HSPH1 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

HSPH1 antibodies (e.g., Proteintech 13383-1-AP, Thermo Fisher 13383-1-AP) are polyclonal antibodies raised against the HSPH1 protein, a member of the HSP70 chaperone family. The immunogen is a fusion protein corresponding to residues 508–858 of human HSPH1 (UniProt ID: Q92598) . The antibody detects both HSP105α (constitutive cytoplasmic isoform) and HSP105β (inducible nuclear isoform) .

Key Features:

PropertyDetails
TargetHSPH1 (heat shock 105kDa/110kDa protein 1)
ReactivityHuman (tested), mouse, rat (cited)
Host/IsotypeRabbit IgG
Observed Molecular Weight~110 kDa (vs. calculated 97 kDa due to post-translational modifications)
Gene ID10808 (NCBI)

2.1. Primary Applications

  • Western Blot (WB): Detects HSPH1 in lysates from MCF7, HeLa, Jurkat, and K-562 cells at dilutions of 1:500–1:2000 .

  • Immunohistochemistry (IHC): Validated in human colon, liver, and pancreatic cancer tissues (1:200–1:1200 dilution) .

  • Immunofluorescence (IF): Demonstrated in stress response studies involving HSP70 complexes .

2.2. Technical Performance

ApplicationRecommended DilutionValidated Tissues/Cells
WB1:500–1:2000MCF7, HeLa, human brain, Jurkat
IHC1:200–1:1200Colon cancer, testis, liver cancer
IFNot specifiedCo-localization with STAT3 in alveolar macrophages

Biological Roles of HSPH1

HSPH1 functions as a molecular chaperone, preventing protein aggregation under stress and regulating proteasomal degradation . Key findings include:

  • Cancer Biology: Overexpressed in melanoma, breast, thyroid, and gastrointestinal cancers, where it exerts anti-apoptotic effects . High HSPH1 levels correlate with poor prognosis in esophageal and prostate cancers .

  • Immune Modulation: Facilitates CD8+ T cell stress responses in prostate cancer, enhancing tumor suppression .

  • Thermotolerance: Cooperates with HSPA1 and DNAJB1 to maintain proteostasis during heat stress, critical for tumor survival .

4.1. Biomarker Potential

  • Identified as a biomarker for lymphoma aggressiveness .

  • Upregulated in prostate cancer extracellular vesicles (EVs), with higher mRNA/protein levels in tumors vs. adjacent tissues .

4.2. Therapeutic Targeting

  • Inhibition Studies: HSPH1 knockdown impedes esophageal tumor growth in mice . KNK437 (HSPH1 inhibitor) reduces STAT3 phosphorylation and lung injury in acute lung injury models .

  • Immune Checkpoint Blockade (ICB): HSPH1+ CD8+ T cells show reduced response to ICB, suggesting dual roles in immune regulation .

Comparative Antibody Profiles

SupplierCatalog #ApplicationsReactivityKey Distinction
Proteintech13383-1-APWB, IHC, IF, ELISAHuman, mouse, ratRecognizes both HSP105α/β isoforms
R&D SystemsAF4029WB, IHCHuman, mouse, ratValidated in rat brain (IHC)
Assay GenieCAB6622WBHumanFocus on proteostasis studies

Recent Research Highlights

  1. Prostate Cancer: HSPH1 in EVs promotes CD8+ T cell-mediated tumor suppression via IL2-MYC-IL2RA signaling .

  2. Thermotolerance: HSPH1 stabilizes denatured proteins during heat stress, enabling rapid protein synthesis recovery .

  3. Acute Lung Injury: IL-1β/KLF2/HSPH1 axis drives STAT3 activation, exacerbating inflammation .

Protocol Considerations

  • Antigen Retrieval: Use TE buffer (pH 9.0) or citrate buffer (pH 6.0) for IHC .

  • Storage: Stable at -20°C in PBS/glycerol . Avoid repeated freeze-thaw cycles .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Stored at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery time may vary depending on your location and the purchasing method. Please consult with your local distributors for specific delivery details.
Synonyms
Antigen NY CO 25 antibody; Antigen NY-CO-25 antibody; DKFZp686M05240 antibody; Heat shock 105kD alpha antibody; Heat shock 105kD antibody; Heat shock 105kD beta antibody; Heat shock 105kDa protein 1 antibody; Heat shock 105kDa protein antibody; Heat shock 105kDa/110kDa protein 1 antibody; Heat shock 110 kDa protein antibody; Heat shock 110kDa protein antibody; Heat shock protein 105 kDa antibody; HS105_HUMAN antibody; HSP105 antibody; HSP105A antibody; HSP105B antibody; HSP110 antibody; HSPH 1 antibody; Hsph1 antibody; KIAA0201 antibody; NY CO 25 antibody
Target Names
Uniprot No.

