AKR1C1 Human

Aldo-Keto Reductase Family 1 Member C1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Structure and Isoform Specificity

AKR1C1 shares high sequence identity with three other isoforms (AKR1C2, AKR1C3, AKR1C4) clustered on chromosome 10. These enzymes exhibit overlapping but distinct substrate specificities, primarily acting on positions 3, 17, and 20 of steroids .

IsoformKey Enzymatic ActivitiesPrimary Tissue Expression
AKR1C120α-hydroxysteroid dehydrogenase, 3α-hydroxysteroid dehydrogenase, dihydrodiol dehydrogenaseLiver, prostate, breast, testes
AKR1C23α-hydroxysteroid dehydrogenase (type 3)Liver, adrenal glands
AKR1C317β-hydroxysteroid dehydrogenase (type 5), 3α-hydroxysteroid dehydrogenase (type 2)Liver, prostate, brain
AKR1C43α-hydroxysteroid dehydrogenase (type 1)Liver, small intestine

AKR1C1’s catalytic activity involves a sequential ordered bi-bi mechanism, with NADPH binding first, followed by substrate reduction . Its structural flexibility allows interactions with diverse steroids, including progesterone, androgens, and bile acids .

Steroid Hormone Metabolism

AKR1C1 regulates the conversion of active hormones to inactive metabolites or vice versa, modulating nuclear receptor activity. Key roles include:

  • Progesterone inactivation: Reduction of progesterone to 20α-hydroxyprogesterone, terminating its luteotropic effects .

  • Neurosteroid biosynthesis: Conversion of pregnenolone to allopregnanolone, a neuroactive steroid with anxiolytic properties .

  • Bile acid synthesis: Participation in bile acid metabolism via interactions with hepatic enzymes .

Cancer Progression and Drug Resistance

AKR1C1 is implicated in tumor invasion, metastasis, and chemoresistance:

  • STAT3 pathway activation: AKR1C1 interacts with STAT3, enhancing its phosphorylation and transcriptional activity, promoting metastasis-related genes (e.g., N-cadherin, fibronectin) in non-small cell lung cancer (NSCLC) .

  • Cisplatin resistance: Upregulation in metastatic bladder cancer cells correlates with reduced cisplatin sensitivity, potentially via oxidative stress modulation or pro-survival signaling .

Cancer-Associated Roles

Cancer TypeMechanismKey Findings
NSCLCSTAT3 activation, EMT promotionAKR1C1 overexpression increases liver/lung metastasis in vivo .
Bladder CancerCisplatin resistance, EMT, IL-1β signalingAKR1C1 knockdown restores cisplatin sensitivity; IL-1β upregulates AKR1C1 .
Prostate CancerAR coactivation, androgen biosynthesisAKR1C3 (paralog) acts as a coactivator for androgen receptor (AR) .

Genetic and Metabolic Disorders

  • Lipedema: A missense mutation (L213Q) in AKR1C1 reduces catalytic efficiency, impairing progesterone metabolism and contributing to sex-limited autosomal dominant lipedema .

  • Bile Acid Deficiency: Mutations in AKR1D1 (a related isoform) disrupt bile acid synthesis, but AKR1C1’s role in this pathway remains under investigation .

Cancer Therapy

  • Flufenamic acid (FFA): A non-steroidal anti-inflammatory drug (NSAID) that inhibits AKR1C1 enzymatic activity, reversing cisplatin resistance in bladder cancer models .

  • ASP9521: A small-molecule inhibitor targeting AKR1C3 (but with potential cross-reactivity), showing anti-tumor activity in preclinical prostate cancer studies .

Neurological Disorders

  • Allopregnanolone modulation: AKR1C1’s role in neurosteroid biosynthesis positions it as a target for treating anxiety or neurodegenerative diseases, though clinical applications remain speculative .

Product Specs

Introduction

Aldo-keto reductase family 1 member C1, also known as AKR1C1, is an enzyme belonging to the aldo/keto reductase family, which comprises over 40 related proteins. AKR1C1 facilitates the conversion of ketones and aldehydes into their corresponding alcohol forms utilizing cofactors like NADH and NADPH. Notably, AKR1C1 plays a role in converting progesterone into its inactive form, 20-alpha-hydroxy-progesterone.

