Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Sci Rep ; 13(1): 19900, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37963997

ABSTRACT

The NFE2L1 transcription factor (aka Nrf1) is a basic leucine zipper protein that performs a critical role in the cellular stress response pathway. Here, we characterized a novel variant of NFE2L1 referred to as NFE2L1-616. The transcript encoding NFE2L1-616 is derived from an intronic promoter, and it has a distinct first exon than other reported full-length NFE2L1 isoforms. The NFE2L1-616 protein constitutively localizes in the nucleus as it lacks the N-terminal amino acid residues that targets other full-length NFE2L1 isoforms to the endoplasmic reticulum. The expression level of NFE2L1-616 is lower than other NFE2L1 isoforms. It is widely expressed across different cell lines and tissues that were examined. NFE2L1-616 showed strong transcriptional activity driving luciferase reporter expression from a promoter containing antioxidant response element. Together, the results suggest that NFE2L1-616 variant can function as a positive regulator in the transcriptional regulation of NFE2L1 responsive genes.


Subject(s)
Antioxidant Response Elements , Gene Expression Regulation , Antioxidant Response Elements/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Cell Line , NF-E2-Related Factor 1/metabolism
2.
Toxicology ; 471: 153173, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35367319

ABSTRACT

Patulin is a mycotoxin produced by a variety of molds that is found in various food products. The adverse health effects associated with exposure to patulin has led to many investigations into the biological basis driving the toxicity of patulin. Nevertheless, the mechanisms through which mammalian cells resists patulin-mediated toxicity is poorly understood. Here, we show that loss of the Nrf1 transcription factor renders cells sensitive to the acute cytotoxic effects of patulin. Nrf1 deficiency leads to accumulation of ubiquitinated proteins and protein aggregates in response to patulin exposure. Nrf1 expression is induced by patulin, and activation of proteasome genes by patulin is Nrf1-dependent. These findings suggest the Nrf1 transcription factor plays a crucial role in modulating cellular stress response against patulin cytotoxicity.

3.
Hum Mutat ; 43(4): 471-476, 2022 04.
Article in English | MEDLINE | ID: mdl-35112409

ABSTRACT

The NFE2L1 transcription factor (also known as Nrf1 for nuclear factor erythroid 2-related factor-1) is a broadly expressed basic leucine zipper protein that performs a critical role in the cellular stress response pathway. Here, we identified a heterozygous nonsense mutation located in the last exon of the gene that terminates translation prematurely, resulting in the production of a truncated peptide devoid of the carboxyl-terminal region containing the DNA-binding and leucine-zipper dimerization interface of the protein. Variant derivatives were well expressed in vitro, and they inhibited the transactivation function of wild-type proteins in luciferase reporter assays. Our studies suggest that this dominant-negative effect of truncated variants is through the formation of inactive heterodimers with wild-type proteins preventing the expression of its target genes. These findings suggest the potential role of diminished NFE2L1 function as an explanation for the developmental delay, hypotonia, hypospadias, bifid scrotum, and failure to thrive observed in the patient.


Subject(s)
Failure to Thrive , Muscle Hypotonia , Gene Expression Regulation , Genitalia , Humans , Male , NF-E2-Related Factor 1/genetics , NF-E2-Related Factor 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
J Biol Chem ; 296: 100732, 2021.
Article in English | MEDLINE | ID: mdl-33933455