Target Background

Function
HSPH1 antibody acts as a nucleotide-exchange factor (NEF) for chaperone proteins HSPA1A and HSPA1B, facilitating the release of ADP from HSPA1A/B and subsequently triggering the release of client/substrate proteins. This function is particularly relevant in situations of severe cellular stress where ATP levels significantly decrease. By preventing the aggregation of denatured proteins, HSPH1 antibody helps maintain cellular homeostasis under stressful conditions. Additionally, it inhibits the ATPase and chaperone activities of HSPA8/HSC70.
Gene References Into Functions
  • HSPH1 expression is not an independent prognostic factor in gastric cancer patients with peritoneal metastasis. However, it may serve as a novel marker for chemosensitivity. PMID: 29204054
  • Research has identified heat shock protein 110 (HSP110) as a prognostic biomarker for colorectal carcinomas (CRCs) with microsatellite instability-high (MSI-H). Notably, the HSP110wt-low MSI-H CRCs exhibited a strong correlation with larger deletions in the HSP110 T17 mononucleotide repeat (>/=4 bp; p < 0.001). PMID: 28971530
  • In analyses employing a pentaplex panel, HSP110 HT17 alone accurately categorized samples that were previously deemed uncertain. PMID: 26831756
  • Deletions within the HSP110 T17 repeat have been observed frequently in microsatellite-unstable advanced gastric cancers. PMID: 28811251
  • Hsp105alpha localizes to the nucleus and interacts with HIF-1alpha, leading to an accumulation of HIF-1a in CoCl2-treated cells. PMID: 28185835
  • Expression of HSP110 in colon cancer contributes to STAT3-dependent tumor growth. PMID: 27819670
  • Approximately 25% of patients with stages II-III colorectal tumors with microsatellite instability exhibit an excellent response to chemotherapy. This positive response is attributed to large, biallelic deletions in the T(17) intron repeat of HSP110 in tumor DNA. PMID: 24512910
  • Depletion of HSP105 disrupts the integration of protein phosphatase 2A into the beta-catenin degradation complex, promoting the hyperphosphorylation and degradation of beta-catenin. PMID: 25645927
  • A receiver operating characteristic curve constructed using HSP105 and TIM yielded a sensitivity of 54.3% and 95% (38/40) specificity in discriminating esophageal squamous cell carcinoma from matched controls. PMID: 24157810
  • Studies have revealed a hierarchy of affinities in the binding of human Hsp72 (HSPA1A) to BAG1, BAG2, BAG3, and the unrelated NEF Hsp105. The order of affinity is BAG3 > BAG1 > Hsp105 >> BAG2. PMID: 24318877
  • HSPH1 and HSPH2 function as bona fide chaperones independently and in collaboration with DNAJA1 and DNAJB1 to hydrolyze ATP and unfold polypeptides. The combination of HSPA1A and HSPH1 forms a potent molecular machinery. PMID: 23737532
  • Elevated HSP105 expression has been associated with Barrett's esophagus. PMID: 22901192
  • HSP105 plays a crucial role in restricting the aggregation of denatured proteins, facilitating protein folding within the cytoplasm, and enhancing the expression of hsp70 in the cell nucleus. PMID: 22712230
  • The multilevel regulation of DeltaF508 CFTR folding and quality control mediated by HSP105 offers valuable insights into how chaperone machinery regulates the homeostasis and functional expression of misfolded proteins within the cell. PMID: 22505710
  • A direct correlation between HSP105 expression and lymphoma aggressiveness has been observed. PMID: 21860023
  • Hsp105alpha and Hsp105beta suppress the expanded polyQ tract-induced protein aggregation and apoptosis by inducing Hsp70. PMID: 20542028
  • Heat shock protein 105 is overexpressed in a wide range of human tumors. PMID: 14534695
  • HSP105 appears to chaperone responses to endoplasmic reticulum (ER) stress through its interactions with GRP78 and GSK3. In the absence of HSP105, cell death following ER stress occurs through a non-caspase-3-dependent mechanism. PMID: 18083346
  • Analysis of HSP105 may serve as a useful tool as a poor prognostic indicator and a diagnostic aid in challenging lesions. PMID: 18477890
  • HSP 105 is considered a more relevant tumor-associated antigen in malignant melanoma compared to HSP 70. PMID: 19476517

Show More

Hide All

Database Links

HGNC: 16969

OMIM: 610703

KEGG: hsa:10808

STRING: 9606.ENSP00000318687

UniGene: Hs.743267

Protein Families
Heat shock protein 70 family
Subcellular Location
Cytoplasm.
Tissue Specificity
Highly expressed in testis. Present at lower levels in most brain regions, except cerebellum. Overexpressed in cancer cells.

Q&A

What is HSPH1 and why is it important in biological research?

