51
|
Ryu D, Lee JH, Kwak MK. NRF2 level is negatively correlated with TGF-β1-induced lung cancer motility and migration via NOX4-ROS signaling. Arch Pharm Res 2020; 43:1297-1310. [PMID: 33242180 DOI: 10.1007/s12272-020-01298-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 11/22/2020] [Indexed: 12/19/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) is a multifaceted factor in cancer biology that regulates cell proliferation and migration. Overactivation of nuclear factor erythroid 2-like 2 (NFE2L2; NRF2) in cancers has been associated with facilitated tumor growth and therapy resistance; however, role in cancer migration has not been clearly explained yet. In this study, we investigated the role of NRF2 on TGF-β1-induced cell motility/migration. In NRF2-silenced lung cancer A549 cells, both basal and TGF-β1-inducible cell motility/migration increased compared to those in A549. SMAD transcription activity and phosphorylated SMAD2/3 levels were higher in TGF-β1-treated NRF2-low A549 cells than those in A549. Notably, the levels of reactive oxygen species (ROS) that were elevated by TGF-β1 treatment were higher in the NRF2-low A549 than those in control cells, and treatment with ROS scavenger blocked TGF-β1-induced cell motility. As an underlying molecular link, NADPH oxidase 4 (NOX4) was associated with higher ROS elevation and cell motility of NRF2-low A549. NOX4 and TGF-β1-inducible NOX4 levels were higher in NRF2-low A549 cells than those in A549. Moreover, the pharmacological inhibition of NOX4 blocked the TGF-β1-induced motility of NRF2-low A549 cells. Collectively, these results indicate that TGF-β1-induced cell motility/migration is facilitated in NRF2-inhibited lung cancer cells and that high levels of NOX4/ROS are associated with enhanced motility/migration.
Collapse
Affiliation(s)
- Dayoung Ryu
- Department of Pharmacy and BK21FOUR Advanced Program for SmartPharma Leaders, Graduate School of the Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Jin-Hee Lee
- Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea
| | - Mi-Kyoung Kwak
- Department of Pharmacy and BK21FOUR Advanced Program for SmartPharma Leaders, Graduate School of the Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea. .,Integrated Research Institute for Pharmaceutical Sciences, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea. .,College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|
52
|
Tamatam CM, Reddy NM, Potteti HR, Ankireddy A, Noone PM, Yamamoto M, Kensler TW, Reddy SP. Preconditioning the immature lung with enhanced Nrf2 activity protects against oxidant-induced hypoalveolarization in mice. Sci Rep 2020; 10:19034. [PMID: 33149211 PMCID: PMC7642393 DOI: 10.1038/s41598-020-75834-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic disease of preterm babies with poor clinical outcomes. Nrf2 transcription factor is crucial for cytoprotective response, whereas Keap1—an endogenous inhibitor of Nrf2 signaling—dampens these protective responses. Nrf2-sufficient (wild type) newborn mice exposed to hyperoxia develop hypoalveolarization, which phenocopies human BPD, and Nrf2 deficiency worsens it. In this study, we used PND1 pups bearing bearing hypomorphic Keap1 floxed alleles (Keap1f/f) with increased levels of Nrf2 to test the hypothesis that constitutive levels of Nrf2 in the premature lung are insufficient to mitigate hyperoxia-induced hypoalveolarization. Both wildtype and Keap1f/f pups at PND1 were exposed to hyperoxia for 72 h and then allowed to recover at room air for two weeks (at PND18), sacrificed, and lung hypoalveolarization and inflammation assessed. Hyperoxia-induced lung hypoalveolarization was remarkably lower in Keap1f/f pups than in wildtype counterparts (28.9% vs 2.4%, wildtype vs Keap1f/f). Likewise, Keap1f/f pups were protected against prolonged (96 h) hyperoxia-induced hypoalveolarization. However, there were no differences in hyperoxia-induced lung inflammatory response immediately after exposure or at PND18. Lack of hypoalveolarization in Keap1f/f pups was accompanied by increased levels of expression of antioxidant genes and GSH as assessed immediately following hyperoxia. Keap1 knockdown resulted in upregulation of lung cell proliferation postnatally but had opposing effects following hyperoxia. Collectively, our study demonstrates that augmenting endogenous Nrf2 activation by targeting Keap1 may provide a physiological way to prevent hypoalveolarization associated with prematurity.
Collapse
Affiliation(s)
- Chandra M Tamatam
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Narsa M Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, 60612, USA
| | - Haranatha R Potteti
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Aparna Ankireddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Patrick M Noone
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University, Sendai, Japan
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Sekhar P Reddy
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
53
|
He F, Antonucci L, Karin M. NRF2 as a regulator of cell metabolism and inflammation in cancer. Carcinogenesis 2020; 41:405-416. [PMID: 32347301 DOI: 10.1093/carcin/bgaa039] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/11/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a master transcriptional regulator of genes whose products defend our cells for toxic and oxidative insults. Although NRF2 activation may reduce cancer risk by suppressing oxidative stress and tumor-promoting inflammation, many cancers exhibit elevated NRF2 activity either due to mutations that disrupt the negative control of NRF2 activity or other factors. Importantly, NRF2 activation is associated with poor prognosis and NRF2 has turned out to be a key activator of cancer-supportive anabolic metabolism. In this review, we summarize the diverse roles played by NRF2 in cancer focusing on metabolic reprogramming and tumor-promoting inflammation.
Collapse
Affiliation(s)
- Feng He
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, San Diego, La Jolla, CA, USA
| | - Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, San Diego, La Jolla, CA, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, San Diego, La Jolla, CA, USA.,Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
54
|
Sun Q, Ye Z, Qin Y, Fan G, Ji S, Zhuo Q, Xu W, Liu W, Hu Q, Liu M, Zhang Z, Xu X, Yu X. Oncogenic function of TRIM2 in pancreatic cancer by activating ROS-related NRF2/ITGB7/FAK axis. Oncogene 2020; 39:6572-6588. [PMID: 32929153 DOI: 10.1038/s41388-020-01452-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/29/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Evidence suggests that tripartite motif-containing 2 (TRIM2) is associated with carcinogenic effects in several malignancies. However, the expression patterns and roles of TRIM2 in pancreatic cancer are rarely studied. Our study demonstrated that TRIM2 was expressed in a high percentage of pancreatic tumors. High TRIM2 expression was negatively correlated with the outcome of pancreatic cancer. TRIM2 silencing significantly inhibited the proliferation, migration, invasion, and in vivo tumorigenicity of pancreatic cancer cells. Regarding the mechanism involved, TRIM2 activated ROS-related E2-related factor 2 (NRF2)/antioxidant response element (ARE) signaling and the integrin/focal adhesion kinase (FAK) pathway. Treatment of pancreatic cancer cells with the antioxidant N-acetyl-L-cysteine decreased ROS activity and expression level of NRF2 and ITGB7. Increased translocation of NRF2 protein into nucleus further rescued the inhibited ITGB7 transcription. Moreover, NRF2 bound to the potential ARE on the promoter region and enhanced the transcriptional activity of ITGB7, indicating the bridging effect of NRF2 between the two signaling pathways. In summary, our study provides evidence that upregulated TRIM2 in pancreatic cancer predicts short survival for pancreatic cancer patients. TRIM2 accelerates pancreatic cancer progression via the ROS-related NRF2/ITGB7/FAK axis.
Collapse
Affiliation(s)
- Qiqing Sun
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Qifeng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Mengqi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 200032, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, 200032, Shanghai, China.
- Pancreatic Cancer Institute, Fudan University, 200032, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, 200032, Shanghai, China.
| |
Collapse
|
55
|
Constitutive Activation of Nrf2 in Mice Expands Enterogenesis in Small Intestine Through Negative Regulation of Math1. Cell Mol Gastroenterol Hepatol 2020; 11:503-524. [PMID: 32896624 PMCID: PMC7797379 DOI: 10.1016/j.jcmgh.2020.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Notch signaling coordinates cell differentiation processes in the intestinal epithelium. The transcription factor Nrf2 orchestrates defense mechanisms by regulating cellular redox homeostasis, which, as shown previously in murine liver, can be amplified through signaling crosstalk with the Notch pathway. However, interplay between these 2 signaling pathways in the gut is unknown. METHODS Mice modified genetically to amplify Nrf2 in the intestinal epithelium (Keap1f/f::VilCre) were generated as well as pharmacological activation of Nrf2 and subjected to phenotypic and cell lineage analyses. Cell lines were used for reporter gene assays together with Nrf2 overexpression to study transcriptional regulation of the Notch downstream effector. RESULTS Constitutive activation of Nrf2 signaling caused increased intestinal length along with expanded cell number and thickness of enterocytes without any alterations of secretory lineage, outcomes abrogated by concomitant disruption of Nrf2. The Nrf2 and Notch pathways in epithelium showed inverse spatial profiles, where Nrf2 activity in crypts was lower than villi. In progenitor cells of Keap1f/f::VilCre mice, Notch downstream effector Math1, which regulates a differentiation balance of cell lineage through lateral inhibition, showed suppressed expression. In vitro results demonstrated Nrf2 negatively regulated Math1, where 6 antioxidant response elements located in the regulatory regions contributed to this repression. CONCLUSIONS Activation of Nrf2 perturbed the dialog of the Notch cascade though negative regulation of Math1 in progenitor cells, leading to enhanced enterogenesis. The crosstalk between the Nrf2 and Notch pathways could be critical for fine-tuning intestinal homeostasis and point to new approaches for the pharmacological management of absorptive deficiencies.
Collapse
|
56
|
Wang F, Hou W, Chitsike L, Xu Y, Bettler C, Perera A, Bank T, Cotler SJ, Dhanarajan A, Denning MF, Ding X, Breslin P, Qiang W, Li J, Koleske AJ, Qiu W. ABL1, Overexpressed in Hepatocellular Carcinomas, Regulates Expression of NOTCH1 and Promotes Development of Liver Tumors in Mice. Gastroenterology 2020; 159:289-305.e16. [PMID: 32171747 PMCID: PMC7387191 DOI: 10.1053/j.gastro.2020.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 01/31/2020] [Accepted: 03/04/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS We investigated whether ABL proto-oncogene 1, non-receptor tyrosine kinase (ABL1) is involved in development of hepatocellular carcinoma (HCC). METHODS We analyzed clinical and gene expression data from The Cancer Genome Atlas. Albumin-Cre (HepWT) mice and mice with hepatocyte-specific disruption of Abl1 (HepAbl-/- mice) were given hydrodynamic injections of plasmids encoding the Sleeping Beauty transposase and transposons with the MET gene and a catenin β1 gene with an N-terminal truncation, which induces development of liver tumors. Some mice were then gavaged with the ABL1 inhibitor nilotinib or vehicle (control) daily for 4 weeks. We knocked down ABL1 with short hairpin RNAs in Hep3B and Huh7 HCC cells and analyzed their proliferation and growth as xenograft tumors in mice. We performed RNA sequencing and gene set enrichment analysis of tumors. We knocked down or overexpressed NOTCH1 and MYC in HCC cells and analyzed proliferation. We measured levels of phosphorylated ABL1, MYC, and NOTCH1 by immunohistochemical analysis of an HCC tissue microarray. RESULTS HCC tissues had higher levels of ABL1 than non-tumor liver tissues, which correlated with shorter survival times of patients. HepWT mice with the MET and catenin β1 transposons developed liver tumors and survived a median 64 days; HepAbl-/- mice with these transposons developed tumors that were 50% smaller and survived a median 81 days. Knockdown of ABL1 in human HCC cells reduced proliferation, growth as xenograft tumors in mice, and expression of MYC, which reduced expression of NOTCH1. Knockdown of NOTCH1 or MYC in HCC cells significantly reduced cell growth. NOTCH1 or MYC overexpression in human HCC cells promoted proliferation and rescued the phenotype caused by ABL1 knockdown. The level of phosphorylated (activated) ABL1 correlated with levels of MYC and NOTCH1 in human HCC specimens. Nilotinib decreased expression of MYC and NOTCH1 in HCC cell lines, reduced the growth of xenograft tumors in mice, and slowed growth of liver tumors in mice with MET and catenin β1 transposons, reducing tumor levels of MYC and NOTCH1. CONCLUSIONS HCC samples have increased levels of ABL1 compared with nontumor liver tissues, and increased levels of ABL1 correlate with shorter survival times of patients. Loss or inhibition of ABL1 reduces proliferation of HCC cells and slows growth of liver tumors in mice. Inhibitors of ABL1 might be used for treatment of HCC.
