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










Publication year range
1.
Biomol Ther (Seoul) ; 30(4): 368-379, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35768333

ABSTRACT

Hyaluronic acid (HA), a ligand of CD44, accumulates in some types of tumors and is responsible for tumor progression. The nuclear factor erythroid 2-like 2 (NRF2) regulates cytoprotective genes and drug transporters, which promotes therapy resistance in tumors. Previously, we showed that high levels of CD44 are associated with NRF2 activation in cancer stem like-cells. Herein, we demonstrate that HA production was increased in doxorubicin-resistant breast cancer MCF7 cells (MCF7-DR) via the upregulation of HA synthase-2 (HAS2). HA incubation increased NRF2, aldo-keto reductase 1C1 (AKR1C1), and multidrug resistance gene 1 (MDR1) levels. Silencing of HAS2 or CD44 suppressed NRF2 signaling in MCF7-DR, which was accompanied by increased doxorubicin sensitivity. The treatment with a HAS2 inhibitor, 4-methylumbelliferone (4-MU), decreased NRF2, AKR1C1, and MDR1 levels in MCF7-DR. Subsequently, 4-MU treatment inhibited sphere formation and doxorubicin resistance in MCF7-DR. The Cancer Genome Atlas (TCGA) data analysis across 32 types of tumors indicates the amplification of HAS2 gene is a common genetic alteration and is negatively correlated with the overall survival rate. In addition, high HAS2 mRNA levels are associated with increased NRF2 signaling and poor clinical outcome in breast cancer patients. Collectively, these indicate that HAS2 elevation contributes to chemoresistance and sphere formation capacity of drug-resistant MCF7 cells by activating CD44/NRF2 signaling, suggesting a potential benefit of HAS2 inhibition.

2.
EBioMedicine ; 76: 103850, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35108666

ABSTRACT

BACKGROUND: Given the lack of visual discrepancy between malignant and surrounding normal tissue, current breast conserving surgery (BCS) is associated with a high re-excision rate. Due to the increasing cases of BCS, a novel method of complete tumour removal at the initial surgical resection is critically needed in the operating room to help optimize the surgical procedure and to confirm tumour-free edges. METHODS: We developed a unique near-infrared (NIR) fluorescence imaging probe, ICG-p28, composed of the clinically nontoxic tumour-targeting peptide p28 and the FDA-approved NIR dye indocyanine green (ICG). ICG-p28 was characterized in vitro and evaluated in multiple breast cancer animal models with appropriate control probes. Our experimental approach with multiple-validations and -blinded procedures was designed to determine whether ICG-p28 can accurately identify tumour margins in mimicked intraoperative settings. FINDINGS: The in vivo kinetics were analysed to optimize settings for potential clinical use. Xenograft tumours stably expressing iRFP as a tumour marker showed significant colocalization with ICG-p28, but not ICG alone. Image-guided surgery with ICG-p28 showed an over 6.6 × 103-fold reduction in residual normalized tumour DNA at the margin site relative to control approaches (i.e., surgery with ICG or palpation/visible inspection alone), resulting in an improved tumour recurrence rate (92% specificity) in multiple breast cancer animal models independent of the receptor expression status. ICG-p28 allowed accurate identification of tumour cells in the margin to increase the complete resection rate. INTERPRETATION: Our simple and cost-effective approach has translational potential and offers a new surgical procedure that enables surgeons to intraoperatively identify tumour margins in a real-time, 3D fashion and that notably improves overall outcomes by reducing re-excision rates. FUNDING: This work was supported by NIH/ National Institute of Biomedical Imaging and Bioengineering, R01EB023924.


Subject(s)
Neoplasm Recurrence, Local , Surgery, Computer-Assisted , Animals , Humans , Indocyanine Green , Margins of Excision , Optical Imaging/methods , Surgery, Computer-Assisted/methods
3.
Methods Mol Biol ; 2394: 857-865, 2022.
Article in English | MEDLINE | ID: mdl-35094362

ABSTRACT

Precise surgical resection directly influences the prognosis and survival of patients with solid tumors. However, it is often difficult to distinguish tumor from normal tissue during resection without any intraoperative imaging guidance. Image-guided surgery particularly when coupled with a near-infrared (NIR) fluorescent agent may improve positive-margin rate thereby improving the overall prognosis. We have developed a unique tumor-targeting fluorescence imaging agent that can aid in the accurate localization of human cancer cells in preclinical settings. The NIR imaging agent, ICG-p28, a water-soluble, nontoxic, and pan-tumor targeting probe consisting of a cell-penetrating peptide (p28) conjugated to indocyanine green (ICG), can accurately localize tumors in vivo. Development of the noninvasive, targeted imaging agent can potentially improve in the resections of tumors by enabling the localization of lesions that are currently difficult or impossible to detect by visual observation or palpation. Here, we describe the methods of preclinical animal imaging models by using NIR fluorescence imager coupled with a new tumor-targeting agent.


Subject(s)
Fluorescent Dyes , Neoplasms , Animals , Humans , Indocyanine Green , Neoplasms/diagnostic imaging , Optical Imaging/methods , Peptides
4.
Free Radic Biol Med ; 138: 33-42, 2019 07.
Article in English | MEDLINE | ID: mdl-31059771

ABSTRACT

Transforming growth factor-ß (TGF-ß) is a potent pathogenic factor of renal injury through the upregulation of extracellular matrix (ECM) expression and facilitation of renal fibrosis. Nuclear factor erythroid 2-like 2 (Nfe2l2; Nrf2), a master regulator of antioxidant and detoxifying systems, is mainly controlled by the binding with cytosolic protein Kelch-like ECH-associated protein 1 (Keap1) and subsequent proteasomal degradation. The protective effect of Nrf2 on renal injury has been attributed to its antioxidant role, where it aids in coping with oxidative stress-associated progression of renal disease. In this study, we investigated the effect of Nrf2 activation on ECM production and TGF-ß/Smad signaling using Keap1-silenced MES-13 cells (a genetic glomerular mesangial cell model with Nrf2 overexpression). The TGF-ß1-inducible expression of fibronectin and α-smooth muscle actin (α-Sma) was suppressed and Smad2/3 phosphorylation was blocked in Nrf2-high mesangial cells as compared with that in control cells. Notably, in these Nrf2-high mesangial cells, levels of TGF-ß1 receptor 1 (TßR1) were substantially diminished, and the protein levels of Smad7, an inhibitor TGF-ß1/Smad signaling, were increased. Nrf2-mediated Smad7 elevation and its anti-fibrotic role in Keap1-silenced cells were confirmed by studies with Nrf2-or Smad7-silencing. As a molecular link for Smad7 elevation in Nrf2-high cells, the reduction of Smad-ubiquitination-regulatory factor 1 (Smurf1), an E3 ubiquitin ligase for Smad7, was notable. Silencing of Smurf1 increased Smad7 in the control mesangial cells; however, forced expression of Smurf1 repressed Smad7 levels in Keap1-silenced cells. Additionally, we demonstrate that bardoxolone (BARD; CDDO-methyl), a pharmacological activator of Nrf2, increased Smad7 levels and attenuated TGF-ß/Smad/ECM expression in MES-13. Moreover, in an aristolochic acid (AA)-mediated nephropathy mouse model, the renal expression of Nrf2 and Smad7 was elevated by BARD treatment, and AA-induced tubular necrosis and interstitial fibrosis were substantially ameliorated by BARD. Collectively, these results indicate that the Nrf2-Smad7 axis plays a key role in the protection of TGF-ß-induced renal fibrosis, and further suggest a novel molecular mechanism of beneficial effect of BARD on renal disease.


Subject(s)
Kelch-Like ECH-Associated Protein 1/genetics , Kidney Diseases/drug therapy , NF-E2-Related Factor 2/genetics , Oleanolic Acid/analogs & derivatives , Smad7 Protein/genetics , Transforming Growth Factor beta1/genetics , Animals , Aristolochic Acids/administration & dosage , Cell Line , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1/deficiency , Kidney Diseases/chemically induced , Kidney Diseases/genetics , Kidney Diseases/pathology , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Smad7 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Toxicol Appl Pharmacol ; 359: 24-33, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30236989

ABSTRACT

Mitochondria play essential roles in cellular bioenergetics, biosynthesis, and apoptosis. During the process of respiration and oxidative phosphorylation, mitochondria utilize oxygen to generate ATP, and at the same time, there is an inevitable generation of reactive oxygen species (ROS). As excess ROS create oxidative stress and damage cells, the proper function of the antioxidant defense system is critical for eukaryotic cell survival under aerobic conditions. Nuclear factor, erythroid 2-like 2 (Nfe2l2/Nrf2) is a master transcription factor for regulating basal as well as inducible expression of multiple antioxidant proteins. Nrf2 has been involved in maintaining mitochondrial redox homeostasis by providing reduced forms of glutathione (GSH); the reducing cofactor NADPH; and mitochondrial antioxidant enzymes such as GSH peroxidase 1, superoxide dismutase 2, and peroxiredoxin 3/5. In addition, recent research advances suggest that Nrf2 contributes to mitochondrial regulation through more divergent intermolecular linkages. Nrf2 has been positively associated with mitochondrial biogenesis through the direct upregulation of mitochondrial transcription factors and is involved in the mitochondrial quality control system through mitophagy activation. Moreover, several mitochondrial proteins participate in regulating Nrf2 to form a reciprocal regulatory loop between mitochondria and Nrf2. Additionally, Nrf2 modulation in cancer cells leads to changes in the mitochondrial respiration system and cancer bioenergetics that overall affect cancer metabolism. In this review, we describe recent experimental observations on the relationship between Nrf2 and mitochondria, and further discuss the effects of Nrf2 on cancer mitochondria and metabolism.


Subject(s)
Mitochondria/drug effects , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Receptor Cross-Talk/drug effects , Animals , Humans , Mitochondria/metabolism , NF-E2-Related Factor 2/drug effects
6.
Cell Death Dis ; 9(9): 896, 2018 08 30.
Article in English | MEDLINE | ID: mdl-30166520

ABSTRACT

Aldehyde dehydrogenase 1A1 (ALDH1A1) is one of cancer stem cell (CSC) markers, and high ALDH1 expression has been related to drug resistance and facilitated tumor growth. In this study, we investigated the potential involvement of nuclear factor erythroid 2-like 2 (NFE2L2/NRF2) in CSC-like properties of ALDH-high ovarian CSCs. Our experimental system, ALDH1A1-high (ALDH-H) subpopulation, was isolated and stabilized using doxorubicin-resistant ovarian cancer A2780 cells. ALDH-H exerted CSC-like properties such as drug resistance, colony/sphere formation, and enhanced tumor growth along with high levels of CSCs markers compared to ALDH1A1-low (ALDH-L). Levels of NRF2 and subsequent target genes substantially increased in ALDH-H cells, and the increase in ALDH1A1 and p62 was associated with NRF2 upregulation. ALDH1A1-silencing blocked increases in NRF2, drug efflux transporters, and p62, along with CSC markers in ALDH-H cells. The inhibition of p62, which was elevated in ALDH-H, suppressed NRF2 activation. High NRF2 level was confirmed in the ALDH1-high subpopulation from colon cancer HCT116 cells. The functional implication of NRF2 activation in ovarian CSCs was verified by two experimental approaches. First, CSC-like properties such as high CSC markers, chemoresistance, colony/sphere formation, and tumor growth were significantly inhibited by NRF2-silencing in ALDH-H cells. Second, all-trans retinoic acid (ATRA) suppressed ALDH1 expression, inhibiting NRF2 activation, which led to the attenuation of CSC-like properties in ALDH-H cells but not in ALDH-L cells. These results provide insight into the molecular basis of the ALDH1A1-mediated development of CSC-like properties such as stress/treatment resistance, and further suggest the therapeutic potential of ATRA in ALDH-high ovarian CSCs.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Antineoplastic Agents/pharmacology , NF-E2-Related Factor 2/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/pathology , Tretinoin/pharmacology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Retinal Dehydrogenase
7.
Biomol Ther (Seoul) ; 26(5): 487-493, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30157616

ABSTRACT

Cluster of differentiation 44 (CD44), a cell surface receptor for hyaluronic acid (HA), is involved in aggressive cancer phenotypes. Herein, we investigated the role of the CD44 standard isoform (CD44s) in hypoxia-inducible factor-1α (HIF-1α) regulation using MCF7 overexpressing CD44s (pCD44s-MCF7). When pCD44s-MCF7 was incubated under hypoxia, levels of HIF-1α, vascular endothelial growth factor, and the HIF-1α response element-derived luciferase activity were significantly increased compared to those in the control MCF7. Incubation of pCD44s-MCF7 cells with HA further increased HIF-1α accumulation, and the silencing of CD44s attenuated HIF-1α elevation, which verifies the role of CD44s in HIF-1α regulation. In addition, the levels of phosphorylated extracellular signal-regulated kinase (ERK) was higher in hypoxic pCD44s-MCF7 cells, and HIF-1α accumulation was diminished by the pharmacological inhibitors of ERK. CD44s-mediated HIF-1α augmentation resulted in two functional outcomes. First, pCD44s-MCF7 cells showed facilitated cell motility under hypoxia via the upregulation of proteins associated with epithelialmesenchymal transition, such as SNAIL1 and ZEB1. Second, pCD44s-MCF7 cells exhibited higher levels of glycolytic proteins, such as glucose transporter-1, and produced higher levels of lactate under hypoxa. As a consequence of the enhanced glycolytic adaptation to hypoxia, pCD44s-MCF7 cells exhibited a higher rate of cell survival under hypoxia than that of the control MCF7, and glucose deprivation abolished these differential responses of the two cell lines. Taken together, these results suggest that CD44s activates hypoxia-inducible HIF-1α signaling via ERK pathway, and the CD44s-ERK-HIF-1α pathway is involved in facilitated cancer cell viability and motility under hypoxic conditions.

8.
Redox Biol ; 17: 246-258, 2018 07.
Article in English | MEDLINE | ID: mdl-29729523

ABSTRACT

Cluster of differentiation 44 (CD44) is the most common cancer stem cell (CSC) marker and high CD44 expression has been associated with anticancer drug resistance, tumor recurrence, and metastasis. In this study, we aimed to investigate the molecular mechanism by which CD44 and nuclear factor erythroid 2-like 2 (NFE2L2; NRF2), a key regulator of antioxidant genes, are linked to CSC resistance using CD44high breast CSC-like cells. NRF2 expression was higher in CD44high cell populations isolated from doxorubicin-resistant MCF7 (ADR), as well as MCF7, MDA-MB231, and A549 cells, than in corresponding CD44low cells. High NRF2 expression in the CD44highCD24low CSC population (ADR44P) established from ADR cells depended on standard isoform of CD44. Silencing of CD44 or overexpression of CD44 resulted in the reduction or elevation of NRF2, respectively, and treatment with hyaluronic acid, a CD44 ligand, augmented NRF2 activation. As functional implications, NRF2 silencing rendered ADR44P cells to retain higher levels of reactive oxygen species and to be sensitive to anticancer drug toxicity. Moreover, NRF2-silenced ADR44P cells displayed tumor growth retardation and reduced colony/sphere formation and invasion capacity. In line with these, CD44 significantly colocalized with NRF2 in breast tumor clinical samples. The molecular mechanism of CD44-mediated NRF2 activation was found to involve high p62 expression. CD44 elevation led to an increase in p62, and inhibition of p62 resulted in NRF2 suppression in ADR44P. Collectively, our results showed that high CD44 led to p62-associated NRF2 activation in CD44high breast CSC-like cells. NRF2 activation contributed to the aggressive phenotype, tumor growth, and anticancer drug resistance of CD44high CSCs. Therefore, the CD44-NRF2 axis might be a promising therapeutic target for the control of stress resistance and survival of CD44high CSC population within breast tumors.


Subject(s)
Breast Neoplasms/drug therapy , Hyaluronan Receptors/genetics , NF-E2-Related Factor 2/genetics , RNA-Binding Proteins/genetics , Antioxidants/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Lineage/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/antagonists & inhibitors , MCF-7 Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology
9.
Oncotarget ; 8(63): 107188-107205, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29291022

ABSTRACT

The nuclear factor (erythroid-derived 2)-like 2 (NFE2L2/NRF2) plays a critical role in the expression of multiple antioxidant and detoxifying enzymes. Herein, we provide evidence of the molecular links between NRF2 and oncogenic signaling hepatocyte growth factor receptor (HGFR/c-MET) and epidermal growth factor receptor (EGFR). Interfering RNA-induced stable inhibition of NRF2 in ovarian carcinoma SKOV3 and renal carcinoma A498 reduced the levels of c-MET and EGFR. MicroRNA-206 (miR-206) that was increased in both NRF2-silenced cells was predicted as a dual regulator of c-MET and EGFR. As experimental evidence, miR-206 decreased c-MET and EGFR levels through a direct binding to the 3'-untranslated region of the c-MET and EGFR genes. The treatment of NRF2-knockdown cells with the miR-206 inhibitor could restore c-MET and EGFR levels. The miR-206-mediated c-MET/EGFR repression resulted in two outcomes. First, presumably through the inhibition of c-MET/EGFR-dependent cell proliferation, overexpression of miR-206 inhibited tumor growth in SKOV3-inoculated nude mice. Second, reduced c-MET/EGFR in NRF2-silenced cells affected breast cancer resistance protein (BCRP/ABCG2) levels. The pharmacological and genetic inhibition of c-MET or EGFR, as well as the miR-206 mimic treatment, repressed BCRP levels and increased cellular accumulation of doxorubicin. In line with these, treatment of NRF2-silenced SKOV3 with the miR-206 inhibitor elevated BCRP levels and consequently made these cells more resistant to doxorubicin treatment. Collectively, our results demonstrated that the NRF2 silencing-inducible miR-206 targeted both c-MET and EGFR, and subsequently suppressed the BCRP level in cancer cells.

10.
Biomol Ther (Seoul) ; 24(5): 482-8, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27582554

ABSTRACT

Cancer stem cells (CSCs) are a subset of tumor cells, which are characterized by resistance against chemotherapy and environmental stress, and are known to cause tumor relapse after therapy. A number of molecular mechanisms underlie the chemoresistance of CSCs, including high expression levels of drug efflux transporters. We investigated the role of the antioxidant transcription factor NF-E2-related factor 2 (NRF2) in chemoresistance development, using a CSC-enriched colonosphere system. HCT116 colonospheres were more resistant to doxorubicin-induced cell death and expressed higher levels of drug efflux transporters such as P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP) compared to HCT116 monolayers. Notably, levels of NRF2 and expression of its target genes were substantially elevated in colonospheres, and these increases were linked to doxorubicin resistance. When NRF2 expression was silenced in colonospheres, Pgp and BCRP expression was downregulated, and doxorubicin resistance was diminished. Collectively, these results indicate that NRF2 activation contributes to chemoresistance acquisition in CSC-enriched colonospheres through the upregulation of drug efflux transporters.

11.
Oxid Med Cell Longev ; 2016: 2428153, 2016.
Article in English | MEDLINE | ID: mdl-26682001

ABSTRACT

Tumors contain a distinct small subpopulation of cells that possess stem cell-like characteristics. These cells have been called cancer stem cells (CSCs) and are thought to be responsible for anticancer drug resistance and tumor relapse after therapy. Emerging evidence indicates that CSCs share many properties, such as self-renewal and quiescence, with normal stem cells. In particular, CSCs and normal stem cells retain low levels of reactive oxygen species (ROS), which can contribute to stem cell maintenance and resistance to stressful tumor environments. Current literatures demonstrate that the activation of ataxia telangiectasia mutated (ATM) and forkhead box O3 (FoxO3) is associated with the maintenance of low ROS levels in normal stem cells such as hematopoietic stem cells. However, the importance of ROS signaling in CSC biology remains poorly understood. Recent studies demonstrate that nuclear factor-erythroid 2-related factor 2 (NRF2), a master regulator of the cellular antioxidant defense system, is involved in the maintenance of quiescence, survival, and stress resistance of CSCs. Here, we review the recent findings on the roles of NRF2 in maintenance of the redox state and multidrug resistance in CSCs, focusing on how NRF2-mediated ROS modulation influences the growth and resistance of CSCs.


Subject(s)
Drug Resistance, Neoplasm , NF-E2-Related Factor 2/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Animals , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , NF-E2-Related Factor 2/genetics , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Reactive Oxygen Species/metabolism
12.
PLoS One ; 10(7): e0134235, 2015.
Article in English | MEDLINE | ID: mdl-26222138

ABSTRACT

Blood monocytes are recruited to injured tissue sites and differentiate into macrophages, which protect against pathogens and repair damaged tissues. Reactive oxygen species (ROS) are known to be an important contributor to monocytes' differentiation and macrophages' function. NF-E2-related factor 2 (NRF2), a transcription factor regulating cellular redox homeostasis, is known to be a critical modulator of inflammatory responses. We herein investigated the role of NRF2 in macrophage differentiation using the human monocytic U937 cell line and phorbol-12-myristate-13-acetate (PMA). In U937 cells with NRF2 silencing, PMA-stimulated cell adherence was significantly facilitated when compared to control U937 cells. Both transcript and protein levels for pro-inflammatory cytokines, including interleukine-1ß (IL-1ß), IL-6, and tumor necrosis factor-α (TNFα) were highly elevated in PMA-stimulated NRF2-silenced U937 compared to the control. In addition, PMA-inducible secretion of monocyte chemotactic protein 1 (MCP-1) was significantly high in NRF2-silenced U937. As an underlying mechanism, we showed that NRF2-knockdown U937 retained high levels of cellular ROS and endoplasmic reticulum (ER) stress markers expression; and subsequently, PMA-stimulated levels of Ca2+ and PKCα were greater in NRF2-knockdown U937 cells, which caused enhanced nuclear accumulation of nuclear factor-Ò¡B (NFÒ¡B) p50 and extracellular signal-regulated kinase (ERK)-1/2 phosphorylation. Whereas the treatment of NRF2-silenced U937 cells with pharmacological inhibitors of NFÒ¡B or ERK1/2 largely blocked PMA-induced IL-1ß and IL-6 expression, indicating that these pathways are associated with cell differentiation. Taken together, our results suggest that the NRF2 system functions to suppress PMA-stimulated U937 cell differentiation into pro-inflammatory macrophages and provide evidence that the ROS-PKCα-ERK-NFÒ¡B axis is involved in PMA-facilitated differentiation of NRF2-silenced U937 cells.


Subject(s)
Macrophages/drug effects , Macrophages/metabolism , Monocytes/drug effects , Monocytes/metabolism , NF-E2-Related Factor 2/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cytokines/metabolism , Endoplasmic Reticulum Stress , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System/drug effects , Macrophages/immunology , Monocytes/immunology , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-kappa B p50 Subunit/metabolism , Protein Kinase C-alpha/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , U937 Cells
13.
Toxicol In Vitro ; 29(5): 884-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25841332

ABSTRACT

The kidney secretes various xenobiotics through a well-established transport system. The transcription factor NF-E2-related factor 2 (NRF2) up-regulates a subset of genes encoding antioxidant and detoxification proteins. Kelch-like ECH-associated protein 1 (KEAP1) down-regulates NRF2 by facilitating continuous degradation of NRF2 protein. Here, we investigated the role of NRF2 in the expression of renal drug transporters by using a stable KEAP1 knockdown renal tubular HK-2 cell line (shKEAP1). KEAP1 knockdown resulted in a significant increase in the expression of four renal transporters, namely, multidrug resistance protein 1 (MDR1; ABCB1), breast cancer resistance protein (BCRP; ABCG2), multidrug resistance-associated protein 2 (MRP2; ABCC2), and MRP3 (ABCC3). In western blot and immunocytochemical analyses, protein levels of these transporters were also significantly higher in the knockdown group. Consequently, shKEAP1 cells released more Hoechst 33342 fluorescent dye and doxorubicin, and they were more resistant to doxorubicin than the control cells. In addition, cisplatin resistance of shKEAP1 decreased upon co-incubation with a transporter inhibitor. Whereas, a short term incubation (24h) with sulforaphane did not show noticeable changes in the expression of transporter. Collectively, these results indicate that NRF2 regulates the expression of MDR1, BCRP, MRP2, and MRP3 in human tubular epithelial cells. Altered expression of these transporters affects drug secretion in these cells, which may result in the renal cellular damage upon exposure to nephrotoxic xenobiotics.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Intracellular Signaling Peptides and Proteins/genetics , NF-E2-Related Factor 2/metabolism , Benzimidazoles/pharmacology , Cell Line , Cisplatin/pharmacology , Doxorubicin/pharmacology , Gene Knockdown Techniques , Humans , Isothiocyanates/pharmacology , Kelch-Like ECH-Associated Protein 1 , Multidrug Resistance-Associated Protein 2 , Propionates/pharmacology , Quinolines/pharmacology , Sulfoxides
14.
Oncotarget ; 6(10): 8167-84, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25717032

ABSTRACT

Cancer stem cells (CSCs) express high levels of drug efflux transporters and antioxidant genes, and are therefore believed to be responsible for cancer recurrence following chemo/radiotherapy intervention. In this study, we investigated the role of NF-E2-related factor 2 (NRF2), a master regulator of antioxidant gene expression, in the growth and stress resistance of CSC-enriched mammosphere. The MCF7 mammospheres expressed significantly higher levels of the NRF2 protein and target gene expression compared to the monolayer. As underlying mechanisms, we observed that proteolytic activity and expression of the proteasome catalytic subunits were decreased in the mammospheres. Additionally, mammospheres retained a high level of p62 and the silencing of p62 was observed to attenuate NRF2 activation. NRF2 increase was confirmed in sphere-cultures of the colon and ovarian cancer cells. The functional implication of NRF2 was demonstrated in NRF2-knockdown mammospheres. NRF2-silenced mammospheres demonstrated increased cell death and retarded sphere growth as a result of target gene repression. Moreover, unlike the control mammospheres, NRF2-knockdown mammospheres did not develop anticancer drug resistance. Collectively, these results indicated that altered proteasome function and p62 expression caused NRF2 activation in CSC-enriched mammospheres. In addition, NRF2 appeared to play a role in CSC survival and anticancer drug resistance.


Subject(s)
Breast Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Neoplastic Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , NF-E2-Related Factor 2/genetics , Neoplastic Stem Cells/pathology , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Transfection , Up-Regulation
15.
Arch Pharm Res ; 38(2): 272-81, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24849033

ABSTRACT

Transforming growth factor-ß1 (TGFß1) induces epithelial-to-mesenchymal transition (EMT) in cultured renal tubular epithelial cells. This phenotypic transition has been known to be involved in the development of chronic kidney diseases by activating profibrotic gene expression. Since oxidative stress has been recognized as one of the contributors to this TGFß1-mediated pathology, we investigated the potential involvement of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which is a key transcription factor for the regulation of multiple antioxidant genes, in TGFß1-stimulated EMT gene changes using the rat proximal tubular epithelial cell line NRK52E. The treatment of NRK52E with TGFß1 led to changes in EMT gene expression, including increased α-Sma and decreased E-cadherin expression. In these cells, the TGFß1 treatment decreased the transcript level of the catalytic subunit of γ-glutamate cysteine ligase (Gclc), a glutathione (GSH) biosynthetic enzyme, and reduced the total GSH content with a concomitant decrease in Nrf2 transcription activity. Accordantly, pre-incubation with the GSH precursor N-acetylcysteine attenuated TGFß1-stimulated EMT gene changes. The involvement of Nrf2 in EMT gene changes has been demonstrated using NRK52E cells with nrf2 knockdown or pharmacological activation. When the expression of Nrf2 was stably silenced in NRK52E cells using interfering RNA administration, Gclc expression was significantly reduced and the increase in the levels of α-Sma and fibronectin-1 by TGFß1 was greater than those in the nonspecific RNA control group. Conversely, Nrf2 activation and subsequent Gclc increase by Nrf2-activating sulforaphane alleviated the TGFß1-stimulated α-Sma increase and E-cadherin decrease. Collectively, these results indicate that Nrf2-GSH signaling can modulate TGFß1-stimulated EMT gene changes and further suggest a beneficial role of Nrf2 inducers in renal pathogenesis.


Subject(s)
Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Glutathione/metabolism , Kidney Tubules, Proximal/drug effects , NF-E2-Related Factor 2/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Cell Line , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression/drug effects , Gene Knockdown Techniques , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , NF-E2-Related Factor 2/genetics , Rats , Recombinant Proteins , Signal Transduction/drug effects , Signal Transduction/genetics
16.
PLoS One ; 9(9): e107158, 2014.
Article in English | MEDLINE | ID: mdl-25226504

ABSTRACT

Photodynamic therapy (PDT) has emerged as an effective treatment for various solid tumors. The transcription factor NRF2 is known to protect against oxidative and electrophilic stress; however, its constitutive activity in cancer confers resistance to anti-cancer drugs. In the present study, we investigated NRF2 signaling as a potential molecular determinant of pheophorbide a (Pba)-based PDT by using NRF2-knockdown breast carcinoma MDA-MB-231 cells. Cells with stable NRF2 knockdown showed enhanced cytotoxicity and apoptotic/necrotic cell death following PDT along with increased levels of singlet oxygen and reactive oxygen species (ROS). A confocal microscopic visualization of fluorogenic Pba demonstrated that NRF2-knockdown cells accumulate more Pba than control cells. A subsequent analysis of the expression of membrane drug transporters showed that the basal expression of BCRP is NRF2-dependent. Among measured drug transporters, the basal expression of breast cancer resistance protein (BCRP; ABCG2) was only diminished by NRF2-knockdown. Furthermore, after incubation with the BCRP specific inhibitor, differential cellular Pba accumulation and ROS in two cell lines were abolished. In addition, NRF2-knockdown cells express low level of peroxiredoxin 3 compared to the control, which implies that diminished mitochondrial ROS defense system can be contributing to PDT sensitization. The role of the NRF2-BCRP pathway in Pba-PDT response was further confirmed in colon carcinoma HT29 cells. Specifically, NRF2 knockdown resulted in enhanced cell death and increased singlet oxygen and ROS levels following PDT through the diminished expression of BCRP. Similarly, PDT-induced ROS generation was substantially increased by treatment with NRF2 shRNA in breast carcinoma MCF-7 cells, colon carcinoma HCT116 cells, renal carcinoma A498 cells, and glioblastoma A172 cells. Taken together, these results indicate that the manipulation of NRF2 can enhance Pba-PDT sensitivity in multiple cancer cells.


Subject(s)
Chlorophyll/analogs & derivatives , Gene Silencing , NF-E2-Related Factor 2/genetics , Neoplasms/genetics , Photochemotherapy , Radiation-Sensitizing Agents/administration & dosage , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chlorophyll/administration & dosage , Chlorophyll/metabolism , Chlorophyll/toxicity , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Female , Gene Knockdown Techniques , Genetic Vectors/genetics , Humans , Laser Therapy , Lasers , Lentivirus/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Peroxiredoxin III/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Radiation-Sensitizing Agents/metabolism , Radiation-Sensitizing Agents/toxicity , Reactive Oxygen Species/metabolism , Transduction, Genetic
17.
PLoS One ; 9(4): e93265, 2014.
Article in English | MEDLINE | ID: mdl-24691097

ABSTRACT

Transforming growth factor ß1 (TGFß1) is a potent stimulator of epithelial-to-mesenchymal transition (EMT) and has been associated with chronic kidney diseases by activating profibrotic gene expression. In this study, we investigated the role of the KEAP1-NRF2 pathway, which is a master regulator of the cellular antioxidant system, in TGFß1-stimulated EMT gene changes using human renal tubular epithelial HK2. Treatment with TGFß1 enhanced the levels of reactive oxygen species (ROS) and TGFß1-stimulated EMT gene changes, including an increase in profibrotic fibronectin-1 and collagen 1A1, were diminished by the antioxidant N-acetylcysteine. In HK2, TGFß1 suppressed NRF2 activity and thereby reduced the expression of GSH synthesizing enzyme through the elevation of ATF3 level. Therefore, the activation of NRF2 signaling with sulforaphane effectively attenuated the TGFß1-stimulated increase in fibronectin-1 and collagen 1A1. Conversely, the TGFß1-EMT gene changes were further enhanced by NRF2 knockdown compared to the control cells. The relationship of NRF2 signaling and TGFß1-EMT changes was further confirmed in a stable KEAP1-knockdown HK2, which is a model of pure activation of NRF2. The TGFß1-mediated increase of collagen 1A1 and fibronectin-1 in KEAP1 knockdown HK2 was suppressed. In particular, TGFß1-SMAD signaling was modulated in KEAP1 knockdown HK2: the TGFß1-stimulated SMAD2/3 phosphorylation and SMAD transcriptional activity were repressed. Additionally, the protein level of SMAD7, an inhibitor of SMAD signaling, was elevated and the level of SMURF1, an E3 ubiquitin ligase for SMAD7 protein, was diminished in KEAP1 knockdown HK2. Finally, the inhibition of SMAD7 expression in KEAP1 knockdown HK2 restored TGFß1 response, indicating that SMURF1-SMAD7 may be a molecular signaling linking the NRF2-GSH pathway to TGFß1-EMT changes. Collectively, these results indicate that the KEAP1-NRF2 antioxidant system can be an effective modulator of TGFß1-stimulated renal epithelial transition to fibroblastic cells through the SMUR1-SMAD7 signaling, and further implies the beneficial role of NRF2 in chronic renal diseases.


Subject(s)
Cell Transdifferentiation/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Cell Transdifferentiation/genetics , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Gene Knockdown Techniques , Glutathione/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kelch-Like ECH-Associated Protein 1 , Kidney/cytology , Kidney/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , NF-E2-Related Factor 2/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Smad Proteins/metabolism
18.
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
19.
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
20.
Toxicol Appl Pharmacol ; 258(1): 89-98, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22036727

ABSTRACT

Silver nanoparticles (nano-Ag) have been widely used in various commercial products including textiles, electronic appliances and biomedical products. However, there remains insufficient information on the potential risk of nano-Ag to human health and environment. In the current study, we have investigated the role of NF-E2-related factor 2 (Nrf2) transcription factor in nano-Ag-induced cytotoxicity. When Nrf2 expression was blocked using interring RNA expression in ovarian carcinoma cell line, nano-Ag treatment showed a substantial decrease in cell viability with concomitant increases in apoptosis and DNA damage compared to the control cells. Target gene analysis revealed that the expression of heme oxygenase-1 (HO-1) was highly elevated by nano-Ag in nonspecific shRNA expressing cells, while Nrf2 knockdown cells (NRF2i) did not increase HO-1 expression. The role of HO-1 in cytoprotection against nano-Ag was reinforced by results using pharmacological inducer of HO-1: cobalt protoporphyrin-mediated HO-1 activation in the NRF2i cells prevented nano-Ag-mediated cell death. Similarly, pharmacological or genetic inhibition of HO-1 in nonspecific control cells exacerbated nano-Ag toxicity. As the upstream signaling mechanism, nano-Ag required the phosphoinositide 3-kinase (PI3K) and p38MAPK signaling cascades for HO-1 induction. The treatment with either PI3K inhibitor or p38MAPK inhibitor suppressed HO-1 induction and intensified nano-Ag-induced cell death. Taken together, these results suggest that Nrf2-dependent HO-1 up-regulation plays a protective role in nano-Ag-induced DNA damage and consequent cell death. In addition, nano-Ag-mediated HO-1 induction is associated with the PI3K and p38MAPK signaling pathways.


Subject(s)
Heme Oxygenase-1/physiology , Metal Nanoparticles/toxicity , NF-E2-Related Factor 2/physiology , Signal Transduction/physiology , Silver/toxicity , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage , Humans , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , p38 Mitogen-Activated Protein Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...