HSPH1 (Heat Shock 105kDa/110kDa Protein 1) belongs to the highly conserved family of heat shock proteins that function primarily as molecular chaperones. These proteins facilitate proper folding of other cellular proteins, prevent protein aggregation, and target improperly folded proteins for degradation . HSPH1 specifically acts as a chaperone to prevent thermal aggregation in mammalian cells, making it crucial for cellular stress response mechanisms . The protein exists in two isoforms: alpha (858 amino acids) and beta (814 amino acids), which share 94% identity but may serve distinct functions under different cellular conditions . Research interest in HSPH1 stems from its fundamental role in protein quality control and potential implications in diseases involving protein misfolding.

What are the key differences between various anti-HSPH1 antibodies available for research?

Anti-HSPH1 antibodies differ primarily in four key aspects:

  • Target epitopes: Antibodies target different regions of the HSPH1 protein, such as those surrounding Pro819, AA 713-733, C-Terminal regions, AA 549-579, AA 659-858, or the full-length protein (AA 1-858) .

  • Host and clonality: Most commercially available antibodies are rabbit polyclonal, though mouse monoclonal options like clone #531104 are also available . The choice between polyclonal and monoclonal impacts specificity and applications.

  • Species reactivity: While many antibodies react with human, mouse, and rat HSPH1, some have broader reactivity including monkey, hamster, cow, sheep, and even Saccharomyces cerevisiae . Cross-reactivity should be verified experimentally as the high conservation (e.g., 92% identity between rat and human, 97% between rat and mouse) can affect specificity .

  • Applications compatibility: Different antibodies are optimized for specific techniques including Western Blotting, Immunohistochemistry, ELISA, Immunofluorescence, and Flow Cytometry .

How should HSPH1 antibodies be stored and handled to maintain optimal activity?

HSPH1 antibodies require proper storage and handling to maintain their specificity and activity. While specific storage conditions may vary slightly between manufacturers, generally these antibodies should be stored at -20°C for long-term preservation. For working solutions, aliquoting is strongly recommended to avoid repeated freeze-thaw cycles, which can progressively degrade antibody performance through protein denaturation and aggregation . Storage buffers typically contain stabilizers like glycerol or bovine serum albumin that help maintain antibody conformation. For daily use, antibodies can be kept at 4°C for short periods (1-2 weeks), but extended storage at this temperature may lead to bacterial contamination or reduced activity. Always centrifuge antibody solutions briefly before opening to collect liquid potentially dispersed on tube walls during shipping or storage. Record dates of reconstitution and track the number of freeze-thaw cycles to maintain experimental reproducibility.

What validation methods should be used to confirm HSPH1 antibody specificity?

Rigorous validation of HSPH1 antibodies requires a multi-approach strategy to ensure experimental results are reliable and reproducible:

  • Positive control testing: Utilize cell lines known to express HSPH1, such as HT-29 human colon adenocarcinoma, NIH-3T3 mouse embryonic fibroblasts, or Nb2-11 rat lymphoma cells, which demonstrate detectable levels of HSPH1 especially after heat shock treatment .

  • Heat shock induction: Compare antibody reactivity in untreated versus heat-shocked samples (e.g., 42°C for 30 minutes followed by recovery at 37°C for 3 hours), as demonstrated in the Western blot validation data from R&D Systems, which shows increased HSPH1 expression after heat stress .

  • Molecular weight verification: Confirm detection of the appropriate band size (~110 kDa on SDS-PAGE) despite calculated molecular weights of 96.4 kDa for alpha and 92 kDa for beta isoforms .

  • Knockdown/knockout controls: Employ siRNA knockdown or CRISPR/Cas9 knockout samples as negative controls to verify antibody specificity.

  • Cross-species reactivity assessment: Test reactivity against expected species targets, noting that rat HSPH1 has 92% identity to human and 97% identity to mouse HSPH1 .

How can researchers differentiate between HSPH1 alpha and beta isoforms in experimental settings?

Differentiating between HSPH1 alpha (858 aa) and beta (814 aa) isoforms presents a significant challenge due to their 94% sequence identity and similar molecular weights . A comprehensive experimental approach includes:

  • Isoform-specific antibodies: Select antibodies targeting regions that differ between isoforms. For example, antibodies recognizing unique sequences in the 44-amino acid region present in alpha but absent in beta isoform.

  • High-resolution gel electrophoresis: Employ gradient gels (e.g., 5-8%) with extended running times to maximize separation between the alpha (apparent MW ~110 kDa) and beta (apparent MW ~105 kDa) isoforms.

  • 2D gel electrophoresis: Combine isoelectric focusing with SDS-PAGE to leverage potential pI differences between isoforms for improved separation.

  • Mass spectrometry: Perform tryptic digestion followed by LC-MS/MS to identify isoform-specific peptides, enabling precise identification and quantification of each isoform.

  • RT-PCR with isoform-specific primers: Design primers spanning the alternative splicing region to quantify mRNA expression of each isoform before protein analysis.

