HIST1H1C (Ab-22) Antibody

Shipped with Ice Packs
In Stock

Description

Target Overview: HIST1H1C/H1.2

HIST1H1C (Histone Cluster 1 H1c) encodes histone H1.2, a member of the H1 histone family that binds linker DNA between nucleosomes to stabilize higher-order chromatin structure. Post-translational modifications (PTMs) like 2-hydroxyisobutyrylation at K22 influence its roles in:

  • Autophagy regulation (via SIRT1/HDAC1-mediated H4K16 deacetylation) .

  • Innate immune response modulation (e.g., interferon-β induction during viral infection) .

  • DNA damage repair (via ATM activation and chromatin destabilization) .

The K22 modification is implicated in diabetes complications, viral pathogenesis, and cancer .

Epigenetic Regulation of Autophagy

  • Overexpression of HIST1H1C increases autophagy markers (ATG12–ATG5, LC3B-II) and inflammation in retinal cells, mimicking diabetic retinopathy .

  • Knockdown of HIST1H1C reduces stress-induced autophagy and glucose toxicity .

  • The Ab-22 antibody enables detection of HIST1H1C’s PTM status in autophagy studies, critical for linking chromatin changes to metabolic diseases .

Antiviral Immune Response

  • HIST1H1C interacts with influenza NS2 protein to suppress IFN-β production, facilitating viral replication .

  • K34/K187 methylation mutants of HIST1H1C enhance IFN-β by freeing IRF3 to bind promoter regions .

  • The Ab-22 antibody helps map PTM-dependent interactions between HIST1H1C and viral proteins .

Technical Validation

  • Western Blot: Detects a ~37 kDa band in wild-type HeLa/A549 cells, absent in HIST1H1C-knockout lines .

  • Immunofluorescence: Localizes HIST1H1C to nuclei, with signal intensity correlating with K22 modification levels .

  • Specificity: No cross-reactivity observed with unmodified H1.2 or other H1 isoforms .

Limitations and Considerations

  • Research Use Only: Not validated for diagnostics .

  • Batch Variability: Polyclonal nature may require revalidation for quantitative studies .

  • Context-Dependent Signals: K22 modification levels vary with cellular stress (e.g., high glucose, viral infection) .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
H1 histone family member 2 antibody; H1.a antibody; H12_HUMAN antibody; H1F2 antibody; H1s-1 antibody; HIST1H1C antibody; Histone 1 H1c antibody; Histone cluster 1 H1c antibody; Histone H1.2 antibody; Histone H1c antibody; Histone H1d antibody; Histone H1s-1 antibody; MGC3992 antibody
Target Names
Uniprot No.

Target Background

Function
Histone H1 protein plays a crucial role in the formation of chromatin fibers. It binds to the linker DNA between nucleosomes, contributing to the compaction of nucleosome chains into higher-order structures. Beyond its structural role, Histone H1 also acts as a regulator of gene transcription by influencing chromatin remodeling, nucleosome spacing, and DNA methylation.
Gene References Into Functions
  1. Research indicates that a network of E2F target genes is susceptible to the regulatory influence of H1.2. H1.2 enhances the global association of pRb with chromatin, amplifies transcriptional repression mediated by pRb, and facilitates pRb-dependent cell cycle arrest. PMID: 28614707
  2. BRG1, a chromatin remodeling protein, participates in gene repression by interacting with H1.2. This interaction promotes the deposition of H1.2 and stabilizes nucleosome positioning around the transcription start site. PMID: 27390128
  3. Studies have shown that histones H1.2 and H1.4 are present in MDA-MB-231 metastatic breast cancer cells. Phosphorylation at specific residues (S173 of H1.2 and S172, S187, T18, T146, and T154 of H1.4) increases significantly during the M phase of the cell cycle, suggesting a cell cycle-dependent regulation. Additionally, the study observed a variant of H1.2 (A18V) in MCF-10A cells. PMID: 26209608
  4. Interactions between linker histone isoforms and apoptotic intermediates, particularly through their C-terminal tail, suggest a broader role for these isoforms in apoptotic cascades. PMID: 24525734
  5. Post-translational modifications at T165 in H1.2 appear to be dispensable for chromatin binding and cell proliferation. However, modifications at K26 in H1.4 are crucial for proper cell cycle progression. PMID: 24873882
  6. H1.2 interacts with Cul4A and PAF1, contributing to the activation of developmental regulatory genes. PMID: 24360965
  7. Compared to other histone H1 variants, H1.2 is less abundant at the transcription start sites of inactive genes. Promoters enriched in H1.2 exhibit distinct characteristics compared to those enriched in other variants and tend to be repressed. PMID: 24476918
  8. Mutations in linker histone genes (HIST1H1 B, C, D, and E), as well as other genes (OCT2, IRF8, and ARID1A), have been implicated in the pathogenesis of follicular lymphoma. PMID: 24435047
  9. Evidence suggests that a cascade involving p53 acetylation and H1.2 phosphorylation serves as a unique mechanism for triggering p53-dependent DNA damage response pathways. PMID: 22249259
  10. Research has confirmed N-terminal acetylation on all isoforms of histone H1, along with a single internal acetylation site. Phosphorylation sites have been identified on peptides containing the cyclin-dependent kinase (CDK) consensus motif. PMID: 15595731
  11. The binding of histone H1 to a general amyloid-like motif suggests a potential common role for histone H1 in diseases associated with amyloid-like fibrils. PMID: 16854430
  12. Histone H1.2 has been observed to translocate from the nucleus to the mitochondria after treatment with bleomycin, where it co-localizes with Bak in mitochondria. PMID: 17879944
  13. Studies indicate that the recruitment of YB1, PURalpha, and H1.2 to the p53 target gene Bax is necessary for the repression of p53-induced transcription. PMID: 18258596

