AKR7A3 is a 331-amino-acid protein (ENSP00000355377) encoded by the AKR7A3 gene on chromosome 1p36. Key structural features include:
Catalytic Activity: NADPH-dependent reduction of aflatoxin B1 dialdehyde to non-binding dialcohol, preventing DNA adduct formation .
Quaternary Structure: Forms dimers via interactions between helices α5–α6 and loop C, distinct from other AKR family members .
Tissue Distribution: Expressed primarily in liver, kidney, stomach, and pancreas .
Functional Partners (from STRING interaction network):
Protein | Score | Key Function |
---|---|---|
AKR7A2 | 0.985 | Reduces succinic semialdehyde to gamma-hydroxybutyrate; broad substrate specificity |
AKR1B1 | 0.874 | Key enzyme in polyol pathway; detoxifies aldehydes and ketones |
AKR1A1 | 0.732 | Reduces toxic aldehydes like methylglyoxal |
AKR7A3 is frequently downregulated in hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC), acting as a tumor suppressor.
Downregulation Mechanisms:
Functional Impact: Overexpression inhibits colony formation, migration, and invasion, while knockdown promotes oncogenicity .
Prognostic Significance: Lower AKR7A3 expression correlates with poor survival and advanced stage .
Mechanisms: Suppresses metastasis and chemoresistance by regulating PHGDH (phosphoglycerate dehydrogenase)-induced autophagy .
AKR7A3 modulates key oncogenic pathways:
Pathway | Effect of AKR7A3 Overexpression | Biological Outcome |
---|---|---|
ERK | Inhibits phosphorylation | Reduced cell proliferation |
c-Jun | Attenuates activation | Suppressed metastasis |
NF-κB | Blocks nuclear translocation | Enhanced chemosensitivity |
These effects explain its role in sensitizing HCC cells to cisplatin and inhibiting tumor growth in vivo .
Biomarker: AKR7A3 expression levels correlate with HCC prognosis and early PDAC detection .
Therapeutic Target: Restoring AKR7A3 activity may mitigate chemoresistance and metastasis, particularly in liver and pancreatic cancers .
Mechanistic Clarity: Further studies needed to elucidate how AKR7A3 regulates PHGDH in PDAC.
Therapeutic Strategies: Development of small-molecule activators or gene therapy to restore AKR7A3 expression in cancers.
AKR7A3, also known as AFAR2 (aflatoxin aldehyde reductase 2), is a protein encoded by the AKR7A3 gene located on chromosome 1p in humans. It belongs to the aldo-keto reductase family, which consists of enzymes involved in the reduction of various aldehydes and ketones in the body . In normal cellular function, AKR7A3 primarily serves in detoxification pathways, particularly in the metabolism and neutralization of potentially harmful aldehydes and ketones .
The protein plays a critical role in liver metabolism, where it helps reduce aflatoxin B1, a potent hepatocarcinogen. This protective function explains its importance in preventing liver damage that could potentially lead to HCC. Additionally, AKR7A3 appears to regulate several signaling pathways crucial for normal cell growth and differentiation.
AKR7A3 expression can be measured at both mRNA and protein levels using several methodologies:
mRNA Level Measurements:
Quantitative Real-Time PCR (qRT-PCR): This technique requires careful primer design due to high sequence homology with other AKR family members. Primers should amplify unique sequences of 100-200 bp length and undergo validation through sequencing .
Transcriptome sequencing (RNA-seq): This provides comprehensive gene expression data and can identify differential expression between tumor and non-tumor tissues .
Protein Level Measurements:
Western blot analysis: Using specific antibodies against AKR7A3 .
Multiple Reaction Monitoring (MRM): This mass spectrometry-based technique uses unique peptides for quantification, though selecting appropriate peptides can be challenging due to sequence homology with other AKR family members .
Immunohistochemistry (IHC): Requires specific monoclonal antibodies developed against full-length or truncated recombinant AKR7A3 .
