HID1 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of HID1 Antibody

The HID1 Antibody (OTI2F4) is a mouse-derived monoclonal IgG1κ antibody targeting human HID1 protein. Key characteristics include:

PropertyDetails
CloneOTI2F4
Host SpeciesMouse
ApplicationsWestern Blot (WB), Immunocytochemistry (ICC/IF), Immunoprecipitation (IP)
Target GeneHID1 (Entrez Gene ID: 283987 in humans)
UniProt IDQ8IV36 (Human)
Protein AliasesDown-regulated in multiple cancers 1, DMC1, C17orf28
ValidationVerified reactivity with human, rat, and dog HID1 homologs

This antibody is widely used to investigate HID1's role in vesicle trafficking, endocrine secretion, and pathological conditions like diabetes or cancer .

Role in Secretory Granule Maturation

  • Mechanistic Insight: HID1 deficiency in pancreatic β cells disrupts homotypic fusion of immature secretory granules (ISGs), leading to defective proinsulin processing and glucose intolerance in mice. HID1 knockout (KO) models showed a 72% reduction in insulin secretion and a 3-fold increase in serum proinsulin/insulin ratio .

  • Subcellular Localization: Immunofluorescence studies using HID1 Antibody revealed that HID1 localizes to the TGN and ISGs in β cells. Its loss caused proinsulin dispersion throughout the cytosol (density increased from 0.58 ± 0.10/µm³ to 0.99 ± 0.07/µm³) .

Clinical Implications

  • Cancer Research: HID1 is downregulated in multiple cancers, and the antibody has been employed to study its tumor-suppressive roles. Reduced HID1 expression correlates with poor prognosis in pancreatic and breast cancers .

  • Neurological Disorders: Biallelic HID1 mutations cause syndromic infantile encephalopathy with hypopituitarism. Patient fibroblasts showed impaired extracellular acidification (↓30% upon KCl stimulation), linking HID1 dysfunction to TGN trafficking defects .

Key Experimental Validations

  • Western Blot: Detects HID1 at ~35 kDa in human cell lysates .

  • Immunocytochemistry: Colocalizes with TGN markers (e.g., TGN46) in pancreatic β cells and neurons, confirming its subcellular distribution .

  • Functional Assays: Used to validate HID1’s role in SG exocytosis via membrane capacitance measurements in β cells, where KO models showed normal Ca²⁺ influx but impaired insulin maturation .

Limitations and Considerations

  • Species Specificity: While reactive with human, rat, and dog homologs, cross-reactivity in non-mammalian systems (e.g., C. elegans) remains unverified .

  • Clinical Utility: Currently labeled "For Research Use Only," limiting diagnostic applications despite its relevance to endocrine disorders .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributor for specific delivery timeframes.
Synonyms
C17orf28 antibody; Chromosome 17 open reading frame 28 antibody; CQ028_HUMAN antibody; DMC1 antibody; Down regulated in multiple cancers 1 antibody; Down-regulated in multiple cancers 1 antibody; Downregulated in multiple cancer 1 antibody; FLJ43526 antibody; HID 1 antibody; HID1 antibody; HID1 domain containing antibody; HID1 domain containing protein antibody; Hypothetical protein LOC283987 antibody; Protein hid 1 homolog antibody; Protein HID1 antibody; UPF0663 transmembrane protein C17orf28 antibody
Target Names
HID1
Uniprot No.

Target Background

Function
HID-1 may play a significant role in the development of cancer across a wide range of tissues.
Gene References Into Functions
  1. Researchers propose that HID-1 influences the early stages of LDCV formation by regulating dense core formation at the trans-Golgi network. PMID: 29074564
  2. Studies have shown that mammalian HID-1 localizes to the medial and trans-Golgi apparatus, as well as the cytosol. PMID: 21337012
Database Links

HGNC: 15736

OMIM: 605752

KEGG: hsa:283987

STRING: 9606.ENSP00000413520

UniGene: Hs.11067

Protein Families
Hid-1 family
Subcellular Location
Cytoplasm. Golgi apparatus membrane; Lipid-anchor. Note=Shuttles between the cytosol and the Golgi apparatus.
Tissue Specificity
Expressed in heart, skeletal muscle, colon, spleen, kidney, liver, small intestine and lung. Highest expression is seen in brain and placenta. Loss of expression is seen in some breast, cervical, hepatocellular, lung, thyroid, gastric and renal cell-cance

Q&A

What is HID1 protein and why is it significant in scientific research?

