HIGD1B Antibody

Shipped with Ice Packs
In Stock

Description

Applications in Research

The HIGD1B antibody is employed across multiple techniques to study protein expression, localization, and functional roles:

Western Blot (WB)

  • Use Case: Quantitative detection of HIGD1B in lysates from cancer cells or hypoxic tissues.

  • Example: PACO42302 detects HIGD1B in human colon cancer tissues .

  • Dilution: 1:300–5000 (bs-15489R-Cy5) .

Immunohistochemistry (IHC)

  • Use Case: Spatial analysis of HIGD1B in paraffin-embedded tissues (e.g., colon cancer) .

  • Protocol: Antigen retrieval with TE buffer (pH 9.0) or citrate buffer (pH 6.0) .

Immunofluorescence (IF)

  • Use Case: Co-localization studies in cell lines (e.g., HepG2) with secondary antibodies like Alexa Fluor 488 .

  • Dilution: 1:50–200 (PACO42302) .

ELISA

  • Use Case: High-throughput screening of HIGD1B levels in serum or lysates.

  • Dilution: 1:2000–1:10,000 (PACO42302) .

Mitochondrial Function and Hypoxia Response

  • Role in Apoptosis: HIGD1B prevents mitochondrial fragmentation under hypoxia by interacting with OPA1, delaying its cleavage and stabilizing mitochondrial integrity. Knockdown of HIGD1B increases caspase-3/-9 activity and apoptosis in cardiomyocytes .

  • Therapeutic Implications: Overexpression of HIGD1B in hypoxic conditions improves cell survival, suggesting potential in ischemia-related diseases .

Cancer Prognosis

  • Gastric Cancer (GC): Elevated HIGD1B expression correlates with poor prognosis, advanced tumor stages, and immune evasion. It serves as a biomarker for predicting chemotherapy and immunotherapy resistance .

  • Mechanism: HIGD1B regulates the tumor microenvironment (TME) and immune cell infiltration, promoting immune escape .

Cross-Species Reactivity

  • Versatility: The Cy5-conjugated HIGD1B antibody (bs-15489R-Cy5) reacts with human, mouse, rat, and other species, enabling comparative studies .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to ship orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. For specific delivery timeframes, please consult your local distributors.
Synonyms
HIGD1B antibody; HIG1 domain family member 1B antibody; Protein CLST 11240 antibody
Target Names
HIGD1B
Uniprot No.

Target Background

Database Links

HGNC: 24318

KEGG: hsa:51751

UniGene: Hs.151787

Subcellular Location
Membrane; Multi-pass membrane protein.

Q&A

What is HIGD1B and why is it important in cancer research?

HIGD1B is a protein-coding gene that belongs to the HIG1 (Hypoxia Inducible Gene 1) family. It has gained significant research interest due to its involvement in the occurrence and progression of various illnesses, particularly gastric cancer. HIGD1B is markedly elevated in gastric cancer tissue and cell lines compared to normal tissues, and patients with high HIGD1B expression demonstrate poorer clinical outcomes . Additionally, HIGD1B expression has been found to correlate with distinct grades, stages, and T stages of cancer, suggesting its potential as both a diagnostic and prognostic biomarker . Beyond gastric cancer, HIGD1B has been implicated in intracranial aneurysms, growth hormone-secreting pituitary adenomas, and lung cancer, indicating its broad relevance to multiple pathological conditions .

What experimental approaches are commonly used to detect HIGD1B expression?

Multiple complementary approaches should be employed for robust HIGD1B detection. Researchers typically begin with mining public databases such as TCGA and GEO to analyze HIGD1B expression patterns across large patient cohorts . For experimental validation, quantitative RT-PCR is used to measure HIGD1B mRNA levels, while Western blotting with specific antibodies detects protein expression in cell lines and tissue samples . Immunohistochemistry (IHC) allows visualization of HIGD1B in tissue contexts, providing information about spatial distribution. For functional studies, researchers may employ siRNA or CRISPR-Cas9 to knock down or knock out HIGD1B expression, respectively. It's important to use multiple detection methods, as studies have shown that mRNA and protein levels don't always correlate perfectly, especially under hypoxic conditions where post-transcriptional regulation may occur .

How do I select an appropriate HIGD1B antibody for my research application?

