HIF1A Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Production

HIF1A Recombinant Monoclonal Antibodies are synthesized in vitro using cloned DNA sequences from immunoreactive hosts (e.g., rabbits). This method eliminates variability between batches and enhances specificity for HIF-1α, a subunit of the heterodimeric HIF-1 transcription factor. HIF-1α is rapidly degraded under normoxia but stabilizes under hypoxia, activating genes involved in angiogenesis, metabolism, and cell survival .

Key Applications and Protocols

HIF1A antibodies are validated for multiple experimental techniques:

ApplicationRecommended UsageSample TypeValidation Highlight
Western Blot (WB)2 µg/mL , 1/1000 dilution HeLa, HepG2 cells treated with DFO 120 kDa band in parental cells, absent in knockouts
Immunocytochemistry (ICC)3–25 µg/mL Fixed HeLa cells treated with DFO Nuclear localization confirmed via DAPI
Immunoprecipitation (IP)5 µg antibody + 0.5 mg lysate HeLa nuclear extracts Bands at 110 kDa under reducing conditions
Chromogenic IHC15 µg/mL Paraffin-embedded human kidney cancer tissue Specific nuclear staining in cancer tissue
Flow Cytometry1/11709 dilution Permeabilized HeLa cells Hypoxia-induced signal vs. isotype control

Validation and Specificity

  • Knockout Validation: HIF1A antibodies show no cross-reactivity in HIF-1α knockout HeLa cells, confirming specificity .

  • Epitope Mapping: Certain clones (e.g., [EP1215Y]) target the oxygen-dependent degradation domain (ODD), critical for HIF-1α regulation .

  • Cross-Species Reactivity: Validated for human, mouse, and rat samples .

Hypoxia Response Studies

  • In HeLa cells treated with deferoxamine (DFO), HIF1A antibodies detect nuclear accumulation of HIF-1α, correlating with hypoxia-induced gene activation .

  • Western Blot Data: A 120 kDa band is observed in wild-type cells under hypoxia, absent in knockouts .

Cancer Research

  • HIF1A antibodies highlight overexpression in human kidney cancer tissue, supporting its role in tumor angiogenesis .

  • In HepG2 hepatocellular carcinoma cells, antibody staining intensity correlates with baicalein concentration, linking HIF-1α to drug response .

Mechanistic Insights

  • Co-immunoprecipitation studies confirm HIF-1α interaction with p300/CBP, essential for transcriptional activation of VEGF and other hypoxia-response genes .

Comparative Analysis of Clones

CloneHostReactivityApplicationsSupplier
16H4L13 RabbitHuman, MouseWB, ICC, IHCThermo Fisher
JE75-33 RabbitHuman, Mouse, RatWB, IF-Cell, IHC-PHuabio
BL-124-3F7 RabbitHumanWB, IP, ChIPBethyl Laboratories

Technical Considerations

  • Storage: Lyophilized antibodies are stable for 1 year at -20°C .

  • Controls: Include knockout cell lines and isotype-matched antibodies to rule out nonspecific binding .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
ARNT interacting protein antibody; ARNT-interacting protein antibody; Basic helix loop helix PAS protein MOP1 antibody; Basic-helix-loop-helix-PAS protein MOP1 antibody; bHLHe78 antibody; Class E basic helix-loop-helix protein 78 antibody; HIF 1A antibody; HIF 1alpha antibody; HIF-1-alpha antibody; HIF-1alpha antibody; HIF-alpha antibody; HIF1 A antibody; HIF1 Alpha antibody; HIF1 antibody; HIF1-alpha antibody; HIF1A antibody; HIF1A_HUMAN antibody; hifla antibody; Hypoxia inducible factor 1 alpha antibody; Hypoxia inducible factor 1 alpha isoform I.3 antibody; Hypoxia inducible factor 1 alpha subunit antibody; Hypoxia inducible factor 1 alpha subunit basic helix loop helix transcription factor antibody; Hypoxia inducible factor 1; alpha subunit (basic helix loop helix transcription factor) antibody; Hypoxia inducible factor1alpha antibody; Hypoxia-inducible factor 1-alpha antibody; Member of PAS protein 1 antibody; Member of PAS superfamily 1 antibody; Member of the PAS Superfamily 1 antibody; MOP 1 antibody; MOP1 antibody; PAS domain-containing protein 8 antibody; PASD 8 antibody; PASD8 antibody
Target Names
Uniprot No.