HID1 (HID1 domain-containing protein 1) is a highly conserved protein involved in vesicle trafficking within the trans-Golgi network (TGN). It plays a crucial role in the secretory pathway, particularly in the biogenesis, maturation, and trafficking of dense core vesicles . The significance of HID1 in research stems from its essential functions in:

  • Homotypic fusion of immature secretory granules (ISGs), a critical step in secretory granule maturation

  • Processing of prohormones to yield active peptides, with implications for endocrine function

  • Potential involvement in cancer development across various tissues

  • Association with severe neurological disorders, as bi-allelic mutations in HID1 cause early infantile encephalopathy with hypopituitarism

Research on HID1 provides valuable insights into fundamental cellular processes and potential therapeutic targets for conditions involving secretory pathway dysfunction.

What are the standard methods for detecting HID1 protein expression in tissue samples?

Detection of HID1 protein expression in tissue samples typically employs immunohistochemical techniques with validated antibodies. The methodological approach includes:

  • Tissue preparation: Fixation with paraformaldehyde followed by paraffin embedding or freezing for cryosections

  • Antigen retrieval: Heat-induced epitope retrieval using citrate buffer (pH 6.0)

  • Primary antibody incubation: Using rabbit polyclonal anti-human HID1 antibody (such as Sigma HPA031406) at 1:100 dilution for 1 hour at room temperature

  • Secondary antibody application: Typically using fluorophore-conjugated secondary antibodies such as donkey anti-rabbit IgG Alexa 488 (1:500)

  • Counterstaining: Often with a Golgi marker (such as mouse monoclonal anti-58k Golgi protein antibody at 1:200) to confirm localization

  • Mounting and visualization: Using Mowiol mounting medium with n-propylgallate as fading agent, followed by confocal microscopy

For optimal results, researchers should verify antibody specificity using appropriate controls, including tissues from HID1 knockout models where available.

What cellular compartments typically show HID1 localization?

HID1 protein demonstrates specific subcellular localization that reflects its functional role in the secretory pathway. Based on immunofluorescence studies:

  • HID1 predominantly localizes to the cytosolic medial- and trans-Golgi apparatus

  • The protein requires its N-terminal myristoylation domain for binding to the trans-Golgi network (TGN)

  • In neuronal cells, HID1 resides on intracellular membranes in the cell soma

  • HID1 shows colocalization with TGN markers but not with markers of mature secretory granules

This localization pattern is consistent across different cell types, though expression levels may vary significantly between tissues. For instance, in pancreatic tissue, HID1 is highly expressed in β cells but only weakly expressed in α cells . When conducting immunofluorescence studies, researchers should employ dual labeling with established organelle markers to confirm proper localization.

How can researchers confirm the specificity of HID1 antibodies?

Confirming antibody specificity is crucial for reliable research outcomes. For HID1 antibodies, researchers should implement the following validation approaches:

  • Genetic validation: Testing the antibody in tissues/cells from HID1 knockout models, which should show absence or significant reduction of signal. For example, in conditional HID1 knockout mouse models, islets showed only 20% of the HID1 expression compared to wild-type controls

  • Peptide competition assays: Pre-incubating the antibody with a synthetic peptide containing the target epitope, which should abolish specific staining

  • Multiple antibody approach: Using different antibodies targeting distinct HID1 epitopes to confirm consistent localization patterns

  • Knockdown validation: Testing in cells with siRNA-mediated HID1 knockdown to confirm signal reduction

  • Recombinant protein controls: Using cells overexpressing tagged HID1 protein to confirm antibody detection capability

  • Western blot analysis: Confirming antibody detects a band of appropriate molecular weight (~89 kDa for human HID1)

  • Cross-reactivity assessment: Testing across multiple species when studying orthologous proteins, as HID1 sequence homology can affect antibody performance

These validation steps should be documented with appropriate controls in all experimental procedures.

How can researchers effectively study HID1's role in secretory granule maturation?

