Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Drug Metab Dispos ; 51(9): 1188-1195, 2023 09.
Article in English | MEDLINE | ID: mdl-37344179

ABSTRACT

Aldo-keto reductase 1C3 (AKR1C3) plays a role in the detoxification and activation of clinical drugs by catalyzing reduction reactions. There are approximately 400 single-nucleotide polymorphisms (SNPs) in the AKR1C3 gene, but their impact on the enzyme activity is still unclear. This study aimed to clarify the effects of SNPs of AKR1C3 with more than 0.5% global minor allele frequency on the reductase activities for its typical substrates. Recombinant AKR1C3 wild-type and R66Q, E77G, C145Y, P180S, or R258C variants were constructed using insect Sf21 cells, and reductase activities for acetohexamide, doxorubicin, and loxoprofen by recombinant AKR1C3s were measured by liquid chromatography-tandem mass spectrometry. Among the variants tested, the C145Y variant showed remarkably low (6%-14% of wild type) intrinsic clearances of reductase activities for all three drugs. Reductase activities of these three drugs were measured using 34 individual Japanese liver cytosols, revealing that heterozygotes of the SNP g.55101G>A tended to show lower reductase activities for three drugs than homozygotes of the wild type. Furthermore, genotyping of the SNP g.55101G>A causing C145Y in 96 Caucasians, 166 African Americans, 192 Koreans, and 183 Japanese individuals was performed by polymerase chain reaction-restriction fragment length polymorphism. This allelic variant was specifically detected in Asians, with allele frequencies of 6.8% and 3.6% in Koreans and Japanese, respectively. To conclude, an AKR1C3 allele with the SNP g.55101G>A causing C145Y would be one of the causal factors for interindividual variabilities in the efficacy and toxicity of drugs reduced by AKR1C3. SIGNIFICANCE STATEMENT: This is the first study to clarify that the AKR1C3 allele with the SNP g.55101G>A causing C145Y results in a decrease in reductase activity. Since the allele was specifically observed in Asians, the allele would be a factor causing an interindividual variability in sensitivity of drug efficacy or toxicity of drugs reduced by AKR1C3 in Asians.


Subject(s)
Doxorubicin , Humans , Alleles , Gene Frequency/genetics , Aldo-Keto Reductase Family 1 Member C3/genetics
2.
Sci Rep ; 13(1): 3280, 2023 02 25.
Article in English | MEDLINE | ID: mdl-36841845

ABSTRACT

The principal pathology of psoriasis is impaired skin barrier function, epidermal thickening, and granular layer loss. Exposure to extrinsic factors such as tobacco smoke and air pollutants is associated with the development of psoriasis. Aryl hydrocarbon receptors (AHRs) are activated by extrinsic factors associated with the development of psoriasis and act as transcriptional regulators. Expression of aldo-keto reductase (AKR) 1C3 in the epidermal spinous layer regulates epidermal keratinocyte differentiation via the AHR signaling pathway. We investigated whether single nucleotide polymorphisms (SNPs) in AKR1C3 are associated with the pathogenesis of psoriasis. The proportions of rs12529 G/C, C/C variants, and rs12387 A/A, A/G variants were twofold higher in Japanese psoriasis patients (n = 231) compared with a Japanese healthy cohort. The SNPs were significantly more common than the majority variants in female patients with disease onset ≤ 22 years of age. Patients with rs12529 G > C and rs12387 A > G SNPs exhibited significantly lower AKR1C3 expression and higher expression of late differentiation markers. In conclusion, AKR1C3 downregulation caused by rs12529 G > C and rs12387 A > G SNPs in the epidermis induces abnormal early differentiation of keratinocytes and skin barrier dysfunction, which may contribute to the genetic pathogenesis of psoriasis in young females.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3 , Polymorphism, Single Nucleotide , Psoriasis , Female , Humans , Epidermal Cells , Epidermis , Keratinocytes , Psoriasis/genetics , Aldo-Keto Reductase Family 1 Member C3/genetics
3.
Oncologist ; 27(11): e870-e877, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36067250