  • Cell-specific expression patterns: Utilize control samples with known differential expression of alpha and beta isoforms (e.g., beta is predominantly expressed under heat stress conditions in specific cell types).

What are the critical parameters for optimizing HSPH1 detection by Western blotting?

Successful detection of HSPH1 by Western blotting requires attention to several critical parameters:

  • Sample preparation: Heat shock treatment (42°C for 30 minutes followed by recovery at 37°C for 3 hours) significantly increases HSPH1 expression, providing stronger signals for detection and validation . Use reducing conditions as demonstrated in R&D Systems' validation data .

  • Protein loading: Load adequate protein (typically 20-50 μg of total protein) to detect endogenous levels of HSPH1. For heat-shocked samples, reduced amounts may be sufficient.

  • Antibody selection and concentration: Select antibodies validated for Western blotting and optimize primary antibody concentration (typically 0.5-1 μg/mL, as used in R&D Systems' protocol) . Different antibodies may require different blocking solutions and incubation conditions.

  • Gel percentage and transfer conditions: Use lower percentage gels (7-10%) for optimal resolution of the high molecular weight HSPH1 (apparent MW 105-110 kDa). Wet transfer is recommended for large proteins, with extended transfer times.

  • Detection system compatibility: Choose secondary antibodies and detection systems compatible with your primary antibody host species. HRP-conjugated secondary antibodies work effectively with chemiluminescent detection systems, as demonstrated in published protocols .

  • Buffer optimization: Use Immunoblot Buffer Group 2 as recommended by R&D Systems or equivalent buffer systems optimized for detection of high molecular weight proteins .

How can researchers troubleshoot non-specific binding or background issues with HSPH1 antibodies in immunohistochemistry?

Non-specific binding and background issues in HSPH1 immunohistochemistry can be addressed through systematic troubleshooting:

  • Antibody dilution optimization: Titrate antibody concentrations to identify the optimal dilution that maximizes specific signal while minimizing background. For example, mouse ESC immunofluorescence protocols have successfully used 1:200 dilution of HSPH1 antibody .

  • Blocking protocol enhancement: Implement robust blocking with a combination of normal serum (1% normal goat serum) and BSA (5%) as successfully demonstrated in mouse embryonic stem cell staining . Extended blocking times (1-2 hours at room temperature) may further reduce background.

  • Fixation method consideration: For HSPH1 detection, 4% paraformaldehyde freshly prepared with NaOH has proven effective, preserving epitope accessibility while maintaining cellular architecture . Compare multiple fixation methods to identify optimal conditions for your specific tissue/cell type.

  • Permeabilization optimization: Adjust permeabilization conditions (e.g., 0.3% Triton X-100 concentration and incubation time) to balance antibody access to intracellular epitopes with preservation of morphology .

  • Antigen retrieval method selection: Compare heat-induced epitope retrieval methods (citrate buffer, pH 6.0 versus EDTA buffer, pH 9.0) to enhance epitope accessibility while maintaining tissue integrity.

  • Secondary antibody controls: Include controls omitting primary antibody to identify non-specific binding of secondary antibodies or endogenous peroxidase/phosphatase activity.

What considerations are important when using HSPH1 antibodies for co-immunoprecipitation experiments?

Co-immunoprecipitation (Co-IP) with HSPH1 antibodies requires careful experimental design:

  • Antibody selection: Choose antibodies affinity-purified using epitope-specific immunogens with demonstrated specificity for endogenous HSPH1. Polyclonal antibodies often perform better in Co-IP due to recognition of multiple epitopes.

  • Lysis buffer composition: Optimize lysis conditions to preserve protein-protein interactions involving HSPH1 while effectively extracting the protein from cellular compartments. For chaperone interactions, non-ionic detergents (0.5-1% NP-40 or Triton X-100) in physiological salt concentrations are recommended.

  • Pre-clearing strategy: Implement sample pre-clearing with appropriate control IgG and protein A/G beads to minimize non-specific binding, particularly important for highly abundant heat shock proteins.

  • Cross-linking consideration: For transient or weak interactions, consider mild cross-linking (0.5-2% formaldehyde for 10 minutes) before lysis to stabilize complexes.

  • Washing stringency balance: Determine optimal washing conditions to remove non-specific interactions while preserving true HSPH1 complexes. A gradient of salt concentrations can help establish the right balance.

  • Control experiments: Include isotype-matched IgG controls, reciprocal Co-IPs (when antibodies to interaction partners are available), and validation in HSPH1-depleted samples to confirm specificity.

  • Detection strategy: For Western blot detection of co-immunoprecipitated complexes, select antibodies recognizing different epitopes than those used for immunoprecipitation to avoid heavy chain interference.

How should experiments be designed to study HSPH1 expression changes under various stress conditions?

