AKR1C3 (Human Aldo-Keto Reductase Family 1 Member C3), also known as 17β-hydroxysteroid dehydrogenase type 5 (17β-HSD5) or prostaglandin F synthase, is a NADPH-dependent oxidoreductase within the AKR superfamily. It regulates steroid hormone metabolism, prostaglandin biosynthesis, and redox balance, playing critical roles in both physiological processes and pathologies such as cancer .
AKR1C3 modulates hormone activity through reductive and oxidative reactions, influencing cellular proliferation, immune response, and stress adaptation.
Redox Regulation: Maintains glutathione (GSH) levels, mitigating oxidative stress .
Cancer Progression: Activates ERK, Akt, and STAT3 pathways, promoting epithelial-mesenchymal transition (EMT) and angiogenesis .
AKR1C3 is implicated in diverse carcinomas, with expression levels correlating with prognosis and therapeutic resistance.
Small-Molecule Inhibitors:
Prodrug Strategy:
Enzalutamide Sensitivity: AKR1C3 inhibition restores AR blockade in castration-resistant prostate cancer (CRPC) .
Immunotherapy Synergy: AKR1C3 expression correlates with immune cell infiltration, suggesting combinatorial potential .
AKR1C3 exhibits organ-specific expression, with implications for targeted therapies.
Aldo-keto reductase family 1 member C3 isoform 1 (AKR1C3) is an enzyme belonging to the aldo-keto reductase family. It facilitates the conversion of ketones and aldehydes to their corresponding alcohol forms using cofactors like NADPH and NADH. AKR1C3 plays a role in reducing prostaglandin (PG), phenanthrenequinone (PQ), and PGH2. Additionally, it is involved in oxidizing 9 alpha, 11 beta-PGF2 to PGD2. AKR1C3 is considered important in the development of allergic conditions such as asthma and might also contribute to the regulation of cell growth and differentiation.
Recombinant human AKR1C3, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 323 amino acids (1-323) with a molecular weight of 36.8 kDa.
The protein undergoes purification using proprietary chromatographic methods.
The AKR1C3 solution is supplied at a concentration of 1mg/ml and contains 10% glycerol, 0.1M NaCl, 1mM DTT, and 20mM Tris-HCl buffer at pH 8.5.
The purity of the product is determined to be greater than 90.0% based on SDS-PAGE analysis.
The specific activity of the enzyme is greater than 1,000 pmol/min/µg, which is defined as the amount of enzyme required to catalyze the oxidation of 1.0 pmole of 1-Acenaphthenol per minute in the presence of NADP at pH 8.8 and a temperature of 25°C.
DD3, DDX, HAKRB, HAKRe, HA1753, HSD17B5, hluPGFS, KIAA0119, AKR1C3, Aldo-keto reductase family 1 member C3, 3-alpha-HSD type 2, 17-beta-HSD 5, PGFS, DD-3.
Escherichia Coli.
MDSKHQCVKL NDGHFMPVLG FGTYAPPEVP RSKALEVTKL AIEAGFRHID SAHLYNNEEQ VGLAIRSKIA DGSVKREDIF YTSKLWSTFH RPELVRPALE NSLKKAQLDY VDLYLIHSPM SLKPGEELSP TDENGKVIFD IVDLCTTWEA MEKCKDAGLA KSIGVSNFNR RQLEMILNKP GLKYKPVCNQ VECHPYFNRS KLLDFCKSKD IVLVAYSALG SQRDKRWVDP NSPVLLEDPV
LCALAKKHKR TPALIALRYQ LQRGVVVLAK SYNEQRIRQN VQVFEFQLTA EDMKAIDGLD RNLHYFNSDS FASHPNYPYS DEY
AKR1C3 (aldo-keto reductase family 1 member C3), also known as type 2 3α-hydroxysteroid dehydrogenase/type 5 17β-hydroxysteroid dehydrogenase, is an enzyme that catalyzes the conversion of aldehydes and ketones to alcohols . It plays crucial roles in:
Androgen and estrogen metabolism, particularly converting androstenedione to testosterone
Prostaglandin metabolism, including the reduction of prostaglandin D2 into PGF2
Regulating ligand access to the androgen receptor, estrogen receptor, and peroxisome proliferator activating receptor γ in hormone target tissues
AKR1C3 shares >86% sequence identity with three highly related human AKRs (AKR1C1, AKR1C2, and AKR1C4) but has distinct catalytic functions . Originally cloned from human prostate and placental cDNA libraries, this enzyme has relatively high 17β-hydroxysteroid dehydrogenase activity .