ABSTRACT

PURPOSE: Aldo-keto reductase family 1 member C3 (AKR1C3) is important in prostate cancer progression, being a potential biomarker in metastatic castration-resistant prostate cancer (mCRPC). Previous explorations of AKR1C3 are mainly based on tissue samples. This study investigates using plasma-based liquid biopsy to validate the prognostic and predictive value of AKR1C3 in patients with mCRPC . MATERIALS AND METHODS: We prospectively recruited 62 patients with mCRPC. All patients received repeated prostate biopsies at the time of mCRPC diagnosis, and immunohistochemistry (IHC) staining was used to detect protein expression of AKR1C3 in the tissues. We took their blood simultaneously and performed digital droplet polymerase chain reaction (ddPCR) to measure expression levels of AKR1C3 in the exosomes. The detected plasma and tissue AKR1C3 expression levels were analyzed for patients' overall survival (OS) and progression-free survival under first-line abiraterone use (ABI-PFS). RESULTS: All other baseline characteristics were balanced between the 2 groups. 15/62 (24.2%) and 25/62 (40.3%) patients showed AKR1C3-EXO positive (≥20 copies/20 µL) and AKR1C3-IHC positive, respectively. Positive AKR1C3-EXO expression was associated with decreased patients' survival [ABI-PFS: 3.9 vs 10.1 months, P < .001; OS: 16.2 vs 32.5 months, P < .001]. AKR1C3-IHC positivity was also correlated with ABI-PFS and OS (P = .010, P = .016). In patients with worse baseline blood tests (including higher alkaline phosphatase (ALP) and lactate dehydrogenase (LDH) level and lower hemoglobin (HB) level), and lower ISUP/WHO group (<4), their OS was significantly worse when showing AKR1C3-EXO positive. CONCLUSION: AKR1C3-EXO is associated with patient prognosis regarding OS and ABI-PFS and can be used as a biomarker in mCRPC.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Aldo-Keto Reductase Family 1 Member C3/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prognosis , Biomarkers , RNA, Messenger
4.
J Steroid Biochem Mol Biol ; 221: 106121, 2022 07.
Article in English | MEDLINE | ID: mdl-35489629

ABSTRACT

Aldo-keto reductase (AKR) 1C3, also known as type 5 17ß-hydroxysteroid dehydrogenase and prostaglandin F synthase, is a member of the AKR superfamily that reduces aldehydes and ketones to primary and secondary alcohols. It plays an essential role in the peripheral formation of androgens and is implicated in several steroid hormone dependent diseases including prostate cancer, breast cancer, and polycystic ovary syndrome (PCOS). AKR1C3 has 14 nonsynonymous single nucleotide polymorphisms (nsSNPs) with different global frequencies and ethnic distributions. Association studies support their role in disease, but a detailed functional genomic analysis of these variants is lacking. One study examined five AKR1C3 nsSNPs for their ability to reduce exemestane, an aromatase inhibitor used to treat breast cancer, to 17ß-dihydroexemestane, and reported a 17-250-fold reduction in catalytic efficiency of H5Q, E77G, K104D, and R258C variants compared to wild type (WT). This observation provided the impetus to examine the impact of these variants on AKR1C3 function. Here, we purified AKR1C3 WT, and the top four most frequently occurring nsSNPs, H5Q, E77G, K104D, and R258C, from E. coli to expand upon their characterization and illuminate functional differences that could affect disease outcome and treatment. While we found negligible deviations in steady state kinetics, the K104D variant showed reduced thermal stability compared to WT. The presence of NAD(P)+ restored the stability of the variant. As it is unlikely that the apoenzyme will exist within the cell without cofactor bound the K104D is not expected to manifest a phenotype.


Subject(s)
Breast Neoplasms , Escherichia coli , 17-Hydroxysteroid Dehydrogenases , 3-Hydroxysteroid Dehydrogenases/metabolism , Aldo-Keto Reductase Family 1 Member C3/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Aldo-Keto Reductases , Escherichia coli/metabolism , Female , Humans , Hydroxyprostaglandin Dehydrogenases , Male , Nucleotides
5.
Cell Biol Int ; 46(6): 965-975, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35257428

ABSTRACT

ARID3A is upregulated in colorectal cancer and can promote the proliferation and metastasis of cancer cells. However, patients with higher level of ARID3A have a better prognosis. This study aimed to uncover the mechanism by which ARID3A benefits the prognosis of colon cancer. Our results indicated that ARID3A upregulation enhanced the chemosensitivity of colon cancer cells to 5-fluorouracil (5-FU), whereas ARID3A downregulation inhibited the chemosensitivity of colon cancer cells to 5-FU. Through database analysis, we found that AKR1C3, a drug resistance-related gene, was the target of ARID3A. Moreover, AKR1C3 was downregulated in colon cancer tissues compared to normal tissues. Next, we assessed the interaction between AKR1C3 and ARID3A, and found that ARID3A inhibited the transcription of AKR1C3, leading to the downregulation of AKR1C3 in colon cancer cells. We also verified that AKR1C3 inhibited the chemosensitivity of colon cancer cells to 5-FU. Moreover, patients with higher ratio of ARID3A to AKR1C3 had a better prognosis. This study suggested that ARID3A promoted chemosensitivity of colon cancer cells by inhibiting AKR1C3 in colon cancer. The ratio of ARID3A to AKR1C3 is a good marker to predict the prognosis of colon cancer patients.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3 , Colonic Neoplasms , DNA-Binding Proteins , Transcription Factors , Aldo-Keto Reductase Family 1 Member C3/antagonists & inhibitors , Aldo-Keto Reductase Family 1 Member C3/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation
6.
Sci Rep ; 12(1): 55, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997089