Description

Recombinant human AKR1C1, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 323 amino acids (residues 1-323). It has a molecular weight of 36.7 kDa.
The purification of AKR1C1 is achieved using proprietary chromatographic methods.

Physical Appearance
A sterile, colorless solution that has been filtered.
Formulation

The AKR1C1 solution is provided at a concentration of 1 mg/ml and contains 20% glycerol, 0.1M NaCl, and 20mM Tris-HCl buffer with a pH of 8.5.

Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C.
To ensure long-term stability, adding a carrier protein (0.1% HSA or BSA) is advised.
Repeated freezing and thawing of the product should be avoided.
Purity

The purity of the product is determined to be greater than 95.0% as assessed by SDS-PAGE analysis.

Biological Activity

The specific activity of the enzyme is greater than 500 pmol per minute per microgram. Specific activity is determined by measuring the enzyme's ability to catalyze the oxidation of 1.0 pmole of 1-Acenaphthenol per minute in the presence of NADP at a pH of 8.8 and a temperature of 25 degrees Celsius.

Synonyms

DDH1, DDH, HAKRC, 20-alpha-HSD, DD1/DD2, HBAB, C9, DD1, H-37, MBAB, MGC8954, 2-ALPHA-HSD, AKR1C1, Aldo-keto reductase family 1 member C1, 20-alpha-hydroxysteroid dehydrogenase, Trans-1,2-dihydrobenzene-1,2-diol dehydrogenase, Indanol dehydrogenase, Dihydrodiol dehydrogenase 1/2, Chlordecone reductase homolog HAKRC, High-affinity hepatic bile acid-binding protein

Source

Escherichia Coli.

Amino Acid Sequence

MDSKYQCVKL NDGHFMPVLG FGTYAPAEVP KSKALEATKL AIEAGFRHID SAHLYNNEEQ VGLAIRSKIA DGSVKREDIF YTSKLWCNSH RPELVRPALE RSLKNLQLDY VDLYLIHFPV SVKPGEEVIP KDENGKILFD TVDLCATWEA VEKCKDAGLA KSIGVSNFNR RQLEMILNKP GLKYKPVCNQ VECHPYFNQR KLLDFCKSKD IVLVAYSALG SHREEPWVDP NSPVLLEDPV
LCALAKKHKR TPALIALRYQ LQRGVVVLAK SYNEQRIRQN VQVFEFQLTS EEMKAIDGLN RNVRYLTLDI FAGPPNYPFS DEY

Q&A

What is AKR1C1 and what are its primary functions in human biology?

AKR1C1 belongs to the aldo-keto reductase superfamily that catalyzes the conversion of aldehydes and ketones to their corresponding alcohols. It functions primarily as a 20α-hydroxysteroid dehydrogenase in steroid metabolism, participating in the reduction of ketosteroids at the 3-, 17-, or 20-position . The enzyme shares high sequence homology (>86%) with other family members (AKR1C2-4) but exhibits distinct tissue expression patterns and substrate preferences. While all isoforms are expressed in the liver, AKR1C1 is also found in lung, prostate, mammary gland, and testis tissues . In steroid metabolism, AKR1C1 stereospecifically favors the formation of 3α,5β-reduced steroids rather than their 3β-isomers, contributing to the regulation of active steroid hormones .

How can researchers distinguish between AKR1C1 and other closely related family members in experimental studies?

Distinguishing between AKR1C family members presents significant challenges due to their high sequence homology (>86%). For RNA-based detection, researchers should design isoform-specific PCR primers targeting unique regions, particularly in the 3' untranslated region, and optimize qRT-PCR conditions with high-stringency parameters. For protein detection, select antibodies raised against unique epitopes of AKR1C1, validated for specificity against all four AKR1C enzymes . Functionally, exploit substrate preferences and reaction stereochemistry, as AKR1C1 functions primarily as a 20α-hydroxysteroid dehydrogenase with distinct stereospecificity patterns . When using genetic manipulation approaches, design CRISPR-Cas9 strategies or siRNAs targeting unique sequences to avoid off-target effects. Expression pattern verification can help, as AKR1C4 is liver-specific while AKR1C1-3 have broader distribution patterns . For definitive identification, combine multiple approaches and include appropriate controls with known expression of specific AKR1C isoforms.

