Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Redox Biol ; 24: 101210, 2019 06.
Article in English | MEDLINE | ID: mdl-31078780

ABSTRACT

Hypoxia, a common element in the tumor environment, leads to Hypoxia-Inducible Factor-1α (HIF-1α) stabilization to modulate cellular metabolism as an adaptive response. In a previous study, we showed that inhibition of the nuclear factor erythroid 2-like-2 (NFE2L2; NRF2), a master regulator of many genes coping with electrophilic and oxidative stress, elevated the level of miR-181c and induced mitochondrial dysfunction in colon cancer cells. In this study, we demonstrate that NRF2-silencing hindered HIF-1α accumulation in hypoxic breast cancer cells and subsequently suppressed hypoxia-inducible expression of glycolysis-associated glucose transporter-1, hexokinase-2, pyruvate dehydrogenase kinase-1, and lactate dehydrogenase A. HIF-1α dysregulation in NRF2-silenced cancer cells was associated with miR-181c elevation. Overexpression of miR-181c in breast cancer cells blocked HIF-1α accumulation and diminished hypoxia-inducible levels of glycolysis enzymes, whereas the inhibition of miR-181c in NRF2-silenced cells restored HIF-1α accumulation. In a subsequent metabolomic analysis, hypoxic incubation increased the levels of metabolites involved in glycolysis and activated the pentose phosphate pathway (PPP) in control cells. However, these elevations were less pronounced in NRF2-silenced cells. In particular, hypoxic incubation increased the levels of amino acids, which implies a shift to catabolic metabolism, and the increased levels were higher in control cells than in NRF2-silenced cells. Concurrently, hypoxia activated BCL2 interacting protein 3 (BNIP3)-mediated autophagy in the control cells and miR-181c was found to be involved in this autophagy activation. Taken together, these results show that hypoxia-induced metabolic changes to glycolysis, the PPP, and autophagy are inhibited by NRF2-silencing through miR-181c-mediated HIF-1α dysregulation. Therefore, targeting NRF2/miR-181c could be an effective strategy to counteract HIF-1α-orchestrated metabolic adaptation of hypoxic cancer cells.


Subject(s)
Adaptation, Biological , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Energy Metabolism , Gene Silencing , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Metabolome , Metabolomics/methods , Mitochondria/genetics , Mitochondria/metabolism , Models, Biological , RNA, Small Interfering/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
2.
Antioxid Redox Signal ; 27(13): 945-961, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28383996

ABSTRACT

AIMS: The nuclear factor (erythroid-derived 2)-like 2 (NFE2L2; NFE2L2/NRF2) pathway contributes to the environmental resistance of cancers by enhancing the antioxidant capacity. Here, we explored the potential connection between NFE2L2/NRF2 and mitochondrial function in cancers. RESULTS: Global miRNA expression analysis of HT29 and HCT116 human colon cancer cells identified that NFE2L2/NRF2 silencing upregulated miR-181c through nuclear factor-κB signaling, and this increase was associated with the reduction in mitochondria-encoded cytochrome c oxidase subunit-1 (MT-CO1), a catalytic core subunit of the complex IV of the electron transport chain (ETC). As a result of ETC dysfunction, NFE2L2/NRF2-silenced cancer cells exhibited the decreases in the mitochondrial membrane potential, oxygen consumption rate, and cellular adenosine triphosphate (ATP) contents. Notably, these changes induced adenosine monophosphate (AMP)-activated protein kinase-α (AMPKα) activation and subsequent metabolic adaptation signaling, including the inhibition of fatty acid and sterol biosynthesis enzymes. As supportive evidence of AMPKα-driven adaption, NFE2L2/NRF2-silenced cells were more vulnerable to AMPKα inhibition-induced growth suppression. Similarly, mouse tumor xenografts derived from NFE2L2/NRF2-silenced HT29 exhibited MT-CO1 reduction and AMPKα activation, thereby increasing responsiveness to the AMPK inhibitor treatment. The association of NFE2L2/NRF2 with MT-CO1 and AMPKα was confirmed in breast cancer cells. INNOVATION: We demonstrated the significance of NFE2L2/NRF2 in cancer mitochondria by elucidating the involvement of miR-181c/MT-CO1 as underlying molecular events. We also provide evidence of the crosstalk between NFE2L2/NRF2 and AMPKα as an adaptive link in cancers. CONCLUSION: Therefore, it may be an effective strategy to inhibit both NFE2L2/NRF2 and AMPKα signaling to overcome adaptive behaviors of cancer. Antioxid. Redox Signal. 27, 945-961.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Colonic Neoplasms/genetics , Electron Transport Complex IV/genetics , MicroRNAs/genetics , NF-E2-Related Factor 2/metabolism , Animals , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice , Mitochondria/metabolism , Neoplasm Transplantation , Signal Transduction
3.
Arch Pharm Res ; 40(3): 391-402, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28084586