ABSTRACT

It is being debated whether prostate-specific antigen (PSA)-based screening effectively reduces prostate cancer mortality. Some of the uncertainty could be related to deficiencies in the age-based PSA cut-off thresholds used in screening. Current study considered 2779 men with prostate cancer and 1606 men without a cancer diagnosis, recruited for various studies in New Zealand, US, and Taiwan. Association of PSA with demographic, lifestyle, clinical characteristics (for cases), and the aldo-keto reductase 1C3 (AKR1C3) rs12529 genetic polymorphisms were analysed using multiple linear regression and univariate modelling. Pooled multivariable analysis of cases showed that PSA was significantly associated with demographic, lifestyle, and clinical data with an interaction between ethnicity and age further modifying the association. Pooled multivariable analysis of controls data also showed that demographic and lifestyle are significantly associated with PSA level. Independent case and control analyses indicated that factors associated with PSA were specific for each cohort. Univariate analyses showed a significant age and PSA correlation among all cases and controls except for the US-European cases while genetic stratification in cases showed variability of correlation. Data suggests that unique PSA cut-off thresholds factorized with demographics, lifestyle and genetics may be more appropriate for prostate cancer screening.


Subject(s)
Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Aldo-Keto Reductase Family 1 Member C3/genetics , Body Mass Index , Case-Control Studies , Cohort Studies , Demography , Early Detection of Cancer , Ethnicity , Humans , Life Style , Linear Models , Male , Mass Screening , Middle Aged , Neoplasm Grading , New Zealand/epidemiology , Polymorphism, Single Nucleotide , Taiwan/epidemiology , United States/epidemiology , Young Adult
7.
Breast Cancer ; 29(1): 38-49, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34351578

ABSTRACT

PURPOSE: Polycyclic aromatic hydrocarbons (PAHs) are a group of environmental pollutants associated with multiple cancers, including female breast cancer. Several xenobiotic metabolism genes (XMGs), including the CYP450 family, play an important role in activating and detoxifying PAHs, and variations in the activity of the enzymes they encode can impact this process. This study aims to examine the association between XMGs and breast cancer, and to assess whether these variants modify the effects of PAH exposure on breast cancer risk. METHODS: In a case-control study in Vancouver, British Columbia, and Kingston, Ontario, 1037 breast cancer cases and 1046 controls had DNA extracted from blood or saliva and genotyped for 138 single nucleotide polymorphisms (SNPs) and tagSNPs in 27 candidate XMGs. Occupational PAH exposure was assessed using a measurement-based job-exposure matrix. RESULTS: An association between genetic variants and breast cancer was observed among six XMGs, including increased risk among the minor allele carriers of AKR1C3 variant rs12387 (OR 2.71, 95% CI 1.42-5.19) and AKR1C4 variant rs381267 (OR 2.50, 95% CI 1.23-5.07). Heterogeneous effects of occupational PAH exposure were observed among carriers of AKR1C3/4 variants, as well as the PTGS2 variant rs5275. CONCLUSION: Our findings support an association between SNPs of XMGs and female breast cancer, including novel genetic variants that modify the toxicity of PAH exposure. These results highlight the interplay between genetic and environmental factors, which can be helpful in understanding the modifiable risks of breast cancer and its complex etiology.


Subject(s)
Breast Neoplasms/epidemiology , Gene-Environment Interaction , Polycyclic Aromatic Hydrocarbons/toxicity , Xenobiotics/metabolism , Adult , Aged , Aged, 80 and over , Aldo-Keto Reductase Family 1 Member C3/genetics , Canada/epidemiology , Case-Control Studies , Cyclooxygenase 2/genetics , Female , Heterozygote , Humans , Middle Aged , Occupational Exposure , Oxidoreductases/genetics , Polymorphism, Single Nucleotide
8.
Int J Mol Sci ; 22(22)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34830394

ABSTRACT

Signet ring cell gastric carcinoma (SRCGC) is a lethal malignancy that has developed drug resistance to cisplatin therapies. The aim of this study was to characterize the acquisition of the cisplatin-resistance SRCGC cell line (KATO/DDP cells) and to understand the molecular mechanisms underlying cisplatin resistance. Transcriptomic and bioinformatic analyses were used to identify the candidate gene. This was confirmed by qPCR and Western blot. Aldoketoreductase1C1 and 1C3 (AKR1C1 and AKR1C3) were the most promising molecules in KATO/DDP cells. A specific inhibitor of AKR1C1 (5PBSA) and AKR1C3 (ASP9521) was used to enhance cisplatin-induced KATO/DPP cell death. Although cisplatin alone induced KATO/DDP apoptosis, a combination treatment of cisplatin and the AKR1C inhibitors had no influence on percent cell apoptosis. In conjunction with the autophagy inhibitor, 3MA, attenuated the effects of 5PBSA or ASP9521 to enhance cisplatin-induced cell death. These results indicated that AKR1C1 and 1C3 regulated cisplatin-induced KATO/DDP cell death via autophagy. Moreover, cisplatin in combination with AKR1C inhibitors and N-acetyl cysteine increased KATO/DDP cells' viability when compared with a combination treatment of cisplatin and the inhibitors. Taken together, our results suggested that AKR1C1 and 1C3 play a crucial role in cisplatin resistance of SRCGC by regulating redox-dependent autophagy.


