HIF1A Human, His mimics endogenous HIF-1α’s role in hypoxia adaptation. Key mechanisms include:
Oxygen sensing: Under hypoxia, HIF-1α escapes VHL-mediated ubiquitination and degradation, enabling dimerization with HIF-1β to form the active HIF-1 complex .
Target gene regulation: Binds hypoxia-response elements (HREs) in promoters of genes like VEGF, EPO, and GLUT1, driving angiogenesis, erythropoiesis, and glycolysis .
Post-translational modifications: Hydroxylation at Pro402/Pro564 by PHD enzymes regulates its stability .
Cancer: Overexpression of HIF1A Human, His in tumor models enhances VEGF production, promoting angiogenesis and metastasis . Polymorphisms in HIF1A (e.g., rs11549465) correlate with increased cancer susceptibility .
Ischemic Injury: Stabilized HIF1A reduces mitochondrial damage in hepatic ischemia-reperfusion injury by upregulating antioxidant genes (e.g., HO-1) .
Acute Lung Injury (ALI): HIF1A knockdown exacerbates pulmonary edema, while its stabilization via DMOG attenuates inflammation .
While HIF1A Human, His is indispensable for hypoxia research, challenges include:
Lack of post-translational modifications: Recombinant protein lacks hydroxylation/acetylation observed in vivo.
Cell-type specificity: Effects vary between cancer, endothelial, and immune cells .
Future studies may focus on isoform-specific roles (HIF-1α vs. HIF-2α) and CRISPR-engineered variants for precision medicine .
HIF1A is the alpha subunit of the heterodimeric transcription factor HIF1, which responds to low oxygen availability. Its structure includes several key functional domains:
An oxygen-dependent degradation domain (ODD)
Two transactivation domains (N-TAD and C-TAD)
A basic helix-loop-helix (bHLH) domain for DNA binding
A PAS (Per-ARNT-Sim) domain for heterodimerization
In normoxia, prolyl hydroxylases modify the ODD domain, marking HIF1A for recognition by von Hippel-Lindau (VHL) protein, leading to ubiquitination and degradation. Under hypoxic conditions, this hydroxylation is inhibited, allowing HIF1A to stabilize, translocate to the nucleus, dimerize with HIF1B, and activate transcription of target genes .
HIF1A acts as a master regulator of the cellular response to hypoxia, controlling multiple physiological processes:
Metabolism and redox homeostasis - glucose catabolism and lipid metabolism regulation
Vascular responses - ischemia-induced angiogenesis and endothelial cell function
Cancer-related processes - tumorigenesis, metastasis, and tumor angiogenesis
Inflammatory responses - regulation in inflammatory cells and myeloid cell function
System-wide hypoxia adaptation mechanisms
Analysis of 98 HIF1A target genes from published studies revealed 20 associated pathways. Additionally, HIF1A regulates both protein-coding genes and non-coding RNA genes, including microRNAs and transcribed-ultraconserved regions .
HIF1A undergoes extensive post-translational regulation through:
Oxygen-dependent mechanisms:
Prolyl hydroxylation by PHD1-3 at Pro402 and Pro564 in normoxia
Asparaginyl hydroxylation by Factor Inhibiting HIF (FIH) at Asn803
Oxygen-independent mechanisms:
Phosphorylation by various kinases (MAPK, Akt, GSK3β)
SUMOylation affecting protein stability
Acetylation modifying transcriptional activity
S-nitrosylation enhancing stability
This multilayered regulation enables precise control of HIF1A function in response to both oxygen levels and other cellular signals .
To investigate oxygen-independent HIF1A activation:
Employ reporter constructs containing HIF1A-responsive elements to measure transcriptional activity
Use Western blotting with phospho-specific antibodies to detect activation-specific modifications
Implement immunoprecipitation followed by mass spectrometry to identify post-translational modifications
Utilize CRISPR-Cas9 to introduce mutations at key regulatory residues
Apply pharmacological inhibitors of specific pathways (PI3K/Akt/mTOR, inflammatory signaling)
When designing experiments, maintain normoxic conditions (21% O₂) while manipulating specific pathways. Include appropriate controls to differentiate between oxygen-dependent and independent mechanisms of HIF1A activation .