What experimental approaches effectively measure AKR1C1 enzyme activity versus protein expression?

Measuring AKR1C1 enzyme activity requires different approaches than detecting protein expression levels. For enzyme activity assessment, spectrophotometric assays tracking NADPH consumption during substrate reduction provide quantitative functional data. Researchers can utilize specific substrates including 20α-hydroxysteroids where AKR1C1 demonstrates preferential activity . HPLC or LC-MS analysis of reaction products offers precise quantification of stereospecific outcomes, particularly important as AKR1C1 stereospecifically produces 3α,5β-reduced steroids . For protein expression, Western blotting with isoform-specific antibodies remains the standard quantitative approach, while immunohistochemistry (IHC) visualizes tissue localization and expression patterns, as demonstrated in studies of nasopharyngeal carcinoma tissues . Flow cytometry enables single-cell resolution analysis of protein levels in heterogeneous populations. When designing experiments, researchers should incorporate appropriate positive controls (tissues/cells with known AKR1C1 expression) and negative controls (AKR1C1-knockout samples), and validate findings using multiple complementary techniques to ensure reliable discrimination between protein abundance and enzymatic function.

How does AKR1C1 expression vary across different cancer types and what are the implications?

AKR1C1 expression demonstrates remarkable heterogeneity across cancer types, with both upregulation and downregulation reported depending on the malignancy:

Cancer TypeAKR1C1 ExpressionClinical Associations
Extrahepatic cholangiocarcinoma (ECC)Highly expressedAssociated with progression and poor prognosis
Non-small cell lung cancer (NSCLC)UpregulatedPromotes metastasis and predicts poor survival
Nasopharyngeal carcinoma (NPC)Significantly downregulatedCorrelates with advanced features but better prognosis

In NPC, a study of 177 NPC tissues and 61 non-cancerous epithelial tissues revealed low AKR1C1 expression in 64.41% of NPC samples compared to only 4.92% of non-cancerous tissues . This variable expression pattern suggests cancer-specific roles for AKR1C1, potentially functioning as an oncogene in some contexts (ECC, NSCLC) while having different roles in others (NPC). These differences likely reflect tissue-specific functions of AKR1C1 in normal physiology or adaptations during oncogenesis. The contradictory expression patterns highlight the importance of cancer-specific analysis when considering AKR1C1 as a diagnostic marker or therapeutic target.

What mechanisms explain AKR1C1's role in cancer cell proliferation and metastasis?

AKR1C1 promotes cancer cell proliferation and metastasis through multiple interconnected mechanisms that vary by cancer type:

  • Metabolic reprogramming via HIF-1α: In NSCLC, AKR1C1 augments hypoxia-inducible factor 1-alpha (HIF-1α) expression, driving tumor metabolic reprogramming that increases lactate production and fuels cancer cell proliferation . This metabolic shift provides energy and intermediates necessary for rapid cell division.

  • STAT3 pathway activation: AKR1C1 directly interacts with STAT3 in NSCLC, facilitating its phosphorylation and reinforcing STAT3 binding to promoter regions of target genes involved in proliferation and metastasis . This interaction may facilitate STAT3's association with its upstream kinase JAK2, enhancing signal transduction that promotes invasive behavior.

  • Ferroptosis regulation: In extrahepatic cholangiocarcinoma, AKR1C1 suppresses ferroptotic cell death by regulating the CYP1B1-cAMP signaling axis . AKR1C1 degrades CYP1B1 protein via ubiquitin-proteasomal degradation and decreases CYP1B1 mRNA through the transcriptional factor aryl-hydrocarbon receptor (AHR) .

  • Chemotherapy resistance: AKR1C1 confers resistance to various chemotherapeutic agents through mechanisms involving reduction of reactive oxygen species, elimination of free radicals, and inactivation of anticancer drugs .

Notably, these mechanisms appear to be cancer-type specific, as evidenced by studies in NPC showing that AKR1C1 silencing had no impact on cell proliferation, migration, or invasion, despite affecting cisplatin sensitivity .

How does AKR1C1 influence response to chemotherapy across different cancer contexts?