ABSTRACT

Multiple comorbidities of metabolic disorders are associated with facilitated chronic kidney disease progression. Anti-platelet cilostazol is used for the treatment of peripheral artery disease. In this study, we investigated the potential beneficial effects of cilostazol and rosuvastatin on metabolic disorder-induced renal dysfunctions. C57BL/6 mice that received high fat diet (HFD) for 22 weeks and a low dose of streptozotocin (STZ, 40 mg/kg) developed albuminuria and had increased urinary cystatin C excretion, and cilostazol treatment (13 weeks) improved these markers. Histopathological changes, including glomerular mesangial expansion, tubular vacuolization, apoptosis, and lipid accumulation were ameliorated by cilostazol treatment. Tubulointerstitial fibrosis that was indicated by the increases in collagen and transforming growth factor-ß1 subsided by cilostazol. Renoprotective effects were also observed in rosuvastatin-treated mice, and combinatorial treatment with cilostazol and rosuvastatin demonstrated enhanced ameliorative effects in histopathological evaluations. Notably, repressed renal heme oxygenase-1 (Ho-1) level in HFD/STZ mice was restored in cilostazol group. Further, we demonstrated that cilostazol enhanced Nrf2/Ho-1 signaling in cultured proximal tubular epithelial cells. Collectively, these results suggest the potential advantageous use of cilostazol as an adjunctive therapy with statins for the amelioration of metabolic disorder-associated renal injury.


Subject(s)
Diet, High-Fat/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/etiology , Rosuvastatin Calcium/therapeutic use , Tetrazoles/therapeutic use , Albuminuria/drug therapy , Albuminuria/etiology , Animals , Anti-Bacterial Agents/toxicity , Cilostazol , Cystatin C/urine , Heme Oxygenase-1/metabolism , Kidney/pathology , Kidney Function Tests , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/urine , Streptozocin/toxicity
4.
Mol Pharmacol ; 87(3): 465-76, 2015.
Article in English | MEDLINE | ID: mdl-25534417

ABSTRACT

Overexpression of BCRP/ABCG2, a xenobiotic efflux transporter, is associated with anticancer drug resistance in tumors. Proto-oncogene c-MET induces cancer cell proliferation, motility, and survival, and its aberrant activation was found to be a prognostic factor in advanced ovarian cancers. In the present study, we investigated the potential crossresistance of doxorubicin-resistant ovarian cancer cells to the pheophorbide a (Pba)-based photodynamic therapy (PDT), and suggest c-MET and BCRP/ABCG2 overexpression as an underlying molecular mechanism. The doxorubicin-resistant A2780 cell line (A2780DR), which was established by incubating A2780 with stepwise increasing concentrations of doxorubicin, showed low levels of cellular Pba accumulation and reactive oxygen species generation, and was more resistant to PDT cytotoxicity than A2780. In a microarray analysis, BCRP/ABCG2 was found to be the only drug transporter whose expression was upregulated in A2780DR; this increase was confirmed by Western blot and immunocytochemical analyses. As functional evidence, the treatment with a BCRP/ABCG2-specific inhibitor reversed A2780DR resistance to both doxorubicin and PDT. We identified that c-MET increase is related to BCRP/ABCG2 activation. The c-MET downstream phosphoinositide 3-kinase (PI3K)/AKT signaling was activated in A2780DR and the inhibition of PI3K/AKT or c-MET repressed resistance to doxorubicin and PDT. Finally, we showed that the pharmacological and genetic inhibition of c-MET diminished levels of BCRP/ABCG2 in A2780DR. Moreover, c-MET inhibition could repress BCRP/ABCG2 expression in breast carcinoma MDA-MB-231 and colon carcinoma HT29, resulting in sensitization to doxorubicin. Collectively, our results provide a novel link of c-MET overexpression to BCRP/ABCG2 activation, suggesting that this mechanism leads to crossresistance to both chemotherapy and PDT.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Doxorubicin/pharmacology , Neoplasm Proteins/biosynthesis , Phosphatidylinositol 3-Kinase/metabolism , Photochemotherapy , Proto-Oncogene Proteins c-met/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Drug Resistance, Neoplasm/radiation effects , Gene Expression Regulation, Neoplastic , Humans , Photochemotherapy/methods , Proto-Oncogene Mas , Signal Transduction/drug effects , Signal Transduction/physiology
5.
Oxid Med Cell Longev ; 2013: 423965, 2013.
Article in English | MEDLINE | ID: mdl-23766854