AKR1C3 shows a specific distribution pattern across human tissues, with both expected and unexpected expression patterns:
In the prostate: Primarily found in stromal cells and a small number of basal cells, with strong immunoreactivity in endothelial cells
In the kidney and bladder: Strong immunoreactivity despite being traditionally considered non-hormone-associated tissues
In the liver: Detectable in hepatocytes and liver tissue as shown by Western blot analysis
In cancer cells: Overexpressed in hepatocellular carcinoma (HCC) and highly expressed in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL)
The presence of AKR1C3 in both hormone-related tissues (prostate, testis) and non-hormone-related tissues (kidney, bladder) suggests that this enzyme has broader physiological roles than previously understood .
When comparing human AKR1C3 and its rat homolog in genitourinary systems, researchers have found that:
The distribution patterns are comparable but not identical between the two species
In human prostate, AKR1C3 is mainly found in stromal cells and some basal cells
In rat prostate, strong immunoreactivity is observed in prostatic epithelial cells but not in stromal or endothelial cells
Both species show expression in non-hormone-associated tissues like kidney and bladder
These differences highlight the importance of careful consideration when using rat models for AKR1C3-related research, particularly for studies related to hormone metabolism in prostate tissue .
Multiple methods have been validated for AKR1C3 detection, each with specific advantages:
Immunohistochemistry (IHC):
Most specific antibody: Sigma/Millipore Anti-AKR1C3 antibody, mouse monoclonal, clone NP6.G6.A6 shows higher specificity compared to rabbit polyclonal antibodies
Quantification: H-score can be used to quantify percent of nuclear immunoreactivity for AKR1C3 with varying disease involvement
Sample preparation: Formalin-fixed, paraffin-embedded tissue sections are typically used
Western Blot:
Effective antibodies include Sheep Anti-Human Aldo-keto Reductase 1C3/AKR1C3 Antigen Affinity-purified Polyclonal Antibody
Quantitative RT-PCR:
Shows concordance with protein detection methods in cases of relapsed/refractory and/or minimal residual disease
Particularly useful for examining mRNA expression levels across different tissues or disease states
For cancer detection, combining these methods provides more comprehensive characterization of AKR1C3 expression patterns .
Given the high sequence similarity (>86%) between AKR1C3 and related enzymes, antibody validation is crucial:
Use appropriate controls:
Cross-reactivity testing:
Multiple detection methods:
Antibody selection:
Proper validation ensures accurate interpretation of results, particularly in tissues where multiple AKR family members may be expressed .
AKR1C3 has been implicated in multiple cancer types through several mechanisms:
Hormone-dependent cancers:
In prostate cancer: Generates testosterone from androstenedione, potentially driving androgen receptor signaling even in the context of androgen deprivation therapy
In breast cancer: May influence estrogen metabolism and signaling pathways
Hepatocellular carcinoma (HCC):
High expression predicts poor prognosis and shorter median survival time
Knockdown of AKR1C3 decreases cell proliferation and inhibits tumorigenesis in HCC cells
Acute lymphoblastic leukemia/lymphoma (ALL):
Highly expressed in T-ALL compared to B-ALL (H-score of 172-190 vs. 30-160)
Can be used to detect minimal residual disease in T-ALL and B-ALL patients
Targeted therapies leveraging AKR1C3 expression (e.g., prodrug ACHM-025) show promising results in T-ALL treatment
The enzyme's role in steroid hormone metabolism, prostaglandin synthesis, and potentially other signaling pathways makes it a significant contributor to cancer progression in multiple contexts .