AKR1C1's influence on chemotherapy response exhibits striking context-dependency across cancer types:

  • Chemoresistance promotion:

    • In bladder, gastric, ovarian, cervical, and lung cancers, AKR1C1 overexpression reduces sensitivity to cisplatin through multiple mechanisms .

    • AKR1C1 can reduce reactive oxygen species production, eliminate free radicals, and inactivate anticancer drugs, thereby reducing DNA damage and inhibiting apoptosis .

    • These protective mechanisms contribute to treatment failure in cancers with high AKR1C1 expression.

  • Chemosensitivity association:

    • Paradoxically, in nasopharyngeal carcinoma (NPC), AKR1C1 downregulation enhances cisplatin sensitivity despite being associated with advanced disease features .

    • In vitro studies demonstrated that AKR1C1 silencing in NPC cells significantly sensitized them to cisplatin cytotoxicity, increasing apoptosis and cell cycle arrest .

  • Experimental evidence:

    • NPC studies showed that while AKR1C1 knockdown had no impact on cell proliferation, migration, or invasion, it specifically enhanced cisplatin sensitivity .

    • This suggests AKR1C1 loss in NPC may be an accompanying molecular event contributing to chemotherapeutic sensitivity rather than driving malignant behavior.

These contradictory roles across cancer types highlight the importance of cancer-specific approaches when considering AKR1C1 as a predictive biomarker for treatment response or as a therapeutic target to enhance chemosensitivity. The findings suggest potential value in testing AKR1C1 expression to guide treatment decisions, particularly for cisplatin-based therapies.

What cell and animal models are most appropriate for studying AKR1C1 in cancer research?

Selection of appropriate experimental models is critical for meaningful AKR1C1 research across different cancer contexts:

  • Cell models:

    • Nasopharyngeal carcinoma: Research identified variable AKR1C1 expression across NPC cell lines - CNE1, CNE2, and S18 express AKR1C1, while HK1-EBV, SUNE1, HONE1, and 5-8F lack expression .

    • NSCLC: Multiple cell lines should be tested as AKR1C1 expression varies; paired cell lines with different metastatic potential provide valuable systems for studying AKR1C1's role in metastasis .

    • Control models: Immortalized nasopharyngeal epithelial cells (NP69, SWSX-1489, HNEpC) serve as non-cancer controls ; NIH-3T3 cells have been used to study malignant transformation upon AKR1C1 ectopic expression .

  • Animal models:

    • Xenograft mouse models have demonstrated that AKR1C1 depletion sensitizes cancer cells to ferroptosis and synergizes with ferroptosis inducers to suppress tumor growth in ECC .

    • Orthotopic models may better recapitulate the tumor microenvironment for studying AKR1C1's context-dependent functions.

    • Patient-derived xenografts could capture the heterogeneity of AKR1C1 expression and function observed in clinical samples.

  • Model selection considerations:

    • Characterize baseline AKR1C1 expression before selection

    • Consider tissue context - expression patterns and roles vary significantly between cancer types

    • For metabolism studies, ensure cells maintain relevant metabolic characteristics

    • Use isogenic cell line pairs (with AKR1C1 knockout/overexpression) to minimize confounding variables

The contradictory roles of AKR1C1 in different cancers highlight the importance of selecting appropriate models that reflect the specific cancer context being studied.

What gene modulation techniques are most effective for studying AKR1C1 function?

Several complementary gene modulation approaches are recommended for comprehensive investigation of AKR1C1 function:

  • RNA interference (siRNA/shRNA):

    • Transient siRNA knockdown: Effective for initial screening and short-term experiments, as demonstrated in studies where siRNA silencing of AKR1C1 in NPC cells revealed effects on cisplatin sensitivity .

    • Stable shRNA expression: Provides longer-term suppression for extended studies and in vivo experiments.

    • Inducible knockdown systems: Allow temporal control of AKR1C1 depletion, as utilized in studies triggering ferroptosis in ECC cells .

  • CRISPR-Cas9 genome editing:

    • Complete knockout: Eliminates confounding effects from residual expression in knockdown approaches.

    • Knock-in mutations: Can introduce specific variants to study structure-function relationships.

    • Base editing or prime editing: For introducing precise modifications without double-strand breaks.

  • Overexpression systems:

    • Constitutive expression vectors: For studying gain-of-function effects in low-expressing cell lines.