Investigating HID1's function in secretory granule maturation requires a sophisticated experimental approach that combines genetic manipulation with advanced imaging and biochemical techniques:

  • Genetic manipulation systems:

    • Conditional knockout models (e.g., RIP-Cre and floxed HID1 for β-cell specific deletion)

    • CRISPR-Cas9 genome editing for introducing patient-specific mutations

    • Inducible knockdown systems for temporal control of HID1 expression

  • Morphological analysis:

    • Large volume three-dimensional electron microscopy to quantify immature and mature secretory granules

    • Correlative light and electron microscopy (CLEM) to track specific granule populations

    • Super-resolution microscopy to detail HID1 localization at the TGN

  • Functional assays:

    • Extracellular acidification rate measurements upon stimulation (e.g., with potassium chloride)

    • Prohormone processing analysis through measurement of proinsulin/insulin ratios

    • Glucose tolerance testing to assess physiological consequences of HID1 dysfunction

  • Vesicle trafficking analysis:

    • Live-cell imaging with tagged cargo proteins to track vesicle movement

    • FRAP (Fluorescence Recovery After Photobleaching) assays to measure protein dynamics

    • Quantification of homotypic fusion events using dual-color labeled vesicles

The combination of these approaches allows for comprehensive assessment of HID1's specific role in secretory granule maturation across different cellular contexts.

What are the technical considerations when using HID1 antibodies for detecting defects in vesicle trafficking?

When using HID1 antibodies to investigate vesicle trafficking defects, researchers should account for several critical technical considerations:

  • Fixation protocol optimization:

    • Membrane proteins like HID1 may require specific fixation protocols to preserve native conformation

    • Comparison of paraformaldehyde (4%) vs. methanol fixation to determine optimal preservation of HID1 epitopes and vesicular structures

    • Limited fixation time (10-15 minutes) may better preserve the delicate TGN architecture

  • Quantitative colocalization analysis:

    • Use of appropriate statistical measures (Manders' coefficient, Pearson's correlation) when assessing HID1 colocalization with TGN markers

    • Implementation of object-based colocalization analysis for discrete vesicular structures

    • Establishment of appropriate thresholds based on control samples

  • Temporal considerations:

    • HID1 function in vesicle trafficking is dynamic, requiring time-resolved imaging approaches

    • Pulse-chase experiments may be necessary to track specific vesicle populations

    • Synchronization protocols can help identify specific trafficking steps affected by HID1 mutations

  • Antibody penetration issues:

    • TGN structures may show limited antibody accessibility in fixed cells

    • Detergent concentration optimization is crucial (typically 0.1-0.3% Triton X-100 or 0.05% saponin)

    • Permeabilization time should be carefully controlled to preserve membrane integrity

  • Controls for pathway specificity:

    • Include assessments of other trafficking pathways to confirm specificity of HID1-related defects

    • Use markers for different vesicle populations (clathrin-coated vesicles, COPI vesicles, etc.)

    • Compare with established TGN trafficking mutants to benchmark the observed phenotypes

These considerations are particularly important when studying subtle trafficking defects associated with pathogenic HID1 mutations identified in patients with syndromic infantile encephalopathy .

How can researchers effectively use HID1 antibodies in studies of disease-causing mutations?

Leveraging HID1 antibodies for investigating disease-causing mutations requires a multifaceted experimental approach:

  • Patient-derived cell models:

    • Fibroblasts from patients with HID1 mutations can be used for direct assessment of protein localization and function

    • iPSC-derived neurons or endocrine cells provide tissue-relevant contexts for mutation studies

    • CRISPR-engineered cell lines with specific mutations enable controlled comparison

  • Structural-functional analysis:

    • Co-immunoprecipitation studies with HID1 antibodies to assess how mutations affect protein-protein interactions

    • Differential centrifugation combined with immunoblotting to determine subcellular fractionation patterns

    • Limited proteolysis accessibility assays to detect mutation-induced conformational changes

  • Comparative phenotype analysis:

    • Systematic comparison of cellular phenotypes across different mutation types:

      • Missense mutations affecting protein folding

      • Truncating mutations leading to loss of functional domains

      • Mutations affecting post-translational modifications

  • Rescue experiments:

    • Re-expression of wild-type HID1 in patient cells to confirm causality

    • Structure-guided design of mutation-specific rescue approaches

    • Quantitative assessment of functional recovery using secretion assays or glucose-stimulated insulin secretion tests in β cells

  • High-content screening:

    • Development of image-based assays using HID1 antibodies to screen for small molecules that rescue trafficking defects

    • Quantification of TGN morphology, HID1 localization, and downstream functional readouts

This comprehensive approach enables researchers to establish clear genotype-phenotype correlations and develop potential therapeutic strategies for HID1-associated disorders.