Subject(s)
20-Hydroxysteroid Dehydrogenases/genetics , Aldo-Keto Reductase Family 1 Member C3/genetics , Carcinoma, Signet Ring Cell/drug therapy , Stomach Neoplasms/drug therapy , Autophagic Cell Death/drug effects , Autophagic Cell Death/genetics , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcriptome/drug effects
9.
Cell Signal ; 84: 110038, 2021 08.
Article in English | MEDLINE | ID: mdl-33984486

ABSTRACT

BACKGROUND: Drug resistance is an important cause of death for most patients with chronic myeloid leukemia (CML). The bone marrow microenvironment is believed to be mainly responsible for resistance to BCR-ABL tyrosine kinase inhibitors. The mechanism involved, however, is still unclear. METHODS: Bioinformatic analysis from GEO database of AKR1C3 was utilized to identify the AKR1C3 expression in CML cells under bone marrow microenvironment. Western blot and qPCR were performed to detect the AKR1C3 expression in two CML cell lines K562 and KU812 cultured +/- bone microenvironment derived stromal cells. CCK-8, soft agar colony assay, and Annexin V/PI assay were performed to detect the sensitivity of CML cells (K562 and KU812) to Imatinib under a gain of or loss of function of AKR1C3 treatment. The CML murine model intravenous inoculated with K562-OE-vector and K562-OE-AKR1C3 cells were established to estimate the effect of AKR1C3 inhibitor Indomethacin on Imatinib resistance. The bioinformatic analysis of miRNA databases was used to predict the potential miRNAs targeting AKR1C3. And the luciferase assay was utilized to validate the target relationship between miR-379-5p and AKR1C3. And, the soft agar colony assay and Annexin V/PI were used to validate the effect of miR-379-5p in AKR1C3 induced Imatinib resistance. RESULTS: In present study, we investigated AKR1C3 was highly expressed in CML under bone marrow microenvironment. AKR1C3 decreased Imatinib activity in K562 and KU812 cells, while inhibition of AKR1C3 could enhance Imatinib sensitivity in vitro study. Furthermore, murine model results showed combination use of AKR1C3 inhibitor Indomethacin effectively prolong mice survival, indicating that AKR1C3 is a promising target to enhance Imatinib treatment. Mechanically, AKR1C3 was found to be suppressed by miR-379-5p, which was down-expression in bone marrow microenvironment. Besides, we found miR-379-5p could bind AKR1C3 3'UTR but not degrade its mRNA level. Further, gain of miR-379-5p rescued the imatinib resistance induced by AKR1C3 overexpression in CML cells. CONCLUSIONS: Altogether, our study identifies a novel signaling regulation of miR-379-5p/AKR1C3/EKR axis in regulating IM resistance in CML cell, and provides a scientific base for exploring AKR1C3 as a biomarker in impeding IM resistance in CML.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3 , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , MicroRNAs , Aldo-Keto Reductase Family 1 Member C3/genetics , Animals , Apoptosis , Bone Marrow/metabolism , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , MicroRNAs/genetics , Tumor Microenvironment
10.
Mol Cell Endocrinol ; 527: 111220, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33675863

ABSTRACT

BACKGROUND: Changes in androgen dynamics within adipose tissue have been proposed as modulators of body fat accumulation. In this context, AKR1C2 likely plays a significant role by inactivating 5α-dihydrotestosterone. AIM: To characterize AKR1C2 expression patterns across adipose depots and cell populations and to provide insight into the link with body fat distribution and genetic regulation. METHODS: We used RNA sequencing data from severely obese patients to assess patterns of AKR1C2 and AKR1C3 expression in abdominal adipose tissue depots and cell fractions. We additionally used data from 856 women to assess AKR1C2 heritability and to link its expression in adipose tissue with body fat distribution. Further, we used public resources to study AKR1C2 genetic regulation as well as reference epigenome data for regulatory element profiling and functional interpretation of genetic data. RESULTS: We found that mature adipocytes and adipocyte-committed adipocyte progenitor cells (APCs) had enriched expression of AKR1C2. We found adipose tissue AKR1C2 and AKR1C3 expression to be significantly and positively associated with percentage trunk fat mass in women. We identified strong genetic regulation of AKR1C2 by rs28571848 and rs34477787 located on the binding sites of two nuclear transcription factors, namely retinoid acid-related orphan receptor alpha and the glucocorticoid receptor. CONCLUSION: We confirm the link between AKR1C2, adipogenic differentiation and adipose tissue distribution. We provide insight into genetic regulation of AKR1C2 by identifying regulatory variants mapping to binding sites for the glucocorticoid receptor and retinoid acid-related orphan receptor alpha which may in part mediate the effect of AKR1C2 expression on body fat distribution.