    • Inducible overexpression: Provides temporal control to study immediate vs. adaptive responses to AKR1C1 upregulation.

    • Structure-function studies: Expression of catalytically inactive mutants has revealed that AKR1C1 exerts pro-metastatic effects in a catalytic-independent manner .

  • Methodological considerations:

    • Include appropriate controls (non-targeting siRNA, empty vectors).

    • Validate knockdown/overexpression at both mRNA and protein levels.

    • Confirm specificity by checking effects on other AKR1C family members.

    • Consider rescue experiments to confirm phenotype specificity.

    • For in vivo studies, use tissue-specific or inducible systems to avoid developmental effects.

When selecting approaches, researchers should consider the cancer-specific context, as AKR1C1 functions vary significantly between cancer types .

What biomarkers and assays should be used to assess AKR1C1-mediated effects on cancer phenotypes?

Comprehensive assessment of AKR1C1-mediated effects requires multiple complementary assays targeting different cancer phenotypes:

  • Proliferation and viability:

    • CCK8/MTT assays: Quantify cell viability and proliferation rates following AKR1C1 modulation, as used in NPC studies .

    • Colony formation assays: Evaluate long-term proliferative capacity and clonogenicity.

    • Cell cycle analysis: Flow cytometry assessment of cell cycle distribution can reveal mechanisms of growth effects.

    • EdU incorporation: Measures DNA synthesis rates more directly than proxy proliferation assays.

  • Invasion and metastasis:

    • Wound healing assays: Quantify collective cell migration capacity .

    • Transwell migration/invasion assays: Assess individual cell motility and basement membrane invasion .

    • 3D spheroid invasion models: Provide more physiologically relevant assessment of invasive behavior.

    • Epithelial-mesenchymal transition (EMT) markers: E-cadherin, N-cadherin, Vimentin, and Snail expression changes.

  • Cell death and therapy response:

    • Apoptosis detection: Flow cytometry with Annexin V/PI staining and DAPI staining to quantify apoptotic cells .

    • Ferroptosis markers: Lipid peroxidation (BODIPY-C11), glutathione depletion, and iron dependency assessments .

    • Drug sensitivity assays: Dose-response curves for chemotherapeutics, particularly cisplatin .

  • Molecular pathway analysis:

    • STAT3 pathway activation: Phospho-STAT3 levels, nuclear localization, and target gene expression .

    • HIF-1α signaling: HIF-1α protein levels and target gene expression to assess metabolic reprogramming .

    • Metabolic profiling: Lactate production, glucose consumption, and oxygen consumption rates .

    • CYP1B1-cAMP axis: CYP1B1 levels and cAMP-PKA signaling components when studying ferroptosis .

  • In vivo assessment:

    • Xenograft tumor growth: Volume and weight measurements .

    • Metastasis quantification: Bioluminescence imaging, histological assessment of distant organs.

    • Response to therapy: Combination with ferroptosis inducers or chemotherapeutics .

Selection of appropriate assays should be guided by the specific cancer context, as AKR1C1 functions vary significantly between cancer types, affecting ferroptosis in ECC , metastasis in NSCLC , and chemosensitivity in NPC .

How does AKR1C1 regulate ferroptosis and what are the implications for cancer therapy?

AKR1C1 emerges as a critical regulator of ferroptosis with significant therapeutic implications:

  • Mechanistic basis:

    • AKR1C1 regulates the stability of cytochrome P450 family member CYP1B1, a newly discovered mediator of ferroptosis .

    • AKR1C1 degrades CYP1B1 protein via ubiquitin-proteasomal degradation and decreases CYP1B1 mRNA levels through modulation of the transcriptional factor aryl-hydrocarbon receptor (AHR) .

    • The AKR1C1-CYP1B1 axis modulates ferroptosis via the cAMP-PKA signaling pathway .

  • Cancer-specific effects:

    • In extrahepatic cholangiocarcinoma (ECC), inducible AKR1C1 knockdown triggers ferroptotic cell death .

    • AKR1C1 depletion sensitizes ECC cells to ferroptosis inducers in xenograft mouse models, synergistically suppressing tumor growth .

  • Therapeutic implications:

    • AKR1C1 inhibition could sensitize cancer cells to ferroptosis inducers, particularly in ECC where AKR1C1 is highly expressed .