When developing antibodies for AKR7A3 detection, validation criteria should include: no cross-reactions with other AKR recombinant proteins, and demonstration of a single band in Western blot analysis against cell lysate .
AKR7A3 down-regulation is significantly associated with several clinicopathological features of HCC:
These associations point to AKR7A3 being not only a potential prognostic marker but also a possible therapeutic target in HCC. The table below summarizes the relationship between AKR7A3 expression and clinicopathological features:
Features | Total | AKR7A3 expression | P value | |
---|---|---|---|---|
Normal expression | Down-regulation | |||
Serum α-fetoprotein level (ng/ml) | ||||
≤ 400 | 64 | 50 (78.1%) | 14 (21.9%) | < 0.001 |
> 400 | 56 | 25 (44.6%) | 31 (55.4%) | |
Differentiation | ||||
Well differentiated (I-II) | 66 | 49 (74.2%) | 17 (25.8%) | 0.011 |
Moderately differentiated (II-III) | 44 | 21 (47.7%) | 23 (52.3%) | |
Poorly differentiated (III-IV) | 3 | 1 (33.3%) | 2 (66.7%) |
Research has identified two primary mechanisms responsible for AKR7A3 down-regulation in HCC:
Epigenetic silencing through promoter hypermethylation:
Bisulfite genomic sequencing and methylation-specific PCR have demonstrated that the promoter region of AKR7A3 is frequently hypermethylated in HCC cell lines with low AKR7A3 expression (such as QGY7703 and PLC8024) .
Cell lines with higher AKR7A3 expression (97L and H2M) show significantly less methylation in the promoter region .
Genetic deletion through Loss of Heterozygosity (LOH):
AKR7A3 is located on chromosome 1p, a region frequently deleted in HCC .
LOH analysis revealed that 43% of HCC samples with down-regulated AKR7A3 showed allelic loss .
The relative down-regulation of AKR7A3 was significantly more pronounced in HCC samples with LOH compared to those without LOH (P < 0.05) .
These findings suggest that both epigenetic and genetic alterations contribute to the silencing of AKR7A3 in HCC, highlighting potential targets for therapeutic intervention.
Overexpression Techniques:
Vector selection: For AKR7A3 overexpression studies, researchers typically use mammalian expression vectors containing full-length AKR7A3 cDNA.
Transfection methods:
Stable transfection: Useful for long-term studies and in vivo experiments, involves selection with antibiotics.
Transient transfection: Suitable for short-term assays.
Validation methods:
Western blot analysis with specific AKR7A3 antibodies
qRT-PCR for mRNA expression confirmation
Knockdown Techniques:
siRNA approach: Small interfering RNAs designed to target specific regions of AKR7A3 mRNA.
shRNA approach: Short hairpin RNAs for stable knockdown of AKR7A3.
CRISPR-Cas9 system: For complete gene knockout studies.
Experimental validation:
For both overexpression and knockdown studies, functional validation should include:
Cell proliferation assays (MTT, BrdU incorporation)
Colony formation assays
Migration and invasion assays
Apoptosis detection (Annexin V/PI staining)
Analysis of downstream signaling pathways (Western blot for ERK, c-Jun, NF-κB phosphorylation)
Based on established research protocols, the following functional assays should be implemented to comprehensively evaluate AKR7A3's tumor suppressive functions:
In vitro assays:
Foci formation: Assess cell contact inhibition and transformation capacity.
Colony formation in soft agar: Evaluate anchorage-independent growth.
Migration assays: Using Boyden chambers or wound healing assays.
Invasion assays: Using Matrigel-coated transwell chambers.
Chemosensitivity assays: Treating cells with cisplatin or other chemotherapeutic agents to assess AKR7A3's role in chemoresistance .
In vivo assays:
Tumor formation in nude mice: Subcutaneous injection of AKR7A3-modified cells.
Metastasis models: Tail vein injection or orthotopic implantation.
Drug response studies: Testing chemotherapeutic agents in tumor-bearing mice.
Molecular signaling analysis:
Western blot: Detection of EMT markers and phosphorylation status of ERK, c-Jun, and NF-κB.