When selecting a HIGD1B antibody, consider these critical factors: First, determine which experimental application you require (Western blot, IHC, flow cytometry, etc.) as antibodies often perform differently across applications. Second, evaluate the antibody's specificity through evidence of validation against positive and negative controls, including knockdown experiments. Third, consider the epitope recognition—whether the antibody targets N-terminal, C-terminal, or internal regions may affect detection depending on protein modifications or isoforms present in your samples. Fourth, assess lot-to-lot consistency, particularly for polyclonal antibodies. Finally, review published literature where the antibody has been successfully used in applications similar to yours. For cutting-edge research on HIGD1B in cancer contexts, especially when measuring expression across different cancer stages and grades, antibodies validated specifically in cancer tissue microarrays would be most appropriate .

What protocols yield optimal results when using HIGD1B antibodies for immunohistochemistry?

For optimal immunohistochemistry results with HIGD1B antibodies, follow this methodological approach: Begin with appropriate tissue fixation, preferably using 10% neutral-buffered formalin for 24-48 hours, as overfixation can mask epitopes. Perform antigen retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0), testing both to determine which works best with your specific HIGD1B antibody. For particularly challenging samples, a combination of heat and enzymatic retrieval may improve results. When staining, use a dilution series (typically 1:100 to 1:500) to determine optimal antibody concentration, and always include positive controls (such as gastric cancer tissues known to express HIGD1B) and negative controls (antibody diluent only) . Incubate primary antibody overnight at 4°C to improve sensitivity while reducing background. For detection, polymer-based systems often provide better signal-to-noise ratio than avidin-biotin methods. Finally, carefully evaluate staining patterns; HIGD1B typically shows cytoplasmic localization with potential membranous enhancement in cancer cells, and scoring should consider both staining intensity and percentage of positive cells to create a composite score for accurate quantification .

How should I design experiments to investigate the relationship between HIGD1B expression and patient prognosis?

To robustly investigate the relationship between HIGD1B expression and patient prognosis, implement this comprehensive experimental design: First, collect a statistically powered cohort of patient samples with complete clinical data including follow-up information, ensuring representation across different cancer stages, grades, and treatment regimens. Use multiple technical approaches to measure HIGD1B expression—immunohistochemistry for spatial information, qRT-PCR for mRNA quantification, and Western blotting for protein levels . Create a standardized scoring system for HIGD1B expression (low vs. high) based on established cutoff values or median expression levels. Conduct Kaplan-Meier survival analysis to compare outcomes between patient groups with different HIGD1B expression levels . Perform both univariate and multivariate Cox regression analyses to determine if HIGD1B is an independent prognostic factor after adjusting for established clinical variables such as age, stage, and grade . Calculate ROC curves to assess the diagnostic efficacy of HIGD1B, with AUC values above 0.7 indicating acceptable performance (studies have reported AUC values of 0.741 for HIGD1B) . Finally, develop and validate a prognostic nomogram incorporating HIGD1B expression with other clinical factors to predict survival probability, as this approach has shown promising results with 5-year prediction accuracy of 0.735 .

What are the key considerations for analyzing HIGD1B in tumor microenvironment studies?

When analyzing HIGD1B in the tumor microenvironment (TME), several methodological considerations are essential. First, employ multiplex immunofluorescence or immunohistochemistry to simultaneously visualize HIGD1B expression alongside key immune cell markers (CD4, CD8, CD68, etc.), enabling spatial correlation between HIGD1B-expressing cells and tumor-infiltrating immune cells . Second, integrate computational deconvolution methods like CIBERSORT or xCell with bulk RNA-seq data to quantitatively estimate immune cell populations in relation to HIGD1B expression levels. Third, perform flow cytometry on fresh tumor samples to precisely quantify and characterize immune populations in HIGD1B-high versus HIGD1B-low tumors . Fourth, assess cytokine and chemokine profiles in the microenvironment using multiplex protein assays, as these may reveal mechanisms by which HIGD1B influences immune recruitment. Fifth, evaluate hypoxia markers (HIF-1α, CA9) alongside HIGD1B, given its association with hypoxic conditions. Research has shown that patients with high HIGD1B expression exhibit elevated levels of tumor-infiltrating immune cells and may be more prone to immune escape mechanisms . Finally, correlate these findings with immune checkpoint expression (PD-1, PD-L1, CTLA-4) to understand how HIGD1B might influence immunotherapy response .

How can I investigate the functional role of HIGD1B in cancer progression using antibody-based approaches?