    • The AKR1C1–CYP1B1–cAMP signaling axis represents a promising therapeutic target for ECC treatment, especially in combination with ferroptosis inducers .

    • This approach might be particularly valuable for cancers resistant to apoptosis-inducing therapies.

  • Experimental considerations:

    • Assessment of ferroptosis markers (lipid peroxidation, iron dependency) is essential when studying AKR1C1's role in cell death.

    • Rescue experiments with ferroptosis inhibitors (e.g., ferrostatin-1) can confirm the specific cell death mechanism.

    • The tissue-specific nature of AKR1C1's role in ferroptosis should be considered when designing experiments or therapeutic approaches.

This connection between AKR1C1 and ferroptosis reveals a novel mechanism by which AKR1C1 contributes to cancer progression and therapy resistance, offering new avenues for therapeutic intervention through targeting the AKR1C1–CYP1B1–cAMP signaling axis .

What is the relationship between AKR1C1 and the STAT3 signaling pathway in metastasis?

AKR1C1 promotes metastasis through complex interactions with the STAT3 pathway, particularly in non-small cell lung cancer (NSCLC):

  • Direct physical interaction:

    • AKR1C1 directly binds to STAT3, as demonstrated through co-immunoprecipitation studies .

    • This physical association is critical for subsequent signaling events that promote metastatic behavior.

  • Enhanced STAT3 phosphorylation:

    • AKR1C1 facilitates STAT3 phosphorylation, a key activation step in the STAT3 signaling cascade .

    • This process may involve promoting interaction between STAT3 and its upstream kinase JAK2 .

  • Reinforced promoter binding:

    • The AKR1C1-STAT3 interaction strengthens STAT3 binding to promoter regions of target genes involved in metastasis .

    • This enhanced binding leads to increased expression of STAT3 target genes that promote epithelial-mesenchymal transition, cell migration, invasion, and survival at distant sites .

  • Catalytic-independent mechanism:

    • Intriguingly, AKR1C1 exerts these pro-metastatic effects independent of its enzymatic activity, suggesting a structural or scaffolding role in STAT3 signaling complexes .

  • Clinical relevance:

    • A significant correlation between AKR1C1 and STAT3 pathway activation is observed in metastatic foci of NSCLC patients .

    • This correlation supports the pathophysiological relevance of this mechanism in human cancer.

The AKR1C1-STAT3 axis represents a potential therapeutic target for preventing metastasis, particularly in NSCLC. Disrupting this interaction could reduce metastatic potential without affecting AKR1C1's enzymatic functions in normal tissues, potentially offering a more selective approach to intervention. For researchers investigating this pathway, approaches should include assessing STAT3 phosphorylation status, nuclear localization, and target gene expression following AKR1C1 modulation .

How does AKR1C1 contribute to metabolic reprogramming in cancer cells?

AKR1C1 orchestrates metabolic reprogramming in cancer cells through a complex interplay with HIF-1α signaling:

  • HIF-1α regulation:

    • AKR1C1 augments hypoxia-inducible factor 1-alpha (HIF-1α) expression in cancer cells, particularly demonstrated in non-small cell lung cancer (NSCLC) .

    • This occurs even under normoxic conditions, suggesting an oxygen-independent mechanism distinct from canonical hypoxia-driven HIF-1α stabilization.

  • Metabolic effects:

    • Enhanced glycolysis: AKR1C1 promotes increased glucose uptake and glycolytic flux, supporting the Warburg effect.

    • Increased lactate production: AKR1C1 knockdown experiments in NSCLC cells demonstrate reduced lactate production, indicating AKR1C1's role in promoting aerobic glycolysis .

    • Upregulation of HIF-1α target genes: AKR1C1 enhances expression of genes involved in glucose metabolism, angiogenesis, and survival.

  • Functional consequences:

    • Enhanced proliferation: The metabolic shift provides energy and intermediates necessary for rapid cancer cell proliferation .

    • Microenvironmental effects: Increased lactate production may contribute to acidification of the tumor microenvironment, potentially affecting immune function and invasion.

    • Therapy resistance: Metabolic adaptations can contribute to resistance against various therapeutic modalities.

  • Clinical significance:

    • AKR1C1 significantly correlates with HIF-1α signaling in clinical samples .