Show More

Hide All

Database Links

HGNC: 4716

OMIM: 142710

KEGG: hsa:3006

STRING: 9606.ENSP00000339566

UniGene: Hs.7644

Protein Families
Histone H1/H5 family
Subcellular Location
Nucleus. Chromosome. Note=Mainly localizes in euchromatin. Distribution goes in parallel with DNA concentration.

Q&A

What is HIST1H1C and what cellular functions does it regulate?

HIST1H1C (Histone H1.2) is an important variant of the linker histone H1 family that compacts nucleosomes into higher-order chromatin fibers. It plays critical roles in genome organization and cellular stress responses . Beyond its structural role in chromatin, HIST1H1C functions as a transcriptional regulator that can interact with phosphorylated RNA polymerase II to enhance gene transcription . Recent studies have demonstrated HIST1H1C's involvement in multiple cellular processes including:

  • Autophagy regulation in retinal cells and diabetic retinopathy

  • Inflammatory responses in stressed cells

  • Cell viability and toxicity under pathological conditions

  • Chromatin organization during cellular mechanical behaviors

Research has shown that HIST1H1C upregulation correlates with disease progression in several pathological conditions, including diabetic retinopathy and cardiac fibrosis, making it a significant target for both basic and translational research .

What are the optimal fixation and permeabilization methods for HIST1H1C immunodetection?

For reliable HIST1H1C detection in immunofluorescence applications, a dual fixation-permeabilization protocol has demonstrated superior results. The recommended methodology includes:

  • Primary fixation with 4% paraformaldehyde for 20 minutes at room temperature to preserve cellular architecture and protein localization

  • Secondary permeabilization with methanol for 10 minutes at room temperature to improve antibody accessibility to nuclear proteins

  • Blocking with 5% bovine serum albumin (BSA) diluted in PBS containing 0.1% Triton X-100

This approach ensures optimal preservation of nuclear architecture while allowing sufficient antibody penetration. For specialized applications studying HIST1H1C distribution during mitosis, researchers should consider cell synchronization protocols (such as Thymidine-Nocodazole synchronization) to increase the percentage of mitotic cells in the sample, which facilitates more robust analysis of dynamic histone distribution patterns .

How can researchers validate the specificity of HIST1H1C (Ab-22) Antibody?

Validating antibody specificity is crucial for generating reliable research findings. For HIST1H1C (Ab-22) Antibody, a multi-faceted validation approach is recommended:

  • Knockdown validation: Establish stable HIST1H1C knockdown cell lines using shRNA targeting HIST1H1C. The significant reduction in both HIST1H1C mRNA and protein levels should result in diminished antibody signal .

  • Overexpression validation: Transfect cells with a plasmid expressing tagged HIST1H1C (e.g., with HA or Flag tags) and confirm co-localization of the antibody signal with the tagged protein . Successful overexpression can be confirmed by detecting the tag and an exogenous HIST1H1C band.

  • Subcellular fractionation: Perform nuclear/cytoplasmic fractionation assays to confirm that the antibody detects HIST1H1C primarily in the nuclear fraction under normal conditions, consistent with its role as a histone protein .

When interpreting validation results, researchers should be aware that HIST1H1C localization may vary between in vitro and in vivo systems, as immunohistochemical studies have demonstrated cytoplasmic staining in some cells of diabetic retinas despite predominantly nuclear localization in cultured cells .

How can the HIST1H1C (Ab-22) Antibody be employed to study autophagy regulation?

HIST1H1C has been identified as a critical regulator of autophagy, particularly in the context of diabetic retinopathy. When designing experiments to study this connection, researchers should consider a comprehensive approach:

  • Monitoring autophagy markers: Use the HIST1H1C antibody in conjunction with antibodies against key autophagy proteins including:

    • ATG12–ATG5 complex

    • ATG7

    • ATG3

    • LC3B (monitoring conversion from LC3B-I to LC3B-II)

    • SQSTM1/p62 (as a substrate of autophagy)

  • Autophagic flux assessment: To determine whether HIST1H1C affects autophagy initiation or progression, combine HIST1H1C immunodetection with autophagy flux analysis using inhibitors such as:

    • Chloroquine (CQ) at 50 μM

    • Bafilomycin A1 (BafA1) at 100 nM

  • Quantification methodology: For accurate quantification of autophagy, use confocal microscopy to analyze GFP-LC3 puncta formation. A cell containing more than 10 cytoplasmic GFP dots should be counted as an autophagic cell, with at least 200 cells analyzed per treatment .

Research has demonstrated that HIST1H1C overexpression significantly increases the percentage of autophagic cells from approximately 8% to 21%, while also promoting autophagy flux as evidenced by decreased SQSTM1 levels and enhanced LC3B-I to LC3B-II conversion .

What is the relationship between HIST1H1C and histone modifications, and how can this be studied?

HIST1H1C regulates autophagy through its influence on histone modifications, particularly H4K16 acetylation. This relationship can be studied using the following methodology:

  • Chromatin immunoprecipitation (ChIP): Use the HIST1H1C (Ab-22) Antibody alongside antibodies against modified histones (particularly H4K16ac) to assess HIST1H1C binding patterns and their correlation with specific histone modifications.

  • Mechanistic analysis: Investigate the relationship between HIST1H1C and histone deacetylases by examining:

    • SIRT1 expression and activity

    • HDAC1 expression and activity

    • H4K16 acetylation levels

Research has shown that HIST1H1C overexpression dramatically reduces the acetylation of H4K16 by upregulating both HDAC1 and SIRT1 . This mechanism represents a novel pathway through which a histone H1 variant regulates core histone modifications, ultimately affecting gene expression patterns related to autophagy.

When studying these relationships, researchers should monitor both the direct effects on histone modifications and the downstream effects on transcription of autophagy-related genes including Becn1, Atg12, Atg7, Atg5, Atg3, and Map1lc3b .

How should researchers approach HIST1H1C analysis in disease models, particularly diabetic retinopathy?

When studying HIST1H1C in disease models, particularly diabetic retinopathy, a multi-parametric approach is essential:

  • In vivo models:

    • For type 1 diabetes: Streptozotocin (STZ)-induced diabetic rats or Akita mice

    • For targeted HIST1H1C manipulation: AAV-mediated HIST1H1C overexpression or siRNA-mediated knockdown

  • Pathological parameters to monitor:

ParameterMethodologySignificance
AutophagyWestern blot for ATG proteins and LC3B-II/I ratioIndicates autophagy activity
InflammationqPCR for Il1b, Il6, Ccl2Measures inflammatory response
Glial activationGFAP immunostainingMarker of retinal stress
Neuronal lossCell counting in GCL; thickness of INL, IPL, GCLIndicates retinal degeneration
  • Validation approach:

    • Compare findings from in vitro high-glucose treated cells with in vivo diabetic retinas

    • Be aware of potential discrepancies between these systems; for example, BECN1 levels increase in diabetic retinas but remain unchanged in high-glucose treated rMC-1 cells

    • Consider differences in HIST1H1C localization between in vitro and in vivo systems

Research has demonstrated that AAV-mediated HIST1H1C overexpression in the retina induces pathological changes similar to those observed in diabetic retinopathy, including increased autophagy, inflammation, and neuronal loss . Conversely, knockdown of HIST1H1C by siRNA in diabetic mice retinas significantly attenuates diabetes-induced pathology, suggesting its potential as a therapeutic target .