To comprehensively investigate HIGD1B's functional role in cancer progression, implement these antibody-dependent techniques: Begin with immunoprecipitation (IP) using validated HIGD1B antibodies to identify protein-protein interactions, revealing potential signaling partners. Combine this with mass spectrometry to create an interaction network. Employ chromatin immunoprecipitation (ChIP) with antibodies against transcription factors like HIF-1α to determine if HIGD1B expression is directly regulated by hypoxia-responsive elements. For spatial understanding, use immunofluorescence co-localization studies to examine HIGD1B distribution relative to organelle markers (mitochondria, endoplasmic reticulum) and signaling molecules, particularly in pathways implicated by GSEA analysis such as TGF-β, Wnt, and Hedgehog signaling . Implement proximity ligation assays (PLA) to confirm direct protein interactions in situ. For functional validation, combine gene knockdown/knockout with rescue experiments using wild-type or mutated HIGD1B, followed by antibody-based detection methods to confirm expression. Utilize phospho-specific antibodies against downstream signaling molecules to map HIGD1B's effect on pathway activation. Finally, employ antibody-based techniques to measure changes in cell cycle proteins, apoptosis markers, and epithelial-mesenchymal transition factors following HIGD1B manipulation, as GSEA studies have linked HIGD1B to these cancer-associated processes .

What approaches should I use to evaluate the potential of HIGD1B as a therapeutic target?

To rigorously evaluate HIGD1B as a therapeutic target, implement this systematic approach: First, perform comprehensive target validation through multiple knockdown/knockout strategies (siRNA, shRNA, CRISPR-Cas9) in diverse cancer cell lines, confirming efficient HIGD1B depletion via antibody-based detection methods. Assess phenotypic consequences including changes in proliferation, invasion, migration, and colony formation. Second, evaluate the druggability of HIGD1B by identifying functionally important domains through structural analysis and creating domain-specific antibodies to determine which regions are essential for its oncogenic functions . Third, develop therapeutic antibodies or small molecules targeting HIGD1B, and evaluate their efficacy in vitro and in patient-derived xenograft models. Fourth, investigate potential resistance mechanisms by analyzing HIGD1B expression in treatment-resistant cell lines and patient samples, particularly as research has shown that high HIGD1B expression correlates with increased likelihood of drug resistance after chemotherapy and immunotherapy . Fifth, explore combination strategies with existing therapies based on pathway analysis, as GSEA studies have linked HIGD1B to multiple cancer-associated pathways including TGF-β, Wnt, and Hedgehog signaling . Finally, evaluate potential biomarkers of response by correlating treatment efficacy with HIGD1B expression levels and related signaling pathway activation markers, creating a companion diagnostic strategy for future clinical applications .

How do I effectively study the relationship between HIGD1B and immunotherapy response?

To effectively study the relationship between HIGD1B and immunotherapy response, implement this methodological framework: First, perform retrospective analysis of HIGD1B expression in pre-treatment tumor samples from immunotherapy-treated patients, correlating expression levels with response rates and survival outcomes. Second, establish predictive biomarker potential by calculating TIDE (Tumor Immune Dysfunction and Exclusion) scores alongside HIGD1B expression, as studies have shown this approach can predict immunotherapy effectiveness . Third, examine correlations between HIGD1B expression and established immunotherapy response markers such as PD-L1 expression, tumor mutational burden (TMB), and microsatellite instability (MSI) status, which have been investigated through computational methods in HIGD1B research . Fourth, create in vitro co-culture systems with tumor cells expressing variable HIGD1B levels and immune cells, then measure T-cell activation, cytokine production, and tumor cell killing in the presence of immune checkpoint inhibitors. Fifth, establish syngeneic mouse models with HIGD1B-modulated tumor cells to evaluate immunotherapy response in vivo. Finally, investigate mechanistic connections by examining how HIGD1B affects the tumor microenvironment, particularly immune cell infiltration and function, as research has demonstrated that high HIGD1B expression correlates with elevated levels of tumor-infiltrating immune cells and potential immune escape mechanisms . This comprehensive approach should clarify whether HIGD1B serves as a predictive biomarker or potential therapeutic target in immunotherapy settings.

What strategies can resolve inconsistent HIGD1B antibody staining results across different samples?