    • This correlation predicts poor prognosis for NSCLC patients .

    • The AKR1C1-HIF-1α axis represents a potential therapeutic target for metabolically targeted cancer therapy.

This metabolic role of AKR1C1 provides insights into how it promotes cancer cell proliferation beyond its classical enzymatic functions in steroid metabolism, highlighting its multifaceted contribution to cancer biology. Research data demonstrate that "AKR1C1 reprograms tumour metabolism to promote NSCLC cells proliferation by activating HIF-1α," establishing a specific role for AKR1C1 in metabolic reprogramming and suggesting it may be a therapeutic target for NSCLC treatment .

How can researchers explain the contradictory prognostic significance of AKR1C1 across different cancer types?

The contradictory prognostic significance of AKR1C1 across cancer types presents a fascinating scientific puzzle requiring nuanced interpretation:

  • Cancer-specific biological functions:

    • In NSCLC and ECC: AKR1C1 actively drives malignant processes (metabolic reprogramming, metastasis, ferroptosis inhibition), making high expression unfavorable .

    • In NPC: AKR1C1 loss has been shown to have no impact on proliferation, migration, or invasion, but specifically enhances cisplatin sensitivity, explaining why low expression correlates with better outcomes despite being associated with advanced disease features .

  • Treatment context considerations:

    • Standard treatment protocols differ between cancer types, influencing how AKR1C1 expression impacts outcomes.

    • In cisplatin-treated cancers like NPC, AKR1C1's role in chemoresistance may predominate (lower expression = better response = improved survival) .

    • In cancers where surgical resection is primary, AKR1C1's effects on proliferation or metastasis may be more important prognostically.

  • Molecular context dependencies:

    • The functional impact of AKR1C1 likely depends on the presence or absence of specific signaling partners (e.g., STAT3, HIF-1α) .

    • Genetic background and mutation profiles of different cancer types may modify AKR1C1's effects.

The NPC example is particularly instructive, where researchers observed that "AKR1C1 was not a driver factor in the process of carcinogenesis and progression in NPC, AKR1C1 loss maybe an accompanying molecular event which contributed only to the chemotherapeutic sensitivity to cisplatin while not the malignant biological behaviours of NPC" . This context-dependent functional specialization may explain seemingly contradictory observations across cancer types.

What methodological challenges exist in studying AKR1C1's enzymatic versus non-enzymatic functions?

Distinguishing between AKR1C1's enzymatic and non-enzymatic functions presents several methodological challenges:

  • Catalytic activity assessment:

    • Enzymatic assays may not capture non-catalytic functions that depend on protein-protein interactions.

    • Standard biochemical approaches measure bulk activity but may miss cell compartment-specific functions.

    • The high sequence similarity between AKR1C family members (>86%) complicates specific activity measurement .

  • Structure-function analysis challenges:

    • Creating catalytically inactive mutants without disrupting protein folding or interaction surfaces is technically demanding.

    • Non-enzymatic functions may depend on specific protein domains that overlap with catalytic regions.

    • Determining whether a phenotype depends on enzymatic activity requires careful controls and rescue experiments.

  • Experimental design considerations:

    • Studies should incorporate catalytically inactive AKR1C1 mutants as controls to distinguish enzymatic from structural roles.

    • Domain mapping approaches can identify regions required for specific interactions independent of catalytic function.

    • Time-resolved experiments may help separate immediate enzymatic effects from delayed non-enzymatic functions.

  • Context-specific functions:

    • The same catalytic activity may have different consequences in different cellular contexts or cancer types.

    • AKR1C1 exerts pro-metastatic effects through STAT3 in a catalytic-independent manner in NSCLC , while its metabolic functions may require enzymatic activity.

    • Ferroptosis regulation by AKR1C1 involves both protein-protein interactions and potentially enzymatic functions .

  • Technical approaches for differentiation:

    • Proximity ligation assays can visualize specific protein interactions in situ.

    • Mass spectrometry-based interactome analysis can identify binding partners independent of catalytic function.

    • Substrate trapping mutants can help distinguish catalytic from scaffolding roles.

These methodological challenges explain why seemingly contradictory observations regarding AKR1C1 function may emerge from different experimental approaches and cellular contexts.