Designing robust experiments to study HSPH1 expression under stress conditions requires:

  • Stress induction protocols: Implement multiple stress paradigms beyond heat shock, including:

    • Heat shock: 42°C for 30 minutes followed by recovery at 37°C for various timepoints (1, 3, 6, 12, 24 hours)

    • Oxidative stress: H₂O₂ treatment (0.1-1 mM for 0.5-3 hours)

    • ER stress: tunicamycin or thapsigargin treatment

    • Hypoxia: 1-5% O₂ exposure

    • Nutrient deprivation: serum starvation or glucose limitation

  • Time course design: Analyze both immediate (0-6 hours) and delayed (12-72 hours) responses to capture both direct stress responses and adaptive mechanisms.

  • Quantification methods: Employ multiple quantification approaches:

    • Western blot with densitometry for protein level quantification

    • qRT-PCR for transcript level analysis of both alpha and beta isoforms

    • Immunofluorescence for subcellular localization changes

  • Cell type considerations: Compare responses in different cell types (e.g., HT-29 human colon adenocarcinoma, NIH-3T3 mouse embryonic fibroblasts, Nb2-11 rat lymphoma) as HSPH1 regulation may vary by cellular context.

  • Controls and normalization: Include appropriate housekeeping controls (β-actin, GAPDH) stable under your chosen stress conditions. For heat shock studies, include known heat shock proteins (HSP70, HSP90) as positive controls.

  • Statistical analysis: Conduct experiments with biological triplicates and technical replicates, applying appropriate statistical tests based on data distribution and experimental design.

What are the key considerations when selecting cell lines for HSPH1 functional studies?

Selecting appropriate cell lines for HSPH1 functional studies involves several critical considerations:

  • Baseline expression profiling: Characterize baseline HSPH1 expression in candidate cell lines using Western blot and qRT-PCR to identify those with detectable but not excessive expression levels. HT-29, NIH-3T3, and Nb2-11 have demonstrated useful HSPH1 expression profiles .

  • Isoform expression ratio: Determine the alpha/beta isoform ratio in candidate cell lines, as this may influence functional outcomes of manipulation experiments.

  • Stress response characteristics: Evaluate how candidate cell lines respond to stress conditions relevant to your research question. Some cell lines exhibit robust heat shock responses (as observed in HT-29, NIH-3T3, and Nb2-11) while others may be more responsive to different stressors.

  • Transfection/transduction efficiency: Assess the amenability of candidate cell lines to your planned genetic manipulation techniques (transfection, viral transduction, CRISPR-Cas9 editing).

  • Functional assay compatibility: Ensure compatibility with downstream functional assays such as viability measurement, protein aggregation quantification, and chaperone activity assessment.

  • Species considerations: Select cell lines appropriate for your research question, noting cross-species conservation (92% identity between rat and human HSPH1; 97% between rat and mouse HSPH1) when translating findings between model systems.

  • Biological relevance: Choose cell lines relevant to the biological context of your research question (e.g., neuron-derived lines for neurodegenerative disease studies, cancer cell lines for oncology applications).

What control experiments are essential when studying HSPH1 in cell culture models?

  • Antibody validation controls:

    • Positive controls: Heat-shocked cells with upregulated HSPH1 expression

    • Negative controls: HSPH1 knockdown/knockout cells

    • Specificity controls: Pre-absorption with immunizing peptide

  • Expression manipulation controls:

    • Empty vector controls for overexpression studies

    • Non-targeting siRNA/shRNA for knockdown experiments

    • Wild-type cells alongside CRISPR-edited lines

  • Stress response controls:

    • Unstressed/untreated cells as baseline controls

    • Time-matched controls for each experimental timepoint

    • Positive control proteins (HSP70, HSP90) that respond to the same stressors

  • Subcellular localization controls:

    • Co-staining with compartment markers (e.g., DAPI for nucleus, phalloidin for actin cytoskeleton)

    • Fractionation quality controls (markers for cytoplasmic, nuclear, membrane fractions)

  • Functional assay controls:

    • Chaperone activity: Known substrate proteins

    • Protein aggregation: Positive aggregation inducers

    • Cell viability: Positive death inducers

  • Colony morphology controls (for stem cell studies):

    • Comparison between round and flattened embryonic stem cell colonies, which show differential HSPH1 expression patterns

How can researchers effectively compare HSPH1 expression across different tissue types?

