HIF1A Human

Hypoxia-Inducible Factor-1 Alpha Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

HIF1A is a basic helix-loop-helix (bHLH) PAS domain-containing protein with distinct functional regions:

  • Domains: N-terminal transactivation domain (NTAD), C-terminal transactivation domain (CTAD), oxygen-dependent degradation (ODD) domain, and nuclear localization signal .

  • Isoforms: Three splice variants exist, but isoform 1 (canonical) is the most extensively studied .

Regulation and Function

HIF1A is constitutively expressed at low levels under normoxia but stabilized during hypoxia. Its activity is modulated by post-translational modifications (hydroxylation, acetylation, phosphorylation) .

Key Target Genes and Pathways:

  • Angiogenesis: VEGF, PDGF .

  • Metabolism: GLUT1, LDHA (enhanced glycolysis) .

  • Erythropoiesis: Erythropoietin (EPO) .

  • Cell Survival: BCL2, survivin .

Role in Cancer

HIF1A overexpression is a hallmark of solid tumors, correlating with aggressive progression and therapy resistance .

Table 2: HIF1A Overexpression in Human Cancers

Cancer TypeClinical AssociationStudy Findings
BreastPoor prognosis, therapy resistanceLinked to rs11549465/rs11549467 SNPs
GlioblastomaNeovascularization, VEGF upregulationCo-expression with VEGF
OvarianMetastasis, microvascular densityPredictive of mortality
ColorectalHypoxic microenvironment adaptationElevated in 80% of tumors

Polymorphisms:

  • rs11549465 (C/T) and rs11549467 (G/A) SNPs are associated with increased breast cancer risk in Asian populations (OR: 1.34–1.72) .

Ischemic Stroke:

  • Adaptive Role: Enhances glycolysis (GLUT1, PDK1) and angiogenesis (VEGF) .

  • Pathological Role: Prolonged activation exacerbates neuronal apoptosis .

Acute Organ Injury:

  • Lung: HIF1A stabilizes Adora2b to reduce inflammation in ALI .

  • Liver: Protects against ischemia-reperfusion injury via antioxidant genes .

  • Kidney: Hypoxic preconditioning reduces cisplatin-induced apoptosis .

Table 3: HIF1A Polymorphisms and Disease Associations

SNP IDPhenotype AssociationPopulationRisk Effect
rs11549465Breast, prostate cancerAsianIncreased (OR: 1.5)
rs11549467Cervical cancer, myocardial infarctionEuropeanMixed
rs2057482No significant associationMeta-analysisNeutral

Cofactors: 201 transcriptional cofactors identified, including STAT3, NF-κB, and SMAD3, which synergize with HIF1A to regulate hypoxia-responsive genes .

Therapeutic Targeting

  • Inhibitors: Echinomycin (blocks HIF1A-DNA binding) and PHD inhibitors (stabilize HIF1A for ischemic conditions) .

  • Preclinical Success: HIF1A knockdown exacerbates lung injury, while stabilization improves hepatic regeneration .

Knockout Models and Developmental Roles

HIF1A-null mice exhibit embryonic lethality (E10.5) due to defective vasculogenesis, underscoring its role in placental and cardiovascular development .