AKR1C3 has emerging value as both a diagnostic and prognostic biomarker:
Diagnostic applications:
Hepatocellular carcinoma: ROC analysis yielded an AUC value of 0.948, indicating excellent diagnostic potential
T-ALL: Higher H-score (172-190) compared to B-ALL cases (H-score 30-160) allows differentiation between leukemia subtypes
Minimal residual disease detection: AKR1C3 expression by IHC, Protein Wes, and RT-qPCR shows concordance in relapsed/refractory T-ALL cases
Prognostic value:
High expression of AKR1C3 predicts poor prognosis and shorter median survival time in HCC
May serve as a predictive biomarker for response to specific therapies, particularly AKR1C3-activated prodrugs
Correlation between AKR1C3 expression levels and in vivo efficacy of therapies like ACHM-025 provides a potential predictive biomarker for treatment response
These findings suggest that AKR1C3 assessment should be considered in the diagnostic workup and prognostic evaluation of certain cancers, particularly HCC and T-ALL .
Several approaches to targeting AKR1C3 have shown promise in research settings:
Direct enzyme inhibition:
Steroidal bile acid fused tetrazoles: C24 bile acids with C-ring fused tetrazoles showed moderate to strong AKR1C3 inhibition (37-88%)
X-ray crystallography at 1.4 Å resolution revealed that the C24 carboxylate anchors to the catalytic oxyanion site (H117, Y55) while the tetrazole interacts with tryptophan (W227) important for steroid recognition
Some inhibitors show specificity for AKR1C3 over AKR1C2, with IC₅₀ values of approximately 7 μM
Prodrug activation approach:
ACHM-025, a third-generation AKR1C3-activated prodrug, is selectively activated by AKR1C3 to a nitrogen mustard DNA alkylating agent
Shows potent in vivo efficacy against T-ALL patient-derived xenografts (PDXs) and eradicated the disease in 7 PDXs
More effective than cyclophosphamide both as a single agent and in combination with cytarabine/6-mercaptopurine
ACHM-025 in combination with nelarabine was curative when used to treat a chemoresistant T-ALL PDX in vivo
The efficacy of these approaches correlates with AKR1C3 expression levels, suggesting the need for patient stratification based on AKR1C3 expression .
Based on the literature, several experimental models have proven valuable for AKR1C3 research:
Cell line models:
HCC cell lines (e.g., Huh-7) for studying AKR1C3 in liver cancer
A549 human lung carcinoma cell line for protein expression studies
Genetic modification of AKR1C3-negative cell lines (e.g., HCT116) for controlled expression studies
Animal models:
Rat models show comparable but not identical distribution of AKR1C3 homolog compared to humans
Patient-derived xenograft (PDX) models in mice for studying therapeutic efficacy of AKR1C3-targeted approaches
Three sentinel mice included for Day 28 time point analyses revealed no detectable human leukemia cells after ACHM-025 treatment
Primary tissue studies:
Human tissue samples from normal and diseased states (prostate, kidney, bladder, testis)
Patient samples for correlation of AKR1C3 expression with clinical outcomes
When selecting models, researchers should consider the known differences in AKR1C3 expression and distribution between species, as well as the specific research question being addressed .
While the provided search results don't directly address post-translational modifications of AKR1C3, structural insights suggest important considerations:
The X-ray crystallography data at 1.4 Å resolution reveals that AKR1C3 forms a complex with NADP+, and specific residues (H117, Y55, W227) are critical for substrate binding and recognition
The catalytic oxyanion site plays a key role in enzyme function
Interactions between specific inhibitors and the enzyme binding pocket highlight the importance of structural integrity for enzymatic activity
Further research into post-translational modifications of AKR1C3 would be valuable, particularly examining:
Phosphorylation states that might alter enzyme activity
Potential redox modifications given the enzyme's role in redox reactions
Changes in subcellular localization that might affect substrate accessibility
Assessment of AKR1C3 enzymatic activity requires careful consideration of substrates and detection methods:
Substrate selection:
Steroid metabolism: Measuring conversion of androstenedione to testosterone
Prostaglandin metabolism: Measuring reduction of prostaglandin D2 to PGF2
Assay approaches:
Fluorescence assays in yeast cells can be used to assess enzymatic activity and specificity
IC₅₀ determination for inhibitors provides quantitative assessment of compounds that modulate AKR1C3 activity
Activity comparison between AKR1C3 and related enzymes (e.g., AKR1C2) helps establish specificity
Structural analyses:
X-ray crystallography at high resolution (e.g., 1.4 Å) provides insights into substrate binding and catalytic mechanisms
Molecular docking can predict binding geometries of substrates and inhibitors
For therapeutic development, correlating enzymatic activity with biological effects (e.g., cell proliferation, tumor growth) is essential to establish functional relevance .