Subject(s)
Abdominal Fat/enzymology , Body Fat Distribution , Hydroxysteroid Dehydrogenases , Polymorphism, Genetic , Response Elements , Adult , Aldo-Keto Reductase Family 1 Member C3/biosynthesis , Aldo-Keto Reductase Family 1 Member C3/genetics , Female , Gene Expression Regulation, Enzymologic , Humans , Hydroxysteroid Dehydrogenases/biosynthesis , Hydroxysteroid Dehydrogenases/genetics
11.
Toxicol Lett ; 342: 50-57, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33581289

ABSTRACT

Carbonyl reduction biotransformation pathway of anthracyclines (doxorubicin, daunorubicin) is a significant process, associated with drug metabolism and elimination. However, it also plays a pivotal role in anthracyclines-induced cardiotoxicity and cancer resistance. Herein, carbonyl reduction of eight anthracyclines, at in vivo relevant concentrations (20 µM), was studied in human liver cytosol, to describe the relationship between their structure and metabolism. Significant differences of intrinsic clearance between anthracyclines, ranging from 0,62-74,9 µL/min/mg were found and associated with data from in silico analyses, considering their binding in active sites of the main anthracyclines-reducing enzymes: carbonyl reductase 1 (CBR1) and aldo-keto reductase 1C3 (AKR1C3). Partial atomic charges of carbonyl oxygen atom were also determined and considered as a factor associated with reaction rate. Structural features, including presence or absence of side-chain hydroxy group, a configuration of sugar chain hydroxy group, and tetracyclic rings substitution, affecting anthracyclines susceptibility for carbonyl reduction were identified.


Subject(s)
Aclarubicin/metabolism , Cytosol/metabolism , Doxorubicin/analogs & derivatives , Hepatocytes/metabolism , Oxidoreductases/metabolism , Aclarubicin/chemistry , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Aldo-Keto Reductase Family 1 Member C3/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Biotransformation , Doxorubicin/chemistry , Doxorubicin/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Protein Conformation
12.
Biochem Biophys Res Commun ; 540: 83-89, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33450484

ABSTRACT

Intracrine androgen synthesis plays a critical role in the development of castration-resistant prostate cancer (CRPC). Aldo-keto reductase family 1 member C3 (AKR1C3) is a vital enzyme in the intracrine androgen synthesis pathway. In this study, mesoporous silica nanoparticles (MSNs) were employed to deliver small interfering RNA targeting AKR1C3 (siAKR1C3) to downregulate AKR1C3 expression in CPRC cells. The optimal weight ratio of MSNs/siAKR1C3 was determined by a gel retardation assay. Prostate cancer cells such as VCaP cells, which intracrinally express AKR1C3, and LNCaP-AKR1C3 cells stably transfected with AKR1C3 were used to investigate the antitumour effect of MSNs-siAKR1C3. Fluorescence detection and Western blot analyses were applied to confirm the entrance of MSNs-siAKR1C3 into the cells. A SRB (Sulforhodamine B) assay was employed to assess the cell viability, and a radioimmunoassay was used to measure the androgen concentration. Moreover, real-time PCR (RT-PCR), Western blot analysis and ELISA were used to determine the transcription and expression of prostate-specific antigen (PSA), AKR1C3 and androgen receptor (AR). Meanwhile, a reporter gene assay was performed to determine the AR activity. Additionally, a castrated nude mouse xenograft tumour model was produced to verify the inhibitory effect of MSNs-siAKR1C3 in vivo. The results showed that the optimal weight ratio of MSNs/siAKR1C3 was 140:1, and the complex could effectively enter cells, downregulate AKR1C3 expression, reduce the androgen concentration, inhibit AR activation, and inhibit CRPC development both in vitro and in vivo. These results indicate that decreasing intracrine androgen synthesis and inactivating AR signals by MSNs-siAKR1C3 may be a potential effective method for CRPC treatment.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/genetics , Androgens/biosynthesis , Nanoparticles/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , RNA, Small Interfering/therapeutic use , Silicon Dioxide/therapeutic use , Aldo-Keto Reductase Family 1 Member C3/deficiency , Aldo-Keto Reductase Family 1 Member C3/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Testosterone/biosynthesis , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
13.
Aging (Albany NY) ; 13(3): 4138-4156, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33493134

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common histological type of primary liver cancer and the majority of patients are diagnosed at an advanced stage and have a poor prognosis. AKR1C3 (Aldo-keto reductase family 1 member C3) and AKR1D1 (Aldo-keto reductase family 1 member D1) catalyze the conversion of aldehydes and ketones to alcohols and play crucial roles in multiple cancers. However, the functions of AKR1C3 and AKR1D1 in HCC remain unclear. In our study, data from the public databases were selected as training and validation sets, then 76 HCC patients in our center were chosen as a test set. Bioinformatics methods suggested AKR1C3 was overexpressed in HCC and AKR1D1 was down-regulated. The receiver operating characteristic curve (ROC) analysis was performed and the area under curve (AUC) values of AKR1C3 and AKR1D1 were above 0.7 (0.948, 0.836, respectively). Also, the high expression of AKR1C3 and low expression of AKR1D1 predicted poor prognosis and short median survival time. Then, the knockdown of AKR1C3 and overexpression of AKR1D1 in HCC cells were achieved with lentivirus. And both decreased cell proliferation, restrained cell viability, and inhibited tumorigenesis. Moreover, the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted and the results showed that AKR1C3 and AKR1D1 might participate in the MAPK/ERK and androgen receptor (AR) signaling pathway. Furthermore, the AR and phosphorylated ERK1/2 were significantly reduced after the suppression of AKR1C3 or overexpression of AKR1D1. Collectively, AKR1C3 and AKR1D1 might serve as candidate diagnostic and prognostic biomarkers for HCC and provide potential targets for HCC treatment.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/genetics , Carcinoma, Hepatocellular/diagnosis , Liver Neoplasms/diagnosis , Oxidoreductases/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Area Under Curve , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Female , Gene Knockdown Techniques , Gene Ontology , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Oxidoreductases/metabolism , Prognosis , Proportional Hazards Models , Protein Interaction Maps , RNA, Messenger/metabolism , ROC Curve , Survival Rate , Up-Regulation
14.
Arch Toxicol ; 95(1): 67-78, 2021 01.
Article in English | MEDLINE | ID: mdl-33025066

