Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Oxid Med Cell Longev ; 2022: 2206167, 2022.
Article in English | MEDLINE | ID: mdl-35126808

ABSTRACT

Nrf2 is a critical regulator of the antioxidant defense systems in cellular protection. Emerging evidence has shown that four-octyl itaconate (OI) activates Nrf2 cascade. In this study, the chondroprotective effects of OI on H2O2-stimulated chondrocytes and DMM-induced osteoarthritis (OA) progression were investigated. In primary murine chondrocytes, OI interrupted the binding of Keap1 and Nrf2, leading to accumulation and nuclear translocation of Nrf2 protein, as well as transcription and expression of Nrf2-dependent genes, such as HO-1, NQO1, and GCLC. Furthermore, OI inhibited cell death and apoptosis, as well as H2O2-stimulated ROS generation, lipid peroxidation, superoxide accumulation, and mitochondrial depolarization in chondrocytes, which were abolished by the silence or depletion of Nrf2. In addition, in vivo experiments revealed the therapeutic effects of OI on OA progression in a DMM mouse model. Collectively, these results suggested that OI might serve as a potential treatment for OA progression.


Subject(s)
NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Succinates/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Disease Models, Animal , Hydrogen Peroxide/pharmacology , Kelch-Like ECH-Associated Protein 1/deficiency , Kelch-Like ECH-Associated Protein 1/genetics , Lipid Peroxidation/drug effects , Male , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , NF-E2-Related Factor 2/genetics , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Reactive Oxygen Species/metabolism , Succinates/chemistry , Succinates/therapeutic use
2.
Sci Rep ; 11(1): 5722, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707458

ABSTRACT

Chondrogenesis and angiogenesis drive endochondral ossification. Using the atmospheric scanning electron microscopy (ASEM) without decalcification and dehydration, we directly imaged angiogenesis-driven ossification at different developmental stages shortly after aldehyde fixation, using aqueous radical scavenger glucose solution to preserve water-rich structures. An embryonic day 15.5 mouse femur was fixed and stained with phosphotungstic acid (PTA), and blood vessel penetration into the hypertrophic chondrocyte zone was visualised. We observed a novel envelope between the perichondrium and proliferating chondrocytes, which was lined with spindle-shaped cells that could be borderline chondrocytes. At postnatal day (P)1, trabecular and cortical bone mineralisation was imaged without staining. Additional PTA staining visualised surrounding soft tissues; filamentous connections between osteoblast-like cells and osteocytes in cortical bone were interpreted as the osteocytic lacunar-canalicular system. By P10, resorption pits had formed on the tibial trabecular bone surface. The applicability of ASEM for pathological analysis was addressed using knockout mice of Keap1, an oxidative-stress sensor. In Keap1-/- femurs, we observed impaired calcification and angiogenesis of epiphyseal cartilage, suggesting impaired bone development. Overall, the quick ASEM method we developed revealed mineralisation and new structures in wet bone tissue at EM resolution and can be used to study mineralisation-associated phenomena of any hydrated tissue.


Subject(s)
Atmosphere , Bone and Bones/pathology , Bone and Bones/ultrastructure , Cartilage/ultrastructure , Kelch-Like ECH-Associated Protein 1/deficiency , Microscopy, Electron, Scanning , Osteogenesis , Osteomalacia/pathology , Animals , Bone and Bones/diagnostic imaging , Calcification, Physiologic , Cartilage/diagnostic imaging , Cartilage/pathology , Chondrogenesis , Cortical Bone/diagnostic imaging , Cortical Bone/ultrastructure , Embryo, Mammalian/diagnostic imaging , Femur/diagnostic imaging , Femur/ultrastructure , Imaging, Three-Dimensional , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred C57BL , Osteocytes/metabolism , Phenotype , Tibia/diagnostic imaging , Tibia/ultrastructure
3.
Mol Cell Biol ; 41(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33139492