Literature reviews have identified 34 HIF1A SNPs tested for associations with 49 phenotypes, with 16 SNPs showing positive associations with 40 phenotypes, including 14 cancer types. The most consistently associated polymorphisms are:
rs11549465 (C1772T, Pro582Ser) - A missense polymorphism in the ODD domain
rs11549467 (G1790A, Ala588Thr) - Another missense polymorphism in the ODD domain
These polymorphisms are associated with breast, lung, colorectal, gastric, prostate, oral cancer, and renal cell carcinoma. Their location within the ODD domain suggests they may affect HIF1A stability and degradation, potentially leading to enhanced activity .
Mechanistically, these polymorphisms may:
Alter protein stability by modifying the ODD domain
Change binding affinity to VHL protein
Modify transcriptional activity through altered cofactor recruitment
Impact interactions with other regulatory proteins
It's worth noting that some studies show conflicting results for the same polymorphism and cancer type, highlighting the complex nature of these associations .
HIF1A polymorphisms have been associated with various non-cancer conditions:
Cardiovascular diseases:
Ischemic heart disease
Coronary artery disease (CAD)
Pre-eclampsia
Acute myocardial infarction
Frequent intradialytic hypotension
The primary polymorphisms implicated include rs10873142, rs11549465, rs11549467, rs2057482, rs2783778, rs41508050, and rs7148720 .
Metabolic disorders:
Diabetic nephropathy
Type 1 diabetes mellitus (T1DM)
Type 2 diabetes mellitus (T2DM)
The rs11549465 polymorphism shows associations with all these metabolic conditions, while rs12434438 and rs1319462 have been linked specifically to T2DM .
The mechanistic basis likely involves the central role of HIF1A in regulating vascular responses to hypoxia, tissue repair mechanisms, and metabolic adaptations. For instance, in cardiovascular diseases, altered HIF1A function may affect ischemic responses, angiogenesis, and vascular remodeling .
HIF1A responses display significant cell-type specificity. Comparative studies show that only 51 genes (canonical HIF-1 targets) were common among different cell types, while many more cell-specific targets were observed .
Several factors contribute to this specificity:
Chromatin accessibility - Cell-specific open chromatin regions at HIF1A binding sites
Interactions with cell-specific transcription factors - For example, enrichment of both HIF1A and OLIG2 in oligodendrocytes
Epigenetic landscape - Distinct histone modification patterns affect HIF1A binding
Expression of cofactors - Cell-specific expression of transcriptional coactivators
Post-translational modifications - Cell-specific enzymes modify HIF1A differently
These mechanisms enable precise, context-dependent responses to hypoxia across different tissues, allowing for specialized adaptation strategies rather than a universal response .
To study cell-specific HIF1A functions:
Select appropriate cellular models:
Use primary cells rather than immortalized lines when possible
Consider 3D culture systems that better preserve tissue architecture
Develop co-culture systems to account for intercellular interactions
Implement genomic approaches:
Perform cell type-specific ChIP-seq for HIF1A binding patterns
Combine with ATAC-seq to assess chromatin accessibility
Use single-cell RNA-seq to capture heterogeneity in responses
Apply genetic strategies:
Develop cell type-specific HIF1A knockout systems
Create reporter systems to monitor HIF1A activity in specific cells
Use CRISPR screens to identify cell-specific cofactors
Physiologically relevant conditions:
Apply tissue-appropriate O₂ concentrations (not 21% or 0%)
Consider physiological nutrient conditions
Account for microenvironmental factors like ECM composition
These approaches help delineate how HIF1A functions uniquely in different cell contexts .