What are the best practices for quantitative analysis of HID1 immunofluorescence patterns?

Quantitative analysis of HID1 immunofluorescence patterns requires rigorous methodology to ensure reproducible and meaningful results:

  • Image acquisition standardization:

    • Consistent microscope settings (laser power, detector gain, pixel size) across all experimental groups

    • Use of confocal microscopy with appropriate airy setting (typically airy 1) and signal averaging (4-8 scans) as used in published protocols

    • Z-stack acquisition to capture the full volume of the Golgi apparatus

  • Image processing workflow:

    • Background subtraction using rolling ball algorithm calibrated to cellular dimensions

    • Deconvolution where appropriate to enhance signal-to-noise ratio

    • Application of consistent thresholding methods across all analyzed images

  • Quantification parameters:

    • Golgi area and morphology (circularity, fragmentation index)

    • Fluorescence intensity distribution (coefficient of variation, intensity histogram)

    • Distance relationships between HID1 and other markers (nearest neighbor analysis)

  • Statistical considerations:

    • Determination of appropriate sample size through power analysis

    • Cell-by-cell analysis rather than image-based averaging

    • Use of histogram analysis of HID1 levels in individual cells to detect population shifts

    • Application of appropriate non-parametric tests for non-normally distributed data

  • Validation controls:

    • Inclusion of cells with known HID1 expression levels (e.g., HID1 knockout cells showing 80% reduction)

    • Technical replicates across multiple experiments

    • Biological replicates from independent samples

By adhering to these best practices, researchers can generate quantitative data suitable for detecting subtle changes in HID1 localization patterns that may be physiologically relevant.

What are common troubleshooting strategies for weak or non-specific HID1 antibody signals?

Researchers frequently encounter challenges with antibody signal quality. For HID1 antibodies, specific troubleshooting approaches include:

ProblemPossible CausesRecommended Solutions
Weak signalInsufficient antibody concentrationTitrate antibody; optimal concentration for anti-human HID1 is typically 1:100
Poor epitope accessibilityOptimize antigen retrieval; test multiple buffers (citrate, EDTA, Tris)
Protein degradationReduce time between sample collection and fixation; add protease inhibitors
Fixation-induced epitope maskingTest alternative fixation methods (PFA vs. methanol) or reduced fixation time
High backgroundExcessive antibody concentrationPerform antibody titration series to determine optimal concentration
Non-specific bindingInclude blocking step with 5% normal serum from secondary antibody species
Insufficient washingIncrease wash duration and volume; use 0.1% Tween-20 in washing buffer
AutofluorescenceInclude quenching step or use far-red fluorophores to avoid tissue autofluorescence
No colocalization with GolgiSample processing artifactsUse fresh samples with minimal processing time
Cell type-specific differencesTest in known HID1-expressing cells (e.g., pancreatic β cells)
Antibody specificity issuesValidate with recombinant HID1 protein expression

Additionally, when working with tissues showing low HID1 expression, signal amplification methods such as tyramide signal amplification may be employed, though these require careful optimization to maintain specificity.

How can researchers accurately interpret conflicting results from different HID1 antibodies?

When faced with discrepancies between results obtained using different HID1 antibodies, researchers should implement a systematic analytical approach:

  • Epitope mapping analysis:

    • Identify the specific epitopes recognized by each antibody

    • Consider whether epitopes may be differentially affected by fixation methods

    • Assess potential post-translational modifications that might mask specific epitopes

  • Cross-validation with complementary approaches:

    • Correlate protein detection results with mRNA expression data

    • Use genetic models (knockout, knockdown) to confirm specificity of each antibody

    • Implement tagged HID1 constructs to directly compare antibody performance against the tag

  • Isoform-specific considerations:

    • Review antibody documentation for specificity to different HID1 isoforms

    • Design experiments to specifically detect potential splice variants

    • Consider species-specific differences in HID1 protein sequence

  • Technical validation matrix:

    • Test all antibodies under identical conditions across multiple techniques

    • Create a validation matrix scoring each antibody on specificity criteria

    • Document batch/lot numbers as antibody performance can vary between production lots

  • Resolution strategies for conflicting data:

    • Prioritize results from antibodies with most comprehensive validation

    • Consider that different antibodies may reveal distinct aspects of HID1 biology

    • Report all findings transparently with appropriate caveats in publications

This structured approach enables researchers to make informed decisions when interpreting seemingly contradictory results and helps advance understanding of HID1 biology despite technical challenges.

What are the most effective protocols for optimizing HID1 antibody performance in co-immunoprecipitation studies?

Co-immunoprecipitation (co-IP) studies with HID1 present unique challenges due to its membrane association and involvement in protein complexes. Optimizing protocols requires attention to several critical factors:

  • Lysis buffer optimization:

    • Use buffers containing mild detergents (0.5-1% NP-40 or 0.5% digitonin)

    • Include physiological salt concentrations (150mM NaCl) to maintain relevant interactions

    • Add protease inhibitor cocktails freshly before use

    • Consider phosphatase inhibitors if studying phosphorylation-dependent interactions

  • Crosslinking considerations:

    • Reversible crosslinkers (DSP, 0.5-2mM) may help stabilize transient interactions

    • Optimize crosslinking time (typically 20-30 minutes at room temperature)

    • Ensure complete quenching before lysis (using 50mM Tris, pH 7.5)

  • Antibody coupling strategies:

    • Direct comparison of different coupling methods:

      • Protein A/G beads with antibody pre-incubation

      • Covalent coupling to activated supports (NHS-activated agarose)

      • Magnetic beads for improved recovery and reduced background

  • Pre-clearing and blocking optimizations:

    • Pre-clear lysates with beads alone to reduce non-specific binding

    • Block beads with irrelevant protein (BSA) and/or competitor DNA if using nuclear extracts

    • Use lysates from HID1-knockout cells as negative controls

  • Washing stringency gradient:

    • Test washing buffers with increasing stringency:

      • Low stringency: PBS with 0.1% detergent

      • Medium stringency: Add 150-300mM NaCl

      • High stringency: Include up to 0.1% SDS for stubborn non-specific interactions

    • Document complexes lost at each stringency level

  • Elution methods:

    • Compare different elution strategies:

      • Gentle: Native elution with excess epitope peptide

      • Denaturing: SDS sample buffer at 70°C (avoid boiling membrane proteins)

      • Acidic: Glycine buffer (pH 2.5) followed by immediate neutralization

This systematic approach allows researchers to optimize HID1 co-IP protocols for their specific experimental questions while maintaining physiologically relevant interactions.

How can researchers effectively study the relationship between HID1 and hormone processing disorders?

Investigating HID1's role in hormone processing disorders requires an integrated experimental approach that spans molecular, cellular, and physiological levels:

  • Patient cohort analysis:

    • Screening for HID1 mutations in patients with early infantile encephalopathy and hypopituitarism

    • Correlation of specific mutation types with severity of hormonal deficiencies

    • Measurement of prohormone/mature hormone ratios in patient samples

  • In vitro hormone processing assays:

    • Pulse-chase experiments with radiolabeled prohormones in cells with wild-type vs. mutant HID1

    • Mass spectrometry analysis of secreted peptides to detect processing intermediates

    • Enzyme activity assays for prohormone convertases in the presence/absence of functional HID1

  • Advanced cellular models:

    • Development of patient-derived organoids (pituitary, pancreatic) to study tissue-specific effects

    • Implementation of microfluidic systems to analyze hormone secretion dynamics

    • Single-cell transcriptomics to identify compensatory mechanisms in HID1-deficient cells

  • Physiological assessment in animal models:

    • Comprehensive hormonal profiling in conditional HID1 knockout models

    • Glucose tolerance testing revealing impaired glucose tolerance in β-cell specific HID1 knockout mice

    • Analysis of proinsulin/insulin ratios to quantify processing defects (significantly elevated in HID1-deficient models)

  • Therapeutic exploration:

    • Small molecule screening to identify compounds that rescue processing defects

    • Gene therapy approaches to restore HID1 function in affected tissues

    • Evaluation of hormone replacement therapies tailored to HID1-deficient phenotypes

This comprehensive approach enables researchers to establish clear mechanistic links between HID1 dysfunction and hormone processing disorders, potentially leading to novel diagnostic and therapeutic strategies.