When encountering inconsistent HIGD1B antibody staining across samples, implement this systematic troubleshooting protocol: First, evaluate pre-analytical variables by standardizing tissue collection, fixation duration (24-48 hours in 10% neutral-buffered formalin), and storage conditions, as these factors significantly impact protein preservation and epitope accessibility. Second, optimize antigen retrieval by testing multiple methods (heat-induced epitope retrieval with citrate buffer pH 6.0 vs. EDTA buffer pH 9.0, or enzymatic retrieval), as different tissue processing can require different retrieval approaches. Third, perform antibody validation by testing multiple HIGD1B antibodies targeting different epitopes, and validate each through Western blot or siRNA knockdown experiments to confirm specificity . Fourth, establish a standardized staining protocol with optimized antibody dilution (titration series), incubation time and temperature, washing steps, and detection system. Fifth, implement quality controls on every staining run, including positive controls (gastric cancer tissues known to express HIGD1B) and negative controls (antibody diluent only, normal gastric tissues) . Sixth, consider biological variables such as hypoxic conditions that may affect HIGD1B expression and epitope accessibility. Finally, establish a standardized scoring system used by multiple trained observers to reduce subjective interpretation, incorporating both staining intensity and percentage of positive cells into a composite score . This comprehensive approach should significantly improve staining consistency and data reliability.

How should I interpret contradictory results between HIGD1B mRNA and protein expression levels?

When facing contradictory results between HIGD1B mRNA and protein expression, implement this interpretive framework: First, recognize that discrepancies often reflect biological reality rather than technical errors, as post-transcriptional and post-translational regulatory mechanisms can cause mRNA and protein levels to diverge. Second, validate both measurements technically—confirm RNA quality through RIN scores and verify antibody specificity through Western blots with appropriate controls . Third, consider time-dependent regulation; mRNA changes typically precede protein changes, so temporal differences in sampling could explain discrepancies. Fourth, examine post-transcriptional regulators such as microRNAs or RNA-binding proteins that may affect HIGD1B translation efficiency, particularly under hypoxic conditions relevant to HIGD1B function. Fifth, investigate protein stability and degradation pathways, as protein half-life significantly impacts steady-state levels independent of transcription rates. Sixth, assess cellular localization and compartmentalization using fractionation approaches followed by Western blotting, as some antibodies may not detect all cellular pools of the protein. Finally, evaluate alternative splicing of HIGD1B through PCR with isoform-specific primers and Western blotting with domain-specific antibodies, as different isoforms may exhibit different functions and expression patterns . This methodical approach will help distinguish technical issues from genuine biological complexity in HIGD1B regulation, advancing understanding of its role in cancer biology rather than dismissing contradictory data as experimental error.

What are the best practices for normalizing and quantifying HIGD1B expression in diverse experimental contexts?

For robust normalization and quantification of HIGD1B expression across diverse experimental contexts, implement these best practices: For Western blotting, use multiple housekeeping proteins (β-actin, GAPDH, and α-tubulin) rather than relying on a single reference, as expression of any individual housekeeping gene may vary across conditions or tissues . Employ densitometry with background subtraction and report HIGD1B as a ratio to housekeeping controls, using biological replicates (n≥3) for statistical validity. For qRT-PCR, validate reference genes for your specific experimental system using algorithms like geNorm or NormFinder, as studies have shown that optimal reference genes vary across tissue types and experimental conditions. Calculate HIGD1B expression using the 2^-ΔΔCt method with multiple validated reference genes . For immunohistochemistry, develop a standardized scoring system incorporating both staining intensity (0-3+) and percentage of positive cells to create a composite score or H-score (0-300), and ensure multiple trained observers score samples independently to minimize subjective bias . In RNA-seq data analysis, use appropriate normalization methods such as TPM (Transcripts Per Million) or FPKM (Fragments Per Kilobase Million) when comparing across samples, and consider batch effect correction when integrating data from multiple sources or cohorts . Finally, for all methods, include appropriate positive and negative controls, and validate findings using orthogonal techniques whenever possible to ensure reproducibility and reliability.

How can HIGD1B antibodies be used to investigate the role of HIGD1B in therapy resistance mechanisms?