ABSTRACT

Midostaurin is an FMS-like tyrosine kinase 3 receptor (FLT3) inhibitor that provides renewed hope for treating acute myeloid leukaemia (AML). The limited efficacy of this compound as a monotherapy contrasts with that of its synergistic combination with standard cytarabine and daunorubicin (Dau), suggesting a therapeutic benefit that is not driven only by FLT3 inhibition. In an AML context, the activity of the enzyme aldo-keto reductase 1C3 (AKR1C3) is a crucial factor in chemotherapy resistance, as it mediates the intracellular transformation of anthracyclines to less active hydroxy metabolites. Here, we report that midostaurin is a potent inhibitor of Dau inactivation mediated by AKR1C3 in both its recombinant form as well as during its overexpression in a transfected cell model. Likewise, in the FLT3- AML cell line KG1a, midostaurin was able to increase the cellular accumulation of Dau and significantly decrease its metabolism by AKR1C3 simultaneously. The combination of those mechanisms increased the nuclear localization of Dau, thus synergizing its cytotoxic effects on KG1a cells. Our results provide new in vitro evidence of how the therapeutic activity of midostaurin could operate beyond targeting the FLT3 receptor.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Daunorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Staurosporine/analogs & derivatives , Aldo-Keto Reductase Family 1 Member C3/genetics , Aldo-Keto Reductase Family 1 Member C3/metabolism , Biotransformation , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Daunorubicin/metabolism , Drug Synergism , HCT116 Cells , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Staurosporine/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
15.
Cancer Res ; 81(5): 1361-1374, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33361392

ABSTRACT

AKR1C3 is an enzyme belonging to the aldo-ketoreductase family, the members of which catalyze redox transformations involved in biosynthesis, intermediary metabolism, and detoxification. AKR1C3 plays an important role in tumor progression and metastasis, however, little is known about the function and the molecular mechanism underlying the role of AKR1C3 in hepatocellular carcinoma (HCC). In this study, we report that AKR1C3 is significantly upregulated in HCC and that increased AKR1C3 is associated with poor survival. AKR1C3 positively regulated HCC cell proliferation and metastasis in vitro and in vivo. AKR1C3 promoted tumor proliferation and metastasis by activating NF-κB signaling. Furthermore, AKR1C3 regulated NF-κB activity by modulating TRAF6 and inducing its autoubiquitination in HCC cells. Activation of NF-κB released proinflammatory factors that facilitated the phosphorylation of STAT3 and increased tumor cell proliferation and invasion. Gain- and loss-of-function experiments showed that AKR1C3 promoted tumor proliferation and invasion via the IL6/STAT3 pathway. STAT3 also directly bound the AKR1C3 promoter and increased transcription of AKR1C3, thereby establishing a positive regulatory feedback loop. Treatment with the AKR1C3 inhibitors indocin and medroxyprogesterone acetate inhibited tumor growth and invasion and promoted apoptosis in HCC cells. Collectively, these results indicate that a AKR1C3/NF-κB/STAT3 signaling loop results in HCC cell proliferation and metastasis and could be a promising therapeutic target in HCC. SIGNIFICANCE: These findings elucidate a novel AKR1C3-driven signaling loop that regulates proliferation and metastasis in HCC, providing potential prognostic and therapeutic targets in this disease.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Aldo-Keto Reductase Family 1 Member C3/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Proliferation , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Male , Mice, Nude , Prognosis , Xenograft Model Antitumor Assays
16.
Cell Oncol (Dordr) ; 44(2): 357-372, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33211282