Target Background

Function
HIF1A functions as a critical transcriptional regulator in the adaptive response to hypoxia. Under hypoxic conditions, it activates the transcription of over 40 genes, including erythropoietin, glucose transporters, glycolytic enzymes, vascular endothelial growth factor (VEGF), HILPDA, and other genes whose protein products enhance oxygen delivery or facilitate metabolic adaptation to hypoxia. HIF1A plays a crucial role in embryonic vascularization, tumor angiogenesis, and the pathophysiology of ischemic disease. It forms a heterodimer with ARNT, and this heterodimer binds to the core DNA sequence 5'-TACGTG-3' within the hypoxia response element (HRE) of target gene promoters. Activation necessitates the recruitment of transcriptional coactivators such as CREBBP and EP300. Its activity is enhanced by interaction with NCOA1 and/or NCOA2. Interaction with the redox regulatory protein APEX1 appears to activate CTAD and potentiates activation by NCOA1 and CREBBP. HIF1A is involved in the axonal distribution and transport of mitochondria in neurons during hypoxia.

In the context of microbial infection, particularly human coronavirus SARS-CoV-2, HIF1A is essential for the induction of glycolysis in monocytes and the subsequent proinflammatory state. In monocytes, HIF1A induces the expression of ACE2 and cytokines like IL1B, TNF, IL6, and interferons. This action promotes human coronavirus SARS-CoV-2 replication and the monocyte inflammatory response.
Gene References Into Functions
  1. Research has demonstrated a notable antitumor synergy when combining CRISPR/Cas9-mediated HIF1alpha knockout with Transarterial embolization (TAE) in mice. This highlights the potential of HIF1a as a therapeutic knockout target in conjunction with TAE for hepatocellular carcinoma treatment. PMID: 30226584
  2. As FIH-1-dependent hydroxylation of NAA10 is oxygen-dependent, NAA10 acetylates HIF-1alpha under normoxia but not under hypoxia. PMID: 30237125
  3. HIF1A is upregulated in breast and bladder tumors exhibiting high NRF2 activity. NRF2 targets a functional antioxidant response element at the HIF1A locus, revealing a direct regulatory link between these two significant oxygen-responsive transcription factors. PMID: 30241031
  4. CPT1A is repressed by HIF1 and HIF2, reducing fatty acid transport into the mitochondria and diverting fatty acids to lipid droplets for storage. PMID: 29176561
  5. The miR-497 approximately 195 cluster regulates angiogenesis during coupling with osteogenesis by maintaining endothelial Notch1 and HIF1A activity. PMID: 28685750
  6. Studies have shown that HIF1alpha is significantly upregulated in gallbladder cancer (GBC) tissues. HIF1alpha overexpression is closely associated with lymph node metastasis and TNM stage. HIF1alpha promotes cell migration in a hypoxic microenvironment by overexpressing VEGF in the GBC cell line. PMID: 30272364
  7. Patients with “positive” HIF-1alpha exhibited decreased overall survival compared to those with “negative” HIF-1alpha, indicating it as an independent adverse prognostic factor for HCC patients with cirrhosis, but not for cirrhosis-free patients. PMID: 30274686
  8. Genetic polymorphisms in HIF1A have not been associated with persistent apical periodontitis. PMID: 29898065
  9. Hypoxia-induced angiogenesis is a complex process involving distinct yet overlapping functions of HIF-1alpha and HIF-2alpha in angiogenesis, bioenergetic adaption, and the redundant transcriptional induction of MIF. PMID: 28993199
  10. High HIF1A expression is correlated with high Collagen I Fibers in Triple Negative Breast Cancer. PMID: 29247885
  11. Cathepsin B (CTSB) is a novel target gene of hypoxia-inducible factor-1-alpha (HIF-1alpha). CTSB mRNA and protein levels can be upregulated in a HIF-1alpha-dependent manner. PMID: 29935187
  12. This is the first case-control study demonstrating that the HIF1A gene rs10873142 polymorphism increases the risk of COPD in a Chinese Han population. PMID: 29339421