How can researchers accurately assess HIST1H1C distribution during cell cycle progression?

HIST1H1C distribution patterns vary throughout the cell cycle, requiring specialized techniques for comprehensive analysis:

  • Cell synchronization: For studying HIST1H1C during mitosis, implement Thymidine-Nocodazole synchronization to increase the percentage of mitotic cells .

  • High-resolution imaging: Utilize confocal microscopy with appropriate z-stack acquisition to capture the three-dimensional distribution of HIST1H1C throughout the nucleus and during mitotic chromosome condensation.

  • Co-localization studies: Combine HIST1H1C (Ab-22) Antibody with markers for different cell cycle phases:

    • Ki-67 for proliferating cells

    • Phospho-histone H3 (Ser10) for mitotic cells

    • PCNA for S-phase cells

  • Differential extraction techniques: To distinguish between loosely and tightly bound HIST1H1C fractions, implement sequential extraction protocols with increasing salt concentrations.

When analyzing HIST1H1C distribution during mitosis, researchers should pay particular attention to potential redistribution patterns, as histone variants often exhibit dynamic localization changes during chromosome condensation and segregation .

What approaches should be used when studying HIST1H1C in mechanical stress responses and tissue fibrosis?

Recent research has revealed HIST1H1C's involvement in cellular mechanical behaviors and tissue fibrosis, particularly in cardiac pathology. When investigating these aspects:

  • Tissue expression correlation analysis: Examine the relationship between HIST1H1C expression and markers of fibroblast activation such as periostin in both animal models and human tissues .

  • Mechanical stress models:

    • In vivo: Isoproterenol (ISO) administration, which stiffens heart muscle through increased fibrosis

    • In vitro: Substrate stiffness modulation or cyclic mechanical stretching

  • Functional parameters to monitor:

ParameterTechniqueRelevance
Left ventricular massEchocardiographyIndicator of cardiac hypertrophy
E and A amplitudeEchocardiographyMeasures of diastolic function
FibrosisHistological staining (Masson's trichrome)Quantifies extracellular matrix deposition
HIST1H1C expressionImmunohistochemistry, Western blotDetermines protein levels in affected tissues

Studies have demonstrated a strong association between histone H1.0 (related to HIST1H1C) levels and metrics of heart muscle pathology, including left ventricular mass and echocardiography parameters that measure the heart's ability to relax during diastole . Similar mechanisms may be relevant for HIST1H1C, and the antibody can be used to investigate these relationships in various fibrotic conditions.

Why might there be discrepancies between in vitro and in vivo HIST1H1C localization patterns?

Researchers frequently observe differences in HIST1H1C localization patterns between cell culture systems and tissue samples. These discrepancies require careful interpretation:

  • Cell type heterogeneity: In vivo tissues contain multiple cell types with potentially different HIST1H1C expression patterns and subcellular distributions. For example, immunohistochemical studies have shown cytoplasmic HIST1H1C staining in some cells of diabetic retinas despite predominantly nuclear localization in cultured cells .

  • Microenvironmental factors: The complex tissue microenvironment in vivo includes factors absent in cell culture:

    • Three-dimensional architecture

    • Cell-cell interactions

    • Extracellular matrix components

    • Circulating factors and metabolites

  • Disease complexity: Diabetic conditions in vivo involve multiple pathological processes beyond high glucose alone, which may affect HIST1H1C localization and function .

To address these discrepancies, researchers should:

  • Compare results across multiple experimental systems

  • Use cell-type specific markers when analyzing tissues

  • Consider the limitations of each model system when interpreting results

  • When possible, validate key findings using both approaches

What controls should be included when studying HIST1H1C's role in autophagy?