ABSTRACT

PURPOSE: Oropharynx squamous cell carcinoma (OPSCC) is a subtype of head and neck squamous cell carcinoma (HNSCC) arising from the base of the tongue, lingual tonsils, tonsils, oropharynx or pharynx. The majority of HPV-positive OPSCCs has a good prognosis, but a fraction of them has a poor prognosis, similar to HPV-negative OPSCCs. An in-depth understanding of the molecular mechanisms underlying OPSCC is mandatory for the identification of novel prognostic biomarkers and/or novel therapeutic targets. METHODS: 14 HPV-positive and 15 HPV-negative OPSCCs with 5-year follow-up information were subjected to gene expression profiling and, subsequently, compared to three extensive published OPSCC cohorts to define robust biomarkers for HPV-negative lesions. Validation of Aldo-keto-reductases 1C3 (AKR1C3) by qRT-PCR was carried out on an independent cohort (n = 111) of OPSCC cases. In addition, OPSCC cell lines Fadu and Cal-27 were treated with Cisplatin and/or specific AKR1C3 inhibitors to assess their (combined) therapeutic effects. RESULTS: Gene set enrichment analysis (GSEA) on the four datasets revealed that the genes down-regulated in HPV-negative samples were mainly involved in immune system, whereas those up-regulated mainly in glutathione derivative biosynthetic and xenobiotic metabolic processes. A panel of 30 robust HPV-associated transcripts was identified, with AKR1C3 as top-overexpressed transcript in HPV-negative samples. AKR1C3 expression in 111 independent OPSCC cases positively correlated with a worse survival, both in the entire cohort and in HPV-positive samples. Pretreatment with a selective AKR1C3 inhibitor potentiated the effect of Cisplatin in OPSCC cells exhibiting higher basal AKR1C3 expression levels. CONCLUSIONS: We identified AKR1C3 as a potential prognostic biomarker in OPSCC and as a potential drug target whose inhibition can potentiate the effect of Cisplatin.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/metabolism , Biomarkers, Tumor/metabolism , Oropharyngeal Neoplasms/metabolism , Aged , Aged, 80 and over , Aldo-Keto Reductase Family 1 Member C3/genetics , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/drug effects , Cisplatin/pharmacology , Down-Regulation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Gene Regulatory Networks , Humans , Male , Middle Aged , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/complications , Prognosis , Up-Regulation/genetics
17.
Prostate ; 80(14): 1223-1232, 2020 10.
Article in English | MEDLINE | ID: mdl-33258507

ABSTRACT

BACKGROUND: Antiandrogens are effective therapies that block androgen receptor (AR) transactivation and signaling in over 50% of castration-resistant prostate cancer (CRPC) patients. However, an estimated 30% of responders will develop resistance to these therapies within 2 years. JNJ-pan-AR is a broad-spectrum AR antagonist that inhibits wild-type AR as well as several mutated versions of AR that have emerged in patients on chronic antiandrogen treatment. In this work, we aimed to identify the potential underlying mechanisms of resistance that may result from chronic JNJ-pan-AR treatment. METHODS: The LNCaP JNJR prostate cancer subline was developed by chronically exposing LNCaP parental cells to JNJ-pan-AR. Transcriptomic and proteomic profiling was performed to identify potential drivers and/or biomarkers of the resistant phenotype. RESULTS: Several enzymes critical to intratumoral androgen biosynthesis, Aldo-keto reductase family 1 member C3 (AKR1C3), UGT2B15, and UGT2B17 were identified as potential upstream regulators of the JNJ-pan-AR resistant cells. While we confirmed the overexpression of all three enzymes in the resistant cells only AKR1C3 expression played a functional role in driving JNJ-pan-AR resistance. We also discovered that AKR1C3 regulates UGT2B15 and UGT2B17 expression in JNJ-pan-AR resistant cells. CONCLUSIONS: This study supports the rationale to further investigate the benefits of AKR1C3 inhibition in combination with antiandrogens to prevent CRPC disease progression.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/metabolism , Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Aldo-Keto Reductase Family 1 Member C3/biosynthesis , Aldo-Keto Reductase Family 1 Member C3/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Genomics , Glucuronosyltransferase/biosynthesis , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Male , Minor Histocompatibility Antigens/biosynthesis , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Proteomics , Receptors, Androgen/metabolism , Transcription, Genetic
18.
Dis Markers ; 2020: 8880004, 2020.
Article in English | MEDLINE | ID: mdl-33354250

ABSTRACT

OBJECTIVE: A growing body of emerging evidence indicates that metabolic processes play a pivotal role in the biological processes underlying acute myocardial infarction (AMI). The aim of the current study was to identify featured metabolism-related genes in patients with AMI using a support vector machine (SVM) and to further explore the value of these genes in the diagnosis of AMI. METHODS: Gene microarray expression data related to AMI were downloaded from the GSE66360 dataset in the Gene Expression Omnibus (GEO) database. This data set consisted of 50 AMI samples and 49 normal controls that were randomly classified into a discovery cohort (21 AMI samples and 22 normal controls) and a validation cohort (28 AMI and 28 normal controls). We applied a machine learning method that combined SVM with recursive feature elimination (RFE) to discriminate AMI patients from normal controls. Based on this, an SVM classifier was constructed. Receiver operating characteristic (ROC) analysis was used to investigate the predictive value for the early diagnosis of AMI in the two cohorts and was then further verified in an independent external cohort. RESULTS: Three metabolism-related genes were identified based on SVM-RFE (AKR1C3, GLUL, and PDE4B). The SVM classifier based on the three genes allowed for excellent discrimination between AMI and healthy samples in both the discovery cohort (AUC = 0.989) and the validation cohort (AUC = 0.964), and this was further confirmed in the GSE68060 dataset (AUC = 0.839). Additionally, the SVM classifier allowed for perfect discrimination between recurrent AMI events and nonrecurrent events in the GSE68060 cohort (AUC = 0.992). GO and KEGG pathway enrichment analysis of the identified featured genes revealed significant enrichment of specific metabolic pathways. CONCLUSION: The identified metabolism-related genes may play important roles in the development of AMI and may represent diagnostic and therapeutic biomarkers of AMI.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Glutamate-Ammonia Ligase/genetics , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Cohort Studies , Databases, Genetic , Gene Expression Profiling , Genetic Markers , Humans , Machine Learning , Metabolic Networks and Pathways/genetics , ROC Curve , Support Vector Machine
19.
BMC Urol ; 20(1): 71, 2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32560654