ABSTRACT

The nuclear factor E2-related factor 1 (Nrf1) transcription factor performs a critical role in regulating cellular homeostasis as part of the cellular stress response and drives the expression of antioxidants and detoxification enzymes among many other functions. Ubiquitination plays an important role in controlling the abundance and thus nuclear accumulation of Nrf1 proteins, but the regulatory enzymes that act on Nrf1 are not fully defined. Here, we identified ubiquitin specific protease 7 (USP7), a deubiquitinating enzyme, as a novel regulator of Nrf1 activity. We found that USP7 interacts with Nrf1a and TCF11-the two long protein isoforms of Nrf1. Expression of wildtype USP7, but not its catalytically defective mutant, resulted in decreased ubiquitination of TCF11 and Nrf1a, leading to their increased stability and increased transactivation of reporter gene expression by TCF11 and Nrf1a. In contrast, knockdown or pharmacologic inhibition of USP7 dramatically increased ubiquitination of TCF11 and Nrf1a and reduction of their steady state levels. Loss of USP7 function attenuated the induction of Nrf1 protein expression in response to treatment with arsenic and other toxic metals, and inhibition of USP7 activity significantly sensitized cells to arsenic treatment. Collectively, these findings suggest that USP7 may act to modulate abundance of Nrf1 protein to induce gene expression in response to toxic metal exposure.


Subject(s)
Metals/metabolism , NF-E2-Related Factor 1/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Animals , Cell Line , HCT116 Cells , HEK293 Cells , Humans , Mice , Protein Interaction Maps , Protein Stability
5.
J Cell Sci ; 130(20): 3467-3480, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28839075

ABSTRACT

The Nrf2 transcription factor is a master regulator of the cellular anti-stress response. A population of the transcription factor associates with the mitochondria through a complex with KEAP1 and the mitochondrial outer membrane histidine phosphatase, PGAM5. To determine the function of this mitochondrial complex, we knocked down each component and assessed mitochondrial morphology and distribution. We discovered that depletion of Nrf2 or PGAM5, but not KEAP1, inhibits mitochondrial retrograde trafficking induced by proteasome inhibition. Mechanistically, this disrupted motility results from aberrant degradation of Miro2, a mitochondrial GTPase that links mitochondria to microtubules. Rescue experiments demonstrate that this Miro2 degradation involves the KEAP1-cullin-3 E3 ubiquitin ligase and the proteasome. These data are consistent with a model in which an intact complex of PGAM5-KEAP1-Nrf2 preserves mitochondrial motility by suppressing dominant-negative KEAP1 activity. These data further provide a mechanistic explanation for how age-dependent declines in Nrf2 expression impact mitochondrial motility and induce functional deficits commonly linked to neurodegeneration.


Subject(s)
Kelch-Like ECH-Associated Protein 1/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Phosphoprotein Phosphatases/metabolism , Animals , Biological Transport , Female , HEK293 Cells , Humans , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Microtubules/metabolism , Mitochondrial Dynamics , Protein Domains , Proteolysis , rho GTP-Binding Proteins/metabolism
6.
Free Radic Biol Med ; 110: 196-205, 2017 09.
Article in English | MEDLINE | ID: mdl-28625484

ABSTRACT

The Nrf1 (Nuclear factor E2-related factor 1) transcription factor performs a critical role in regulating cellular homeostasis. Using a proteomic approach, we identified Host Cell Factor-1 (HCF1), a co-regulator of transcription, and O-GlcNAc transferase (OGT), the enzyme that mediates protein O-GlcNAcylation, as cellular partners of Nrf1a, an isoform of Nrf1. Nrf1a directly interacts with HCF1 through the HCF1 binding motif (HBM), while interaction with OGT is mediated through HCF1. Overexpression of HCF1 and OGT leads to increased Nrf1a protein stability. Addition of O-GlcNAc decreases ubiquitination and degradation of Nrf1a. Transcriptional activation by Nrf1a is increased by OGT overexpression and treatment with PUGNAc. Together, these data suggest that OGT can act as a regulator of Nrf1a.


Subject(s)
Host Cell Factor C1/metabolism , N-Acetylglucosaminyltransferases/metabolism , Nuclear Respiratory Factor 1/metabolism , Protein Processing, Post-Translational , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/pharmacology , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Gene Expression , Glycosylation , HEK293 Cells , Host Cell Factor C1/chemistry , Host Cell Factor C1/genetics , Humans , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/genetics , Nuclear Respiratory Factor 1/chemistry , Nuclear Respiratory Factor 1/genetics , Oximes/pharmacology , Phenylcarbamates/pharmacology , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcriptional Activation/drug effects , Transfection , Ubiquitination
7.
Gene ; 584(1): 17-25, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-26947393

ABSTRACT

Nrf1 (also referred to as NFE2L1) is a member of the CNC-bZIP family of transcription factors that are characterized by a highly conserved CNC-domain, and a basic-leucine zipper domain required for dimerization and DNA binding. Nrf1 is ubiquitously expressed across tissue and cell types as various isoforms, and is induced by stress signals from a broad spectrum of stimuli. Evidence indicates that Nrf1 plays an important role in regulating a range of cellular functions including oxidative stress response, differentiation, inflammatory response, metabolism, and maintaining proteostasis. Thus, Nrf1 has been implicated in the pathogenesis of various disease processes including cancer development, and degenerative and metabolic disorders. This review summarizes our current understanding of Nrf1 and the molecular mechanism underlying its regulation and action in different cellular functions.


Subject(s)
Gene Expression Regulation , NF-E2-Related Factor 1/physiology , Animals , Homeostasis/genetics , Humans , NF-E2-Related Factor 1/chemistry , NF-E2-Related Factor 1/genetics , Protein Conformation
8.
J Biol Chem ; 291(14): 7754-66, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26841864

ABSTRACT

The NRF2 (also known as NFE2L2) transcription factor is a critical regulator of genes involved in defense against oxidative stress. Previous studies suggest thatNrf2plays a role in adipogenesisin vitro, and deletion of theNrf2gene protects against diet-induced obesity in mice. Here, we demonstrate that resistance to diet-induced obesity inNrf2(-/-)mice is associated with a 20-30% increase in energy expenditure. Analysis of bioenergetics revealed thatNrf2(-/-)white adipose tissues exhibit greater oxygen consumption. White adipose tissue showed a >2-fold increase inUcp1gene expression. Oxygen consumption is also increased nearly 2.5-fold inNrf2-deficient fibroblasts. Oxidative stress induced by glucose oxidase resulted in increasedUcp1expression. Conversely, antioxidant chemicals (such asN-acetylcysteine and Mn(III)tetrakis(4-benzoic acid)porphyrin chloride) and SB203580 (a known suppressor ofUcp1expression) decreasedUcp1and oxygen consumption inNrf2-deficient fibroblasts. These findings suggest that increasing oxidative stress by limitingNrf2function in white adipocytes may be a novel means to modulate energy balance as a treatment of obesity and related clinical disorders.


Subject(s)
Adipogenesis , Gene Expression Regulation , Ion Channels/biosynthesis , Mitochondrial Proteins/biosynthesis , NF-E2-Related Factor 2/deficiency , Obesity/metabolism , Oxidative Stress , Animals , Diet/adverse effects , Fibroblasts/metabolism , Fibroblasts/pathology , Free Radical Scavengers/pharmacology , Ion Channels/genetics , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Oxygen Consumption/drug effects , Uncoupling Protein 1
9.
Reprod Toxicol ; 58: 24-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26247513

ABSTRACT

Polycyclic aromatic hydrocarbons, like benzo[a]pyrene (BaP), are ubiquitous environmental pollutants and potent ovarian toxicants. The transcription factor NRF2 is an important regulator of the cellular response to electrophilic toxicants like BaP and to oxidative stress. NRF2 regulates transcription of genes involved in the detoxification of reactive metabolites of BaP and reactive oxygen species. We therefore hypothesized that Nrf2-/- mice have accelerated ovarian aging and increased sensitivity to the ovarian toxicity of BaP. A single injection of BaP dose-dependently depleted ovarian follicles in Nrf2+/+ and Nrf2-/- mice, but the effects of BaP were not enhanced in the absence of Nrf2. Similarly, Nrf2-/- mice did not have increased ovarian BaP DNA adduct formation compared to Nrf2+/+ mice. Ovarian follicle numbers did not differ between peripubertal Nrf2-/- and Nrf2+/+ mice, but by middle age, Nrf2-/- mice had significantly fewer primordial follicles than Nrf2+/+ mice, consistent with accelerated ovarian aging.


Subject(s)
Benzo(a)pyrene/toxicity , Cellular Senescence/drug effects , Environmental Pollutants/toxicity , Epithelial Cells/drug effects , Gene Deletion , NF-E2-Related Factor 2/deficiency , Ovarian Follicle/drug effects , Ovary/drug effects , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Genotype , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Ovarian Reserve/drug effects , Ovary/metabolism , Ovary/pathology , Oxidative Stress/drug effects , Phenotype
10.
FEBS Lett ; 589(5): 615-20, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25637874

ABSTRACT

Herpud1 is an ER-localized protein that contributes to endoplasmic reticulum (ER) homeostasis by participating in the ER-associated protein degradation pathway. The Nrf1 transcription factor is important in cellular stress pathways. We show that loss of Nrf1 function results in decreased Herpud1 expression in cells and liver tissues. Expression of Herpud1 increases in response to ER stress, but not in Nrf1 knockout cells. Transactivation studies show that Nrf1 acts through antioxidant response elements located in the Herpud1 promoter, and chromatin immunoprecipitation demonstrates that Herpud1 is a direct Nrf1 target gene. These results indicate that Nrf1 is a transcriptional activator of Herpud1 expression during ER stress, and they suggest Nrf1 is a key player in the regulation of the ER stress response in cells.


Subject(s)
Membrane Proteins/metabolism , Nuclear Respiratory Factor 1/metabolism , Blotting, Western , Chromatin Immunoprecipitation , Endoplasmic Reticulum Stress , Humans , Membrane Proteins/genetics , Nuclear Respiratory Factor 1/genetics , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Transcriptional Activation/genetics , Transcriptional Activation/physiology
11.
Cell Transplant ; 24(11): 2273-83, 2015.
Article in English | MEDLINE | ID: mdl-25581574

ABSTRACT

Oxidative stress is a major cause of islet damage and loss during the islet isolation process. The Nrf2 pathway plays a critical role in protecting the cells against oxidative stress. The aim of this study was to investigate the effect of an Nrf2 activator (dh404) on islet isolation and transplantation in a rodent model. Islet isolation was conducted using Nrf2-deficient and wild-type mice and vehicle-treated and Nrf2 activator (dh404)-treated rats. Islet yield, viability, and Nrf2 pathway activity were determined. An in vivo islet potency test was done. Islet yield and viability in Nrf2-deficient mice was significantly lower compared to wild-type (p < 0.05) mice. Furthermore, administration of dh404 to normal Sprague-Dawley rats enhanced nuclear translocation of Nrf2 and elevated HO-1 expression in the pancreas. Islet yield and viability in dh404-treated rats was significantly higher compared to the vehicle-treated group (p < 0.05). The diabetes cure rate in nude mice with chemically induced diabetes was significantly greater in those transplanted with islets from the dh404-treated group (6/9) than vehicle-treated rats (2/9, p < 0.05). The Nrf2 pathway plays a significant role in protecting islets against stress caused by the isolation process. Pharmacological activation of the Nrf2 pathway significantly increased HO-1 expression, improved islet yield, viability, and function after transplantation.


Subject(s)
Diabetes Mellitus/therapy , Islets of Langerhans Transplantation , Signal Transduction , Animals , Cell Survival , Diabetes Mellitus/chemically induced , Islets of Langerhans/drug effects , Male , Mice , Mice, Nude , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Oxidative Stress , Rats , Rats, Sprague-Dawley , Streptozocin
12.
Dig Dis Sci ; 60(5): 1215-22, 2015 May.
Article in English | MEDLINE | ID: mdl-25399330

ABSTRACT

BACKGROUND: Gut inflammation is prevalent in chronic kidney disease (CKD) and likely contributes to systemic inflammation via disruption of the epithelial tight junction with subsequent endotoxin and bacterial translocation. AIMS: To study the expression profile of inflammatory and tight junction proteins in the colon from CKD rats compared to healthy controls, and demonstrate the role of Nrf2 (transcription factor nuclear factor erythroid 2-related factor 2) using a potent Nrf2 activator. METHODS: CKD was induced via 5/6 nephrectomy in Sprague-Dawley rats, and dh404 (2 mg/kg/day) was used to study the effects of systemic Nrf2 activation. The experimental groups included sham, CKD and CKD+ dh404 rats. Blood and colon tissues were analyzed after a 10-week study period. RESULTS: Colon from CKD rats showed histological evidence of colitis, depletion of epithelial tight junction proteins, significant reduction of Nrf2 and its measured target gene products (NQO1, catalase, and CuZn SOD), activation of NFkB, and upregulation of pro-inflammatory molecules (COX-2, MCP-1, iNOS, and gp91(phox)). Treatment with dh404 attenuated colonic inflammation, restored Nrf2 activity and levels of NQO1, catalase and CuZn SOD, decreased NFkB and lowered expression of COX-2, MCP-1, iNOS, and gp91(phox). This was associated with restoration of colonic epithelial tight junction proteins (occludin and claudin-1). CONCLUSIONS: CKD rats exhibited colitis, disruption of colonic epithelial tight junction, activation of inflammatory mediators, and impairment of Nrf2 pathway. Treatment with an Nrf2 activator restored Nrf2 activity, attenuated colonic inflammation, and restored epithelial tight junction proteins.


Subject(s)
Colitis/etiology , Colon/metabolism , Intestinal Mucosa/metabolism , NF-E2-Related Factor 2/metabolism , Renal Insufficiency, Chronic/complications , Uremia/etiology , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers/metabolism , Case-Control Studies , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , Colitis/physiopathology , Colon/drug effects , Colon/physiopathology , Disease Models, Animal , Inflammation Mediators/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Male , NF-E2-Related Factor 2/agonists , Nephrectomy , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/pharmacology , Oxidative Stress , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Signal Transduction , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Uremia/metabolism , Uremia/physiopathology
13.
Am J Physiol Gastrointest Liver Physiol ; 308(4): G262-8, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25524062

ABSTRACT

The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates various cellular activities, including redox balance, detoxification, metabolism, autophagy, proliferation, and apoptosis. Several studies have demonstrated that Nrf2 regulates hepatocyte proliferation during liver regeneration. The aim of this study was to investigate how Nrf2 modulates the cell cycle of replicating hepatocytes in regenerating livers. Wild-type and Nrf2 null mice were subjected to 2/3 partial hepatectomy (PH) and killed at multiple time points for various analyses. Nrf2 null mice exhibited delayed liver regrowth, although the lost liver mass was eventually restored 7 days after PH. Nrf2 deficiency did not affect the number of hepatocytes entering the cell cycle but did delay hepatocyte mitosis. Mechanistically, the lack of Nrf2 resulted in increased mRNA and protein levels of hepatic cyclin A2 when the remaining hepatocytes were replicating in response to PH. Moreover, Nrf2 deficiency in regenerating livers caused dysregulation of Wee1, Cdc2, and cyclin B1 mRNA and protein expression, leading to decreased Cdc2 activity. Thus, Nrf2 is required for timely M phase entry of replicating hepatocytes by ensuring proper regulation of cyclin A2 and the Wee1/Cdc2/cyclin B1 pathway during liver regeneration.


Subject(s)
Cell Division , Hepatocytes/metabolism , Liver Regeneration , Liver/metabolism , Mitosis , NF-E2-Related Factor 2/metabolism , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin A2/genetics , Cyclin A2/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Gene Expression Regulation , Hepatectomy , Hepatocytes/pathology , Kinetics , Liver/pathology , Liver/surgery , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism
14.
PLoS One ; 9(9): e107423, 2014.
Article in English | MEDLINE | ID: mdl-25222179