Product Specs

Introduction
Hypoxia-inducible factor-1 (HIF-1) is a transcription factor crucial for cellular and systemic oxygen homeostasis. It functions as a heterodimer composed of HIF-1b and one of three HIF-1a subunits. HIF-1 activation is strongly linked to various tumors and oncogenic pathways. Structurally, HIF-1 comprises a DNA binding domain (DBD), a dimerization domain, and C-terminal regulatory domains. These regulatory domains include two transactivation domains (TAD), an oxygen-dependent degradation (ODD) domain, and inhibitory domains. Under hypoxic conditions, HIF1A activates the transcription of over 40 genes, including those encoding erythropoietin, glucose transporters, glycolytic enzymes, VEGF, and others that enhance oxygen delivery or facilitate metabolic adaptation to hypoxia. HIF-1A is also essential for embryonic vascularization, tumor angiogenesis, and the pathophysiology of ischemic disease. It binds to the core DNA sequence 5'-[AG]CGTG-3' within the hypoxia response element (HRE) of target gene promoters. Activation involves recruiting transcriptional coactivators like CREBPB and EP300. Interaction with NCOA1 or NCOA2 enhances its activity. Interaction with the redox regulatory protein APEX activates CTAD and potentiates activation by NCOA1 and CREBBP. Induction occurs under reduced oxygen tension. HIF1A is also induced, though less intensely than hypoxia, by receptor-mediated factors such as growth factors (PDGF, EGF, FGF-2, IGF-2, TGF-1 beta, HGF), cytokines (TNF alpha, IL-1 beta), and circulatory factors (angiotensin-2, thrombin).
Description
HIF1A Human Recombinant, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 298 amino acids (530-826). It has a molecular mass of 32.8 kDa and migrates as a 32.8 kDa band on SDS-PAGE. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless solution.
Formulation
The HIF1A recombinant Human solution (1 mg/ml) is formulated in 20 mM Tris-HCl pH 7.5 and 1 mM DTT.
Stability
For short-term storage (2-4 weeks), store at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 90.0% purity as determined by SDS-PAGE analysis.
Synonyms
Hypoxia-inducible factor 1 alpha, HIF-1 alpha, HIF1 alpha, ARNT-interacting protein, Member of PAS protein 1, Basic-helix-loop-helix-PAS protein MOP1, HIF1A, MOP1, HIF1, PASD8, HIF-1A.
Source
Escherichia Coli.
Amino Acid Sequence
MEFKLELVEK LFAEDTEAKN PFSTQDTDLD LEMLAPYIPM DDDFQLRSFD QLSPLESSSA SPESASPQST VTVFQQTQIQ EPTANATTTT ATTDELKTVT KDRMEDIKIL IASPSPTHIH KETTSATSSP YRDTQSRTAS PNRAGKGVIE QTEKSHPRSP NVLSVALSQR TTVPEEELNP KILALQNAQR KRKMEHDGSL FQAVGIGTLL QQPDDHAATT SLSWKRVKGC KSSEQNGMEQ KTIILIPSDL ACRLLGQSMD ESGLPQLTSY DCEVNAPIQG SRNLLQGEEL LRALDQVN.

Q&A

What is HIF1A and what is its primary function in human cells?

HIF1A is the alpha subunit of transcription factor hypoxia-inducible factor-1 (HIF-1), which functions as a master regulator of cellular and systemic homeostatic responses to hypoxia. It activates numerous genes involved in energy metabolism, angiogenesis, apoptosis, and other processes that increase oxygen delivery or facilitate metabolic adaptation to hypoxic conditions. HIF1A plays essential roles in embryonic vascularization, tumor angiogenesis, and pathophysiology of ischemic diseases . The protein forms a heterodimer with a beta subunit to create the functional HIF-1 complex that drives transcriptional responses to low oxygen environments .

What are the key structural domains of HIF1A and their functions?

HIF1A contains several critical functional domains:

  • bHLHe78 domain: Basic helix-loop-helix DNA binding motif

  • PAS (Per-Arnt-Sim) domains: Facilitate protein-protein interactions, particularly dimerization with HIF1B

  • ODD (Oxygen-Dependent Degradation) domain: Contains proline residues targeted for hydroxylation under normoxic conditions

  • N-TAD and C-TAD: N-terminal and C-terminal transactivation domains required for transcriptional activity

These domains collectively enable HIF1A to sense oxygen levels, interact with cofactors, bind DNA, and activate gene transcription .

How is HIF1A expression and activity regulated at different biological levels?

HIF1A regulation occurs through multiple mechanisms:

  • Transcriptional regulation: The gene contains binding sites for several transcription factors.

  • Post-translational modification: Most notably, oxygen-dependent hydroxylation of specific proline residues by prolyl hydroxylases, leading to ubiquitination and proteasomal degradation under normoxia.

  • Protein-protein interactions: HIF1A interacts with over 200 potential cofactors that modify its function .

  • Epigenetic mechanisms: HIF1A binding is associated with H3K27ac modifications, suggesting epigenetic regulation of its target genes .

What are the most significant HIF1A polymorphisms linked to human diseases?