ABSTRACT

BACKGROUND: Androgen deprivation therapy (ADT) is a standard treatment for advanced prostate cancer (PCa). However, PCa recurrence and progression rates during ADT are high. Until now, there has been no evidence regarding when progression begins. This study evaluated the gene expression of intraprostatic androgen receptor (AR) and steroidogenic enzymes in the early stages of ADT. METHODS: Prostate tissue samples were taken from PCa patients with urinary retention who received ADT (ADT-PCa; n = 10) and were further subgrouped into ADT ≤12 months (n = 4) and ADT > 12 months (n = 6). The ADT-PCa tissues were then compared with BPH (n = 12) and primary (no treatment) PCa tissues (n = 16). mRNA for gene expression analysis of AR and steroidogenic enzymes was extracted from formalin-fixed paraffin embedded (FFPE) tissues and analyzed by real-time PCR. Protein expression was evaluated by immunohistochemistry with specific antibodies. RESULTS: AR gene expression was higher in the ADT-PCa group than in the BPH or primary PCa group. Both the ADT ≤12 and > 12 months subgroups had significantly higher relative gene expression levels of AR (p < 0.01 and 0.03, respectively) than the primary PCa group. In the ADT-PCa group, AR protein expression showed an increasing trend in the ADT ≤12 months subgroup and was significantly elevated in the ADT > 12 months subgroup compared with the PCa group (100%; p < 0.01). Half (50%) of the patients in the ADT ≤12 months subgroup were found to have upregulation of AR, and one showed upregulation beginning at 3 months of ADT. A trend toward elevated relative gene expression of SRD5A3 was also apparent in the ADT groups. CONCLUSION: AR and steroidogenic enzymes are upregulated in ADT-PCa patients as early as 3 months, without PSA elevation. Steroidogenic enzymes, particularly SRD5A3, were also upregulated before PSA rose.


Subject(s)
Androgen Antagonists/therapeutic use , Gonadotropin-Releasing Hormone/agonists , Orchiectomy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/therapy , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/analysis , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/biosynthesis , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Aged , Aged, 80 and over , Aldo-Keto Reductase Family 1 Member C3/analysis , Aldo-Keto Reductase Family 1 Member C3/biosynthesis , Aldo-Keto Reductase Family 1 Member C3/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Proteins/analysis , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Middle Aged , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/genetics , Receptors, Androgen/analysis , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Time Factors , Up-Regulation
20.
Mol Cancer Ther ; 19(8): 1708-1718, 2020 08.
Article in English | MEDLINE | ID: mdl-32430485

ABSTRACT

The next-generation antiandrogen drugs, XTANDI (enzalutamide), ZYTIGA (abiraterone acetate), ERLEADA (apalutamide) and NUBEQA (darolutamide) extend survival times and improve quality of life in patients with advanced prostate cancer. Despite these advances, resistance occurs frequently and there is currently no definitive cure for castration-resistant prostate cancer. Our previous studies identified that similar mechanisms of resistance to enzalutamide or abiraterone occur following treatment and cross-resistance exists between these therapies in advanced prostate cancer. Here, we show that enzalutamide- and abiraterone-resistant prostate cancer cells are further cross-resistant to apalutamide and darolutamide. Mechanistically, we have determined that the AKR1C3/AR-V7 axis confers this cross-resistance. Knockdown of AR-V7 in enzalutamide-resistant cells resensitize cells to apalutamide and darolutamide treatment. Furthermore, targeting AKR1C3 resensitizes resistant cells to apalutamide and darolutamide treatment through AR-V7 inhibition. Chronic apalutamide treatment in C4-2B cells activates the steroid hormone biosynthesis pathway and increases AKR1C3 expression, which confers resistance to enzalutamide, abiraterone, and darolutamide. In conclusion, our results suggest that apalutamide and darolutamide share similar resistant mechanisms with enzalutamide and abiraterone. The AKR1C3/AR-V7 complex confers cross-resistance to second-generation androgen receptor-targeted therapies in advanced prostate cancer.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/metabolism , Alternative Splicing , Androgen Receptor Antagonists/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/chemistry , Aldo-Keto Reductase Family 1 Member C3/genetics , Androgen Receptor Antagonists/classification , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Humans , Male , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL