Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Biol Interact ; 351: 109746, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34780792

ABSTRACT

Human aldo-keto reductase family 1C1 (AKR1C1) is an important enzyme involved in human hormone metabolism, which is mainly responsible for the metabolism of progesterone in the human body. AKR1C1 is highly expressed and has an important relationship with the occurrence and development of various diseases, especially some cancers related to hormone metabolism. Nowadays, many inhibitors against AKR1C1 have been discovered, including some synthetic compounds and natural products, which have certain inhibitory activity against AKR1C1 at the target level. Here we briefly reviewed the physiological and pathological functions of AKR1C1 and the relationship with the disease, and then summarized the development of AKR1C1 inhibitors, elucidated the interaction between inhibitors and AKR1C1 through molecular docking results and existing co-crystal structures. Finally, we discussed the design ideals of selective AKR1C1 inhibitors from the perspective of AKR1C1 structure, discussed the prospects of AKR1C1 in the treatment of human diseases in terms of biomarkers, pre-receptor regulation and single nucleotide polymorphisms, aiming to provide new ideas for drug research targeting AKR1C1.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/physiology , Enzyme Inhibitors/pharmacology , 20-Hydroxysteroid Dehydrogenases/chemistry , 20-Hydroxysteroid Dehydrogenases/metabolism , Animals , Catalytic Domain , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Molecular Docking Simulation , Protein Binding
2.
J Steroid Biochem Mol Biol ; 198: 105527, 2020 04.
Article in English | MEDLINE | ID: mdl-31733346

ABSTRACT

The clinical use of the steroidal aromatase inhibitor Formestane (4-hydroxandrostenedione, 4-OHA) in the treatment of advanced ER+ breast cancer has been discontinued, and therefore, interest in this remarkable drug has vanished. As a C-19 sterol, 4-OHA can undergo extensive intracellular metabolism depending on the expression of specific enzymes in the corresponding cells. We used the metabolites 4ß-hydroxyandrosterone, 4ß-hydroxyepiandrosterone and its 17ß-reduced derivative as standards for the proof of catalytic activity present in the cell culture medium and expressed by the isolated enzymes. All of the aldo-keto reductases AKR1C1, AKR1C2, AKR1C3 and AKR1C4 catalysed the reduction of the 3-keto-group and the Δ4,5 double bond of 4-OHA at the same time. Molecular docking experiments using microscale thermophoresis and the examination of the kinetic behaviour of the isolated enzymes with the substrate 4-OHA proved that AKR1C3 had the highest affinity for the substrate, whereas AKR1C1 was the most efficient enzyme. Both enzymes (AKR1C1and AKR1C3) are highly expressed in adipose tissue and lungs, exhibiting 3ß-HSD activity. The possibility that 4-OHA generates biologically active derivatives such as the androgen 4-hydroxytestosterone or some 17ß-hydroxy derivatives of the 5α-reduced metabolites may reawaken interest in Formestane, provided that a suitable method of administration can be developed, avoiding oral or intramuscular depot-injection administration.


Subject(s)
3-alpha-Hydroxysteroid Dehydrogenase (B-Specific)/physiology , Androstenedione/analogs & derivatives , Steroids/pharmacokinetics , 20-Hydroxysteroid Dehydrogenases/physiology , Administration, Oral , Aldo-Keto Reductase Family 1 Member C3/physiology , Androstenedione/chemical synthesis , Androstenedione/pharmacokinetics , Animals , COS Cells , Chlorocebus aethiops , Humans , Hydroxysteroid Dehydrogenases/physiology , Kinetics , Molecular Docking Simulation , Oxidoreductases/physiology , Protein Binding , Protein Isoforms , Recombinant Proteins/chemistry , Solvents , Steroids/chemical synthesis
3.
Br J Cancer ; 118(7): 985-994, 2018 04.
Article in English | MEDLINE | ID: mdl-29515258

ABSTRACT

BACKGROUND: Despite chemotherapy intensification, a subgroup of high-risk paediatric T-cell acute lymphoblastic leukemia (T-ALL) patients still experience treatment failure. In this context, we hypothesised that therapy resistance in T-ALL might involve aldo-keto reductase 1C (AKR1C) enzymes as previously reported for solid tumors. METHODS: Expression of NRF2-AKR1C signaling components has been analysed in paediatric T-ALL samples endowed with different treatment outcomes as well as in patient-derived xenografts of T-ALL. The effects of AKR1C enzyme modulation has been investigated in T-ALL cell lines and primary cultures by combining AKR1C inhibition, overexpression, and gene silencing approaches. RESULTS: We show that T-ALL cells overexpress AKR1C1-3 enzymes in therapy-resistant patients. We report that AKR1C1-3 enzymes play a role in the response to vincristine (VCR) treatment, also ex vivo in patient-derived xenografts. Moreover, we demonstrate that the modulation of AKR1C1-3 levels is sufficient to sensitise T-ALL cells to VCR. Finally, we show that T-ALL chemotherapeutics induce overactivation of AKR1C enzymes independent of therapy resistance, thus establishing a potential resistance loop during T-ALL combination treatment. CONCLUSIONS: Here, we demonstrate that expression and activity of AKR1C enzymes correlate with response to chemotherapeutics in T-ALL, posing AKR1C1-3 as potential targets for combination treatments during T-ALL therapy.


Subject(s)
Aldo-Keto Reductases/physiology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , 20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 20-Hydroxysteroid Dehydrogenases/physiology , Age of Onset , Aldo-Keto Reductase Family 1 Member C3/antagonists & inhibitors , Aldo-Keto Reductase Family 1 Member C3/physiology , Aldo-Keto Reductases/antagonists & inhibitors , Animals , Child , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Hydroxysteroid Dehydrogenases/physiology , Isoenzymes/physiology , Medroxyprogesterone Acetate/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Vincristine/administration & dosage , Xenograft Model Antitumor Assays
4.
Curr Med Chem ; 18(17): 2554-65, 2011.
Article in English | MEDLINE | ID: mdl-21568892

ABSTRACT

The AKR1C aldo-keto reductases (AKR1C1-AKR1C4) are enzymes that interconvert steroidal hormones between their active and inactive forms. In this manner, they can regulate the occupancy and trans-activation of the androgen, estrogen and progesterone receptors. The AKR1C isoforms also have important roles in the production and inactivation of neurosteroids and prostaglandins, and in the metabolism of xenobiotics. They thus represent important emerging drug targets for the development of agents for the treatment of hormone-dependent forms of cancer, like breast, prostate and endometrial cancers, and other diseases, like premenstrual syndrome, endometriosis, catamenial epilepsy and depressive disorders. We present here the physiological roles of these enzymes, along with their structural properties and an overview of the recent developments regarding their inhibitors. The most important strategies of inhibitor design are described, which include the screening of banks of natural compounds (like cinnamic acids, flavonoids, jasmonates, and related compounds), the screening of and structural modifications to non-steroidal anti-inflammatory drugs, the substrate-inspired design of steroidal and nonsteroidal inhibitors, and computer-assisted structure-based inhibitor design.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , 20-Hydroxysteroid Dehydrogenases/physiology , Amino Acid Sequence , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Catalytic Domain , Cinnamates/pharmacology , Drug Design , Drug Discovery , Flavonoids/pharmacology , Gonadal Steroid Hormones/metabolism , Humans , Models, Molecular , Neurotransmitter Agents/metabolism , Protein Structure, Secondary , Salicylates/pharmacology , Sequence Alignment
5.
Cancer Res ; 69(11): 4769-75, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487289

ABSTRACT

Members of the aldo-keto reductase (AKR) superfamily, particularly the AKR1C subfamily, are emerging as important mediators of the pathology of cancer. Agents that inhibit these enzymes may provide novel agents for either the chemoprevention or treatment of diverse malignancies. Recently, jasmonates, a family of plant stress hormones that bear a structural resemblance to prostaglandins, have been shown to elicit anticancer activities both in vitro and in vivo. In this study, we show that jasmonic acid (JA) and methyl jasmonate (MeJ) are capable of inhibiting all four human AKR1C isoforms. Although JA is the more potent inhibitor of recombinant AKR1C proteins, including the in vitro prostaglandin F synthase activity of AKR1C3, MeJ displayed greater potency in cellular systems that was, at least in part, due to increased cellular uptake of MeJ. Moreover, using the acute myelogenous leukemia cell lines HL-60 and KG1a, we found that although both jasmonates were able to induce high levels of reactive oxygen species in a dose-dependent fashion, only MeJ was able to induce high levels of mitochondrial superoxide (MSO), possibly as an epiphenomenon of mitochondrial damage. There was a strong correlation observed between MSO formation at 24 hours and reduced cellularity at day 5. In conclusion, we have identified AKR1C isoforms as a novel target of jasmonates in cancer cells and provide further evidence of the promise of these compounds, or derivatives thereof, as adjunctive therapies in the treatment of cancer.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Acetates/pharmacology , Cyclopentanes/pharmacology , Drug Delivery Systems , Mitochondria/drug effects , Oxylipins/pharmacology , 20-Hydroxysteroid Dehydrogenases/metabolism , 20-Hydroxysteroid Dehydrogenases/physiology , Acetates/pharmacokinetics , Cell Survival/drug effects , Cells, Cultured , Cyclopentanes/pharmacokinetics , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , HL-60 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Hydroxyprostaglandin Dehydrogenases/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Isoenzymes/physiology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mitochondria/physiology , Models, Biological , Oxylipins/pharmacokinetics , Prostaglandin D2/metabolism , Reactive Oxygen Species/metabolism
6.
Gen Comp Endocrinol ; 163(3): 259-69, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19401204

ABSTRACT

20beta-hydroxysteroid dehydrogenase (20beta-HSD) synthesizes 17alpha,20beta-dihdroxy-4-pregnen-3-one, the steroid required for resumption of prophase-I arrested oocytes in teleosts. Though 20beta-HSD cDNAs have been cloned from few fish species, its role in final oocyte maturation (FOM) is still questionable. To study the role of 20beta-HSD in FOM more explicitly, we cloned and characterized 20beta-HSD from ovary of air-breathing catfish, Clarias gariepinus. Interestingly, Escherichia coli expressed recombinant proteins, both full-length and an N-terminal truncated proteins catalyzed the reduction of steroids and xenobiotics, however there was significant difference between them. Semi-quantitative RT-PCR and Western blots demonstrated the presence of 20beta-HSD transcript and protein in various tissues with relatively higher level in gonads, gill, kidney and brain. A positive correlation of 20beta-HSD expression was observed in different phases of ovarian cycles. Immunocytochemical/immunofluoroscence analysis with specific antibody identified presence of 20beta-HSD in follicular layer of ovary. Real-time RT-PCR and Western blotting showed an induction of 20beta-HSD expression during human chorionic gonadotropin (hCG)-induced oocyte maturation, in vitro and in vivo. Concomitantly, a rise in 20beta-HSD enzyme activity was also noticed. Specific inhibitors of carbonyl reductase inhibited not only recombinant protein catalytic activity but also hCG-induced oocyte maturation in a dose-dependent manner as evidenced by blocking of germinal vesicle break down. These results together provide new evidences for the involvement of 20beta-HSD in the FOM/meiotic maturation.


Subject(s)
20-Hydroxysteroid Dehydrogenases/physiology , Catfishes/physiology , Oocytes/enzymology , Oocytes/growth & development , 20-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Southern , Blotting, Western , Catfishes/growth & development , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Molecular Sequence Data , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
7.
Biochem Pharmacol ; 75(2): 414-26, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17945194

ABSTRACT

While studying differentially expressed genes between sensitive and 10(-5)M Methotrexate (MTX) resistant HT29 human colon cancer cells, we identified some members of the aldo-keto reductase (AKR) superfamily. The study was followed with the member AKR1C1 (EC 1.1.1.213), validating its increase in mRNA and protein levels in MTX resistant cells. The genomic content for AKR1C1 remained unchanged between sensitive and resistant cells, thereby excluding a mechanism of AKR1C1 gene amplification. Thus, we cloned the AKR1C1 human promoter and performed luciferase experiments that revealed a transcriptional regulation of the gene in the resistant cells. Computational studies showed a putative binding site for the transcription factor Sp1. The co-transfection of Sp1 or Sp3 with different constructs of AKR1C1 promoter deletions, including and excluding the proximal GC-box, demonstrated a key role for these factors in regulating AKR1C1 transcriptional activity. Gel-shift assays revealed an increase in Sp1 and Sp3 binding in resistant compared to sensitive cells, without differences in Sp1 protein levels. Dephosphorylation of the extracts coincided with a decrease in Sp1 binding, which is consistent with a process of regulation of Sp1 by phosphorylation. We also investigated the possible relationship between AKR1C1 expression and MTX action. Overexpression of AKR1C1 counteracted the S-phase accumulation of cells and apoptosis caused by MTX treatment. This suggests a role of AKR1C1 in cell proliferation. Finally, overexpression of AKR1C1 in MTX sensitive HT29 cells conferred resistance to the chemotherapeutic agent and silencing of AKR1C1 by means of iRNA technology sensitized the cells to MTX.


Subject(s)
20-Hydroxysteroid Dehydrogenases/genetics , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Gene Expression Regulation, Neoplastic , Methotrexate/pharmacology , 20-Hydroxysteroid Dehydrogenases/physiology , Base Sequence , Drug Resistance, Neoplasm , HT29 Cells , Humans , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Sp1 Transcription Factor/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic
8.
Int J Cancer ; 120(9): 2019-27, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17266043