ABSTRACT

Nrf2, a central regulator of the cellular defense against oxidative stress and inflammation, participates in modulating hepatocyte proliferation during liver regeneration. It is not clear, however, whether Nrf2 regulates hepatocyte growth, an important cellular mechanism to regain the lost liver mass after partial hepatectomy (PH). To determine this, various analyses were performed in wild-type and Nrf2-null mice following PH. We found that, at 60 h post-PH, the vast majority of hepatocytes lacking Nrf2 reduced their sizes, activated hepatic progenitor markers (CD133, TWEAK receptor, and trefoil factor family 3), depleted HNF4α protein, and downregulated the expression of a group of genes critical for their functions. Thus, the identity of hepatocytes deficient in Nrf2 was transiently but massively impaired in response to liver mass loss. This event was associated with the coupling of protein depletion of hepatic HNF4α, a master regulator of hepatocyte differentiation, and concomitant inactivation of hepatic Akt1 and p70S6K, critical hepatocyte growth signaling molecules. We conclude that Nrf2 participates in maintaining newly regenerated hepatocytes in a fully differentiated state by ensuring proper regulation of HNF4α, Akt1, and p70S6K during liver regeneration.


Subject(s)
Hepatocytes/metabolism , Liver Regeneration/physiology , Liver/metabolism , Liver/physiology , NF-E2-Related Factor 2/metabolism , Animals , Blotting, Western , Immunohistochemistry , Liver Regeneration/genetics , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
Free Radic Biol Med ; 72: 134-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24746615

ABSTRACT

Age-related cataracts are a leading cause of blindness. Previously, we have demonstrated the association of the unfolded protein response with various cataractogenic stressors. However, DNA methylation alterations leading to suppression of lenticular antioxidant protection remains unclear. Here, we report the methylglyoxal-mediated sequential events responsible for Keap1 promoter DNA demethylation in human lens epithelial cells, because Keap1 is a negative regulatory protein that regulates the Nrf2 antioxidant protein. Methylglyoxal induces endoplasmic reticulum stress and activates the unfolded protein response leading to overproduction of reactive oxygen species before human lens epithelial cell death. Methylglyoxal also suppresses Nrf2 and DNA methyltransferases but activates the DNA demethylation pathway enzyme TET1. Bisulfite genomic DNA sequencing confirms the methylglyoxal-mediated Keap1 promoter DNA demethylation leading to overexpression of Keap1 mRNA and protein. Similarly, bisulfite genomic DNA sequencing shows that human clear lenses (n = 15) slowly lose 5-methylcytosine in the Keap1 promoter throughout life, at a rate of 1% per year. By contrast, diabetic cataractous lenses (n = 21) lose an average of 90% of the 5-methylcytosine regardless of age. Overexpressed Keap1 protein is responsible for decreasing Nrf2 by proteasomal degradation, thereby suppressing Nrf2-dependent stress protection. This study demonstrates for the first time the associations of unfolded protein response activation, Nrf2-dependent antioxidant system failure, and loss of Keap1 promoter methylation because of altered active and passive DNA demethylation pathway enzymes in human lens epithelial cells by methylglyoxal. As an outcome, the cellular redox balance is altered toward lens oxidation and cataract formation.


Subject(s)
Cataract/metabolism , DNA Methylation/physiology , Endoplasmic Reticulum Stress/physiology , Intracellular Signaling Peptides and Proteins/genetics , Lens, Crystalline/metabolism , Promoter Regions, Genetic , Pyruvaldehyde/metabolism , Aging/genetics , Aging/metabolism , Animals , Blotting, Western , Cataract/genetics , Cells, Cultured , DNA Methylation/drug effects , Diabetes Mellitus, Experimental , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Lens, Crystalline/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyruvaldehyde/pharmacology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Unfolded Protein Response/drug effects
16.
Free Radic Biol Med ; 65: 1134-1142, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24029383