The most extensively studied HIF1A polymorphisms include:

SNP IDCommon NameGenomic LocationAssociated ConditionsRisk Assessment
rs115494651772 C/TODD domainCOPDC allele: protective (OR < 1)
rs115494651772 C/TODD domainSkin diseaseC allele: risk factor (OR > 1)
rs115494651772 C/TODD domainDiabetic complicationsC allele: risk factor (OR > 1)
rs115494671790 G/AODD domainCardiovascular diseaseA allele: protective
rs115494671790 G/AODD domainDiabetic complicationsA allele: risk factor

These polymorphisms affect the stability and transcriptional activity of the HIF1A protein, potentially altering downstream physiological responses to hypoxia .

How do researchers resolve contradictory findings regarding HIF1A polymorphisms and disease risk?

Contradictory findings are common in HIF1A polymorphism studies. For example, opposing results have been reported for rs11549465 in prostate cancer susceptibility studies, with some showing association and others showing no association . These inconsistencies typically arise from:

  • Small sample sizes

  • Heterogeneous populations

  • Different study designs and methodologies

  • Varying environmental factors

Researchers typically address these contradictions through:

  • Statistical meta-analysis combining multiple studies with appropriate weighting

  • Large-scale genome-wide association studies (GWAS)

  • Stratification by ethnic background and environmental factors

  • Functional validation studies of polymorphism effects

What techniques are most effective for studying HIF1A-DNA interactions?

Several complementary approaches are commonly used:

  • ChIP-seq (Chromatin Immunoprecipitation-sequencing): Maps genome-wide HIF1A binding sites, revealing that HIF1A predominantly binds enhancers with a smaller portion at promoters .

  • HiChIP (H3K27ac): Identifies three-dimensional chromatin interactions, allowing researchers to detect long-range interactions between HIF1A-bound enhancers and target gene promoters .

  • Dual luciferase reporter assay: Verifies direct interactions between HIF1A and specific promoter regions. For example, this technique has been used to confirm HIF1A binding to the ANGPTL2 promoter in cardiomyocytes .

  • ChIP followed by qPCR: Targets specific genomic regions to quantify HIF1A occupancy at candidate target genes.

How can researchers effectively modulate HIF1A activity in experimental systems?

Multiple approaches for manipulating HIF1A expression and activity include:

  • Genetic approaches:

    • siRNA/shRNA-mediated knockdown to reduce HIF1A expression

    • Overexpression using expression vectors (e.g., pcDNA3.1-HIF1A)

    • CRISPR-Cas9 genome editing for permanent genetic modification

  • Pharmacological approaches:

    • PHD inhibitors to stabilize HIF1A (e.g., FG-4592, dimethyloxalylglycine)

    • HIF1A inhibitors to block its activity (e.g., acriflavine, YC-1)

    • Hypoxia chambers to simulate physiological hypoxia

  • Cellular model selection:

    • Cell lines with differential HIF1A expression

    • Primary cells from different tissue origins

    • 3D organoid cultures to better mimic tissue architecture

What methodologies provide the most comprehensive assessment of HIF1A target genes?

A multi-omics approach yields the most complete picture:

  • Transcriptomics:

    • RNA-seq before and after HIF1A modulation

    • Single-cell RNA-seq to capture cell-type-specific responses

    • Analysis of temporal gene expression changes

  • Epigenomics:

    • ChIP-seq for HIF1A and histone modifications (particularly H3K27ac and H3K4me3)

    • ATAC-seq to identify changes in chromatin accessibility

    • DNA methylation analysis of HIF1A target gene promoters

  • Integration approaches:

    • Combined analysis of HIF1A ChIP-seq with RNA-seq to identify direct targets

    • Intersection of binding data with differentially expressed genes

    • Computational motif mining tools like SIOMICS to systematically identify HIF1A cofactors

How does HIF1A function differ between normal cells and cancer cells?

The activity and regulation of HIF1A show significant differences between normal and cancer cells:

  • Expression levels: Cancer cells often show higher baseline expression of HIF1A, even under normoxic conditions, due to genetic alterations, increased synthesis, or decreased degradation.

  • Cofactor interactions: Cancer cells exhibit unique patterns of HIF1A cofactor interactions. A systematic study identified 201 potential HIF1A cofactors in cancer cell lines, including 21 previously known and 180 novel cofactors .