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (NRF2) is the transcription factor that regulates an array of antioxidant/detoxifying genes for cellular defense. The conformational changes of Kelch-like ECH-associated protein 1 (KEAP1), a cytosolic repressor protein of NRF2, by various stimuli result in NRF2 liberation and accumulation in the nucleus. In the present study, we aimed to investigate the effect of KEAP1 knockdown on NRF2 target gene expression and its toxicological implication using human colon cancer cells. The stable KEAP1-knockdown HT29 cells exhibit elevated levels of NRF2 and its target gene expressions. In particular, the mRNA levels of aldo-keto reductases (AKR1C1, 1C2, 1C3, 1B1, and 1B10) were substantially increased in KEAP1 silenced HT29 cells. These differential AKRs expressions appear to contribute to protection against oxidative stress. The KEAP1-knockdown cells were relatively more resistant to hydrogen peroxide (H2O2) and 4-hydroxynonenal (4HNE) compared to the control cells. Accordantly, we observed accumulation of 4HNE protein adducts in H2O2- or 4HNE-treated control cells, whereas KEAP1-knockdown cells did not increase adduct formation. The treatment of KEAP1-silenced cells with AKR1C inhibitor flufenamic acid increased 4HNE-induced cellular toxicity and protein adduct formation. Taken together, these results indicate that AKRs, which are NRF2-dependent highly inducible gene clusters, play a role in NRF2-mediated cytoprotection against lipid peroxide toxicity.


Subject(s)
Aldehydes/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Silencing , Intracellular Signaling Peptides and Proteins/metabolism , Aldehyde Reductase/metabolism , Aldo-Keto Reductases , Cell Death/drug effects , Cell Survival/drug effects , Cell Survival/genetics , DNA Adducts/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Silencing/drug effects , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Vitamin K 3/pharmacology
6.
Toxicol Lett ; 218(1): 39-49, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23305850

ABSTRACT

Transcription factor NF-E2-related factor 2 (NRF2) plays a crucial role in the cellular defense against oxidative/electrophilic stress by up-regulating multiple antioxidant genes. Numerous studies with genetically modified animals have demonstrated that Nrf2 is a sensitivity determining factor upon the exposure to environmental chemicals including carcinogens. Moreover, recent studies have demonstrated that polymorphism in the human NRF2 promoter is associated with higher risks for developing acute lung injury, gastric mucosal inflammation, and nephritis. Therefore, the identification of reliable and effective human target genes of NRF2 may allow the monitoring of NRF2 activity and to predict individual sensitivity to environmental stress-induced damage. For this purpose, we investigated genes that are tightly controlled by NRF2 to establish markers for NRF2 activity in human cells. Firstly, in the normal human renal epithelial HK-2 cells, the measurement of the expression of 30 previously reported NRF2 target genes in response to NRF2 inducers (sulforaphane, tert-butylhydroquinone, cinnamic aldehyde, and hydrogen peroxide) showed that the aldo-keto reductase (AKR) 1C1 is highly inducible by all treatments. Accordantly, the basal and inducible expressions of AKRs were significantly attenuated in NRF2-silenced HK-2 cells. Whereas, cells with stable KEAP1 knockdown, which causes a modest NRF2 activation, demonstrated substantially increased levels of AKR1A1, 1B1, 1B10, 1C1, 1C2, and 1C3. Secondly, the linkage between NRF2 and the AKRs was confirmed in human monocytic leukemia cell line U937, which can be a model of peripherally available blood cells. The treatment of U937 cells with NRF2 inducers including sulforaphane effectively elevated the expression of AKR1B1, 1B10, 1C1, 1C2, and 1C3. Whereas, the levels of both the basal and sulforaphane-inducible expression of AKR1C1 were significantly reduced in NRF2-silenced stable U937 cells compared to the control cells. Similarly, the inducible expression of AKR1C1 was observed in another human monocytic leukemia cell line THP-1 as well as in human primary blood CD14(+) monocytes. In conclusion, together with the high inducibility and NRF2 dependency shown in renal epithelial cells as well as in peripherally available blood cells, current findings suggest that AKRs can be utilized as a marker of NRF2 activity in human cells.