To investigate HIGD1B's role in therapy resistance using antibody-based approaches, implement this multifaceted research strategy: First, develop paired sensitive/resistant cell line models by exposing cancer cells to increasing concentrations of chemotherapeutic agents or targeted therapies, then use HIGD1B antibodies in Western blotting and immunocytochemistry to compare expression levels between sensitive and resistant populations . Second, perform temporal analysis of HIGD1B expression during resistance development using antibody-based techniques to determine whether HIGD1B upregulation precedes or follows resistance acquisition. Third, collect pre-treatment and post-progression biopsies from patients undergoing therapy and analyze HIGD1B expression by immunohistochemistry, correlating changes with treatment outcomes and resistance emergence . Fourth, use proximity ligation assays (PLA) with HIGD1B antibodies to identify novel protein interactions that emerge specifically in resistant cells. Fifth, implement ChIP-seq using antibodies against transcription factors identified through GSEA analysis (such as those involved in TGF-β, Wnt, and Hedgehog pathways) to determine whether HIGD1B transcriptional regulation changes during resistance development . Finally, combine HIGD1B knockdown/overexpression with drug sensitivity assays to establish causal relationships, confirming protein modulation using antibody-based detection methods. Research has shown that patients with high HIGD1B expression are more likely to experience drug resistance after chemotherapy and immunotherapy, making this investigation particularly relevant for developing strategies to overcome treatment resistance .

What are the most promising approaches for studying the relationship between HIGD1B and cancer metabolism?

To investigate the relationship between HIGD1B and cancer metabolism, implement these cutting-edge approaches: First, perform metabolic profiling using techniques such as Seahorse XF analysis to measure oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) in cells with modulated HIGD1B expression, confirming protein levels via Western blotting with validated antibodies . Second, use stable isotope-resolved metabolomics (SIRM) with 13C-labeled glucose, glutamine, and fatty acids to trace metabolic flux through glycolysis, TCA cycle, and fatty acid metabolism in HIGD1B-high versus HIGD1B-low cells. Third, implement proximity labeling techniques such as BioID or APEX2 fused to HIGD1B to identify proximal proteins in metabolic compartments, followed by mass spectrometry analysis. Fourth, investigate mitochondrial dynamics and function using antibodies against HIGD1B and mitochondrial markers in confocal microscopy and electron microscopy with immunogold labeling. Fifth, examine metabolic adaptation to hypoxia, as GSEA analyses have shown that HIGD1B expression correlates with differential regulation of oxidative phosphorylation and other metabolic pathways . Sixth, analyze metabolic gene expression signatures in patient cohorts stratified by HIGD1B expression levels, integrating transcriptomic and metabolomic data. Finally, investigate potential metabolic vulnerabilities in HIGD1B-high tumors using drug screening with metabolism-targeting compounds, which could reveal synthetic lethal interactions for therapeutic exploitation. This comprehensive approach should clarify how HIGD1B contributes to the metabolic reprogramming that supports cancer progression and therapy resistance .

How can I develop and validate novel HIGD1B-targeted therapeutic approaches?

To develop and validate novel HIGD1B-targeted therapeutic approaches, implement this systematic research pipeline: First, perform comprehensive target validation through multiple knockdown methods (siRNA, shRNA, CRISPR-Cas9) in diverse cancer cell lines, confirming HIGD1B depletion via validated antibodies in Western blotting and immunohistochemistry . Second, develop therapeutic antibodies against HIGD1B by generating monoclonal antibodies through hybridoma technology or phage display, with epitopes selected based on structural analysis and functional studies. Third, create antibody-drug conjugates (ADCs) by linking cytotoxic payloads to HIGD1B-targeting antibodies, optimizing drug-antibody ratio and linker chemistry for maximum efficacy and stability. Fourth, for intracellular targeting, develop proteolysis-targeting chimeras (PROTACs) or molecular glues that induce HIGD1B degradation, validating target engagement through Western blotting with HIGD1B antibodies . Fifth, identify smallmolecule HIGD1B inhibitors through high-throughput screening or structure-based drug design, focusing on disrupting key protein interactions identified through immunoprecipitation studies. Sixth, test candidate therapeutics in patient-derived xenograft models, confirming target engagement through immunohistochemistry and pharmacodynamic biomarker analysis. Finally, develop combination strategies with existing therapies based on pathway analysis, as GSEA studies have linked HIGD1B to multiple cancer-associated pathways including TGF-β, Wnt, and Hedgehog signaling . This pathway-informed approach is particularly promising as research has demonstrated that high HIGD1B expression correlates with poor prognosis and potential therapy resistance, suggesting it as a valuable therapeutic target for improving cancer treatment outcomes .

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.