When investigating HIST1H1C's role in autophagy regulation, the following controls are essential:

  • Autophagy pathway controls:

    • Positive controls: Starvation (serum deprivation) or rapamycin treatment, which are classical inducers of autophagy

    • Negative controls: Autophagy inhibitors such as chloroquine (50 μM) and bafilomycin A1 (100 nM)

  • HIST1H1C manipulation controls:

    • For overexpression: Empty vector (e.g., pCI) transfection control

    • For knockdown: Non-targeting shRNA control

    • For localization: Nuclear/cytoplasmic fractionation to confirm expected distribution

  • Autophagy flux assessment: To distinguish between increased autophagosome formation and impaired autophagosome clearance, monitor:

    • LC3B-II levels with and without lysosomal inhibitors

    • SQSTM1/p62 degradation kinetics

    • Tandem fluorescent-tagged LC3 (tfLC3) for assessing autophagosome-lysosome fusion

Research has shown that histone HIST1H1C knockdown significantly reduces both basal and stress-induced autophagy, including high glucose-induced autophagy . These controls help ensure that observed effects on autophagy are specifically attributable to HIST1H1C manipulation rather than experimental artifacts.

How can researchers optimize transfection protocols for HIST1H1C overexpression or knockdown studies?

Effective genetic manipulation of HIST1H1C requires optimized protocols tailored to the specific research context:

  • Overexpression approach:

    • Vector selection: Use a vector with appropriate tags (e.g., HA, Flag) for detection and appropriate restriction sites (e.g., NotI and EcoRI)

    • Transfection reagent: Lipofectamine LTX has shown good efficacy for HIST1H1C plasmid delivery

    • Expression verification: Confirm successful overexpression by detecting both the tag and an exogenous HIST1H1C band by Western blot at 48 hours post-transfection

  • Knockdown approach:

    • For transient knockdown: siRNA targeting HIST1H1C

    • For stable knockdown: shRNA in vectors like pSuper with puromycin selection

    • Inducible systems: Doxycycline-inducible shRNA (2.5 μg/ml for 6 days) for temporal control

    • Selection conditions: For puromycin selection, use 1 μg/ml concentration

  • Validation parameters:

    • mRNA level: qPCR for HIST1H1C transcript

    • Protein level: Western blot using the HIST1H1C (Ab-22) Antibody

    • Functional readouts: Changes in target gene expression or processes (e.g., autophagy markers)

When establishing stable knockdown cell lines, researchers should monitor cells for potential compensatory mechanisms that might emerge over extended culture periods, as these could confound experimental results.

How should researchers interpret changes in HIST1H1C expression across different disease models?

When analyzing HIST1H1C expression patterns across various disease contexts, consider these interpretation frameworks:

  • Tissue-specific expression patterns: HIST1H1C expression varies between tissues and may have context-dependent functions. For example:

    • In retinal cells: Associated with diabetic retinopathy progression

    • In cardiac tissue: Correlates with fibroblast activation and fibrosis

    • In other contexts: May have distinct roles depending on the tissue microenvironment

  • Correlation with disease markers: Assess correlations between HIST1H1C levels and established disease parameters:

Disease ContextRelevant CorrelationsInterpretation Approach
Diabetic RetinopathyAutophagy markers, inflammation, neuronal lossHigher HIST1H1C suggests more severe pathology
Cardiac FibrosisPeriostin expression, LV mass, diastolic functionHIST1H1C correlates with fibroblast activation
Other ConditionsContext-dependent markersRequires disease-specific analysis
  • Causality assessment: Distinguish between correlation and causation through intervention studies:

    • HIST1H1C overexpression should recapitulate disease phenotypes

    • HIST1H1C knockdown should ameliorate disease features if it is causally involved

Research has shown that knockdown of HIST1H1C by siRNA in diabetic mice significantly attenuated diabetes-induced autophagy, inflammation, glial activation, and neuron loss, suggesting a causal role in diabetic retinopathy pathogenesis .

What approaches should be used to analyze the genome-wide distribution of HIST1H1C?

Understanding the genomic distribution of HIST1H1C requires sophisticated analytical approaches:

  • Chromatin immunoprecipitation sequencing (ChIP-seq):

    • Use HIST1H1C (Ab-22) Antibody for immunoprecipitation

    • Include appropriate controls (input DNA, IgG control)

    • Analyze enrichment patterns relative to genomic features (promoters, enhancers, gene bodies)

  • Integration with gene expression data:

    • Correlate HIST1H1C binding patterns with RNA-seq data

    • Identify genes whose expression changes correlate with HIST1H1C occupancy

    • Focus on autophagy-related genes that show consistent regulation

  • Histone modification correlation:

    • Compare HIST1H1C distribution with histone modifications, particularly H4K16ac

    • Analyze how HIST1H1C overexpression or knockdown affects these modification patterns

Recent imaging analysis has revealed universal properties in the genomic distribution of human histone H1 variants . Researchers should consider these distribution patterns when interpreting ChIP-seq data and designing experiments to study HIST1H1C's genomic functions.

How can researchers reconcile contradictory findings about HIST1H1C function in different experimental systems?

When faced with contradictory results regarding HIST1H1C function across different experimental systems, consider these reconciliation approaches:

  • Context-dependent functions: HIST1H1C may have distinct roles depending on:

    • Cell type (e.g., neuronal cells vs. fibroblasts)

    • Physiological state (normal vs. stressed conditions)

    • Disease context (diabetes vs. other pathologies)

  • Methodological differences: Examine how methodological variations might explain discrepancies:

    • Antibody specificity and epitope accessibility

    • Fixation and permeabilization protocols

    • Detection systems and sensitivity thresholds

  • Experimental timeframes: Consider temporal aspects of HIST1H1C function:

    • Acute vs. chronic responses

    • Developmental stage-specific effects

    • Disease progression timeline

  • Potential compensatory mechanisms: In stable knockdown or knockout systems, compensatory upregulation of other histone H1 variants might mask HIST1H1C's functions .

Research has noted inconsistencies between high-glucose treated cell lines and diabetic retinas, such as increased BECN1 in diabetic retinas but unchanged levels in high-glucose treated cells . These differences likely reflect the complex nature of diabetic conditions compared to high glucose exposure alone, highlighting the importance of validating findings across multiple experimental systems.

What emerging applications of HIST1H1C (Ab-22) Antibody should researchers consider?

As HIST1H1C research evolves, several innovative applications of the HIST1H1C (Ab-22) Antibody show particular promise:

  • Single-cell analysis: Implementing the antibody in single-cell protein profiling technologies could reveal cell-type specific roles of HIST1H1C in heterogeneous tissues, particularly in disease contexts where cell populations may respond differently.

  • Live-cell imaging: Developing non-invasive labeling approaches using antibody fragments could enable dynamic tracking of HIST1H1C distribution during cellular processes like mitosis, stress responses, and autophagy induction.

  • Therapeutic target validation: Using the antibody to validate HIST1H1C as a potential therapeutic target in diseases like diabetic retinopathy, where knockdown studies have shown promising results in ameliorating pathological changes .

  • Biomarker development: Exploring HIST1H1C expression patterns across patient samples to evaluate its potential as a diagnostic or prognostic biomarker for conditions like diabetic retinopathy or cardiac fibrosis .

  • Chromatin dynamics studies: Investigating HIST1H1C's role in chromatin remodeling during cellular mechanical stress responses, which could reveal new insights into mechanotransduction pathways .

These emerging applications represent significant opportunities to expand our understanding of HIST1H1C biology and its relevance to human disease.

How might HIST1H1C (Ab-22) Antibody be used to study interactions between autophagy and epigenetic regulation?

The intersection between autophagy and epigenetic regulation represents an exciting frontier in HIST1H1C research:

  • Chromatin-autophagy crosstalk: Investigate how HIST1H1C-mediated chromatin changes affect autophagy gene accessibility and expression through:

    • Sequential ChIP (re-ChIP) to identify co-occupancy with autophagy transcription factors

    • ATAC-seq to assess chromatin accessibility at autophagy gene loci

    • Chromosome conformation capture techniques to identify long-range interactions

  • Mechanistic studies: The HIST1H1C antibody can help elucidate the molecular pathway connecting histone modifications and autophagy:

    • Co-immunoprecipitation to identify HIST1H1C-interacting proteins

    • Proximity ligation assays to visualize interactions with HDAC1 and SIRT1

    • Mass spectrometry to identify post-translational modifications on HIST1H1C that might regulate its function

  • Therapeutic modulation: Explore how compounds that affect histone acetylation (such as HDAC inhibitors) impact HIST1H1C distribution and function in autophagy regulation, which could reveal new therapeutic approaches for conditions like diabetic retinopathy .

This research direction could potentially uncover novel mechanisms by which nuclear events control cytoplasmic autophagy, bridging two fundamental cellular processes through HIST1H1C's dual role in chromatin organization and autophagy 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 2025 TheBiotek. All Rights Reserved.