematopoietic stem cells, which represent rare populations within their respective tissues .

How can researchers distinguish between direct and indirect effects of ZNHIT1 manipulation?

Distinguishing direct from indirect effects of ZNHIT1 manipulation requires systematic experimental design:

  • Temporal analysis strategies:

    • Acute vs. chronic deletion studies

    • Time-course experiments after ZNHIT1 depletion

    • Inducible systems for temporal control

    • Early changes (0-24h) likely represent direct effects

  • Genomic approaches:

    • ChIP-seq to identify direct ZNHIT1 binding sites

    • Integration with H2A.Z incorporation patterns

    • Motif analysis of ZNHIT1-associated regions

    • Changes in H2A.Z localization represent primary effects

  • Molecular validation:

    • Site-directed mutagenesis of ZNHIT1 binding sites

    • Reporter assays for direct transcriptional effects

    • Rapid degradation systems (e.g., auxin-inducible degron)

    • Targeted epigenome editing of ZNHIT1-dependent loci

  • Rescue experiments:

    • Domain-specific ZNHIT1 mutants for function separation

    • Sequential restoration of downstream pathways

    • Expression of ZNHIT1-independent H2A.Z targeting systems

  • Network analysis:

    • Computational modeling of primary vs. secondary responses

    • Causality inference from time-series data

    • Identification of immediate transcriptional targets

These approaches help establish direct molecular roles of ZNHIT1 in stem cell maintenance, distinguishing them from secondary consequences of altered stem cell homeostasis .

What are the best approaches for analyzing ZNHIT1 mutations in human disease contexts?

Analyzing ZNHIT1 mutations in human disease contexts requires integrated genomic and functional approaches:

  • Mutation identification strategies:

    • Targeted sequencing of ZNHIT1 in relevant patient cohorts

    • Analysis of ZNHIT1 in existing whole-exome/genome datasets

    • Focus on hematological disorders and intestinal pathologies

    • Correlation with epigenomic signatures

  • Structural and functional classification:

    • Domain-specific mutation mapping

    • Computational prediction of functional impacts

    • Conservation analysis across species

    • Structural modeling of mutation effects on protein interactions

  • Functional validation:

    • CRISPR-based recreation of patient mutations

    • Isogenic cell line comparisons

    • Patient-derived organoid models

    • H2A.Z incorporation assays for mutant ZNHIT1

  • Disease relevance assessment:

    • Stem cell functional assays (self-renewal, differentiation)

    • Pathway analysis (PI3K-Akt, TGF-β)

    • Gene expression profiling

    • Stress response characteristics

  • Therapeutic implications:

    • Identification of synthetic lethal interactions

    • Epigenetic modulator sensitivity profiling

    • Development of mutation-specific interventions

    • Stem cell transplantation strategies for ZNHIT1-related disorders

While direct evidence for ZNHIT1 mutations in human disease is still emerging, its fundamental roles in stem cell maintenance suggest potential contributions to developmental disorders, cancer, and degenerative conditions involving stem cell dysfunction .

Product Science Overview

Introduction

Zinc Finger HIT-Type Containing 1 (ZNHIT1) is a protein encoded by the ZNHIT1 gene in humans. This protein is part of the zinc finger protein family, which is characterized by the presence of zinc finger domains that facilitate binding to DNA, RNA, or other proteins. The HIT (Histidine Triad) domain is a specific type of zinc finger domain that plays a crucial role in various cellular processes.

Gene and Protein Structure

The ZNHIT1 gene is located on chromosome 7 and is responsible for encoding the ZNHIT1 protein. The protein itself contains a HIT domain, which is essential for its function. The HIT domain is known for its ability to bind to nucleic acids and other proteins, making it a versatile component in cellular mechanisms.

Function

ZNHIT1 plays a significant role in chromatin remodeling, a process that alters the structure of chromatin to regulate gene expression. It promotes the incorporation of the histone variant H2AZ1 into the genome, which is crucial for regulating gene expression . This incorporation is mediated by the SRCAP complex, which ZNHIT1 helps to recruit to specific genomic sites.

Additionally, ZNHIT1 is involved in maintaining hematopoietic stem cell (HSC) quiescence by determining chromatin accessibility at distal enhancers of HSC quiescence genes such as PTEN, FSTL1, and KLF4 . This regulation ensures the sustained transcription of these genes and restricts PI3K-AKT signaling inhibition.

Clinical Significance

Mutations or dysregulation of the ZNHIT1 gene have been associated with certain diseases. For instance, ZNHIT1 has been linked to Spastic Paraplegia 27, an autosomal recessive disorder characterized by progressive weakness and spasticity of the lower limbs . Understanding the function and regulation of ZNHIT1 can provide insights into the mechanisms underlying such diseases and potentially lead to therapeutic interventions.

Recombinant ZNHIT1

Recombinant ZNHIT1 is a form of the protein that is produced through recombinant DNA technology. This involves inserting the ZNHIT1 gene into an expression system, such as bacteria or yeast, to produce the protein in large quantities. Recombinant ZNHIT1 is used in various research applications to study its function and interactions with other molecules.

The recombinant protein is typically purified and supplied in a solution containing buffers and stabilizers to maintain its activity. For example, a common preparation of recombinant ZNHIT1 includes 20mM Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.4M urea .

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.