  • Target gene specificity: The set of genes activated by HIF1A varies between normal and cancer cells, with cancer cells showing preferential activation of genes promoting survival, angiogenesis, and metabolic reprogramming.

  • Response to hypoxia: Cancer cells often show enhanced HIF1A stabilization and nuclear localization in response to hypoxia compared to normal cells.

What experimental approaches can identify novel HIF1A cofactors in specific cancer types?

Researchers can employ multiple strategies:

  • Computational approaches:

    • Motif mining tools like SIOMICS to analyze HIF1A ChIP-seq data and identify co-occurring transcription factor binding motifs

    • Protein-protein interaction network analysis

  • Biochemical approaches:

    • Co-immunoprecipitation followed by mass spectrometry

    • Proximity labeling techniques (BioID, APEX)

    • Yeast two-hybrid screening

  • Functional genomics:

    • CRISPR screens to identify genes that modify HIF1A activity

    • siRNA/shRNA screens targeting potential cofactors

The study by Nature Scientific Reports identified 37 top HIF1A cofactors in cancer cell lines, with 19 directly validated in literature and 18 representing novel interactions .

How can researchers address the challenge of targeting HIF1A in cancer treatment?

HIF1A presents unique challenges for therapeutic targeting:

  • Selectivity issues:

    • Differential targeting between cancer cells and normal cells undergoing physiological hypoxia

    • Distinguishing between HIF1A and related family members (HIF2A, HIF3A)

  • Experimental approaches:

    • Testing combination therapies targeting both HIF1A and its key cofactors

    • Developing cancer-specific delivery systems for HIF1A inhibitors

    • Targeting downstream effectors with greater tissue specificity

  • Biomarker development:

    • Identification of HIF1A-specific gene signatures for patient stratification

    • Analysis of HIF1A polymorphisms as predictors of treatment response

    • Monitoring HIF1A activity in real-time during treatment

How does HIF1A contribute to cardiovascular disease pathophysiology?

HIF1A influences cardiovascular disease through multiple mechanisms:

  • Genetic associations: HIF1A polymorphisms are associated with:

    • Peripheral artery disease

    • Premature coronary artery disease

    • Ischemic heart disease (coronary artery collaterals)

  • Cellular mechanisms:

    • In cardiomyocytes, knockdown of HIF1A has been shown to downregulate ANGPTL2, alleviating high glucose-stimulated hypoxia/reoxygenation injury

    • HIF1A regulates metabolic adaptation during cardiac ischemia

    • Influences angiogenic responses after myocardial infarction

  • Therapeutic implications:

    • Transient HIF1A activation may be protective in acute cardiac events

    • Chronic HIF1A activation might contribute to adverse cardiac remodeling

What is the role of HIF1A in inflammatory and immune responses?

HIF1A has emerged as a critical regulator of inflammation:

  • Gene regulation: HIF1A controls the expression of pro-inflammatory cytokines and chemokines:

    • Regulates IL-6 and CXCL8 expression

    • Controls RUNX1, which is involved in pro-inflammatory processes

  • Immune cell function:

    • Influences metabolic reprogramming in immune cells

    • Affects macrophage polarization and function

    • Modulates T-cell differentiation and activity

  • Disease implications:

    • HIF1A polymorphisms are associated with inflammatory skin diseases

    • Contributes to pathology in inflammatory conditions like COPD

    • Influences tissue repair processes through modulation of extracellular matrix genes

How does HIF1A interact with epigenetic mechanisms to regulate gene expression?

HIF1A extensively cooperates with epigenetic machinery:

  • Histone modification interactions:

    • HIF1A binding correlates with increased H3K27ac (activating mark) but not H3K27me3 (repressive mark)

    • Predominantly binds regions with enhancer-associated H3K4me1 modifications

  • Chromatin structure impacts:

    • Facilitates long-range chromatin interactions connecting distal enhancers to gene promoters

    • HIF1A binding observed at super-enhancers (SEs) that regulate key genes like RUNX1

  • Genome organization:

    • Three-dimensional genome technology (HiChIP) has identified genes remotely regulated by HIF1A, extending research beyond promoter regions

    • HIF1A influences enhancer-promoter interactions under hypoxic conditions

What are promising future research directions for HIF1A in human disease contexts?