Co-immunoprecipitation: Investigating protein-protein interactions.
Reporter gene assays: Evaluating transcriptional activity of target pathways.
AKR7A3's tumor suppressive function operates through its interaction with several key signaling pathways:
ERK Signaling Pathway:
c-Jun Pathway:
NF-κB Signaling:
EMT Regulation:
To further elucidate these interactions, researchers should consider combining pharmacological inhibitors of these pathways with AKR7A3 modulation, and employ co-immunoprecipitation to identify direct protein interactions.
AKR7A3 plays a significant role in modulating chemotherapeutic response, particularly to cisplatin treatment:
Sensitization to apoptosis:
Molecular mechanisms:
The chemosensitizing effect of AKR7A3 may be mediated through its inhibitory effect on ERK, c-Jun, and NF-κB signaling pathways .
These pathways are known to promote cell survival and resistance to apoptosis when activated.
By attenuating these pro-survival signals, AKR7A3 may enhance the efficacy of chemotherapeutic agents.
Implications for therapy:
The correlation between AKR7A3 expression and chemosensitivity suggests potential clinical applications:
AKR7A3 could serve as a predictive biomarker for chemotherapy response.
Restoring AKR7A3 expression or function might enhance the efficacy of conventional chemotherapy.
Combination therapies targeting both AKR7A3-related pathways and using conventional chemotherapy might improve treatment outcomes.
Researchers investigating this relationship should design studies that combine AKR7A3 modulation with various chemotherapeutic agents, analyze apoptotic signaling pathways, and consider drug efflux mechanisms that might be affected by AKR7A3.
The high sequence homology among AKR family members presents significant challenges for specific detection and analysis of AKR7A3. Researchers should employ the following strategies:
Primer design for qRT-PCR:
Antibody selection and validation:
Use monoclonal antibodies specifically developed against AKR7A3.
Validate antibody specificity through dot blotting against recombinant AKR proteins .
Confirm specificity with Western blot analysis in cell lysates, demonstrating a single band of the expected molecular weight .
Perform antibody validation using both positive controls (AKR7A3-expressing cells) and negative controls (AKR7A3-knockdown cells).
Mass spectrometry approaches:
Bioinformatic analysis:
Conduct thorough sequence alignment to identify unique regions of AKR7A3.
Use tools like BLAST and protein databases to ensure that selected regions do not cross-react with other AKR members.
By implementing these methodological precautions, researchers can ensure specific and accurate analysis of AKR7A3 despite the high sequence homology challenges.
Analysis of AKR7A3 in clinical samples requires special considerations to ensure accurate results and meaningful clinical correlations:
Sample collection and processing:
Collect paired tumor and adjacent non-tumor tissue samples to allow for direct comparison.
Process samples immediately or preserve appropriately to prevent RNA/protein degradation.
Consider using laser capture microdissection for heterogeneous tumor samples.
Expression analysis:
mRNA level: Perform qRT-PCR with validated primers targeting unique regions of AKR7A3.
Protein level:
Immunohistochemistry using validated antibodies is ideal for tissue samples.
Western blot analysis for semi-quantitative protein detection.
Consider tissue microarrays for high-throughput analysis of multiple samples.
Genetic and epigenetic analysis:
Methylation analysis:
Bisulfite sequencing or methylation-specific PCR to assess promoter methylation status.
Compare methylation patterns with expression levels.
LOH detection:
Clinical correlation analysis:
Statistical considerations:
Use appropriate statistical tests based on data distribution.
Adjust for multiple testing when necessary.
Include multivariate analysis to identify independent prognostic factors.
By employing these comprehensive approaches, researchers can generate reliable data on AKR7A3 in clinical samples that may have significant implications for patient stratification and treatment planning.
Based on current understanding of AKR7A3's tumor suppressive functions, several therapeutic strategies are being explored:
Epigenetic modulation:
Gene therapy approaches:
Delivery of AKR7A3 via viral vectors to tumor cells.