ABSTRACT

Nrf2 is a transcription factor that protects against inflammatory diseases, but the underlying mechanism of this effect remains unclear. Here, we report that Nrf2 uses lipocalin-prostaglandin D synthase (L-PGDS) as a mechanism for suppressing inflammation. Exogenously added prostaglandin D2 (PGD2) induced L-PGDS expression in bone-marrow-derived macrophages (BMDMs), suggesting a positive feedback loop between L-PGDS expression and PGD2. Unlike lipopolysaccharide (LPS)-induced L-PGDS expression, PGD2-mediated expression was independent of MAPK, PU.1, or TLR4. Sequence analysis located a putative Nrf2 binding site in the murine L-PGDS promoter, to which Nrf2 bound when treated with PGD2. Chemical activation, or overexpression, of Nrf2 was sufficient to induce L-PGDS expression in macrophages, BMDMs, or lungs of Nrf2-knockout (KO) mice, but treatment with PGD2 failed to do so, suggesting a pivotal role for Nrf2 in the expression of L-PGDS. Consistent with this, expression of Nrf2 in the lungs of Nrf2-KO mice was sufficient to induce the expression of L-PGDS and to reduce neutrophilic lung inflammation elicited by LPS. Furthermore, expression of L-PGDS in mouse lungs decreased neutrophilic infiltration, ameliorating lung inflammation in mice. Together, our results show that Nrf2, activated by PGD2, induced L-PGDS expression, resulting in decreased inflammation. We suggest that the positive feedback induction of L-PGDS by PGD2 is part of the mechanism by which Nrf2 regulates inflammation.


Subject(s)
Intramolecular Oxidoreductases/biosynthesis , Lipocalins/biosynthesis , NF-E2-Related Factor 2/metabolism , Neutrophil Infiltration/immunology , Pneumonia/immunology , Prostaglandin D2/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Cyclooxygenase 2/genetics , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Macrophages/enzymology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Neutrophils/immunology , Pneumonia/genetics , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering , Sequence Analysis, DNA , Toll-Like Receptor 4/genetics
17.
FEBS J ; 280(15): 3609-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23702335

ABSTRACT

The ubiquitin-proteasome system is important in maintaining protein homeostasis. NFE2-related factor 1 (Nrf1), a transcription factor in the cap 'n' collar basic-leucine zipper family, regulates expression of cytoprotective genes. It was previously shown that liver-specific knockout of Nrf1 (Nrf1LKO) leads to hepatic cell death, steatohepatitis and cancer. However, the mechanisms underlying these pathologies are not clear. Here, we report that Nrf1 is critical for proteasome gene expression in the liver. Liver-specific knockout of Nrf1 results in impaired basal and induced expression of proteasome genes, and diminished proteasome activity in hepatocytes. In addition, our findings demonstrated that endoplasmic reticulum stress signaling pathway was also activated in Nrf1LKO livers. Inhibition of proteasome activity leads to endoplasmic reticulum stress in Nrf1-deficient hepatocytes, prompting the development of steatosis in the liver. Our results indicate that Nrf1 plays an integral role in the maintenance of proteasome function in hepatocytes and in the prevention of liver steatosis development. Moreover, these results highlight an association between proteasome dysfunction, endoplasmic reticulum stress and steatosis.


Subject(s)
Endoplasmic Reticulum Stress , Fatty Liver/metabolism , Hepatocytes/enzymology , NF-E2-Related Factor 1/physiology , Proteasome Endopeptidase Complex/genetics , Animals , Boronic Acids , Bortezomib , Fatty Liver/chemically induced , Gene Expression , Gene Expression Regulation, Enzymologic , Liver/metabolism , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Pyrazines
18.
Exp Cell Res ; 319(13): 1922-1931, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23623971