ABSTRACT

Dihydrodiol dehydrogenase (DDH) is a member of the aldo-keto reductases superfamily (AKR1C1-AKR1C4), which plays central roles in the metabolism of steroid hormone, prostaglandin and xenobiotics. We have previously detected overexpression of DDH as an indicator of poor prognosis and chemoresistance in human non-small lung cancer (NSCLC). We also found DDH expression to be closely related to chronic inflammatory conditions. The aim of this study was to investigate the links between inflammation, DDH expression and drug resistance in NSCLC cells. We showed that pro-inflammatory mediators including interleukin-6 (IL-6) could induce AKR1C1/1C2 expression in NSCLC cells and increase cellular resistance to cisplatin and adriamycin. This effect was nullified by Safingol, a protein kinase C inhibitor. Moreover, the expression of AKR1C1/1C2 was inversely correlated to NBS1 and apoptosis-inducing factor (AIF). We also showed that IL-6-induced AKR1C1/1C2 expression and drug resistance were inhibited by wogonin and chrysin, which are major flavonoids in Scutellaria baicalensis, a widely used traditional Chinese and Japanese medicine. In conclusion, this study demonstrated novel links of pro-inflammatory signals, AKR1C1/1C2 expression and drug resistance in NSCLC. The protein kinase C pathway may play an important role in this process. Overexpression of AKR1C1/1C2 may serve as a marker of chemoresistance. Further studies are warranted to evaluate wogonin and chrysin as a potential adjuvant therapy for drug-resistant NSCLC, especially for those with AKR1C1/1C2 overexpression.


Subject(s)
20-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Flavanones/pharmacology , Flavonoids/pharmacology , Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Lung Neoplasms/drug therapy , 20-Hydroxysteroid Dehydrogenases/physiology , Apoptosis , Cell Cycle , Cell Line, Tumor , DNA Repair , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Humans , Hydroxysteroid Dehydrogenases/physiology , Interleukin-6/pharmacology
9.
J Pharmacol Exp Ther ; 316(3): 1300-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16339391

ABSTRACT

Tibolone [[7alpha,17alpha]-17-hydroxy-7-methyl-19-norpregn-5(10)-en-20-yn-3-one] is used to treat climacteric symptoms and prevent osteoporosis. It exerts tissue-selective effects via site-specific metabolism into 3alpha- and 3beta-hydroxymetabolites and a Delta4-isomer. Recombinant human cytosolic aldo-keto reductases 1C1 and 1C2 (AKR1C1 and AKR1C2) produce 3beta-hydroxytibolone, and the liver-specific AKR1C4 produces predominantly 3alpha-hydroxytibolone. These observations may account for the appearance of 3beta-hydroxytibolone in target tissues and 3alpha-hydroxytibolone in the circulation. Using liver autopsy samples (which express AKR1C1-AKR1C4), tibolone was reduced via 3alpha- and 3beta-hydroxysteroid dehydrogenase (HSD) activity. 3beta-Hydroxytibolone was exclusively formed in the cytosol and was inhibited by the AKR1C2-specific inhibitor 5beta-cholanic acid-3alpha, 7alpha-diol. The cytosolic formation of 3alpha-hydroxytibolone was inhibited by an AKR1C4-selective inhibitor, phenolphthalein. The ratio of these stereoisomers was 4:1 in favor of 3beta-hydroxytibolone. In HepG2 cell cytosol and intact cells (which do not express AKR1C4), tibolone was exclusively reduced to 3beta-hydroxytibolone and was blocked by the AKR1C1-AKR1C3 inhibitor flufenamic acid. In primary hepatocytes (which express AKR1C1-AKR1C4), time-dependent reduction of tibolone into 3beta- and 3alpha-hydroxytibolone was observed again in a 4:1 ratio. 3beta-HSD activity was inhibited by both 5beta-cholanic acid-3alpha,7alpha-diol and flufenamic acid, implicating a role for AKR1C2 and AKR1C1. By contrast, the formation of 3alpha-hydroxytibolone was exclusively inhibited by phenolphthalein implicating AKR1C4 in this reaction. 3beta- and 3alpha-Hydroxytibolone were rapidly metabolized into polar metabolites (>85%). The formation of minor amounts of tibolone was also observed followed by AKR1C-catalyzed epimerization. The low hepatic formation of 3alpha-hydroxytibolone suggests that AKR1C4 is not the primary source of this metabolite and instead it maybe formed by an intestinal or enterobacterial 3alpha-HSD.


Subject(s)
17-Hydroxysteroid Dehydrogenases/physiology , 20-Hydroxysteroid Dehydrogenases/physiology , 3-Hydroxysteroid Dehydrogenases/physiology , Hydroxyprostaglandin Dehydrogenases/physiology , Hydroxysteroid Dehydrogenases/physiology , Liver/metabolism , Norpregnenes/metabolism , Oxidoreductases/physiology , Aldo-Keto Reductase Family 1 Member C3 , Bile Acids and Salts/pharmacology , Catalysis , Cells, Cultured , Flufenamic Acid/pharmacology , Hepatocytes/enzymology , Humans , Phenolphthalein/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...