What are the most effective experimental designs for studying HID1's impact on cancer development?

Given HID1's potential role in cancer development across various tissues , designing robust experiments to elucidate its mechanism requires careful consideration:

  • Clinical correlation studies:

    • Analysis of HID1 expression across tumor types using tissue microarrays

    • Correlation of expression levels with patient outcomes and clinicopathological features

    • Genomic analysis to identify HID1 mutations, copy number variations, or epigenetic silencing

  • Mechanistic investigation models:

    • Inducible HID1 knockdown/overexpression in relevant cancer cell lines

    • CRISPR-Cas9 engineering of cancer-associated HID1 mutations

    • 3D organoid cultures to study HID1's role in a more physiologically relevant context

  • Functional readouts:

    • Cell proliferation and apoptosis assays under various stress conditions

    • Migration and invasion assays to assess metastatic potential

    • Anchorage-independent growth assays (soft agar colony formation)

  • Secretome analysis:

    • Quantitative proteomics of secreted factors in HID1-modulated cells

    • Analysis of extracellular vesicle content and release dynamics

    • Assessment of growth factor processing and maturation

  • In vivo tumor models:

    • Xenograft studies comparing growth rates of tumors with modified HID1 expression

    • Genetically engineered mouse models with tissue-specific HID1 alterations

    • Analysis of tumor microenvironment interactions

  • Therapeutic vulnerability assessment:

    • Synthetic lethality screens to identify context-dependent vulnerabilities

    • Drug sensitivity profiling based on HID1 status

    • Combination approaches targeting both HID1 and compensatory pathways

These experimental designs allow for comprehensive evaluation of HID1's role in cancer development while providing insights into potential therapeutic strategies based on HID1 status.

What are the considerations for developing new HID1 antibodies with improved specificity and sensitivity?

Development of next-generation HID1 antibodies requires sophisticated approaches that leverage structural biology and advanced screening methodologies:

  • Epitope selection strategy:

    • Structural analysis of HID1 protein to identify accessible, unique epitopes

    • Bioinformatic comparison across species to identify conserved vs. divergent regions

    • Consideration of known post-translational modifications and protein interaction sites

  • Advanced immunization approaches:

    • Use of structured peptide antigens that maintain native conformation

    • DNA immunization with optimized HID1 expression constructs

    • Prime-boost strategies combining different antigen formats

  • Novel antibody discovery platforms:

    • Implementation of the Golden Gate-based dual-expression vector system for rapid antibody screening

    • Single B-cell isolation with DNA barcode antigen technology

    • Next-generation sequencing of immunoglobulin variable-region genes for high-throughput identification

  • Recombinant antibody engineering:

    • Conversion of conventional antibodies to recombinant formats for improved consistency

    • Affinity maturation through directed evolution approaches

    • Development of bispecific formats targeting HID1 plus Golgi markers for improved specificity

  • Comprehensive validation matrix:

    • Testing across multiple techniques (Western blot, immunofluorescence, immunoprecipitation)

    • Evaluation in tissues from multiple species to confirm cross-reactivity

    • Quantitative assessment of sensitivity using defined concentrations of recombinant protein

  • Format diversification:

    • Development of directly conjugated primary antibodies to eliminate secondary antibody steps

    • Creation of nanobody formats for improved penetration in tissue sections

    • Generation of proximity labeling antibody conjugates for improved detection of protein-protein interactions

By implementing these considerations, researchers can develop HID1 antibodies with substantially improved performance characteristics for diverse experimental applications.

How can researchers effectively study the role of HID1 in glucose homeostasis?