The gut microbiota influences HIF1A signaling through several sophisticated mechanisms:
Production of short-chain fatty acids (SCFAs):
Butyrate, produced by gut bacteria, stabilizes HIF1A in intestinal epithelial cells
This occurs even under normoxic conditions, suggesting oxygen-independent activation
Butyrate inhibits histone deacetylase activity, increasing chromatin accessibility at HIF1A target genes
Maintenance of physiological hypoxia:
The microbiota/SCFA axis is essential for maintaining the normal oxygen gradient in intestinal tissue
This physiological hypoxia gradient is critical for intestinal epithelial function
Germ-free mice show altered oxygen distribution in the intestine
Epigenetic modulation:
Conversely, HIF1A affects host-microbiome interactions by regulating:
Antimicrobial peptide production
Mucin secretion
Intestinal barrier integrity
Local immune responses
This reciprocal relationship forms a homeostatic circuit important for intestinal health .
The microbiota-HIF1A axis offers several therapeutic intervention points:
Microbiota manipulation strategies:
Probiotics containing butyrate-producing bacteria
Prebiotics to selectively promote growth of beneficial microbes
Fecal microbiota transplantation to restore healthy microbial communities
Metabolite-based approaches:
Direct supplementation with SCFAs, particularly butyrate
Development of SCFA analogs with improved pharmacokinetics
Targeted delivery systems for intestinal release
HIF1A pathway modulation:
PHD inhibitors to stabilize HIF1A protein
Epigenetic modifiers to enhance HIF1A target gene accessibility
Cell-specific HIF1A activation strategies
Combined approaches:
Synergistic therapies targeting both microbiota composition and HIF1A activity
Personalized interventions based on individual microbiome profiles
For implementation, researchers should consider:
Timing of intervention (acute vs. chronic)
Disease-specific approaches (IBD vs. colorectal cancer)
Cell-type specific effects in the complex intestinal ecosystem
Potential systemic effects beyond the gut
Research methodologies should include both reductionist approaches and systems-level analysis to capture the complexity of this axis .
Studying HIF1A polymorphisms requires rigorous methodological approaches:
Study design:
Power calculations based on expected effect sizes and polymorphism frequencies
Careful matching of cases and controls for confounding variables
Consideration of multiple genetic models (dominant, recessive, additive)
Multi-ethnic approach to account for population differences
Technical approaches:
Selection of appropriate genotyping methods with adequate quality control
Validation of key findings using alternative techniques
Standardized phenotype definitions
Adjustment for multiple testing
Functional validation:
Reporter assays to test effects on transcriptional activity
CRISPR-Cas9 to introduce specific polymorphisms into cell models
Assessment of protein stability, localization, and interactions
Measurement of target gene expression
Data integration:
Combining results from multiple studies through meta-analysis
Integration with functional genomic data (eQTLs, epigenetic marks)
Pathway-level analysis rather than single-gene focus
Consideration of gene-environment interactions
This comprehensive approach is essential given the complex relationship between HIF1A variants and disease phenotypes .
Addressing contradictory findings in HIF1A research requires systematic approaches:
Experimental condition standardization:
Precise control and reporting of oxygen levels
Standardization of hypoxia duration
Consistent cell densities in culture systems
Detailed documentation of culture media and supplements
Model system considerations:
Recognize cellular context specificity of HIF1A responses
Account for species differences in the HIF pathway
Consider compensatory mechanisms (HIF2A, HIF3A upregulation)
Evaluate the influence of genetic background
Analytical approaches:
Meta-analysis of multiple studies
Systematic reviews with predefined inclusion criteria
Bayesian analysis to integrate diverse data types
Publication bias assessment
Validation strategies:
Independent replication in different laboratories
Use of multiple complementary techniques
Both gain-of-function and loss-of-function approaches
Application of newer technologies (CRISPR-Cas9, single-cell analysis)
When encountering contradictory literature, researchers should systematically evaluate methodological differences, consider biological context, and design experiments that can directly address the specific contradictions .