ABSTRACT

Activating mutations in the KEAP1-NRF2 pathway are found in approximately 25% of lung tumors, where the hijacking of NRF2's cytoprotective functions results in aggressive tumor growth, chemoresistance, and a poor prognosis for patients. There are currently no approved drugs which target aberrant NRF2 activation, which means that there is an urgent clinical need to target this orphan oncogenic pathway in human tumors. In this study, we used an isogenic pair of wild-type and Keap1 knockout cells to screen a range of chemotherapeutic and pathway-targeted anticancer drugs in order to identify compounds which display enhanced toxicity toward cells with high levels of Nrf2 activity. Through this approach, complemented by validation across a panel of eight human cancer cell lines from a range of different tissues, we identified the DNA-damaging agent mitomycin C to be significantly more toxic in cells with aberrant Nrf2 activation. Mechanistically, we found that the NRF2 target genes for cytochrome P450 reductase, NQO1, and enzymes in the pentose phosphate pathway are all responsible for the NRF2-dependent enhanced bioactivation of mitomycin C. As mitomycin C is already approved for clinical use, it represents as excellent drug repositioning candidate to target the currently untreatable NRF2 activation in human tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Kelch-Like ECH-Associated Protein 1/genetics , Mitomycin/pharmacology , NADP/metabolism , NF-E2-Related Factor 2/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/deficiency , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , NADPH-Ferrihemoprotein Reductase/metabolism , NF-E2-Related Factor 2/deficiency , Oxidative Stress , Paclitaxel/pharmacology , Pentose Phosphate Pathway/drug effects , Pentose Phosphate Pathway/genetics , Signal Transduction , Red Fluorescent Protein
4.
Thyroid ; 31(1): 23-35, 2021 01.
Article in English | MEDLINE | ID: mdl-32689903

ABSTRACT

Background: Familial nontoxic multinodular goiter (MNG) is a rare disease. One of the associated genes is Kelch-like ECH-associated protein 1 (KEAP1), which encodes the main inhibitor of nuclear factor erythroid 2-related transcription factor 2 (Nrf2), a central mediator of antioxidant responses. The association of KEAP1 with familial MNG is based on only two loss-of-function mutations identified in two families, only one of which included proper phenotyping and adequate demonstration of co-segregation of the phenotype and the mutation. There is no experimental evidence from model organisms to support that decreased Keap1 levels can, indeed, cause goiter. This study used mice hypomorphic for Keap1 to test whether decreased Keap1 expression can cause goiter, and to characterize the activation status of Nrf2 in their thyroid. Methods: C57BL/6J Keap1flox/flox (Keap1 knock-down [Keap1KD]) mice were studied at 3 and 12 months of age. Plasma and thyroid glands were harvested for evaluation of thyroid function tests and for gene and protein expression by real-time polymerase chain reaction and immunoblotting, respectively. Results: Keap1KD mice showed diffuse goiter that began to develop in early adult life and became highly prominent and penetrant with age. The goiter was characterized by a markedly increased size of thyroid follicles, most notably of the colloid compartment, and by absence of thyroid nodules or hyperplasia. Keap1KD mice also showed decreased T4 levels in early adult life that were eventually well compensated over time by increased thyrotropin (TSH) levels. Nrf2 was activated in the thyroid of Keap1KD mice. Despite a known stimulatory effect of Nrf2 on thyroglobulin (Tg) gene transcription and Tg protein abundance, the expression levels were decreased in the thyroid of Keap1KD mice. No clear patterns were observed in the expression profiles of other thyroid hormone synthesis-specific factors, with the exception of Tg-processing and Tg-degrading cathepsins, including an increase in mature forms of cathepsins D, L, and S. Conclusions: Keap1KD mice develop age-dependent diffuse goiter with elevated TSH levels. The precise mechanism accounting for the thyroidal phenotype remains to be elucidated, but it may involve enhanced Tg solubilization and excessive lysosomal Tg degradation.