Collapse
Affiliation(s)
- Fang Wang
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Wei Hou
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Lennox Chitsike
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Yingchen Xu
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University
| | - Carlee Bettler
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Aldeb Perera
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Thomas Bank
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Scott J. Cotler
- Department of Medicine, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Asha Dhanarajan
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Mitchell F. Denning
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Peter Breslin
- Departments of Molecular/Cellular Physiology and Oncology Institute, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA,Department of Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Wenan Qiang
- Department of Obstetrics and Gynecology and Pathology, Northwestern University
| | - Jun Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame
| | | | - Wei Qiu
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois.
| |
Collapse
|
57
|
Vargas-Mendoza N, Morales-González Á, Morales-Martínez M, Soriano-Ursúa MA, Delgado-Olivares L, Sandoval-Gallegos EM, Madrigal-Bujaidar E, Álvarez-González I, Madrigal-Santillán E, Morales-Gonzalez JA. Flavolignans from Silymarin as Nrf2 Bioactivators and Their Therapeutic Applications. Biomedicines 2020; 8:122. [PMID: 32423098 PMCID: PMC7277158 DOI: 10.3390/biomedicines8050122] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Silymarin (SM) is a mixture of flavolignans extracted from the seeds of species derived from Silybum marianum, commonly known as milk thistle or St. Mary'sthistle. These species have been widely used in the treatment of liver disorders in traditional medicine since ancient times. Several properties had been attributed to the major SM flavolignans components, identified as silybin, isosilybin, silychristin, isosilychristin, and silydianin. Previous research reported antioxidant and protective activities, which are probably related to the activation of the nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2), known as a master regulator of the cytoprotector response. Nrf2 is a redox-sensitive nuclear transcription factor able to induce the downstream-associated genes. The disruption of Nrf2 signaling has been associated with different pathological conditions. Some identified phytochemicals from SM had shown to participate in the Nrf2 signaling pathway; in particular, they have been suggested as activators that disrupt interactions in the Keap1-Nrf2 system, but also as antioxidants or with additional actions regarding Nrf2 regulation. Thus, the study of these molecules makes them appear attractive as novel targets for the treatment or prevention of several diseases.
Collapse
Affiliation(s)
- Nancy Vargas-Mendoza
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México Escuela Superior de Medicina, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Ángel Morales-González
- Escuela Superior de Cómputo, Instituto Politécnico Nacional, Av. Juan de Dios Bátiz s/n esquina Miguel Othón de Mendizabal, Unidad Profesional Adolfo López Mateos, Mexico City CP 07738, Mexico;
| | - Mauricio Morales-Martínez
- Licenciatura en Nutrición, Universidad Intercontinental, Insurgentes Sur 4303, Santa Úrsula Xitla, Alcaldía Tlalpan, Mexico City CP 14420, Mexico;
| | - Marvin A. Soriano-Ursúa
- Academia de Fisiología Humana, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Del. Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Luis Delgado-Olivares
- Centro de Investigación Interdisciplinario, Área Académica de Nutrición, Instituto de Ciencias de la Salud. Universidad Autónoma del Estado de Hidalgo. Circuito Actopan-Tilcuauttla, s/n, Ex hacienda La Concepción, San Agustín Tlaxiaca, Hidalgo CP 42160, Mexico; (L.D.-O.); (E.M.S.-G.)
| | - Eli Mireya Sandoval-Gallegos
- Centro de Investigación Interdisciplinario, Área Académica de Nutrición, Instituto de Ciencias de la Salud. Universidad Autónoma del Estado de Hidalgo. Circuito Actopan-Tilcuauttla, s/n, Ex hacienda La Concepción, San Agustín Tlaxiaca, Hidalgo CP 42160, Mexico; (L.D.-O.); (E.M.S.-G.)
| | - Eduardo Madrigal-Bujaidar
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, “Unidad Profesional A. López Mateos”. Av. Wilfrido Massieu. Col., Zacatenco, Mexico City 07738, Mexico; (E.M.-B.); (I.Á.-G.)
| | - Isela Álvarez-González
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, “Unidad Profesional A. López Mateos”. Av. Wilfrido Massieu. Col., Zacatenco, Mexico City 07738, Mexico; (E.M.-B.); (I.Á.-G.)
| | - Eduardo Madrigal-Santillán
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México Escuela Superior de Medicina, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico;
| | - José A. Morales-Gonzalez
- Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México Escuela Superior de Medicina, Plan de San Luis y Díaz Mirón, Col. Casco de Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico;
| |
Collapse
|
58
|
Fragoulis A, Schenkel J, Herzog M, Schellenberg T, Jahr H, Pufe T, Trautwein C, Kensler TW, Streetz KL, Wruck CJ. Nrf2 Ameliorates DDC-Induced Sclerosing Cholangitis and Biliary Fibrosis and Improves the Regenerative Capacity of the Liver. Toxicol Sci 2020; 169:485-498. [PMID: 30825315 DOI: 10.1093/toxsci/kfz055] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The Nrf2 pathway protects against oxidative stress and induces regeneration of various tissues. Here, we investigated whether Nrf2 protects from sclerosing cholangitis and biliary fibrosis and simultaneously induces liver regeneration. Diet containing 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) was fed to Nrf2-KO mice (Nrf2-/-), mice with liver-specific hyperactivated Nrf2 (HKeap1-/-) and wild-type (WT) littermates to induce cholangitis, liver fibrosis, and oval cell expansion. HKeap1-/--mice were protected from almost all DDC-induced injury compared with WT and Nrf2-/-. Liver injury in Nrf2-/- and WT mice was mostly similar, albeit Nrf2-/- suffered more from DDC diet as seen for several parameters. Nrf2 activity was especially important for the expression of the hepatic efflux transporters Abcg2 and Abcc2-4, which are involved in hepatic toxin elimination. Surprisingly, cell proliferation was more enhanced in Nrf2-/-- and HKeap1-/--mice compared with WT. Interestingly, Nrf2-/--mice failed to sufficiently activate oval cell expansion after DDC treatment and showed almost no resident oval cell population under control conditions. The resident oval cell population of untreated HKeap1-/--mice was increased and DDC treatment resulted in a stronger oval cell expansion compared with WT. We provide evidence that Nrf2 activation protects from DDC-induced sclerosing cholangitis and biliary fibrosis. Moreover, our data establish a possible role of Nrf2 in oval cell expansion.