Investigation of HID1's role in glucose metabolism requires integrated approaches spanning from molecular mechanisms to whole-organism physiology:

  • Conditional knockout models:

    • Pancreatic β-cell specific deletion using RIP-Cre and floxed HID1 alleles

    • Tissue-specific knockouts in liver and muscle to assess insulin sensitivity

    • Inducible systems to distinguish developmental from acute effects

  • Physiological characterization:

    • Glucose tolerance testing (GTT) reveals significant glucose intolerance in Hid1-betaKO mice

    • Insulin tolerance testing (ITT) to assess peripheral insulin sensitivity

    • Hyperinsulinemic-euglycemic clamp studies for detailed insulin action assessment

  • Islet function analysis:

    • Ex vivo glucose-stimulated insulin secretion assays

    • Perifusion studies to assess first and second phase insulin release

    • Calcium imaging to evaluate β-cell excitability and signaling

  • Molecular mechanism investigation:

    • Quantification of mature insulin vs. proinsulin secretion ratios

    • Assessment of dense core vesicle maturation using electron microscopy

    • Analysis of homotypic fusion of immature secretory granules using live-cell imaging

  • Translational relevance:

    • Screening for HID1 variants in patients with monogenic diabetes

    • Development of biomarkers based on proinsulin/insulin ratios

    • Exploration of therapeutic approaches to enhance insulin processing

This comprehensive approach has already yielded significant insights, demonstrating that HID1 deficiency in β cells leads to glucose intolerance despite normal insulin sensitivity, likely due to defective proinsulin processing and secretory granule maturation .

What methodological approaches can effectively detect alterations in HID1-dependent vesicle trafficking?

Detecting subtle changes in vesicle trafficking dynamics requires sophisticated methodological approaches:

  • Live-cell imaging techniques:

    • Spinning disk confocal microscopy for high-speed acquisition

    • Total internal reflection fluorescence (TIRF) microscopy to visualize membrane-proximal events

    • Lattice light-sheet microscopy for extended 3D imaging with minimal phototoxicity

  • Cargo-specific tracking strategies:

    • pH-sensitive fluorescent protein fusions (pHluorin) to monitor vesicle fusion events

    • Photoactivatable/photoconvertible fluorescent proteins for pulse-chase visualization

    • Multi-color labeling to distinguish different vesicle populations

  • Quantitative analytical frameworks:

    • Automated particle tracking algorithms for vesicle movement analysis

    • Mean square displacement calculations to characterize motion types

    • Dwell time analysis at different subcellular compartments

  • Functional secretion assays:

    • Measurement of extracellular acidification rate upon stimulation with potassium chloride

    • Time-resolved analysis of cargo release using fluorescence-based reporters

    • Total internal reflection fluorescence (TIRF) microscopy to visualize individual exocytic events

  • Ultrastructural approaches:

    • Large volume three-dimensional electron microscopy to quantify vesicle populations

    • Correlative light and electron microscopy to connect dynamic events with ultrastructure

    • Immuno-electron microscopy for precise localization of HID1 and cargo proteins

  • Biochemical fractionation:

    • Isolation of different vesicle populations using density gradient centrifugation

    • Proteomic analysis of vesicle composition in normal vs. HID1-deficient cells

    • In vitro reconstitution of vesicle fusion using isolated fractions

These methodological approaches have revealed that HID1 deficiency results in accumulation of immature secretory granules due to impaired homotypic fusion , providing mechanistic insight into the functional role of HID1 in the secretory pathway.

How can HID1 antibodies be effectively utilized in studies of neurodevelopmental disorders?

Given the association of HID1 mutations with early infantile encephalopathy , HID1 antibodies can be strategically employed in neurodevelopmental disorder research:

  • Neuropathological analysis:

    • Assessment of HID1 expression patterns in developing vs. mature human brain tissues

    • Comparison of HID1 localization in patient-derived brain tissues with controls

    • Correlation of HID1 distribution with markers of synaptogenesis and neurite outgrowth

  • Cellular model systems:

    • Immunostaining in iPSC-derived neuronal cultures from patients with HID1 mutations

    • Analysis of neuronal morphology and synaptic development in relation to HID1 expression

    • Time-course studies during neuronal differentiation to identify critical developmental windows

  • Functional correlations:

    • Co-labeling with markers of neurotransmitter release machinery

    • Analysis of dense core vesicle distribution in neuronal processes

    • Assessment of activity-dependent changes in HID1 localization

  • Comparative disease studies:

    • Immunohistochemical comparison across multiple neurodevelopmental disorders

    • Assessment of HID1 expression in models of epilepsy and encephalopathy

    • Correlation with other trans-Golgi network proteins implicated in neurological disorders

  • Therapeutic monitoring:

    • Evaluation of treatment effects on HID1 localization and function

    • Development of high-content screening assays using HID1 antibodies

    • Biomarker development for stratification of patients with vesicular trafficking defects

Postmortem examination of patients with HID1 mutations has confirmed cerebral atrophy with enlarged lateral ventricles , highlighting the importance of studying HID1's role in neurodevelopment and potential therapeutic interventions for associated disorders.