Comparing HSPH1 expression across tissues requires standardized approaches to overcome technical and biological variability:

  • Tissue collection and preservation standardization:

    • Implement consistent collection protocols minimizing ischemia time

    • Standardize fixation/preservation methods (e.g., flash freezing, fixed time in formalin)

    • Document and control for relevant variables (e.g., age, sex, treatment status)

  • Multiple detection methodologies:

    • Western blot for protein level quantification

    • IHC/IF for localization and cell-type specific expression

    • qRT-PCR for transcript level analysis

    • Proteomics for unbiased quantification

  • Normalization strategy optimization:

    • Identify stable reference genes/proteins specific to each tissue type

    • Consider global normalization methods (total protein staining) to overcome housekeeping protein variability between tissues

    • Calculate relative rather than absolute expression when appropriate

  • Semi-quantitative IHC approach:

    • Implement standardized scoring systems (H-score, Allred score)

    • Use digital pathology quantification with consistent thresholding

    • Include standardized positive controls on each slide

  • Cross-reactivity assessment:

    • Verify antibody specificity in each tissue type

    • Consider species-specific optimization for multi-species studies, accounting for the known conservation rates (e.g., 92% identity between rat and human HSPH1)

  • Technical replicate planning:

    • Include biological triplicates minimum

    • Analyze multiple tissue sections/extracts per sample

    • Consider batch effects in experimental design

How should researchers interpret differences in HSPH1 isoform expression patterns between experimental conditions?

Interpreting HSPH1 isoform expression differences requires sophisticated analysis:

  • Isoform-specific quantification:

    • Distinguish between alpha (858 aa) and beta (814 aa) isoforms through high-resolution Western blotting or isoform-specific qRT-PCR

    • Consider the 94% sequence identity between isoforms when interpreting antibody-based detection methods

  • Context-dependent expression patterns:

    • Evaluate whether observed changes reflect typical stress responses (e.g., heat shock typically induces both isoforms but with different kinetics)

    • Consider cell-type specific regulatory mechanisms based on established patterns (e.g., higher expression in round versus flattened embryonic stem cell colonies)

  • Functional correlation analysis:

    • Correlate isoform ratio changes with functional outcomes (e.g., protein aggregation, cell survival)

    • Determine whether observed changes are upstream drivers or downstream consequences of the biological process under study

  • Temporal dynamics evaluation:

    • Analyze time course data to distinguish between early regulatory events and secondary adaptive responses

    • Consider differential stability of isoforms when interpreting steady-state measurements

  • Statistical robustness assessment:

    • Apply appropriate statistical tests accounting for data distribution

    • Implement correction for multiple testing when examining isoform expression across numerous conditions

  • Evolutionary conservation consideration:

    • Interpret species differences in context of known conservation rates (92% human-rat identity, 97% mouse-rat identity)

    • Evaluate whether observed regulatory patterns are conserved across species or represent species-specific mechanisms

What statistical approaches are most appropriate for analyzing HSPH1 expression data from different experimental designs?

Selecting appropriate statistical methods for HSPH1 studies depends on experimental design:

  • Before applying statistical tests:

    • Evaluate data distribution (normal vs. non-normal) using Shapiro-Wilk or Kolmogorov-Smirnov tests

    • Check for homogeneity of variance with Levene's test

    • Identify and address outliers through principled approaches

  • For comparing two groups:

    • Normally distributed data: Independent t-test (unpaired samples) or paired t-test (paired samples)

    • Non-normally distributed data: Mann-Whitney U test (unpaired) or Wilcoxon signed-rank test (paired)

  • For comparing multiple groups:

    • Normally distributed data: One-way ANOVA with appropriate post-hoc tests (Tukey for all pairwise comparisons, Dunnett for comparisons to control)

    • Non-normally distributed data: Kruskal-Wallis with Dunn's post-hoc test

    • For factorial designs: Two-way or multi-way ANOVA with interaction term analysis

  • For time course experiments:

    • Repeated measures ANOVA for normally distributed data

    • Mixed-effects models for handling missing timepoints or irregular sampling

    • Area under curve (AUC) analysis to compress temporal data for simplified comparison

  • For correlation analyses:

    • Pearson correlation for normally distributed continuous variables

    • Spearman correlation for non-normally distributed or ordinal data

    • Multiple regression for controlling confounding variables

  • For high-dimensional data:

    • Correction for multiple testing (Bonferroni, Benjamini-Hochberg)

    • Dimension reduction techniques (PCA, t-SNE) for visualizing patterns

    • Hierarchical clustering for identifying expression pattern similarities

How can researchers reconcile contradictory findings between different detection methods for HSPH1?

Reconciling contradictory HSPH1 detection results requires systematic investigation:

  • Method-specific limitations assessment:

    • Western blot: Protein extraction efficiency, antibody specificity, linear detection range

    • qRT-PCR: Primer specificity, amplification efficiency, reference gene stability

    • IHC/IF: Fixation effects, epitope accessibility, background fluorescence

    • Mass spectrometry: Peptide recovery, ionization efficiency, isoform-specific peptide detection

  • Technical vs. biological variability differentiation:

    • Replicate experiments to distinguish random variation from consistent methodological differences

    • Control for batch effects through experimental design and statistical adjustment

    • Implement standardized protocols to minimize technical variables

  • Antibody-specific considerations:

    • Compare results using antibodies targeting different epitopes on HSPH1

    • Consider epitope accessibility differences between native and denatured protein