  13. ST6Gal-I activity augments HIF-1alpha accumulation in cells grown in a hypoxic environment or treated with two chemical hypoxia mimetics, deferoxamine and dimethyloxalylglycine. PMID: 29475939
  14. mRNA expression levels of both HIF1A and LACC1 are upregulated in the skin lesions of individuals with leprosy. PMID: 29706348
  15. Data suggests a significant role of miR-210 in sustaining HIF-1alpha activity by suppressing HIF-3alpha, thereby regulating cell growth and chemotherapeutic drug resistance in cholangiocarcinoma. PMID: 29953500
  16. HOXA9 inhibits HIF1A-mediated glycolysis by interacting with CRIP2 to repress cutaneous squamous cell carcinoma development. PMID: 29662084
  17. HIF-1alpha inhibits the NCR1/NKp46 pathway by upregulating miR-224, impacting the killing capability of NK cells on prostate cancer, thus inducing immune escape of tumor cells. PMID: 29885835
  18. Research indicates that AGPAT2, which is mutated in patients with congenital generalized lipodystrophy and overexpressed in various types of cancer, is a direct transcriptional target of HIF-1. This suggests that HIF-1-mediated upregulation of lipid storage plays a critical role in the adaptation and survival of cancer cells under low oxygen conditions. PMID: 29908837
  19. HIF1A overexpression leads to radioresistance of cervical cancer. PMID: 30355300
  20. Knockdown of DEC2 resulted in a significant (26.7%) reduction of VEGF expression in MIO-M1 cells under hypoxia-mimicking conditions induced by DFO (P < .05). Levels of HIF1alpha protein were also significantly reduced, by 60.2%, in MIO-M1 cells treated with siRNA against the DEC2 gene (P < .05). Moreover, HIF1alpha levels showed a significant (2.5-fold) increase in MIO-M1 cells overexpressing DEC2 (P < .05). PMID: 30250985
  21. High HIF1A expression is associated with salivary adenoid cystic carcinoma. PMID: 30015895
  22. Studies have shown that Hif-1alpha directly promotes H19 expression by binding to the H19 promoter and indirectly through SP1-mediated H19 transcriptional activation under hypoxia in glioblastoma cells. PMID: 28327666
  23. The IRIS-driven metastatic mechanism arises from IRIS-dependent suppression of phosphatase and tensin homolog (PTEN) transcription, which disrupts the PI3K/AKT/GSK-3beta pathway, leading to prolyl hydroxylase-independent HIF-1alpha stabilization and activation in a normoxic environment. PMID: 30254159
  24. Data indicates that von Hippel-Lindau-binding protein 1 (VBP1) enhances the stability of von Hippel-Lindau tumor suppressor protein (pVHL) and facilitates pVHL-mediated ubiquitination of hypoxia-inducible factor 1, alpha subunit (HIF-1alpha). PMID: 29121446
  25. Multifunctional proteins epigenetically modulating HIF1A stability and activity have been described. (Review) PMID: 29032501
  26. Studies have demonstrated that both HIF1alpha and HIF2alpha contribute to the regulation of cellular adaptation to hypoxia and resistance to cancer therapies. They have the potential to exert significant effects on the maintenance and evolution of cancer stem cells. Additionally, HIF1alpha and HIF2alpha appear to hold significant prognostic and predictive value. [review] PMID: 29845228
  27. HIF-1 was overexpressed in osteosarcoma tissues and cell lines, promoting cell proliferation, clone formation, migration, invasion, and inhibiting cell apoptosis. PMID: 29807229
  28. Hypoxia-induced expression of CXCR4 promoted trophoblast cell migration and invasion via the activation of HIF1alpha, which is crucial during placentation. PMID: 29786753
  29. Data suggests that NRF2/NFE2L2 promotes breast cancer progression by enhancing glycolysis through co-activation of HIF1A; NRF2 and HIF1A mRNA and protein levels are significantly upregulated in breast cancer cells compared to benign breast epithelial cells. (NRF2/NFE2L2 = nuclear factor erythroid 2-related factor 2; HIF1A = hypoxia inducible factor 1 subunit alpha) PMID: 29275212
  30. The expression of MMIF, HIF-1alpha, and VEGF in the serum and endometrial tissues can be used to assess the stage of endometriosis and the severity of dysmenorrhea. PMID: 30074218