While the provided search results don't specifically address AKR1C3 polymorphisms, researchers should consider:
Genetic screening:
Sequencing of AKR1C3 in study populations to identify relevant polymorphisms
Correlation of polymorphisms with enzyme activity and clinical outcomes
Functional characterization:
Expression of polymorphic variants to assess differences in catalytic activity
Structural studies to understand how polymorphisms might affect substrate binding or catalysis
Population considerations:
Clinical correlations:
Given the emerging therapeutic applications targeting AKR1C3, understanding polymorphic variations will be increasingly important for personalized medicine approaches .
Based on current findings, several areas warrant further investigation:
Expanded therapeutic applications:
Development of next-generation AKR1C3-activated prodrugs building on the success of ACHM-025
Exploration of combination therapies leveraging AKR1C3 targeting with existing treatment modalities
Investigation of AKR1C3 inhibitors in hormone-dependent cancers beyond prostate cancer
Mechanistic understanding:
Elucidation of AKR1C3's roles in non-hormone-associated tissues like kidney and bladder
Investigation of how AKR1C3 influences cancer cell metabolism beyond steroid hormone production
Understanding the regulation of AKR1C3 expression in different tissue and disease contexts
Biomarker development:
Standardization of AKR1C3 detection methods for clinical application
Validation of AKR1C3 as a minimal residual disease marker in leukemia
Integration of AKR1C3 assessment into multiparameter diagnostic and prognostic models
Structural biology:
Further refinement of AKR1C3 structural understanding to design more potent and specific inhibitors
Investigation of protein-protein interactions involving AKR1C3
These research directions have potential to advance both basic understanding of AKR1C3 biology and clinical applications in cancer diagnosis and treatment .
Several challenges and limitations in current AKR1C3 research require attention:
Technical challenges:
Need for standardized detection methods with high specificity given the >86% sequence identity with related AKR enzymes
Difficulty in distinguishing AKR1C3 activity from other related enzymes in complex biological samples
Requirement for multiple validation approaches when characterizing new antibodies or detection methods
Knowledge gaps:
Incomplete understanding of AKR1C3's functions in non-hormone-associated tissues
Limited data on how AKR1C3 expression is regulated in different physiological and pathological contexts
Need for more comprehensive characterization of species differences when using animal models
Therapeutic development:
Challenge of developing inhibitors with sufficient specificity for AKR1C3 over related enzymes
Potential off-target effects of AKR1C3-activated prodrugs in tissues with high AKR1C3 expression
Need for biomarkers to identify patients most likely to benefit from AKR1C3-targeted therapies
Addressing these limitations will be essential for advancing both fundamental understanding of AKR1C3 biology and its clinical applications in cancer diagnosis and treatment .
The AKR1C3 gene is located on chromosome 10p15-p14 and shares high sequence identity with three other gene members clustered in the same region . The gene encodes a protein that is involved in the reduction of prostaglandin D2, prostaglandin H2, and phenanthrenequinone, as well as the oxidation of prostaglandin F2α to prostaglandin D2 . The enzyme is also capable of metabolizing estrogen and progesterone .
AKR1C3 plays a crucial role in steroid metabolism. It catalyzes oxidation/reduction reactions at the 3-alpha, 20-alpha, and 17-beta positions of steroids . This enzyme is involved in the conversion of adrenal androgens, such as dehydroepiandrosterone and androstenedione, into androstenediol and testosterone . Additionally, AKR1C3 is known as Prostaglandin F Synthase because it reduces prostaglandin D2 to F2, which may play a role in allergic conditions such as asthma .
AKR1C3 is overexpressed in prostate cancer and is associated with the development of castration-resistant prostate cancer (CRPC) . Its overexpression may serve as a promising biomarker for prostate cancer progression . Furthermore, AKR1C3 may play an important role in the pathogenesis of allergic diseases such as asthma and may also have a role in controlling cell growth and/or differentiation .
Recombinant human AKR1C3 is produced using recombinant DNA technology, which involves inserting the human AKR1C3 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in various research applications to study its function, structure, and role in disease processes.