Collapse
Affiliation(s)
- Athanassios Fragoulis
- Department of Anatomy and Cell Biology.,Molecular Tumor Biology, Department of General, Visceral and Transplantation Surgery
| | | | | | | | | | | | - Christian Trautwein
- Department of Medicine III, Medical Faculty, Uniklinik RWTH Aachen University, 52074 Aachen, Germany
| | - Thomas W Kensler
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205
| | - Konrad L Streetz
- Department of Medicine III, Medical Faculty, Uniklinik RWTH Aachen University, 52074 Aachen, Germany
| | | |
Collapse
|
59
|
Bi Z, Zhang Q, Fu Y, Wadgaonkar P, Zhang W, Almutairy B, Xu L, Rice M, Qiu Y, Thakur C, Chen F. Nrf2 and HIF1α converge to arsenic-induced metabolic reprogramming and the formation of the cancer stem-like cells. Theranostics 2020; 10:4134-4149. [PMID: 32226544 PMCID: PMC7086359 DOI: 10.7150/thno.42903] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/06/2020] [Indexed: 01/02/2023] Open
Abstract
In this report, we demonstrated that inorganic arsenic (iAs) induces generation of the cancer stem-like cells (CSCs) through Nrf2-dependent HIF1α activation, and the subsequent metabolic reprogramming from mitochondrial oxidative phosphorylation to glycolysis in epithelial cells. Methods: Genome-wide ChIP-seq analysis was performed to investigate the global binding of Nrf2 and/or HIF1α on the genome in the cells treated with iAs. Both untargeted metabolomics and UDP-13C-glucose flux were applied to determine metabolic reprogramming in the iAs-induced CSCs. The role of Nrf2 on iAs-induced HIF1α and other stemness gene expression was validated by lentiviral transfection of Nrf2 inhibitor Keap1 and CRISPR-Cas9-mediated Nrf2 gene knockout, respectively. Results: The CSCs induced by iAs exhibit a diminished mitochondrial oxidative phosphorylation and an enhanced glycolysis that is actively shunted to the hexosamine biosynthetic pathway (HBP) and serine/glycine pathway. ChIP-seq data revealed that treatment of the cells with iAs amplified Nrf2 enrichment peaks in intergenic region, promoter and gene body. In contrast, a shift of the HIF1α peaks from distal intergenic region to gene promoter and the first exon was noted. Both Nrf2 and HIF1α are responsible for the iAs-induced expression of the glycolytic genes and the genes important for the stemness of the CSCs. Intriguingly, we also discovered a mutual transcriptional regulation between Nrf2 and HIF1α. Inhibition of Nrf2 by lentiviral infection of Keap1, or knockout of Nrf2 by CRISPR-Cas9 gene editing, not only blocked iAs-induced HIF1α activation, but reduced the expression of the key stemness genes for the formation of CSCs also. Conclusion: We demonstrated that Nrf2 activation is an initiating signal for iAs-induced HIF1α activation, and Nrf2 and HIF1α played a concerted role on inducing metabolic reprogramming and the CSCs.
Collapse
Affiliation(s)
- Zhuoyue Bi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
- School of Health Sciences, Wuhan University, 115 Donghu Road, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, 8 Zhudaoquanbei Road, Wuhan 430079, China
| | - Qian Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Yao Fu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Priya Wadgaonkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Wenxuan Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Bandar Almutairy
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
- College of Pharmacy, Al-Dawadmi Campus, Shaqra University, P.O.Box 11961, Riyadh, Kingdom of Saudi Arabia
| | - Liping Xu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - M'Kya Rice
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Yiran Qiu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Chitra Thakur
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Fei Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| |
Collapse
|
60
|
Zhou XL, Wu X, Zhu RR, Xu H, Li YY, Xu QR, Liu S, Lai SQ, Xu X, Wan L, Wu QC, Liu JC. Notch1-Nrf2 signaling crosstalk provides myocardial protection by reducing ROS formation. Biochem Cell Biol 2020; 98:106-111. [PMID: 32069075 DOI: 10.1139/bcb-2018-0398] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Both the Notch1 and Keap1-Nrf2 signaling pathways have cardioprotective effects, but the role of Notch1-Nrf2 crosstalk in myocardial ischemia-reperfusion injury is unclear. In this study, we established hypoxia-reoxygenation in neonate rat myocardial cells and employed γ-secretase inhibitor and curcumin to inhibit and activate the Notch1 and Keap1-Nrf2 signaling pathways, respectively. We found that the combined action of the Notch1 and Keap1-Nrf2 signaling pathways significantly increased cardiomyocyte viability, inhibited cardiomyocyte apoptosis, reduced the formation of reactive oxygen species, and increased antioxidant activities. In conclusion, these findings suggest that Notch1-Nrf2 crosstalk exerts myocardial protection by reducing the formation of reactive oxygen species.
Collapse
Affiliation(s)
- Xue-Liang Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Xia Wu
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Rong-Rong Zhu
- Department of Obstetrics and Gynecology, Jiangxi Province hospital of integrated traditional Chinese and Western medicine, Nanchang, China
| | - Hua Xu
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Yun-Yun Li
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Qi-Rong Xu
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Sheng Liu
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Song-Qing Lai
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Xinping Xu
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Li Wan
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Qi-Cai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Ji-Chun Liu
- Department of Cardiac Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, China
| |
Collapse
|
61
|
Ostrand-Rosenberg S, Beury DW, Parker KH, Horn LA. Survival of the fittest: how myeloid-derived suppressor cells survive in the inhospitable tumor microenvironment. Cancer Immunol Immunother 2020; 69:215-221. [PMID: 31501954 PMCID: PMC7004852 DOI: 10.1007/s00262-019-02388-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are present in most cancer patients where they are significant contributors to the immune suppressive tumor microenvironment (TME). The TME is a hostile locale due to deficiencies in oxygen (hypoxia) and nutrients, and the presence of reactive oxygen species (ROS). The survival of tumor cells within the TME is partially governed by two mechanisms: (1) Activation of the transcription factor Nuclear Factor Erythroid-derived 2-like 2 (Nrf2) which turns on genes that attenuate oxidative stress; and (2) The presence of High Mobility Group Box Protein-1 (HMGB1), a damage-associated molecular pattern molecule (DAMP) that induces autophagy and protects against apoptosis. Because Nrf2 and HMGB1 promote tumor cell survival, we speculated that Nrf2 and HMGB1 may facilitate MDSC survival. We tested this hypothesis using Nrf2+/+ and Nrf2-/- BALB/c and C57BL/6 mice and pharmacological inhibitors of HMGB1. In vitro and in vivo studies demonstrated that Nrf2 increased the suppressive potency and quantity of tumor-infiltrating MDSC by up-regulating MDSC production of H2O2 and decreasing MDSC apoptosis. Decreased apoptosis was accompanied by a decrease in the production of MDSC, demonstrating that MDSC levels are homeostatically regulated. Pharmacological inhibition of autophagy increased MDSC apoptosis, indicating that autophagy increases MDSC half-life. Inhibition of HMGB1 also increased MDSC apoptosis and reduced MDSC autophagy. These results combined with our previous findings that HMGB1 drives the accumulation of MDSC demonstrate that HMGB1 maintains MDSC viability by inducing autophagy. Collectively, these findings identify Nrf2 and HMGB1 as important factors that enable MDSC to survive in the TME.
Collapse
Affiliation(s)
- Suzanne Ostrand-Rosenberg
- Department of Biological Sciences, University of Maryland Baltimore County (UMBC), Baltimore, MD, USA.
- Department of Pathology, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, 15 North Medical Drive East, Ste. #1100, Salt Lake City, UT, 84112, USA.
| | - Daniel W Beury
- Department of Biological Sciences, University of Maryland Baltimore County (UMBC), Baltimore, MD, USA
| | - Katherine H Parker
- Department of Biological Sciences, University of Maryland Baltimore County (UMBC), Baltimore, MD, USA
| | - Lucas A Horn
- Department of Biological Sciences, University of Maryland Baltimore County (UMBC), Baltimore, MD, USA
| |
Collapse
|
62
|
Zhou Y, Xu X, Wu J, Xu L, Zhang M, Li Z, Wang D. Allyl isothiocyanate treatment alleviates chronic obstructive pulmonary disease through the Nrf2-Notch1 signaling and upregulation of MRP1. Life Sci 2020; 243:117291. [PMID: 31927049 DOI: 10.1016/j.lfs.2020.117291] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/06/2020] [Accepted: 01/06/2020] [Indexed: 12/17/2022]
Abstract
AIMS Chronic obstructive pulmonary disease (COPD) is a disease with high morbidity and mortality worldwide, which can cause serious social and economic burdens. Allyl isothiocyanate (AITC) is one of the most common natural isothiocyanates and has been shown to have anti-inflammatory and antioxidant biological activities. The purpose of this study was to investigate whether AITC regulated Multidrug resistance-associated protein 1 (MRP1), reactive oxide species (ROS) and reduced glutathione (GSH) levels via Nrf2 and Notch1 signaling pathways to treat COPD and whether there was an interaction between these two pathways. MAIN METHODS Lung function indexes and histopathological changes in mice were determined by lung function instrument and HE staining, respectively. The protein expression was analyzed using immunohistochemistry and Western blotting. The mRNA expression was measured by RT-PCR in human bronchial epithelial cell line 16HBE. The contents of ROS, GSH and GSSG were detected by kits in 16HBE cells. KEY FINDINGS The protein expression of Notch1, Hes1, MRP1, Nrf2, and HO-1 in lung tissues of WT mice and untransfected cells were significantly down-regulated in COPD, then significantly ameliorated in treatment groups. The protein expression of MRP1, Notch1 and Hes1 in lung tissues of Nrf2-/- mice were markedly reduced. There was a significant reduction in expression of Nrf2, HO-1 and MRP1 in si-Notch1 transfected cells. Pretreatment with AITC markedly improved oxidative stress and GSH-redox disorder in COPD. SIGNIFICANCE Our study demonstrates that there is a potential interaction between Nrf2 and Notch1 signaling pathways during treatment of COPD.
Collapse
Affiliation(s)
- Yuanyuan Zhou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Xiaoya Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; Department of Pharmacy, Hospital of Armed Police of Anhui Province, Heifei 230061, Anhui, China
| | - Jie Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Lingling Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Min Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
| | - Zegeng Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China.
| | - Dianlei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, Anhui, China.
| |
Collapse
|
63
|
Escoll M, Lastra D, Pajares M, Robledinos-Antón N, Rojo AI, Fernández-Ginés R, Mendiola M, Martínez-Marín V, Esteban I, López-Larrubia P, Gargini R, Cuadrado A. Transcription factor NRF2 uses the Hippo pathway effector TAZ to induce tumorigenesis in glioblastomas. Redox Biol 2020; 30:101425. [PMID: 31918259 PMCID: PMC7016245 DOI: 10.1016/j.redox.2019.101425] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/22/2019] [Accepted: 12/31/2019] [Indexed: 11/29/2022] Open
Abstract
Transcription factor NRF2 orchestrates a cellular defense against oxidative stress and, so far, has been involved in tumor progression by providing a metabolic adaptation to tumorigenic demands and resistance to chemotherapeutics. In this study, we discover that NRF2 also propels tumorigenesis in gliomas and glioblastomas by inducing the expression of the transcriptional co-activator TAZ, a protein of the Hippo signaling pathway that promotes tumor growth. The expression of the genes encoding NRF2 (NFE2L2) and TAZ (WWTR1) showed a positive correlation in 721 gliomas from The Cancer Genome Atlas database. Moreover, NRF2 and TAZ protein levels also correlated in immunohistochemical tissue arrays of glioblastomas. Genetic knock-down of NRF2 decreased, while NRF2 overexpression or chemical activation with sulforaphane, increased TAZ transcript and protein levels. Mechanistically, we identified several NRF2-regulated functional enhancers in the regulatory region of WWTR1. The relevance of the new NRF2/TAZ axis in tumorigenesis was demonstrated in subcutaneous and intracranial grafts. Thus, intracranial inoculation of NRF2-depleted glioma stem cells did not develop tumors as determined by magnetic resonance imaging. Forced TAZ overexpression partly rescued both stem cell growth in neurospheres and tumorigenicity. Hence, NRF2 not only enables tumor cells to be competent to proliferate but it also propels tumorigenesis by activating the TAZ-mediated Hippo transcriptional program. Expression of NRF2 and TAZ positively correlate in gliomas and glioblastomas. NRF2 regulates the expression of WWTR1 encoding the transcription co-activator TAZ in glioma stem cells. TAZ provides a redox-independent mechanism of NRF2 induction of glioblastomas. Downregulation of the new NRF2/TAZ axis may provide a novel therapy for glioblastomas.
Collapse
Affiliation(s)
- Maribel Escoll
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain; Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Diego Lastra
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain; Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Marta Pajares
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain; Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Natalia Robledinos-Antón
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain; Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Ana I Rojo
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain; Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Raquel Fernández-Ginés
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain; Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Marta Mendiola
- Laboratory of Pathology and Translational Oncology, Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
| | - Virginia Martínez-Marín
- Department of Pathology, Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
| | - Isabel Esteban
- Department of Pathology, Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain
| | - Ricardo Gargini
- Centro de Biología Molecular "Severo Ochoa" UAM-CSIC, Autonomous University of Madrid, Madrid, Spain
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Spain; Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain.
| |
Collapse
|
64
|
Abstract
Cullin 3 (Cul3) family of ubiquitin ligases comprises three components, the RING finger protein RBX1, the Cul3 scaffold, and a Bric-a-brac/Tramtrack/Broad complex (BTB) protein. The BTB protein serves as a bridge to connect Cul3 to substrate and is functionally equivalent to the combination of substrate adaptor and linker in other Cullin complexes. Human genome encodes for ~180 BTB proteins, implying a broad spectrum of ubiquitination signals and substrate repertoire. Accordingly, Cul3 ubiquitin ligases are involved in diverse cellular processes, including cell division, differentiation, cytoskeleton remodeling, stress responses, and nerve cell functions. Emerging evidence has pointed to the prominent role of Cul3 ubiquitin ligases in cancer. This chapter will describe recent advances on the roles of Cul3 E3 ligase complexes in regulating various cancer hallmarks and therapeutic responses and the mutation/dysregulation of Cul3 substrate adaptors in cancer. In particular, we will focus on several extensively studied substrate adaptors, such as Keap1, SPOP, KLHL20, and LZTR1, and will also discuss other recently identified Cul3 adaptors with oncogenic or tumor-suppressive functions. We conclude that Cul3 ubiquitin ligases represent master regulators of human malignancies and highlight the importance of developing modulating agents for oncogenic/tumor-suppressive Cul3 E3 ligase complexes to prevent or intervene tumorigenesis.
Collapse
Affiliation(s)
- Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
65
|
Keratin 5-Cre-driven deletion of Ncstn in an acne inversa-like mouse model leads to a markedly increased IL-36a and Sprr2 expression. Front Med 2019; 14:305-317. [DOI: 10.1007/s11684-019-0722-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/07/2019] [Indexed: 10/25/2022]
|
66
|
Taguchi K, Kensler TW. Nrf2 in liver toxicology. Arch Pharm Res 2019; 43:337-349. [PMID: 31782059 DOI: 10.1007/s12272-019-01192-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 11/19/2019] [Indexed: 12/14/2022]
Abstract
Liver plays essential roles in the metabolism of many endogenous chemicals and exogenous toxicants. Mechanistic studies in liver have been at the forefront of efforts to probe the roles of bioactivation and detoxication of environmental toxins and toxicants in hepatotoxicity. Moreover, idiosyncratic hepatoxicity remains a key barrier in the clinical development of drugs. The now vast Nrf2 field emerged in part from biochemical and molecular studies on chemical inducers of hepatic detoxication enzymes and subsequent characterization of the modulation of drug/toxicant induced hepatotoxicities in mice through disruption of either Nrf2 or Keap1 genes. In general, loss of Nrf2 increases the sensitivity to such toxic chemicals, highlighting a central role of this transcription factor and its downstream target genes as a modifier to chemical stress. In this review, we summarize the impact of Nrf2 on the toxicology of multiple hepatotoxicants, and discuss efforts to utilize the Nrf2 response in predictive toxicology.
Collapse
Affiliation(s)
- Keiko Taguchi
- Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba, Sendai, 980-8575, Japan.
| | - Thomas W Kensler
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N, Seattle, WA, 98109, USA
| |
Collapse
|
67
|
Jeong Y, Hellyer JA, Stehr H, Hoang NT, Niu X, Das M, Padda SK, Ramchandran K, Neal JW, Wakelee H, Diehn M. Role of KEAP1/NFE2L2 Mutations in the Chemotherapeutic Response of Patients with Non-Small Cell Lung Cancer. Clin Cancer Res 2019; 26:274-281. [PMID: 31548347 DOI: 10.1158/1078-0432.ccr-19-1237] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/12/2019] [Accepted: 09/16/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Activation of NFE2L2 has been linked to chemoresistance in cell line models. Recently, somatic mutations that activate NFE2L2, including mutations in NFE2L2, KEAP1, or CUL3, have been found to be associated with poor outcomes in patients with non-small cell lung cancer (NSCLC). However, the impact of these mutations on chemoresistance remains incompletely explored. EXPERIMENTAL DESIGN We investigated the effect of Keap1 deletion on chemoresistance in cell lines from Trp53-based mouse models of lung squamous cell carcinoma (LSCC) and lung adenocarcinoma (LUAD). Separately, we identified 51 patients with stage IV NSCLC with KEAP1, NFE2L2, or CUL3 mutations and a matched cohort of 52 wild-type patients. Time to treatment failure after first-line platinum doublet chemotherapy and overall survival was compared between the two groups. RESULTS Deletion of Keap1 in Trp53-null murine LUAD and LSCC resulted in increased clonogenic survival upon treatment with diverse cytotoxic chemotherapies. In patients with NSCLC, median time to treatment failure (TTF) after first-line chemotherapy for the KEAP1/NFE2L2/CUL3-mutant cohort was 2.8 months compared with 8.3 months in the control group (P < 0.0001). Median overall survival (OS) was 11.2 months in the KEAP1/NFE2L2/CUL3-mutant group and 36.8 months in the control group (P = 0.006). CONCLUSIONS Keap1 deletion confers chemoresistance in murine lung cancer cells. Patients with metastatic NSCLC with mutations in KEAP1, NFE2L2, or CUL3 have shorter TTF and OS after first-line platinum doublet chemotherapy compared with matched controls. Novel approaches for improving outcomes in this subset of patients with NSCLC are therefore needed.
Collapse
Affiliation(s)
- Youngtae Jeong
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California.,Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Jessica A Hellyer
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Henning Stehr
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Ngoc T Hoang
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California.,Department of Biology, San Francisco State University, San Francisco, California
| | - Xiaomin Niu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.,Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Millie Das
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.,Department of Medicine, VA Palo Alto Health Care System, Palo Alto, California
| | - Sukhmani K Padda
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Kavitha Ramchandran
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Joel W Neal
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Heather Wakelee
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California.,Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
68
|
Burgener AV, Bantug GR, Meyer BJ, Higgins R, Ghosh A, Bignucolo O, Ma EH, Loeliger J, Unterstab G, Geigges M, Steiner R, Enamorado M, Ivanek R, Hunziker D, Schmidt A, Müller-Durovic B, Grählert J, Epple R, Dimeloe S, Lötscher J, Sauder U, Ebnöther M, Burger B, Heijnen I, Martínez-Cano S, Cantoni N, Brücker R, Kahlert CR, Sancho D, Jones RG, Navarini A, Recher M, Hess C. SDHA gain-of-function engages inflammatory mitochondrial retrograde signaling via KEAP1-Nrf2. Nat Immunol 2019; 20:1311-1321. [PMID: 31527833 DOI: 10.1038/s41590-019-0482-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/31/2019] [Indexed: 12/15/2022]
Abstract
Whether screening the metabolic activity of immune cells facilitates discovery of molecular pathology remains unknown. Here we prospectively screened the extracellular acidification rate as a measure of glycolysis and the oxygen consumption rate as a measure of mitochondrial respiration in B cells from patients with primary antibody deficiency. The highest oxygen consumption rate values were detected in three study participants with persistent polyclonal B cell lymphocytosis (PPBL). Exome sequencing identified germline mutations in SDHA, which encodes succinate dehydrogenase subunit A, in all three patients with PPBL. SDHA gain-of-function led to an accumulation of fumarate in PPBL B cells, which engaged the KEAP1-Nrf2 system to drive the transcription of genes encoding inflammatory cytokines. In a single patient trial, blocking the activity of the cytokine interleukin-6 in vivo prevented systemic inflammation and ameliorated clinical disease. Overall, our study has identified pathological mitochondrial retrograde signaling as a disease modifier in primary antibody deficiency.
Collapse
Affiliation(s)
- Anne-Valérie Burgener
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Glenn R Bantug
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland.,Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Benedikt J Meyer
- Immunodeficiency Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Rebecca Higgins
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Adhideb Ghosh
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland.,Competence Center for Personalized Medicine University of Zürich/Eidgenössische Technische Hochschule, Zürich, Switzerland
| | - Olivier Bignucolo
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Eric H Ma
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, USA.,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Jordan Loeliger
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Gunhild Unterstab
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Marco Geigges
- Epigenomics Group, D-BSSE, Eidgenössische Technische Hochschule, Basel, Switzerland
| | - Rebekah Steiner
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Michel Enamorado
- Immunobiology Laboratory, entro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Washington DC, USA
| | - Robert Ivanek
- Bioinformatics Facility, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Danielle Hunziker
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Bojana Müller-Durovic
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Jasmin Grählert
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Raja Epple
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Sarah Dimeloe
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Jonas Lötscher
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Ursula Sauder
- Electron Microscopy Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Monika Ebnöther
- Division of Hematology and Oncology, Claraspital, Basel, Switzerland
| | - Bettina Burger
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Ingmar Heijnen
- Division Medical Immunology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Sarai Martínez-Cano
- Immunobiology Laboratory, entro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Nathan Cantoni
- Division of Hematology, Cantonal Hospital of Aarau, Aargau, Switzerland
| | - Rolf Brücker
- Division of Internal Medicine and Rheumatology, Hospital St. Anna, Luzern, Switzerland
| | - Christian R Kahlert
- Division of Infectious Diseases, Children's Hospital of St. Gallen, St. Gallen, Switzerland
| | - David Sancho
- Immunobiology Laboratory, entro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Russell G Jones
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, USA.,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Alexander Navarini
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Mike Recher
- Immunodeficiency Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Christoph Hess
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland. .,Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
69
|
Qin JJ, Cheng XD, Zhang J, Zhang WD. Dual roles and therapeutic potential of Keap1-Nrf2 pathway in pancreatic cancer: a systematic review. Cell Commun Signal 2019; 17:121. [PMID: 31511020 PMCID: PMC6740038 DOI: 10.1186/s12964-019-0435-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 09/02/2019] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most fatal diseases with a very high rate of metastasis and low rate of survival. Despite the advances in understanding this devastating disease, PC still accounts for 3% of all cancers and causes almost 7% of death of cancer patients. Recent studies have demonstrated that the transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) and its key negative regulator Kelch-like ECH-associated protein 1 (Keap1) are dysregulated in PC and the Keap1-Nrf2 pathway is an emerging target for PC prevention and therapy. Indeed, Nrf2 plays an either tumor-suppressive or promoting function in PC, which depends on the developmental stages of the disease and the cellular context. Several natural-product Nrf2 activators have been developed to prevent pancreatic carcinogenesis, while the Nrf2 inhibitors have been examined for their efficacy in inhibiting PC growth and metastasis and reversing chemoresistance. However, further preclinical and clinical studies for determining the effectiveness and safety of targeting the Keap1-Nrf2 pathway for PC prevention and therapy are warranted. In this review, we comprehensively discuss the dual roles of the Keap1-Nrf2 signaling pathway in PC as well as the current targeting strategies and known activators and inhibitors of Nrf2. We also propose new strategies that may be used to address the current issues and develop more specific and more effective Nrf2 activator/inhibitors for PC prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China. .,Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | | | - Jia Zhang
- Shanxi Institute of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Wei-Dong Zhang
- School of Pharmacy, Naval Medical University, 325 Guohe Road, Yangpu District, Shanghai, 200433, China. .,Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
70
|
Galiè M, Covi V, Tabaracci G, Malatesta M. The Role of Nrf2 in the Antioxidant Cellular Response to Medical Ozone Exposure. Int J Mol Sci 2019; 20:E4009. [PMID: 31426459 PMCID: PMC6720777 DOI: 10.3390/ijms20164009] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023] Open
Abstract
Ozone (O3) is a natural, highly unstable atmospheric gas that rapidly decomposes to oxygen. Although not being a radical molecule, O3 is a very strong oxidant and therefore it is potentially toxic for living organisms. However, scientific evidence proved that the effects of O3 exposure are dose-dependent: high dosages stimulate severe oxidative stress resulting in inflammatory response and tissue injury, whereas low O3 concentrations induce a moderate oxidative eustress activating antioxidant pathways. These properties make O3 a powerful medical tool, which can be used as either a disinfectant or an adjuvant agent in the therapy of numerous diseases. In this paper, the cellular mechanisms involved in the antioxidant response to O3 exposure will be reviewed with special reference to the activation of nuclear factor erythroid 2-related factor 2 (Nrf2) and its role in the efficacy of ozone therapy.
Collapse
Affiliation(s)
- Mirco Galiè
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy
| | - Viviana Covi
- San Rocco Clinic, Via Monsignor G. V. Moreni 95, I-25018 Montichiari (BS), Italy
| | - Gabriele Tabaracci
- San Rocco Clinic, Via Monsignor G. V. Moreni 95, I-25018 Montichiari (BS), Italy
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, I-37134 Verona, Italy.
| |
Collapse
|
71
|
Metabolic reprogramming and Notch activity distinguish between non-small cell lung cancer subtypes. Br J Cancer 2019; 121:51-64. [PMID: 31114017 PMCID: PMC6738087 DOI: 10.1038/s41416-019-0464-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 02/01/2019] [Accepted: 03/07/2019] [Indexed: 01/12/2023] Open
Abstract
Background Previous studies suggested that the metabolism is differently reprogrammed in the major subtypes of non-small cell lung cancer (NSCLC), squamous cell carcinomas (SCC) and adenocarcinomas (AdC). However, a comprehensive analysis of this differential metabolic reprogramming is lacking. Methods Publicly available gene expression data from human lung cancer samples and cell lines were analysed. Stable isotope resolved metabolomics were performed on SCC and ADC tumours in human patients and in freshly resected tumour slices. Results Analysis of multiple transcriptomics data from human samples identified a SCC-distinguishing enzyme gene signature. SCC tumours from patients infused with [U-13C]-glucose and SCC tissue slices incubated with stable isotope tracers demonstrated differential glucose and glutamine catabolism compared to AdCs or non-cancerous lung, confirming increased activity through pathways defined by the SCC metabolic gene signature. Furthermore, the upregulation of Notch target genes was a distinguishing feature of SCCs, which correlated with the metabolic signature. Notch and MYC-driven murine lung tumours recapitulated the SCC-distinguishing metabolic reprogramming. However, the differences between SCCs and AdCs disappear in established cell lines in 2D culture. Conclusions Our data emphasise the importance of studying lung cancer metabolism in vivo. They also highlight potential targets for therapeutic intervention in SCC patients including differentially expressed enzymes that catalyse reactions in glycolysis, glutamine catabolism, serine, nucleotide and glutathione biosynthesis.