Subject(s)
Aldehyde Reductase/genetics , Gene Expression Regulation/physiology , Kidney Tubules, Proximal/enzymology , NF-E2-Related Factor 2/genetics , 20-Hydroxysteroid Dehydrogenases/biosynthesis , 20-Hydroxysteroid Dehydrogenases/drug effects , Acrolein/analogs & derivatives , Acrolein/pharmacology , Aldehyde Reductase/metabolism , Biomarkers/metabolism , Enzyme Induction/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Gene Silencing , Genetic Markers , Humans , Hydrogen Peroxide/pharmacology , Hydroquinones/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Isothiocyanates , Kelch-Like ECH-Associated Protein 1 , Kidney Tubules, Proximal/drug effects , Leukemia, Monocytic, Acute/enzymology , Leukemia, Monocytic, Acute/genetics , Monocytes/enzymology , Monocytes/pathology , NF-E2-Related Factor 2/biosynthesis , Oxidative Stress/physiology , Sulfoxides , Thiocyanates/pharmacology , U937 Cells
7.
Toxicol Appl Pharmacol ; 264(3): 431-8, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22959925

ABSTRACT

The ubiquitin-proteasome system plays a central role in protein quality control through endoplasmic reticulum (ER)-associated degradation (ERAD) of unfolded and misfolded proteins. NF-E2-related factor 2 (Nrf2) is a transcription factor that controls the expression of an array of phase II detoxification and antioxidant genes. Nrf2 signaling has additionally been shown to upregulate the expression of the proteasome catalytic subunits in several cell types. Here, we investigated the role of Nrf2 in tunicamycin-induced ER stress using a murine insulinoma ß-cell line, ßTC-6. shRNA-mediated silencing of Nrf2 expression in ßTC-6 cells significantly increased tunicamycin-induced cytotoxicity, elevated the expression of the pro-apoptotic ER stress marker Chop10, and inhibited tunicamycin-inducible expression of the proteasomal catalytic subunits Psmb5 and Psmb6. The effects of 3H-1,2-dithiole-3-thione (D3T), a small molecule Nrf2 activator, on ER stress were also examined in ßTC-6 cells. D3T pretreatment reduced tunicamycin cytotoxicity and attenuated the tunicamycin-inducible Chop10 and protein kinase RNA-activated-like ER kinase (Perk). The protective effect of D3T was shown to be associated with increased ERAD. D3T increased the expression of Psmb5 and Psmb6 and elevated chymotrypsin-like peptidase activity; proteasome inhibitor treatment blocked D3T effects on tunicamycin cytotoxicity and ER stress marker changes. Similarly, silencing of Nrf2 abolished the protective effect of D3T against ER stress. These results indicate that the Nrf2 pathway contributes to the ER stress response in pancreatic ß-cells by enhancing proteasome-mediated ERAD.


Subject(s)
Endoplasmic Reticulum/metabolism , Insulin-Secreting Cells/metabolism , NF-E2-Related Factor 2/metabolism , Stress, Physiological/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Endoplasmic Reticulum/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Mice , NF-E2-Related Factor 2/genetics , Proteasome Endopeptidase Complex , RNA/genetics , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tunicamycin/toxicity
8.
Free Radic Biol Med ; 52(9): 1773-85, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22387177

ABSTRACT

NF-E2-related factor 2 (NRF2) is a transcription factor that regulates the expression of various antioxidant and detoxifying enzymes. Although the benefit of NRF2 in cancer prevention is well established, its role in cancer pathobiology was recently discovered. In this study, the role of NRF2 in tumor growth and docetaxel sensitivity was investigated in ErbB2-overexpressing ovarian carcinoma SKOV3 cells. Interfering RNA-mediated stable inhibition of NRF2 in SKOV3 cells repressed NRF2 signaling, resulting in cell growth arrest at G(0)/G(1) phase and tumor growth retardation in mouse xenografts. Microarray analysis revealed that ErbB2 expression is substantially reduced in NRF2-inhibited SKOV3 and this was further confirmed by RT-PCR and immunoblot analysis. Repression of ErbB2 led to a decrease in phospho-AKT and enhanced p27 protein, reinforcing the effect of NRF2 knockdown on SKOV3 growth. Furthermore, NRF2 inhibition-mediated ErbB2 repression increases the sensitivity of these cells to docetaxel cytotoxicity and apoptosis. The linkage between NRF2 and ErbB2 was confirmed in the ErbB2-positive breast cancer cell line BT-474: NRF2 knockdown suppressed ErbB2 expression and enhanced docetaxel sensitivity. Our results provide insight into the coordinated regulation of signaling molecules responding to environmental stress and suggest that NRF2 modulation might be a therapeutic strategy to limit tumor growth and enhance sensitivity to taxane-based chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division , NF-E2-Related Factor 2/antagonists & inhibitors , Ovarian Neoplasms/pathology , Receptor, ErbB-2/metabolism , Signal Transduction/physiology , Taxoids/pharmacology , Animals , Base Sequence , Cell Line, Tumor , DNA Primers , Docetaxel , Female , Glutathione/metabolism , Humans , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/physiology , Ovarian Neoplasms/metabolism , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
9.
Molecules ; 15(10): 7266-91, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20966874