ABSTRACT

Nuclear factor E2-related factor-1 (Nrf1) is a basic leucine zipper transcription factor that is known to regulate antioxidant and cytoprotective gene expression. It was recently shown that Nrf1 is regulated by SCF-Fbw7 ubiquitin ligase. However our knowledge of upstream signals that targets Nrf1 for degradation by the UPS is not known. We report here that Nrf1 expression is negatively regulated by glycogen synthase kinase 3 (GSK3) in Fbw7-dependent manner. We show that GSK3 interacts with Nrf1 and phosphorylates the Cdc4 phosphodegron domain (CPD) in Nrf1. Mutation of serine residue in the CPD of Nrf1 to alanine (S350A), blocks Nrf1 from phosphorylation by GSK3, and stabilizes Nrf1. Knockdown of Nrf1 and expression of a constitutively active form of GSK3 results in increased apoptosis in neuronal cells in response to ER stress, while expression of the GSK3 phosphorylation resistant S350A-Nrf1 attenuates apoptotic cell death. Together these data suggest that GSK3 regulates Nrf1 expression and cell survival function in response to stress activation.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Nuclear Respiratory Factor 1/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Cell Survival/genetics , Cells, Cultured , Down-Regulation/genetics , F-Box Proteins/metabolism , F-Box Proteins/physiology , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , Humans , Mice , Nuclear Respiratory Factor 1/antagonists & inhibitors , Nuclear Respiratory Factor 1/metabolism , Protein Binding/physiology , Proteolysis , Stress, Physiological/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology
19.
J Cell Sci ; 126(Pt 7): 1618-25, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23418358

ABSTRACT

Pregnancy induces widespread adaptive responses in maternal organ systems including the liver. The maternal liver exhibits significant growth by increasing the number and size of hepatocytes, by largely unknown mechanisms. Nrf2 mediates cellular defense against oxidative stress and inflammation and also regulates liver regeneration. To determine whether Nrf2 is involved in the regulation of maternal hepatic adaptations to pregnancy, we assessed the proliferation and size of maternal hepatocytes and the associated molecular events in wild-type and Nrf2-null mice at various stages of gestation. We found that wild-type maternal hepatocytes underwent proliferation and size reduction during the first half, and size increase without overt replication during the second half, of pregnancy. Although pregnancy decreased Nrf2 activity in the maternal liver, Nrf2 deficiency caused a delay in maternal hepatocyte proliferation, concomitant with dysregulation of the activation of Cyclin D1, E1, and, more significantly, A2. Remarkably, as a result of Nrf2 absence, the maternal hepatocytes were largely prevented from reducing their sizes during the first half of pregnancy, which was associated with an increase in mTOR activation. During the second half of pregnancy, maternal hepatocytes of both genotypes showed continuous volume increase accompanied by persistent activation of mTOR. However, the lack of Nrf2 resulted in dysregulation of the activation of the mTOR upstream regulator AKT1 and the mTOR target p70SK6 and thus disruption of the AKT1/mTOR/p70S6K pathway, which is known to control cell size. This suggests an mTOR-dependent and AKT1- and p70S6K-independent compensatory mechanism when Nrf2 is deficient. In summary, our study demonstrates that Nrf2 is required for normal maternal hepatic adjustments to pregnancy by ensuring proper regulation of the number and size of maternal hepatocytes.


Subject(s)
Liver/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Blotting, Western , Cyclin A2/metabolism , Cyclin D1/metabolism , Cyclin E/metabolism , Female , Hepatocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Oncogene Proteins/metabolism , Pregnancy
20.
Adv Nutr ; 4(1): 62-6, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23319124

ABSTRACT

Maintenance of a balanced redox state within the cell is of critical importance to a wide variety of biological systems. Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a critical regulator of key aspects of the antioxidant defense pathway and has long been a subject of interest regarding conditions of chronic stress such as inflammation and cancer. Recent data have emerged demonstrating that oxidative stress and Nrf2 also play critical roles in the biology of adipose tissue. This review examines data identifying the roles of Nrf2 and oxidative stress in the biological process of adipose cell differentiation as well as the implications of Nrf2 modulation on obesity. Working to understand the complex interplay among Nrf2, oxidative stress, and adipose biology could lead to a variety of possible treatments for obesity and other related disorders.


Subject(s)
Adipocytes/metabolism , NF-E2-Related Factor 2/metabolism , Obesity/metabolism , Oxidative Stress , Adipogenesis/genetics , Adipose Tissue/metabolism , Animals , Cell Differentiation , Humans , Inflammation , Mice , Mice, Knockout , Models, Animal , NF-E2-Related Factor 2/genetics , Obesity/genetics , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...