Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 898: 173972, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33652058

ABSTRACT

Dehydrogenase/reductase member 2 (DHRS2) belongs to the short-chain dehydrogenase/reductase (SDR) family. It was initially isolated from the nuclear extract of hepatocellular carcinoma HepG2 cells and was identified as a specific cell cycle regulator. DHRS2 is a reduced nicotinamide adenine dinucleotide phosphate (NADPH)-dependent carbonyl reductase and catalyzes the reduction of dicarbonyl compounds. It is also functionally active in lipid metabolism and acts as a metabolic enzyme of hormones. Recent studies have shown that DHRS2 reprograms lipid metabolism and redox homeostasis to regulate proliferation, migration, invasion, and drug resistance of cancer cells. Here, we describe the structure, organelle localization and function of DHRS2, and also highlight its roles in the pathologic progression of diseases.


Subject(s)
Carbonyl Reductase (NADPH)/metabolism , Lipid Metabolism , Neoplasms/enzymology , Animals , Antineoplastic Agents/pharmacology , Carbonyl Reductase (NADPH)/antagonists & inhibitors , Carbonyl Reductase (NADPH)/chemistry , Enzyme Inhibitors/pharmacology , Humans , Lipid Metabolism/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Protein Conformation , Structure-Activity Relationship
2.
Oncol Rep ; 42(5): 1725-1734, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31436301

ABSTRACT

Oxaliplatin (Oxa)­based chemotherapy is widely used as the first­line treatment for colorectal cancer (CRC). However, Oxa­resistance is common for many postoperative CRC patients. To explore drug resistance in CRC, an Oxa­resistant cell line, HCT116/Oxa, was established from parental HCT116 cells. These Oxa­resistant cells exhibited characteristics of epithelial­mesenchymal transition (EMT) and a higher migratory capacity than parental cells. Protein profiles of HCT116/Oxa and HCT116 cells were compared using a tandem mass tag­based quantitative proteomics technique. The protein dehydrogenase/reductase SDR family member 2 (DHRS2) was revealed to be highly expressed in HCT116/Oxa cells. Silencing of DHRS2 in HCT116/Oxa cells effectively restored Oxa­sensitivity by suppressing the expression of excision repair cross­complementing group 1 protein via a p53­dependent pathway, and reversed the EMT phenotype. Overall, the suppression of DHRS2 expression may be a promising strategy for the prevention of Oxa­resistance in CRC.


Subject(s)
Carbonyl Reductase (NADPH)/antagonists & inhibitors , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Endonucleases/metabolism , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Cadherins/metabolism , Carbonyl Reductase (NADPH)/genetics , Cell Movement/drug effects , Chromatography, Liquid , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Down-Regulation , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , HCT116 Cells , Humans , Oxaliplatin/pharmacology , Proteomics , Signal Transduction/drug effects , Tandem Mass Spectrometry
3.
Biomaterials ; 210: 62-69, 2019 07.
Article in English | MEDLINE | ID: mdl-31075724

ABSTRACT

Nanomedicine has become a promising approach to improve cancer chemotherapy. It remains a major challenge how to enhance anti-drug efficacy and reduce side effects of anti-cancer drugs. Herein, we report a self-assembled nanoplatform (FDEP NPs) by integration of doxorubicin (DOX) and epigallocatechin-3-O-gallate (EGCG) with the help of coordination between Fe3+ ions and polyphenols. The EGCG from FDEP NPs could inhibit the expression of carbonyl reductase 1 (CBR1) protein and thereby inhibit the doxorubicinol (DOXOL) generation from DOX both in vitro and in vivo, thus the efficacy of DOX to cancerous cells is improved significantly. More importantly, the FDEP NPs could reduce cardiac toxicity and the DOX mediated toxicity to blood cells due to the repression of DOXOL production. Moreover, the blood half-life of FDEP NPs is longer than 23 h as determined by positron emission tomography (PET) imaging of biodistribution of radiolabelled NPs and HPLC measurement of plasma level of DOX, ensuring high tumor accumulation of FDEP NPs by enhanced permeability and retention (EPR) effect. The FDEP NPs also exhibited much improved antitumor effect over free drugs. Our work sheds new light on the engineering of nanomaterials for combination chemotherapy and may find unique clinical applications in the near future.


Subject(s)
Carbonyl Reductase (NADPH)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Nanostructures/chemistry , Polyphenols/pharmacology , Tea/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carbonyl Reductase (NADPH)/metabolism , Cell Survival/drug effects , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Drug Liberation , Endocytosis/drug effects , Enzyme Inhibitors/pharmacokinetics , HEK293 Cells , Humans , Mice, Inbred BALB C , Optical Imaging , Positron-Emission Tomography , Tissue Distribution/drug effects
4.
Chem Biol Interact ; 300: 40-48, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30611789

ABSTRACT

One of the causes of therapeutic failure of chemotherapy is cancer cell resistance. In the case of anthracyclines, many resistance mechanisms have been described. One of them assumes the role of carbonyl reductase 1 (CBR1), a cytosolic enzyme that is responsible for the biotransformation process of anthracyclines to less active, undesirable metabolites. Therefore, CBR1 inhibitors are considered for use as a chemosensitizing agents. In the present study, piperlongumine (PL), a Piper longum L. alkaloid that has previously been described as a CBR1 inhibitor, was investigated for its chemosensitizing properties in co-treatment with doxorubicin (DOX). The biotransformation process of DOX in the presence of PL was tracked using human cytosol fraction and LC-MS, then a molecular modeling study was conducted to predict the interaction of PL with the active site of the CBR1. The biological interaction between DOX and PL was investigated using DU-145 prostate cancer cells. Cytotoxic and antiproliferative properties of DOX and PL were examined, and the type and potency of interaction was quantified by Combination Index. The mechanism of the cell death induced by the agents was investigated by flow cytometry and the anti-invasive properties of the drugs were determined by monitoring the movement of individual cells. PL showed dose-dependent inhibition of DOX metabolism in cytosol, which resulted in less doxorubicinol (DOXol) metabolite being formed. The possible mechanism of CBR1 inhibition was explained through molecular modeling studies by prediction of PL's binding mode in the active site of the enzyme's crystal structure-based model. DOX and PL showed a synergistic antiproliferative and proapoptotic effect on cancer cells. Significant anti-invasive properties of the combination of DOX and PL were found, but when the drugs were used separately they did not alter the cancer cells' motility. Cell motility inhibition was accompanied by significant changes in cytoskeleton architecture. DOX and PL used in co-treatment showed significant synergistic anticancer properties. Inhibition of DOX metabolism by PL was found to be a mechanism that was likely to be responsible for the observed interaction.


Subject(s)
Carbonyl Reductase (NADPH)/metabolism , Cell Proliferation/drug effects , Dioxolanes/pharmacology , Doxorubicin/pharmacology , Apoptosis/drug effects , Binding Sites , Biotransformation , Carbonyl Reductase (NADPH)/antagonists & inhibitors , Catalytic Domain , Cell Line, Tumor , Cell Movement/drug effects , Doxorubicin/metabolism , Drug Synergism , Humans , Hydrogen Bonding , Male , Molecular Docking Simulation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
5.
Invest New Drugs ; 35(3): 375-385, 2017 06.
Article in English | MEDLINE | ID: mdl-28283780

ABSTRACT

Anthracycline antibiotics (ANT), such as doxorubicin or daunorubicin, are a class of anticancer drugs that are widely used in oncology. Although highly effective in cancer therapy, their usefulness is greatly limited by their cardiotoxicity. Possible mechanisms of ANT cardiotoxicity include their conversion to secondary alcohol metabolites (i.e. doxorubicinol, daunorubicinol) catalyzed by carbonyl reductases (CBR) and aldo-keto reductases (AKR). These metabolites are suspected to be more cardiotoxic than their parent compounds. Moreover, overexpression of ANT-reducing enzymes (CBR and AKR) are found in many ANT-resistant cancers. The secondary metabolites show decreased cytotoxic properties and are more susceptible to ABC-mediated efflux than their parent compounds; thus, metabolite formation is considered one of the mechanisms of cancer resistance. Inhibitors of CBR and AKR were found to reduce the cardiotoxicity of ANT and the resistance of cancer cells, and therefore are being investigated as prospective cardioprotective and chemosensitizing drug candidates. In this review, the significance of a two-electron reduction of ANT, including daunorubicin, epirubicin, idarubicin, valrubicin, amrubicin, aclarubicin, and especially doxorubicin, is described with respect to toxicity and efficacy of therapy. Additionally, CBR and AKR inhibitors, including monoHER, curcumin, (-)-epigallocatechin gallate, resveratrol, berberine or pixantrone, and their modulating effect on the activity of ANT is characterized and discussed as potential mechanism of action for novel therapeutics in cancer treatment.


Subject(s)
Aldo-Keto Reductases/antagonists & inhibitors , Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Carbonyl Reductase (NADPH)/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Cardiotoxicity/prevention & control , Animals , Anthracyclines/pharmacokinetics , Antibiotics, Antineoplastic/pharmacokinetics , Cardiotoxicity/etiology , Drug Resistance, Neoplasm , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...