What are the optimal protocols for combining HID1 immunodetection with other secretory pathway markers?

Multiparameter analysis of the secretory pathway requires careful protocol optimization for simultaneous detection of HID1 and other markers:

  • Antibody compatibility assessment:

    • Systematic testing of different fixation protocols compatible with all target epitopes

    • Evaluation of primary antibody species combinations to avoid cross-reactivity

    • Titration of antibody concentrations to achieve balanced signal intensities

  • Multiplexed staining strategies:

    • Sequential immunostaining with careful stripping between rounds

    • Tyramide signal amplification for detecting low-abundance markers

    • Use of directly labeled primary antibodies to expand multiplexing capacity

  • Recommended marker combinations:

    • TGN markers: TGN46, Golgin-97, or 58K Golgi protein (as demonstrated effective)

    • Vesicle markers: Chromogranin A, VAMP2, Secretogranin II

    • Compartment markers: GM130 (cis-Golgi), Syntaxin 6 (trans-Golgi network)

  • Imaging considerations:

    • Spectral unmixing for closely overlapping fluorophores

    • Sequential scanning to minimize bleed-through

    • Super-resolution techniques (STED, STORM) for detailed colocalization analysis

  • Quantitative colocalization workflow:

    • Channel alignment verification using multicolor beads

    • Background subtraction tailored to each channel

    • Application of appropriate colocalization algorithms and statistical analyses

A recommended starter protocol includes:

  • Fixation with 4% paraformaldehyde for 15 minutes

  • Permeabilization with 0.1% Triton X-100 for 10 minutes

  • Blocking with 5% normal serum

  • Co-incubation with rabbit polyclonal anti-human HID1 antibody (1:100) and mouse monoclonal anti-58k Golgi protein antibody (1:200)

  • Detection with species-specific secondary antibodies with minimal cross-reactivity

This approach has been successfully used to demonstrate normal colocalization of HID1 with the TGN in fibroblasts of patients with HID1 mutations compared to controls .

How can researchers integrate HID1 antibody-based approaches with functional secretion assays?

Combining immunodetection of HID1 with functional secretion assays provides powerful mechanistic insights:

  • Integrated live-cell imaging platforms:

    • Design of experimental chambers allowing antibody access after functional recordings

    • Development of fixable activity reporters for correlation with post-hoc immunostaining

    • Implementation of genetically encoded tags for live imaging followed by super-resolution microscopy

  • Correlative functional-structural analysis:

    • Recording of extracellular acidification rates in response to stimulation

    • Fixation and immunostaining of the same cells for HID1 localization

    • Computational registration to correlate functional responses with HID1 distribution patterns

  • Cell population segmentation strategies:

    • Use of microwell arrays to track individual cells through functional assays and immunostaining

    • Implementation of machine learning algorithms for phenotype classification

    • Correlation of HID1 expression levels with secretory capacity on a cell-by-cell basis

  • Temporal coordination approaches:

    • Synchronization of secretory activity prior to fixation and immunostaining

    • Time-series collection with fixation at defined points after stimulation

    • Pulse-chase cargo labeling combined with HID1 immunodetection

  • Validation in disease models:

    • Comparison of wild-type vs. HID1-mutant cells in glucose-stimulated insulin secretion assays

    • Correlation of proinsulin/insulin ratios with HID1 expression patterns

    • Rescue experiments reintroducing wild-type HID1 in knockout models

This integrated approach has revealed that HID1 deficiency results in significantly reduced extracellular acidification rates upon stimulation with potassium chloride in patient fibroblasts compared to controls , establishing a direct link between HID1 expression and secretory function.

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.