Several emerging areas show particular promise:

  • Single-cell analysis:

    • Single-cell transcriptomics and spatial approaches to understand cell-type-specific HIF1A functions

    • Cell-specific responses to hypoxia in heterogeneous tissues

  • Systems biology approaches:

    • Network analysis of HIF1A interactions with other transcription factors

    • Mathematical modeling of HIF1A regulatory networks

  • Translational opportunities:

    • HIF1A polymorphisms as biomarkers for disease risk stratification

    • Precision medicine approaches based on patient-specific HIF1A function

    • Development of cell-type specific HIF1A modulators

  • Role in emerging disease areas:

    • HIF1A functions in metabolic reprogramming during aging

    • Contributions to neurodegenerative diseases

    • Involvement in infectious disease response

How can researchers integrate multi-omics data to better understand HIF1A function?

Integrative approaches offer the most comprehensive understanding:

  • Data integration strategies:

    • Combining HIF1A ChIP-seq, RNA-seq, and histone modification data

    • Integrating proteomics and transcriptomics to identify post-transcriptional regulation

    • Correlating genetic variation with functional outcomes

  • Computational methods:

    • Machine learning approaches to predict context-specific HIF1A binding

    • Network analysis to identify key regulatory hubs

    • Pathway enrichment to understand biological consequences

  • Validation approaches:

    • Functional genomics to confirm computational predictions

    • Targeted experiments based on multi-omics insights

    • In vivo models to validate findings from cell-based systems

Product Science Overview

Introduction

Hypoxia-Inducible Factor-1 Alpha (HIF-1α) is a crucial transcription factor that plays a significant role in the cellular response to low oxygen levels (hypoxia). It is a subunit of the heterodimeric transcription factor Hypoxia-Inducible Factor-1 (HIF-1), which is composed of HIF-1α and the aryl hydrocarbon receptor nuclear translocator (ARNT), also known as HIF-1β .

Structure and Isoforms

HIF-1α is a basic helix-loop-helix (bHLH) PAS domain-containing protein. It consists of several functional domains, including:

  • bHLH domain: Involved in DNA binding and dimerization.
  • PAS domains: Important for dimerization with HIF-1β.
  • Nuclear localization signal: Directs the protein to the nucleus.
  • Transactivation domains (CTAD and NTAD): Essential for transcriptional activation.
  • Inhibitory domain (ID): Represses the transcriptional activities of CTAD and NTAD .

There are three known isoforms of HIF-1α, formed by alternative splicing. Isoform 1 is the most extensively studied and is considered the canonical structure .

Biological Functions

HIF-1α is the master regulator of the cellular and developmental response to hypoxia. It activates the transcription of various genes involved in:

  • Energy metabolism: Enhances glycolysis and reduces mitochondrial oxygen consumption.
  • Angiogenesis: Promotes the formation of new blood vessels by upregulating vascular endothelial growth factor (VEGF).
  • Cell survival: Induces the expression of genes that help cells survive under low oxygen conditions.
  • Tumor invasion: Facilitates cancer cell adaptation and invasion in hypoxic tumor microenvironments .
Expression and Regulation

The expression of HIF-1α is tightly regulated by oxygen levels. Under normoxic conditions (normal oxygen levels), HIF-1α is rapidly degraded by the proteasome. However, under hypoxic conditions, HIF-1α is stabilized and translocates to the nucleus, where it dimerizes with HIF-1β and activates target gene transcription .

Role in Disease

Dysregulation and overexpression of HIF-1α are implicated in various pathophysiological conditions, including:

  • Cancer: HIF-1α promotes tumor growth and metastasis by enhancing angiogenesis and metabolic adaptation.
  • Ischemic diseases: HIF-1α plays a protective role in conditions such as stroke and myocardial infarction by promoting cell survival and tissue repair.
  • Placental development: HIF-1α is essential for normal placental development during early gestation .
Recombinant HIF-1α

Recombinant human HIF-1α is produced using recombinant DNA technology, which involves inserting the HIF1A gene into an expression vector and introducing it into a host cell (e.g., E. coli or mammalian cells) to produce the protein. This recombinant protein is used in various research applications to study its structure, function, and role in disease .

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.