    • Validate antibody specificity in the specific experimental context (e.g., after heat shock treatment)

  • Sample preparation impact:

    • Evaluate how sample processing affects detection (e.g., protein extraction methods)

    • Consider how stress induction protocols (42°C for 30 minutes followed by recovery) affect protein conformation or subcellular localization

  • Resolution limitations:

    • Consider whether methods differ in ability to distinguish alpha and beta isoforms (94% identity)

    • Assess whether apparent contradictions reflect different subcellular pools of HSPH1

  • Integrated data analysis approach:

    • Implement weighted analysis considering the relative strengths/limitations of each method

    • Triangulate results by adding orthogonal methods when two approaches conflict

What are the key considerations when comparing HSPH1 antibody performance across different research groups?

Comparing HSPH1 antibody performance across research groups requires careful analysis:

  • Antibody source and validation variability:

    • Determine whether identical antibody clones/catalog numbers were used or just similar target regions

    • Compare validation protocols between groups (e.g., knockout controls, specificity testing)

    • Evaluate lot-to-lot variability in antibody production that might explain discrepancies

  • Protocol differences assessment:

    • Compare key protocol parameters (antibody concentration, incubation time/temperature, blocking conditions)

    • Identify different buffer systems used (e.g., Immunoblot Buffer Group 2 as used in some Western blot protocols)

    • Examine sample preparation methods (cell lysis, tissue processing, antigen retrieval)

  • Cell/tissue variability consideration:

    • Analyze baseline HSPH1 expression differences in experimental models between groups

    • Consider passage number effects in cell lines

    • Evaluate genetic background differences in model organisms

  • Detection system comparison:

    • Assess sensitivity differences between detection methods (chemiluminescence vs. fluorescence)

    • Compare digital image acquisition and processing approaches

    • Evaluate quantification strategies (densitometry methods, normalization approaches)

  • Stress induction standardization:

    • Compare heat shock protocols (temperature, duration, recovery time)

    • Evaluate other stress induction methods that might affect HSPH1 expression or conformation

    • Consider non-standardized environmental stressors (incubator CO₂ variations, media differences)

  • Reporting standards evaluation:

    • Assess completeness of methods reporting (antibody catalog numbers, dilutions, exposure times)

    • Consider whether negative results might be underreported

    • Implement standardized reporting using established guidelines (e.g., ARRIVE guidelines for animal studies)

How should researchers interpret HSPH1 localization data from immunofluorescence studies?

Interpreting HSPH1 localization from immunofluorescence requires detailed analysis:

  • Subcellular pattern characterization:

    • Document predominant localization patterns (cytoplasmic, nuclear, membrane-associated)

    • Note whether observed patterns match expected HSPH1 localization (primarily cytoplasmic, as observed in mouse embryonic stem cells)

    • Quantify distribution patterns using intensity profiles or compartment ratio measurements

  • Co-localization analysis with organelle markers:

    • Implement robust co-localization quantification (Pearson's correlation, Manders' overlap coefficient)

    • Control for random overlap in densely packed cellular regions

    • Consider three-dimensional distribution using confocal z-stacks rather than single optical sections

  • Physiological state consideration:

    • Compare unstressed versus stressed conditions (heat shock typically triggers redistribution)

    • Evaluate cell cycle-dependent changes in localization

    • Consider cell density effects (as observed in embryonic stem cells, where HSPH1 expression varies between round and flattened colonies)

  • Fixation and permeabilization artifacts:

    • Compare multiple fixation methods (4% paraformaldehyde with NaOH as used successfully in ESCs)

    • Evaluate permeabilization effects (0.3% Triton X-100 versus alternative methods)

    • Include appropriate controls for each fixation/permeabilization combination

  • Antibody penetration assessment:

    • Verify antibody access to different subcellular compartments

    • Consider epitope masking in protein complexes or aggregates

    • Implement antigen retrieval optimization when necessary

  • Technical parameters standardization:

    • Control image acquisition settings (exposure, gain, offset)

    • Implement consistent thresholding in intensity quantification

    • Address potential bleed-through or cross-talk in multi-channel imaging

What emerging technologies are changing how researchers study HSPH1 function and expression?