Translational opportunities in HIF1A research include:
Therapeutic targeting:
PHD inhibitors for ischemic diseases, anemia, and inflammatory conditions
HIF1A inhibitors for cancer, where it drives malignant progression
Cell-type specific HIF1A modulation to minimize off-target effects
Combinatorial approaches targeting HIF1A alongside other pathways
Biomarker development:
HIF1A polymorphisms as predictive markers for disease risk
HIF1A activity signatures as prognostic indicators
Downstream targets as surrogate markers for treatment response
Integration into multi-marker panels for personalized medicine
Microbiome-based interventions:
Targeting the microbiota-HIF1A axis for intestinal diseases
Development of prebiotics/probiotics to modulate HIF1A activation
Dietary approaches to optimize the gut environment
Emerging technologies:
RNA therapeutics targeting HIF1A or its regulators
Gene editing to correct pathogenic HIF1A variants
Nanotechnology-based delivery systems for tissue-specific targeting
These translational approaches hold promise for addressing conditions with hypoxic components, including cardiovascular disease, cancer, and inflammatory disorders .
Key unresolved questions in HIF1A biology include:
Cell-type specificity:
How do chromatin structure and epigenetic modifications direct cell-specific HIF1A responses?
What determines which genes HIF1A activates in different cellular contexts?
How do tissue-specific transcription factors interact with HIF1A?
Temporal dynamics:
How does acute versus chronic HIF1A activation affect cellular outcomes?
What mechanisms govern adaptation to prolonged hypoxia?
How do cells "remember" previous hypoxic episodes (hypoxic memory)?
Non-canonical functions:
What are the roles of HIF1A beyond transcriptional regulation?
How does cytoplasmic HIF1A influence cellular processes?
Are there functions independent of the canonical HIF1A/HIF1B heterodimer?
Regulatory complexity:
How do different oxygen sensors coordinate responses?
What is the significance of HIF1A regulation by non-coding RNAs?
How do post-translational modifications fine-tune HIF1A activity?
Microbiota interactions:
What mechanisms beyond SCFAs link the microbiome to HIF1A?
How does the oxygen gradient in different intestinal regions affect this relationship?
Can microbiota-derived signals activate HIF1A systemically?
Addressing these questions will require interdisciplinary approaches combining molecular biology, genetics, computational biology, and systems-level analysis .
HIF-1α is a basic helix-loop-helix PAS domain-containing protein. It is considered the master transcriptional regulator of cellular and developmental responses to hypoxia . The protein structure includes a nuclear localization signal motif, two transactivating domains (CTAD and NTAD), and an intervening inhibitory domain (ID) that can repress the transcriptional activities of CTAD and NTAD .
Under normal oxygen conditions, HIF-1α is rapidly degraded. However, in hypoxic conditions, it stabilizes and translocates to the nucleus, where it dimerizes with HIF-1β. This complex then binds to hypoxia-responsive elements (HREs) in the promoter regions of target genes, activating the transcription of genes involved in various adaptive responses, including angiogenesis, erythropoiesis, and glycolysis .
The dysregulation and overexpression of HIF-1α have been heavily implicated in cancer biology and other pathophysiologies. It plays a critical role in areas such as vascularization and angiogenesis, energy metabolism, cell survival, and tumor invasion . Additionally, HIF-1α is essential for normal placental development in early gestation .
The recombinant form of HIF-1α, tagged with a His (histidine) tag, is used in various research applications. The His tag facilitates the purification of the protein using affinity chromatography, making it easier to study its structure and function in detail. This recombinant protein is crucial for investigating the regulatory mechanisms of HIF-1α and its role in hypoxia-related diseases .
Research on HIF-1α has significant therapeutic potential, particularly in cancer treatment. By understanding how HIF-1α regulates the cellular response to hypoxia, scientists can develop targeted therapies to inhibit its activity in tumors, potentially limiting cancer progression and metastasis .
In conclusion, Hypoxia-Inducible Factor-1 Alpha (Human Recombinant, His Tag) is a vital tool in hypoxia research, providing insights into the molecular mechanisms of oxygen homeostasis and its implications in various diseases.