  31. Research revealed an HIF-1alpha/IL-1beta signaling loop between cancer cells and tumor-associated macrophages in a hypoxic microenvironment, leading to cancer cell epithelial-mesenchymal transition and metastasis. Importantly, these findings suggest a potential role for an anti-inflammatory strategy in hepatocellular carcinoma treatment. PMID: 29171040
  32. Overexpression of HIF-1alpha and P4HB is associated with poor prognosis in patients with gastric cancer. PMID: 29904245
  33. Hypoxic stress in hepatocellular carcinoma (HCC) cells promotes YAP binding to HIF-1a in the nucleus, sustaining HIF-1a protein stability to bind to the PKM2 gene and directly activate PKM2 transcription to accelerate glycolysis. PMID: 30180863
  34. Overexpression of VHL was more effective at inhibiting fibrosis compared to silencing HIF-1a plus HIF-2a. Normoxia-active HIF-1a or HIF-2a prevented the inhibitory effect of VHL on liver fibrosis, indicating that attenuating fibrosis via VHL is, to some extent, HIF-1a- and HIF-2a-dependent. PMID: 28112200
  35. Results indicate that knockdown of HIF-1alpha reduced hypoxia-induced SENP1 expression, suggesting that the induction of SENP1 expression is mediated by hypoxia-inducible factor HIF-1alpha. PMID: 28796315
  36. ADM was an upstream molecule of HIF-1alpha/VEGF and it promoted angiogenesis by upregulating HIF-1alpha/VEGF in epithelial ovarian cancer. PMID: 28091613
  37. HIF-1alpha expression correlates with the expression level of IL-8, as evidenced by the downregulation of IL-8 in response to silencing of HIF-1alpha in HCC cell lines under hypoxic conditions. PMID: 29881400
  38. MOLP8/R cells display a very high overexpression of LCP1 gene (l-Plastin) controlled by HIF1&alpha. PMID: 29882856
  39. The role of FTH1 in the FIH control of HIF-1 activity is reported. PMID: 29580991
  40. The current study demonstrates the presence of the IDH1 R132H mutation in primary human glioblastoma cell lines with upregulated HIF-1alpha expression. This mutation downregulates c-MYC activity, resulting in a consequential decrease in miR-20a, which is responsible for cell proliferation and resistance to standard temozolomide treatment. PMID: 29625108
  41. The interplay between HIF1A, NRF2, and NF-kappaB is crucial for adapting to changes in oxygen availability. (Review) PMID: 29485192
  42. PKD1 regulates the hypoxic glycolytic metabolism of cancer cells by modulating the expression of HIF-1alpha and glycolytic enzymes. PMID: 29901206
  43. Findings provide evidence that HIF1alpha overexpression promotes the radioresistance of prostate cancer cells through enhanced betacatenin nuclear translocation. PMID: 29658569
  44. Collectively, the present study demonstrated mitochondrial fission as a tumor suppression process that is regulated by the HIF/miR125a/Mfn2 pathways. These pathways act to restrict PANC1 cell survival, energy metabolism, and migration, with potential implications for novel approaches for PC therapy. PMID: 29749475
  45. The results of the present study demonstrated that hypoxia-induced cytoprotective autophagy counteracted gemcitabine-induced apoptosis by increasing HIF1alpha expression. PMID: 29693166
  46. CD40 is a key molecule for the upregulation of HIF-1alpha and PTEN underlying the severity of microangiopathy. PMID: 29549140
  47. Enhanced expression of HIF-1alpha may be linked to autophagy activation in SH-SY5Y cells, thus contributing to ischemic/hypoxic brain damage. PMID: 29724989
  48. Data shows that deletion of hypoxia inducible factor 1 subunit alpha (HIF-1alpha) in NK cells inhibited tumor growth despite impaired tumor cell killing. PMID: 29150606
  49. AEG-1 was found to be significantly associated with hypoxia in ovarian cancer by regulating the HIF-1alpha/NF-kappaB/VEGF pathway. PMID: 29770329
  50. No significant differences were observed in the serum levels between early-onset pre-eclampsia, late-onset pre-eclampsia, and controls. PMID: 28574293