The study of HSPH1 is being transformed by several cutting-edge technologies:

  • CRISPR-based approaches:

    • CRISPR-Cas9 knockout models for precise functional analysis

    • CRISPRi/CRISPRa for tunable expression modulation

    • CRISPR-based endogenous tagging to visualize HSPH1 without antibodies

  • Advanced imaging technologies:

    • Super-resolution microscopy (STORM, PALM) for nanoscale localization

    • Live-cell imaging with photoconvertible fluorescent protein fusions

    • Correlative light and electron microscopy for ultrastructural context

  • Single-cell analysis platforms:

    • Single-cell RNA-seq for expression heterogeneity assessment

    • Single-cell proteomics for protein level variance

    • Spatial transcriptomics for tissue context preservation

  • Interactome mapping tools:

    • Proximity labeling (BioID, APEX) for identifying dynamic interaction partners

    • Thermal proteome profiling to assess protein stability changes

    • Cross-linking mass spectrometry for structural interaction details

  • Structural biology advances:

    • Cryo-EM for high-resolution structures of HSPH1 complexes

    • Hydrogen-deuterium exchange mass spectrometry for conformational dynamics

    • AlphaFold2 predictions to guide functional domain analysis

  • Organoid and tissue-chip systems:

    • 3D organoid cultures for physiologically relevant expression studies

    • Microphysiological systems for tissue-specific HSPH1 function

    • Patient-derived models for personalized disease relevance

What are the most promising research directions for understanding HSPH1's role in disease pathology?

Several promising research directions are emerging for HSPH1 in disease contexts:

  • Neurodegenerative disorders:

    • HSPH1's interaction with misfolded proteins in Alzheimer's, Parkinson's, and ALS

    • Differential roles of alpha vs. beta isoforms in protein aggregation prevention

    • Potential therapeutic strategies enhancing HSPH1 chaperone activity

  • Cancer biology:

    • HSPH1 contribution to proteostasis in rapidly dividing tumor cells

    • Role in therapy resistance mechanisms

    • Potential as a biomarker or therapeutic target, particularly in heat shock protein-dependent cancers

  • Inflammatory conditions:

    • HSPH1's potential immunomodulatory functions

    • Role in cellular responses to inflammatory stressors

    • Interaction with damage-associated molecular patterns (DAMPs)

  • Developmental biology:

    • Functional significance of differential HSPH1 expression in embryonic stem cell states (round versus flattened colonies)

    • Role in cellular differentiation and fate determination

    • Importance during embryonic development and tissue remodeling

  • Aging research:

    • Decline in chaperone function as a contributor to age-related proteostasis collapse

    • HSPH1 as a potential intervention target for age-related pathologies

    • Relationship between stress resistance and longevity through HSPH1 pathways

  • Drug discovery approaches:

    • High-throughput screening for HSPH1 modulators

    • Structure-based drug design targeting specific functional domains

    • Exosome-based delivery of functional HSPH1 for therapeutic applications

Comparison of Common Anti-HSPH1 Antibodies and Their Applications

Antibody Target RegionHost/ClonalitySpecies ReactivityValidated ApplicationsEpitope LocationNotable Features
Region surrounding Pro819 Rabbit/PolyclonalHuman, MouseWB, IHC(p), EIAC-terminal regionDetects endogenous levels of HSP105
AA 713-733 Rabbit/PolyclonalHuman, Mouse, RatIHCNear C-terminusUseful for localization studies
C-Terminal region Rabbit/PolyclonalHuman, Mouse, RatWB, IHC(p), IHC(fro)C-terminusVersatile for multiple applications
AA 549-579 Rabbit/PolyclonalHuman, Mouse, RatWB, IHC, ELISA, IF, ICCMiddle regionBroad application compatibility
AA 659-858 Rabbit/PolyclonalHumanWB, IHC(p), FACSLarge C-terminal fragmentUseful for flow cytometry
Full length (AA 1-858) Rabbit/PolyclonalHumanWB, IHC, ELISAMultiple epitopesRecognizes multiple domains
Lys388-Glu505 Mouse/Monoclonal (Clone #531104)Human, Mouse, RatWBMiddle regionWell-characterized for Western blot

Troubleshooting Guide for Common HSPH1 Antibody Application Issues

IssuePotential CausesRecommended SolutionsValidation Approach
No signal in Western blotInsufficient protein loading, Poor transferIncrease protein amount, Optimize transfer for high MW proteinsInclude heat-shocked positive control samples (42°C, 30 min)
Multiple bands in Western blotCross-reactivity, Protein degradationTry alternative antibody, Add protease inhibitorsCompare with predicted MW (~110 kDa)
High background in IHC/IFInsufficient blocking, Non-specific bindingEnhance blocking (1% normal goat serum + 5% BSA) , Optimize antibody dilutionInclude secondary-only controls
Inconsistent staining intensityVariable fixation, Epitope maskingStandardize fixation (4% paraformaldehyde with NaOH) , Optimize permeabilization (0.3% Triton) Include positive control sections on each slide
Poor immunoprecipitationEpitope inaccessibility, Weak antibody bindingTry antibodies against different epitopes, Modify lysis conditionsValidate pull-down with Western blot
Discrepancy between protein and mRNA levelsPost-transcriptional regulation, Method sensitivity differencesCompare multiple antibodies, Analyze time courseCorrelate with functional readouts
Inability to distinguish isoformsSimilar molecular weights, High sequence identity (94%) Use high-resolution gels, Isoform-specific antibodiesValidate with recombinant protein controls

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.