How should inconsistencies in AKR1C1 expression and function be addressed in cancer research?

Addressing inconsistencies in AKR1C1 research requires systematic approaches to reconcile conflicting data:

  • Cancer type stratification:

    • Explicitly separate findings by cancer type rather than generalizing across malignancies.

    • Consider AKR1C1 as having cancer-specific functions rather than universal oncogenic or tumor-suppressive roles.

    • Data from NPC shows AKR1C1 downregulation is associated with advanced features but better prognosis, while other cancers show opposite trends .

  • Methodology standardization:

    • Adopt consistent detection methods for AKR1C1 expression (specific antibodies, primer designs).

    • Standardize functional assays to enable direct comparison between studies.

    • Report detailed methodological parameters to facilitate reproduction and comparison.

  • Contextual analysis:

    • Examine AKR1C1 in the context of treatment modalities (e.g., cisplatin-based therapy in NPC) .

    • Consider microenvironmental factors that may modify AKR1C1 function.

    • Evaluate co-expression of interacting partners (STAT3, CYP1B1, HIF-1α) that may explain contextual differences .

  • Multi-dimensional data integration:

    • Combine genomic, transcriptomic, and proteomic data to capture the full complexity of AKR1C1 regulation.

    • Correlate expression with multiple endpoints (proliferation, metastasis, therapy response) simultaneously.

    • Use patient-derived models that maintain the genetic and epigenetic context of original tumors.

  • Mechanism-based reconciliation:

    • Investigate whether AKR1C1 has biphasic effects depending on expression level.

    • Consider post-translational modifications or alternative splicing that might explain functional differences.

    • Examine subcellular localization differences that could reconcile contradictory observations.

Research in NPC provides an instructive example, where investigators demonstrated that "AKR1C1 silencing had no impact on cell proliferation, migration and invasion, while AKR1C1 silencing sensitized NPC cells to the cytotoxicity of cisplatin" . This finding reconciles the apparent contradiction between AKR1C1 downregulation being associated with both advanced disease features and better survival outcomes in this specific cancer context.

Product Science Overview

Introduction

Aldo-Keto Reductase Family 1 Member C1 (AKR1C1) is an enzyme that belongs to the aldo-keto reductase (AKR) superfamily, which consists of more than 40 known enzymes and proteins. These enzymes catalyze the conversion of aldehydes and ketones to their corresponding alcohols by utilizing NADH and/or NADPH as cofactors . AKR1C1 is also known by several other names, including 20α-hydroxysteroid dehydrogenase, 3α-hydroxysteroid dehydrogenase, and dihydrodiol dehydrogenase 1/2 .

Gene and Protein Structure

The AKR1C1 gene is located on chromosome 10 at the position 10p15.1 in humans . The gene encodes a protein that is involved in various biological processes, including steroid metabolism, bile acid metabolism, and the metabolism of xenobiotics . The protein structure of AKR1C1 includes several key domains that are essential for its enzymatic activity, including the NADP(H)-binding domain and the substrate-binding domain .

Biological Functions

AKR1C1 plays a significant role in the metabolism of steroids, particularly in the conversion of progesterone to its inactive form, 20α-hydroxyprogesterone (20α-OHP) . This enzyme is also involved in the metabolism of bile acids and the regulation of intrahepatic bile acid concentration . Additionally, AKR1C1 participates in the formation of myelin, which is crucial for the proper functioning of the nervous system .

Clinical Significance

AKR1C1 has been implicated in various clinical conditions, including cancer and metabolic disorders. The enzyme’s ability to metabolize steroids and other compounds makes it a potential target for therapeutic interventions . For example, AKR1C1 has been shown to regulate the osteogenic differentiation of human adipose-derived mesenchymal stromal/stem cells (hASCs) by targeting the progesterone receptor . This finding suggests that AKR1C1 could be used as a molecular target for modifying hASCs to enhance their osteogenic capacity in bone tissue engineering .

Research and Applications

Research on AKR1C1 has focused on understanding its role in various biological processes and its potential applications in medicine. Studies have shown that AKR1C1 acts as a negative regulator of osteogenesis and a positive regulator of adipogenesis in hASCs . This dual role highlights the enzyme’s importance in tissue engineering and regenerative medicine .

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.