Subject(s)
Goiter, Nodular/genetics , Kelch-Like ECH-Associated Protein 1/deficiency , NF-E2-Related Factor 2/metabolism , Thyroid Gland/metabolism , Thyrotropin/blood , Age Factors , Animals , Biomarkers/blood , Genetic Predisposition to Disease , Goiter, Nodular/blood , Goiter, Nodular/pathology , Kelch-Like ECH-Associated Protein 1/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Oxidative Stress , Phenotype , Thyroglobulin/metabolism , Thyroid Gland/pathology , Up-Regulation
5.
Cancer Res ; 80(16): 3331-3344, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32636316

ABSTRACT

The transcription factor Nrf2 activates transcription of cytoprotective genes during oxidative and electrophilic insults. Nrf2 activity is regulated by Keap1 in a stress-dependent manner in normal cells, and somatic loss-of-function mutations of Keap1 are known to induce constitutive Nrf2 activation, especially in lung adenocarcinomas, conferring survival and proliferative benefits to tumors. Therefore, several therapeutic strategies that aim to inhibit Nrf2 in tumors have been developed for the treatment of Nrf2-activated cancers. Here we addressed whether targeting Nrf2 activation in the microenvironment can suppress the progression of Nrf2-activated tumors. We combined two types of Keap1-flox mice expressing variable levels of Keap1 with a Kras-driven adenocarcinoma model to generate Keap1-deficient lung tumors surrounded by normal or Keap1-knockdown host cells. In this model system, activation of Nrf2 in the microenvironment prolonged the survival of Nrf2-activated tumor-bearing mice. The Nrf2-activated microenvironment suppressed tumor burden; in particular, preinvasive lesion formation was significantly suppressed. Notably, loss of Nrf2 in bone marrow-derived cells in Nrf2-activated host cells appeared to counteract the suppression of Nrf2-activated cancer progression. Thus, these results demonstrate that microenvironmental Nrf2 activation suppresses the progression of malignant Nrf2-activated tumors and that Nrf2 activation in immune cells at least partially contributes to these suppressive effects. SIGNIFICANCE: This study clarifies the importance of Nrf2 activation in the tumor microenvironment and in the host for the suppression of malignant Nrf2-activated cancers and proposes new cancer therapies utilizing inducers of Nrf2.


Subject(s)
Adenocarcinoma of Lung/metabolism , Disease Progression , Kelch-Like ECH-Associated Protein 1/deficiency , Lung Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Tumor Microenvironment , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/therapy , Alleles , Animals , Base Sequence , Bone Marrow Transplantation/methods , CD8-Positive T-Lymphocytes , Flow Cytometry , Gene Knockdown Techniques , Gene Silencing , Genes, ras , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Organ Specificity , RNA, Messenger/metabolism , Recombination, Genetic , Stress, Physiological , Transcription, Genetic , Transcriptional Activation , Tumor Burden , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
6.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G419-G427, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31961719

ABSTRACT

The activation of the Kelch-like ECH-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) pathway contributes to cancer progression in addition to oxidative stress responses. Loss-of-function Keap1 mutations were reported to activate Nrf2, leading to cancer progression. We examined the effects of Keap1 deletion in a cholangiocarcinoma mouse model using a mutant K-ras/p53 mouse. Introduction of the Keap1 deletion into liver-specific mutant K-ras/p53 expression resulted in the formation of invasive cholangiocarcinoma. Comprehensive analyses of the gene expression profiles identified broad upregulation of Nrf2-target genes such as Nqo1 and Gstm1 in the Keap1-deleted mutant K-ras/p53 expressing livers, accompanied by upregulation of cholangiocyte-related genes. Among these genes, the transcriptional factor Sox9 was highly expressed in the dysplastic bile duct. The Keap-Nrf2-Sox9 axis might serve as a novel therapeutic target for cholangiocarcinoma.NEW & NOTEWORTHY The Keap1-Nrf2 system has a wide variety of effects in addition to the oxidative stress response in cancer cells. Addition of the liver-specific Keap1 deletion to mice harboring mutant K-ras and p53 accelerated cholangiocarcinoma formation, together with the hallmarks of Nrf2 activation. This process involved the expansion of Sox9-positive cells, indicating increased differentiation toward the cholangiocyte phenotype.


Subject(s)
Bile Duct Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Cholangiocarcinoma/genetics , Gene Deletion , Genes, ras , Kelch-Like ECH-Associated Protein 1/genetics , Mutation , Tumor Suppressor Protein p53/genetics , Animals , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Kelch-Like ECH-Associated Protein 1/deficiency , Male , Mice, Transgenic , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasm Invasiveness , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Time Factors , Transcriptome , Tumor Suppressor Protein p53/metabolism
7.
Cell Res ; 30(2): 146-162, 2020 02.
Article in English | MEDLINE | ID: mdl-31949285

ABSTRACT

Ferroptosis, a form of regulated cell death caused by lipid peroxidation, was recently identified as a natural tumor suppression mechanism. Here, we show that ionizing radiation (IR) induces ferroptosis in cancer cells. Mechanistically, IR induces not only reactive oxygen species (ROS) but also the expression of ACSL4, a lipid metabolism enzyme required for ferroptosis, resulting in elevated lipid peroxidation and ferroptosis. ACSL4 ablation largely abolishes IR-induced ferroptosis and promotes radioresistance. IR also induces the expression of ferroptosis inhibitors, including SLC7A11 and GPX4, as an adaptive response. IR- or KEAP1 deficiency-induced SLC7A11 expression promotes radioresistance through inhibiting ferroptosis. Inactivating SLC7A11 or GPX4 with ferroptosis inducers (FINs) sensitizes radioresistant cancer cells and xenograft tumors to IR. Furthermore, radiotherapy induces ferroptosis in cancer patients, and increased ferroptosis correlates with better response and longer survival to radiotherapy in cancer patients. Our study reveals a previously unrecognized link between IR and ferroptosis and indicates that further exploration of the combination of radiotherapy and FINs in cancer treatment is warranted.


Subject(s)
Ferroptosis/radiation effects , Neoplasms/pathology , Radiation, Ionizing , Amino Acid Transport System y+/metabolism , Animals , Cell Line, Tumor , Coenzyme A Ligases/metabolism , DNA Damage , DNA Repair/radiation effects , Glutathione Peroxidase/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/deficiency , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Inbred NOD , Mice, SCID , Neoplasms/radiotherapy , Neoplasms/ultrastructure , Radiation Tolerance/radiation effects , Up-Regulation/radiation effects
8.
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
9.
Cell Rep ; 27(3): 699-707.e4, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995469

ABSTRACT

Kelch-like ECH-associated protein 1 (Keap1) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) proteins work in concert to regulate the levels of reactive oxygen species (ROS). The Keap1-Nrf2 antioxidant system also participates in T cell differentiation and inflammation, but its role in innate T cell development and functions remains unclear. We report that T cell-specific deletion of Keap1 results in defective development and reduced numbers of invariant natural killer T (NKT) cells in the thymus and the peripheral organs in a cell-intrinsic manner. The frequency of NKT2 and NKT17 cells increases while NKT1 decreases in these mice. Keap1-deficient NKT cells show increased rates of proliferation and apoptosis, as well as increased glucose uptake and mitochondrial function, but reduced ROS, CD122, and Bcl2 expression. In NKT cells deficient in Nrf2 and Keap1, all these phenotypic and metabolic defects are corrected. Thus, the Keap1-Nrf2 system contributes to NKT cell development and homeostasis by regulating cell metabolism.


Subject(s)
Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Natural Killer T-Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Kelch-Like ECH-Associated Protein 1/deficiency , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Thymus Gland/metabolism
11.
PLoS One ; 13(8): e0200004, 2018.
Article in English | MEDLINE | ID: mdl-30114225

ABSTRACT

The transcription factor Nrf2 (encoded by Nfe2l2) induces expression of numerous detoxifying and antioxidant genes in response to oxidative stress. The cytoplasmic protein Keap1 interacts with and represses Nrf2 function. Computational approaches were developed to identify factors that modulate Nrf2 in a mouse liver gene expression compendium. Forty-eight Nrf2 biomarker genes were identified using profiles from the livers of mice in which Nrf2 was activated genetically in Keap1-null mice or chemically by a potent activator of Nrf2 signaling. The rank-based Running Fisher statistical test was used to determine the correlation between the Nrf2 biomarker genes and a test set of 81 profiles with known Nrf2 activation status demonstrating a balanced accuracy of 96%. For a large number of factors examined in the compendium, we found consistent relationships between activation of Nrf2 and feminization of the liver transcriptome through suppression of the male-specific growth hormone (GH)-regulated transcription factor STAT5b. The livers of female mice exhibited higher Nrf2 activation than male mice in untreated or chemical-treated conditions. In male mice, Nrf2 was activated by treatment with ethinyl estradiol, whereas in female mice, Nrf2 was suppressed by treatment with testosterone. Nrf2 was activated in 5 models of disrupted GH signaling containing mutations in Pit1, Prop1, Ghrh, Ghrhr, and Ghr. Out of 59 chemical treatments that activated Nrf2, 36 exhibited STAT5b suppression in the male liver. The Nrf2-STAT5b coupling was absent in in vitro comparisons of chemical treatments. Treatment of male and female mice with 11 chemicals that induce oxidative stress led to activation of Nrf2 to greater extents in females than males. The enhanced basal and inducible levels of Nrf2 activation in females relative to males provides a molecular explanation for the greater resistance often seen in females vs. males to age-dependent diseases and chemical-induced toxicity.


Subject(s)
Liver/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , STAT5 Transcription Factor/metabolism , Animals , Disease Resistance , Female , Hormones/metabolism , Kelch-Like ECH-Associated Protein 1/deficiency , Kelch-Like ECH-Associated Protein 1/genetics , Male , Mice, Transgenic , NF-E2-Related Factor 2/genetics , Oxidants/adverse effects , Sex Characteristics , Transcriptome
12.
Am J Physiol Gastrointest Liver Physiol ; 314(1): G65-G74, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28971839

ABSTRACT

The Kelch-like ECH-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) system has a wide variety of effects in addition to the oxidative stress response, such as growth promotion and chemoresistance of cancer cells. Nrf2 is constitutively activated in most cancer cells. However, the activation of Nrf2 together with oncogenic mutations does not always result in cancer promotion. K-rasLSL-G12D/+:: p53LSL-R172H/+:: Pdx-1-Cre (KPC) mice are an established model of pancreatic cancer that specifically express mutants of both K-ras and p53 in the pancreas by using Pdx-1-Cre. We here generated Pdx-1-Cre::K-rasLSL-G12D/+:: Keap1fl/fl (KC::Keap1) and KPC:: Keap1fl/fl (KPC::Keap1) mice in which Nrf2 is constitutively activated by Keap1 deletion. KC::Keap1 and KPC::Keap1 mice started to die or showed obvious weakness at approximately around 40 days after birth. Histological examination revealed that KC::Keap1 and KPC::Keap1 mice did not develop pancreatic cancer but, instead, progressive atrophy of the pancreatic parenchyma. In these mice, amylase-positive acinar cells as well as insulin- and glucagon-positive islet cells were decreased and surrounded by fibrotic tissues. KC::Keap1 and KPC::Keap1 mice presented lower body weight and glucose levels than C::Keap1 mice, presumably resulting from pancreatic exocrine insufficiency. Histological changes were not obvious in C::Keap1 and PC::Keap1 mice. The presence of the p53 mutation did not affect the phenotypes in KC::Keap1 mice. Heterologous or homologous Nrf2 deletion ( Nrf2+/- or Nrf2-/-) rescued the pancreatic phenotypes, weight loss, and hypoglycemia in KC::Keap1 mice, suggesting that Nrf2 is a major downstream target of Keap1. In conclusion, simultaneous K-ras activation and Keap1 deletion caused progressive atrophy of the pancreatic parenchyma in mice. NEW & NOTEWORTHY Aberrant activation of the Kelch-like ECH-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) system usually promotes carcinogenesis, and we assumed that simultaneous activation of K-ras and Nrf2 might promote pancreatic carcinogenesis. Conditional expression of mutant K-ras and Keap1 deletion did not result in pancreatic cancer development. Instead, these mice developed progressive loss of pancreatic parenchyma, accompanied by body weight loss and hypoglycemia, presumably because of pancreatic exocrine insufficiency. Nrf2 activation by Keap1 deletion concomitant with K-ras activation cause pancreatic atrophy.


Subject(s)
Gene Deletion , Genes, ras , Kelch-Like ECH-Associated Protein 1/deficiency , Pancreas/metabolism , Pancreatitis, Chronic/metabolism , Animals , Atrophy , Blood Glucose/metabolism , Disease Models, Animal , Female , Genes, p53 , Genetic Predisposition to Disease , Glucagon/blood , Homeodomain Proteins/genetics , Insulin/blood , Integrases/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Male , Mice, Knockout , Mutation , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Pancreas/pathology , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/pathology , Parenchymal Tissue/metabolism , Parenchymal Tissue/pathology , Phenotype , Trans-Activators/genetics
13.
Stem Cell Res ; 19: 52-54, 2017 03.
Article in English | MEDLINE | ID: mdl-28413007

ABSTRACT

Kelch-like ECH-associated protein 1 (keap1) is a cysteine-rich protein that interacts with transcription factor Nrf2 in a redox-sensitive manner, leading to the degradation of Nrf2 (Kim et al., 2014a). Disruption of Keap1 results in the induction of Nrf2-related signaling pathways involving the expression of a set of anti-oxidant and anti-inflammatory genes. We generated biallelic mutants of the Keap1 gene using a CRISPR-Cas9 genome editing method in the H9 human embryonic stem cell (hESC). The Keap1 homozygous-knockout H9 cell line retained normal morphology, gene expression, and in vivo differentiation potential.


Subject(s)
CRISPR-Cas Systems/genetics , Human Embryonic Stem Cells/cytology , Kelch-Like ECH-Associated Protein 1/genetics , Base Sequence , Cell Line , Gene Knockout Techniques , Homozygote , Human Embryonic Stem Cells/metabolism , Humans , Karyotype , Kelch-Like ECH-Associated Protein 1/deficiency , Microscopy, Fluorescence
14.
BMC Nephrol ; 17(1): 110, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27484495

ABSTRACT

BACKGROUND: Transcription factor Nrf2 protects from experimental acute kidney injury (AKI) and is promising to limit progression in human chronic kidney disease (CKD) by upregulating multiple antioxidant genes. We recently demonstrated that deletion of Keap1, the endogenous inhibitor of Nrf2, in T lymphocytes significantly protects from AKI. In this study, we investigated the effect of Keap1 deletion on Nrf2 mediated antioxidant response in the renal tubular epithelial cells. METHODS: We deleted Keap1 exon 2 and 3 in the renal tubular epithelial cells by crossing Ksp-Cre mice with Keap1 floxed (Keap1 (f/f)) mice. Deletion of Keap1 gene in the kidney epithelial cells of Ksp-Keap1 (-/-) mice and its effect on Nrf2 target gene expression was performed using PCR and real-time PCR respectively. Histological evaluation was performed on H&E stained sections. Complete blood count, serum and urine analysis were performed to assess systemic effects of defective kidney development. Student's T test was used to determine statistical difference between the groups. RESULTS: Ksp-Cre resulted in the deletion of Keap1 exon 2 and 3 and subsequent upregulation of Nrf2 target genes, Nqo1, Gclm and Gclc in the kidney epithelial cells of Ksp-Keap1 (-/-) mice at baseline. Renal epithelial cell specific deletion of Keap1 in Ksp-Keap1 (-/-) mice caused marked renal pelvic expansion and significant compression of medullary parenchyma consistent with hydronephrosis in both (3 month-old) males and females. Kidneys from 6 month-old Ksp-Keap1 (-/-) mice showed progressive hydronephrosis. Hematological, biochemical and urinary analysis showed significantly higher red blood cell count (p = 0.04), hemoglobin (p = 0.01), hematocrit (p = 0.02), mean cell volume (p = 0.02) and mean cell hemoglobin concentration (p = 0.003) in Ksp-Keap1 (-/-) mice in comparison to Keap1 (f/f) mice. CONCLUSIONS: These unexpected findings demonstrate that Keap1 deletion in renal tubular epithelial cells results in an abnormal kidney development consistent with hydronephrosis and reveals a novel Keap1 mediated signaling pathway in renal development.


Subject(s)
Epithelium/metabolism , Hydronephrosis/genetics , Hydronephrosis/metabolism , Kelch-Like ECH-Associated Protein 1/deficiency , Kelch-Like ECH-Associated Protein 1/genetics , Kidney/metabolism , Animals , Gene Deletion , Hydronephrosis/pathology , Kidney/growth & development , Kidney/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...