ABSTRACT

Oxidative stress causes damage to multiple cellular components such as DNA, proteins, and lipids, and is implicated in various human diseases including cancer, neurodegeneration, inflammatory diseases, and aging. In response to oxidative attack, cells have developed an antioxidant defense system to maintain cellular redox homeostasis and to protect cells from damage. The thiol-containing small molecules (e.g. glutathione), reactive oxygen species-inactivating enzymes (e.g. glutathione peroxidase), and phase 2 detoxifying enzymes (e.g. NAD(P)H: quinine oxidoreductase 1 and glutathione-S-transferases) are members of this antioxidant system. NF-E2-related factor 2 (Nrf2) is a CNC-bZIP transcription factor which regulates the basal and inducible expression of a wide array of antioxidant genes. Following dissociation from the cytosolic protein Keap1, a scaffolding protein which binds Nrf2 and Cul3 ubiquitin ligase for proteasome degradation, Nrf2 rapidly accumulates in the nucleus and transactivates the antioxidant response element in the promoter region of many antioxidant genes. The critical role of Nrf2 has been demonstrated by various animal studies showing that mice with a targeted disruption of the nrf2 gene are prone to develop lesions in response to environmental toxicants/carcinogens, drugs, and inflammatory insults. In this review, we discuss the role of the Nrf2 system, with particular focus on Nrf2-controlled target genes and the potential pleiotropic effects of Nrf2 activation of indirect antioxidants.


Subject(s)
Antioxidants/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Animals , Antioxidants/chemistry , Gene Expression Regulation , Humans , Molecular Structure , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Reactive Oxygen Species/metabolism
10.
Free Radic Biol Med ; 48(8): 1051-63, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20096777

ABSTRACT

Epithelial-mesenchymal transition (EMT) is an underlying mechanism of tissue fibrosis, generating myofibroblasts, which serve as the primary source of extracellular matrix production from tissue epithelial cells. Recently, EMT has been implicated in immunosuppressive cyclosporin A (CsA)-induced renal fibrosis. In this study, the potential role of NRF2, which is the master regulator of genes associated with the cellular antioxidant defense system, in CsA-induced EMT renal fibrosis has been investigated. Pretreatment of rat tubular epithelial NRK-52E cells with sulforaphane, an activator of NRF2, could prevent EMT gene changes such as the loss of E-cadherin and the increase in alpha-smooth muscle actin (alpha-SMA) expression. Conversely, genetic inhibition of NRF2 in these cells aggravated changes in CsA-induced EMT markers. These in vitro observations could be confirmed in vivo: CsA treatment resulted in severe renal damage and fibrosis with increased expression of alpha-SMA in NRF2-deficient mice compared to wild-type mice. NRF2-mediated amelioration of CsA-caused EMT changes could be accounted for in part by the regulation of heme oxygenase-1 (HO-1). CsA treatment increased HO-1 expression in an NRF2-dependent manner in NRK cells as well as in murine fibroblasts. Induction of HO-1 by CsA seems to be advantageous in that it counteracts EMT gene changes: specific increase in HO-1 expression caused by cobalt protoporphyrin prevented CsA-mediated alpha-SMA induction, whereas genetic inhibition of HO-1 by siRNA substantially enhanced alpha-SMA induction compared to control cells. Collectively, our results suggest that the NRF2-HO-1 system plays a protective role against CsA-induced renal fibrosis by modulating EMT gene changes.


Subject(s)
Cyclosporine/pharmacology , Epithelial Cells/physiology , Heme Oxygenase-1/metabolism , Kidney/pathology , Mesoderm/physiology , NF-E2-Related Factor 2/metabolism , Animals , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibrosis , Isothiocyanates , Male , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Mice, Inbred ICR , Rats , Sulfoxides , Thiocyanates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...