Show More

Hide All

Database Links

HGNC: 4910

OMIM: 603348

KEGG: hsa:3091

STRING: 9606.ENSP00000338018

UniGene: Hs.597216

Subcellular Location
Cytoplasm. Nucleus. Nucleus speckle.
Tissue Specificity
Expressed in most tissues with highest levels in kidney and heart. Overexpressed in the majority of common human cancers and their metastases, due to the presence of intratumoral hypoxia and as a result of mutations in genes encoding oncoproteins and tumo

Q&A

What is HIF-1α and why is it important in research?

HIF-1α (hypoxia-inducible factor 1 subunit alpha) functions as a master transcriptional regulator of the adaptive response to hypoxia. This 826-amino acid protein is expressed in most tissues, with highest expression levels in the kidney and heart . The protein undergoes various post-translational modifications including glycosylation, ubiquitination, sumoylation, acetylation, and phosphorylation . HIF-1α is critically important in research because it regulates genes involved in adaptation to insufficient oxygen environments, which has broad implications in various diseases including vascular and pulmonary conditions as well as cancer progression . The protein forms a heterodimer (HIF-1) that binds to hypoxia-response elements (HREs) in gene promoters, coordinating cellular adaptation to hypoxic conditions .

What are the key applications for HIF-1α antibodies in research?

HIF-1α antibodies are utilized across multiple experimental platforms in research settings. The primary applications include:

  • Western Blotting: For detecting protein expression levels in cell and tissue lysates, particularly after hypoxic treatments

  • Immunocytochemistry: For visualizing subcellular localization, especially nuclear translocation under hypoxic conditions

  • Chromatin Immunoprecipitation (ChIP): For investigating DNA-protein interactions

  • CUT & RUN: For high-resolution chromatin profiling

  • ELISA: For quantitative protein detection

  • Simple Western: For automated capillary-based immunodetection

These applications enable researchers to investigate HIF-1α expression, localization, and function in diverse experimental contexts, particularly in hypoxia-related studies.

Which cell lines are commonly used as positive controls for HIF-1α antibody validation?

Several human cancer cell lines serve as reliable positive controls for validating HIF-1α antibodies:

Cell LineCancer TypeTreatmentApplicationReference
HepG2Hepatocellular carcinomaDFO (1 mM, overnight)Western Blot, Simple Western
HeLaCervical epithelial carcinomaDFOImmunocytochemistry
MCF-7Breast cancerNone specifiedImmunocytochemistry

These cell lines consistently show HIF-1α induction, particularly after treatment with deferoxamine (DFO), which mimics hypoxic conditions by inhibiting prolyl hydroxylases that normally target HIF-1α for degradation . The nuclear localization of HIF-1α in these treated cells provides an excellent positive control for antibody specificity testing .

How should I design experiments to induce and detect HIF-1α expression?

Designing experiments for HIF-1α detection requires careful consideration of induction conditions and detection methods:

For induction:

  • Chemical induction: Treatment with 1 mM deferoxamine (DFO) overnight is a standard protocol that stabilizes HIF-1α by inhibiting prolyl hydroxylases

  • True hypoxia: Culture cells in hypoxic chambers (1-2% O2) for 4-24 hours to physiologically induce HIF-1α

For detection:

  • Western blot: Use 2 μg/mL of anti-HIF-1α antibody with appropriate HRP-conjugated secondary antibodies under reducing conditions

  • Immunocytochemistry: Apply 1-3 μg/mL of primary antibody for 3 hours at room temperature, followed by fluorescent secondary antibody detection

  • Simple Western: Use 20 μg/mL antibody concentration with 0.2 mg/mL protein loading

Include both untreated and treated samples side-by-side to demonstrate induction. The expected molecular weight for detection is approximately 116-120 kDa . Always include appropriate loading controls such as GAPDH for Western blot normalization .

Validating antibody specificity is crucial for reliable research outcomes. Implement these strategies:

  • Knockout/knockdown validation: Compare antibody reactivity between parental cell lines and HIF-1α knockout lines

    • HeLa parental vs. HIF-1α knockout HeLa lines treated with DFO show clear differences in signal

    • The absence of signal in knockout lines confirms specificity

  • Induction comparison: Run parallel samples with and without HIF-1α induction

    • The significant increase in signal after DFO treatment confirms detection of the regulated protein

  • Molecular weight verification: Confirm detection at the expected 116-120 kDa range

  • Subcellular localization: Verify nuclear accumulation upon induction in immunocytochemistry

    • Nuclear staining pattern in DFO-treated cells but not in untreated controls

  • Antibody cross-reactivity: Review the species cross-reactivity data to ensure suitability for your model system

    • Many HIF-1α antibodies react with human, mouse, rat, and monkey samples

Implementing these validation steps ensures reliable and reproducible results across different experimental platforms.

What are common issues with HIF-1α detection and how can they be resolved?

Several challenges can arise when detecting HIF-1α in experimental systems:

  • Weak or absent signal:

    • Cause: Insufficient induction or rapid protein degradation

    • Solution: Extend DFO treatment time (12-24 hours), increase DFO concentration to 1 mM, or use proteasome inhibitors (MG132) in combination with hypoxia

  • High background:

    • Cause: Non-specific antibody binding or insufficient blocking

    • Solution: Increase blocking time/concentration, optimize antibody dilution, use a different secondary antibody, or try a validated antibody clone like D2U3T or 2443B

  • Multiple bands in Western blot:

    • Cause: Degradation products, splice variants, or post-translational modifications

    • Solution: Use fresh samples with protease inhibitors, optimize sample preparation, and run a positive control (DFO-treated HepG2 cells)

  • Inconsistent results across experiments:

    • Cause: Variable HIF-1α stabilization conditions or cell culture differences

    • Solution: Standardize hypoxia induction protocols, control cell density, and validate antibody performance in your specific cell line

  • Discrepancies between protein and mRNA levels:

    • Cause: HIF-1α is primarily regulated post-translationally

    • Solution: Focus on protein detection methods rather than mRNA analysis for HIF-1α studies

Addressing these common issues systematically will significantly improve detection reliability and experimental reproducibility.

How should I interpret differences in HIF-1α band intensity and localization patterns?

Interpreting HIF-1α detection data requires consideration of several factors:

For Western blot band intensity:

  • Strong bands at 116-120 kDa after hypoxic treatment indicate successful HIF-1α stabilization

  • Absence of bands in untreated samples is expected due to rapid degradation under normoxic conditions

  • Quantitative comparison requires normalization to loading controls like GAPDH

  • Varying intensity between cell lines may reflect different hypoxia response capacities or genetic backgrounds

For immunocytochemistry localization patterns:

  • Nuclear localization (co-localization with DAPI) indicates active HIF-1α that has translocated to regulate gene expression

  • Cytoplasmic staining may indicate either newly synthesized protein or non-specific antibody binding

  • Heterogeneous staining within a population may reflect cell cycle differences or microenvironmental variations

  • Intensity differences between nuclear and cytoplasmic compartments can provide insights into HIF-1α activation status

Differences in detection patterns may also reflect post-translational modifications that affect antibody recognition or protein stability. When comparing across experimental conditions, maintain consistent antibody concentrations, exposure times, and image acquisition settings to enable meaningful quantitative analysis.

How can HIF-1α antibodies be utilized in cancer research models?

HIF-1α antibodies enable sophisticated investigations in cancer research:

  • Tumor hypoxia mapping:

    • Immunohistochemical detection of HIF-1α in tumor sections reveals hypoxic regions

    • Correlation with distance from blood vessels provides insights into oxygen diffusion gradients

    • Co-staining with proliferation and apoptosis markers helps understand hypoxic cell fate

  • Drug resistance mechanisms:

    • Monitoring HIF-1α levels before and after chemotherapy treatment

    • Correlation of HIF-1α expression with therapeutic response in patient-derived xenografts

    • Investigation of HIF-1α-dependent metabolic adaptations using metabolomics combined with immunoprecipitation

  • Metastasis research:

    • Tracking HIF-1α activation in circulating tumor cells

    • Evaluating HIF-1α expression at invasion fronts using immunofluorescence

    • ChIP-seq analysis to identify HIF-1α transcriptional targets driving metastatic programs

  • Therapeutic targeting:

    • Screening compounds that modulate HIF-1α stability or activity

    • Assessing HIF-1α nuclear translocation after drug treatment

    • Monitoring HIF-1α target gene expression following pathway inhibition

Cancer cell lines that reliably express HIF-1α upon induction include MCF-7 (breast cancer), HepG2 (liver cancer), and HeLa (cervical cancer) , making them valuable models for these advanced applications.

What approaches enable investigation of HIF-1α interaction with other proteins and signaling pathways?

Investigating HIF-1α interactions with other cellular components requires specialized techniques:

  • Co-immunoprecipitation (Co-IP):

    • Use anti-HIF-1α antibodies to pull down protein complexes

    • Western blot analysis of precipitates for suspected interaction partners

    • Reverse Co-IP with antibodies against suspected partners to confirm interactions

  • Proximity ligation assay (PLA):

    • Visualize and quantify protein-protein interactions in situ

    • Combine HIF-1α antibodies with antibodies against potential interactors

    • Fluorescent signals indicate close proximity (<40 nm) between proteins

  • ChIP-seq analysis:

    • Identify genome-wide binding sites of HIF-1α

    • Integrate with transcriptomic data to correlate binding with gene expression

    • Compare binding patterns under different oxygen tensions or treatment conditions

  • Mass spectrometry-based interactomics:

    • Immunoprecipitate HIF-1α from cells under different conditions

    • Analyze protein complexes by mass spectrometry

    • Identify condition-specific interaction partners

  • FRET/BRET analyses:

    • Engineer fluorescent fusion proteins for real-time interaction monitoring

    • Validate interactions discovered through antibody-based methods

    • Track dynamic interactions in living cells

These approaches provide complementary insights into how HIF-1α integrates with cellular signaling networks and transcriptional machinery to orchestrate adaptive responses to hypoxia.

How can post-translational modifications of HIF-1α be studied using specific antibodies?

Studying HIF-1α post-translational modifications (PTMs) requires specialized approaches:

  • Modification-specific antibodies:

    • Use antibodies that specifically recognize phosphorylated, hydroxylated, or acetylated forms of HIF-1α

    • Compare levels of modified and total HIF-1α protein under various conditions

    • Correlate modifications with protein stability and transcriptional activity

  • 2D gel electrophoresis:

    • Separate HIF-1α protein spots based on charge and mass

    • Western blot with total HIF-1α antibodies

    • Identify shifts in migration patterns indicative of specific modifications

  • Mass spectrometry following immunoprecipitation:

    • Enrich HIF-1α using validated antibodies

    • Perform tryptic digestion and analyze peptide fragments

    • Map modifications to specific amino acid residues

  • Pharmacological inhibitor studies:

    • Treat cells with inhibitors of specific modifying enzymes (kinases, deacetylases, etc.)

    • Monitor changes in HIF-1α stability, localization, and function

    • Correlate with changes in specific modifications

  • CRISPR-based mutagenesis:

    • Generate cell lines with mutations at key modification sites

    • Assess HIF-1α function using validated antibodies

    • Compare wild-type and mutant responses to hypoxia

HIF-1α undergoes multiple PTMs including glycosylation, ubiquitination, sumoylation, acetylation, and phosphorylation , each potentially affecting its stability, localization, or transcriptional activity. Investigating these modifications provides deeper insights into the complex regulation of hypoxic response pathways.

How should I optimize immunofluorescence protocols for HIF-1α detection in different cell types?

Optimizing immunofluorescence for HIF-1α requires systematic adjustment of multiple parameters:

  • Fixation methods:

    • 4% paraformaldehyde for 10-15 minutes preserves protein structure while maintaining antigenicity

    • Methanol fixation (ice-cold, 10 minutes) may improve nuclear antigen accessibility

    • Test both methods to determine optimal preservation of HIF-1α epitopes in your specific cell type

  • Permeabilization:

    • 0.1-0.5% Triton X-100 for adherent cells

    • 0.1% saponin for more delicate cell types

    • Optimize time (5-15 minutes) to balance antigen accessibility with structural preservation

  • Antibody concentration and incubation:

    • Test concentration range (1-25 μg/mL) for optimal signal-to-noise ratio

    • Extended incubation (3 hours at room temperature or overnight at 4°C)

    • PBS with 1-3% BSA as diluent to minimize background

  • Signal amplification:

    • Use high-sensitivity detection systems like fluorescent-conjugated secondary antibodies

    • NorthernLights™ 557-conjugated Anti-Rabbit IgG has been validated for HIF-1α detection

    • Counterstain nuclei with DAPI to confirm nuclear localization

  • Cell-specific considerations:

    • Increase induction time for cells with slower hypoxic response

    • Adjust cell density to 50-70% confluence for optimal visualization

    • Consider three-dimensional culture systems for more physiologically relevant results

Examples of successful detection parameters include 1.7 μg/mL antibody for MCF-7 cells and 3 μg/mL for HeLa cells , highlighting the importance of cell-specific optimization.

Studying dynamic hypoxic responses with HIF-1α antibodies requires specialized approaches:

  • Time-course experiments:

    • Harvest cells at multiple time points after hypoxia induction (30 min, 1h, 2h, 4h, 8h, 24h)

    • Process parallel samples for Western blot and immunofluorescence

    • Quantify nuclear accumulation rate and total protein levels

  • Live-cell imaging:

    • Use cell-permeable fluorescently tagged antibody fragments (Fabs)

    • Engineer cell lines expressing HIF-1α fused to fluorescent proteins

    • Combine with hypoxia-indicating dyes for correlative analysis

  • Microfluidic hypoxia chambers:

    • Create oxygen gradients while enabling real-time microscopy

    • Apply immunofluorescence at fixed time points across the gradient

    • Correlate HIF-1α levels with precise oxygen measurements

  • Pulse-chase experiments:

    • Label newly synthesized proteins during hypoxia

    • Immunoprecipitate HIF-1α at different chase times

    • Determine protein synthesis and degradation rates

  • Reoxygenation studies:

    • Monitor HIF-1α levels during hypoxia followed by normoxia

    • Quantify degradation kinetics upon reoxygenation

    • Correlate with recovery of cellular functions

These approaches leverage the specificity of validated HIF-1α antibodies to provide insights into the temporal aspects of hypoxic adaptation, revealing how quickly cells respond to and recover from oxygen deprivation events.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.