CRISPR-based gene editing to correct mutations or enhance expression.
mRNA-based therapeutics to temporarily restore AKR7A3 function.
Small molecule activators:
Development of compounds that enhance AKR7A3 enzymatic activity or stability.
Drugs that mimic AKR7A3's effects on downstream signaling pathways (ERK, c-Jun, NF-κB inhibitors).
Combination therapies:
Personalized medicine approaches:
Stratifying patients based on AKR7A3 expression levels to guide treatment decisions.
Developing companion diagnostics to identify patients most likely to benefit from AKR7A3-targeted therapies.
Researchers investigating these therapeutic strategies should consider both direct approaches targeting AKR7A3 itself and indirect approaches targeting the pathways it regulates, while carefully evaluating specificity, efficacy, and potential side effects.
While AKR7A3's role in HCC is well-documented, its potential functions in other cancer types warrant investigation:
Expression patterns across cancer types:
Researchers should conduct comprehensive analysis of AKR7A3 expression in various cancer types using:
Cancer genome databases (TCGA, ICGC)
Tissue microarrays of multiple cancer types
Cell line panels representing diverse cancer origins
Potential mechanisms in other cancers:
Detoxification function: AKR7A3's role in neutralizing carcinogens may be relevant in smoking-related cancers (lung, bladder, head and neck).
Signaling pathway regulation: The inhibitory effects on ERK, c-Jun, and NF-κB pathways may have broad implications across cancer types .
Chemoresistance modulation: AKR7A3's influence on drug sensitivity could extend to various cancer types treated with platinum-based therapies.
Tissue-specific functions:
AKR7A3 may have additional, tissue-specific functions depending on the metabolic context.
Its interaction with tissue-specific transcription factors or metabolites might reveal novel mechanisms.
Correlation with environmental exposures:
Given AKR7A3's role in detoxification, its relevance in cancers associated with specific environmental carcinogens should be explored.
The relationship between AKR7A3 expression and exposure to aflatoxins or other environmental toxins in different cancer types.
Research methodologies:
Comparative analysis of AKR7A3 function across multiple cancer cell lines.
Transgenic mouse models with tissue-specific AKR7A3 modulation.
Systems biology approaches to identify cancer-specific interactions and functions.
This broader investigation of AKR7A3 beyond HCC might reveal previously unknown roles in cancer biology and identify additional cancer types where AKR7A3-targeted therapies could be beneficial.
Aldo-Keto Reductase Family 7 Member A3, commonly referred to as AKR7A3, is a protein-coding gene that plays a significant role in the detoxification of aldehydes and ketones. This protein is part of the aldo-keto reductase superfamily, which consists of enzymes that catalyze the reduction of aldehydes and ketones to their corresponding alcohols using NADPH as a cofactor .
The AKR7A3 gene is located on chromosome 1 and encodes a protein that is 331 amino acids long. The recombinant form of this protein, tagged with a polyhistidine (His) tag, is expressed in Escherichia coli for research and industrial purposes. The His tag facilitates the purification of the protein using affinity chromatography .
AKR7A3 is known for its ability to reduce the dialdehyde protein-binding form of aflatoxin B1 to the non-binding aflatoxin B1 dialcohol. This activity is crucial in protecting the liver from the toxic and carcinogenic effects of aflatoxin B1, a potent hepatocarcinogen . The enzyme’s activity is measured by its ability to catalyze the oxidation of NADPH, and its specific activity is expressed in units per milligram of protein .
The detoxification role of AKR7A3 is vital in various metabolic pathways, including the biotransformation of xenobiotics and the metabolism of endogenous compounds. The enzyme’s ability to neutralize harmful aldehydes and ketones helps maintain cellular homeostasis and protect against oxidative stress .
Mutations or dysregulation of the AKR7A3 gene have been associated with certain diseases, including congenital symmetric circumferential skin creases and multiple benign circumferential skin creases on limbs . Understanding the function and regulation of AKR7A3 can provide insights into the development of therapeutic strategies for these conditions.