Collapse
|
72
|
Yang Y, Zeng B, Sun Z, Esfahani AM, Hou J, Jiao ND, Liu L, Chen L, Basson MD, Dong L, Yang R, Xi N. Optimization of Protein-Protein Interaction Measurements for Drug Discovery Using AFM Force Spectroscopy. IEEE TRANSACTIONS ON NANOTECHNOLOGY 2019; 18:509-517. [PMID: 32051682 PMCID: PMC7015265 DOI: 10.1109/tnano.2019.2915507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Increasingly targeted in drug discovery, protein-protein interactions challenge current high throughput screening technologies in the pharmaceutical industry. Developing an effective and efficient method for screening small molecules or compounds is critical to accelerate the discovery of ligands for enzymes, receptors and other pharmaceutical targets. Here, we report developments of methods to increase the signal-to-noise ratio (SNR) for screening protein-protein interactions using atomic force microscopy (AFM) force spectroscopy. We have demonstrated the effectiveness of these developments on detecting the binding process between focal adhesion kinases (FAK) with protein kinase B (Akt1), which is a target for potential cancer drugs. These developments include optimized probe and substrate functionalization processes and redesigned probe-substrate contact regimes. Furthermore, a statistical-based data processing method was developed to enhance the contrast of the experimental data. Collectively, these results demonstrate the potential of the AFM force spectroscopy in automating drug screening with high throughput.
Collapse
Affiliation(s)
- Yongliang Yang
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, 48823, USA
| | - Bixi Zeng
- Departments of Surgery and Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Zhiyong Sun
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, 48823, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, NE 68588 USA
| | - Jing Hou
- School of Information and Control Engineering, Shenyang Jianzhu University, Shenyang 110168, China
| | - Nian-Dong Jiao
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110006, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110006, China
| | - Liangliang Chen
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, 48823, USA
| | - Marc D Basson
- Departments of Surgery and Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Lixin Dong
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, 48823, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska Lincoln, NE 68588 USA
| | - Ning Xi
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, 48823, USA
| |
Collapse
|
73
|
Cuadrado A, Rojo AI, Wells G, Hayes JD, Cousin SP, Rumsey WL, Attucks OC, Franklin S, Levonen AL, Kensler TW, Dinkova-Kostova AT. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat Rev Drug Discov 2019; 18:295-317. [PMID: 30610225 DOI: 10.1038/s41573-018-0008-x] [Citation(s) in RCA: 932] [Impact Index Per Article: 155.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The transcription factor NF-E2 p45-related factor 2 (NRF2; encoded by NFE2L2) and its principal negative regulator, the E3 ligase adaptor Kelch-like ECH-associated protein 1 (KEAP1), are critical in the maintenance of redox, metabolic and protein homeostasis, as well as the regulation of inflammation. Thus, NRF2 activation provides cytoprotection against numerous pathologies including chronic diseases of the lung and liver; autoimmune, neurodegenerative and metabolic disorders; and cancer initiation. One NRF2 activator has received clinical approval and several electrophilic modifiers of the cysteine-based sensor KEAP1 and inhibitors of its interaction with NRF2 are now in clinical development. However, challenges regarding target specificity, pharmacodynamic properties, efficacy and safety remain.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
- Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Ana I Rojo
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
- Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Geoffrey Wells
- UCL School of Pharmacy, University College London, London, UK
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK
| | | | | | | | | | - Anna-Liisa Levonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK.
- Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
74
|
Dodson M, de la Vega MR, Cholanians AB, Schmidlin CJ, Chapman E, Zhang DD. Modulating NRF2 in Disease: Timing Is Everything. Annu Rev Pharmacol Toxicol 2019; 59:555-575. [PMID: 30256716 PMCID: PMC6538038 DOI: 10.1146/annurev-pharmtox-010818-021856] [Citation(s) in RCA: 358] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The transcription factor nuclear factor erythroid 2 (NF-E2)-related factor 2 (NRF2) is a central regulator of redox, metabolic, and protein homeostasis that intersects with many other signaling cascades. Although the understanding of the complex nature of NRF2 signaling continues to grow, there is only one therapeutic targeting NRF2 for clinical use, dimethyl fumarate, used for the treatment of multiple sclerosis. The discovery of new therapies is confounded by the fact that NRF2 levels vary significantly depending on physiological and pathological context. Thus, properly timed and targeted manipulation of the NRF2 pathway is critical in creating effective therapeutic regimens. In this review, we summarize the regulation and downstream targets of NRF2. Furthermore, we discuss the role of NRF2 in cancer, neurodegeneration, and diabetes as well as cardiovascular, kidney, and liver disease, with a special emphasis on NRF2-based therapeutics, including those that have made it into clinical trials.
Collapse
Affiliation(s)
- Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, USA;
| | - Montserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, USA;
| | - Aram B Cholanians
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, USA;
| | - Cody J Schmidlin
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, USA;
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, USA;
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, USA;
- Arizona Cancer Center, University of Arizona, Tucson, Arizona 85724, USA
| |
Collapse
|
75
|
Scharf C, Eymann C, Emicke P, Bernhardt J, Wilhelm M, Görries F, Winter J, von Woedtke T, Darm K, Daeschlein G, Steil L, Hosemann W, Beule A. Improved Wound Healing of Airway Epithelial Cells Is Mediated by Cold Atmospheric Plasma: A Time Course-Related Proteome Analysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7071536. [PMID: 31223425 PMCID: PMC6541959 DOI: 10.1155/2019/7071536] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/28/2019] [Indexed: 01/08/2023]
Abstract
The promising potential of cold atmospheric plasma (CAP) treatment as a new therapeutic option in the field of medicine, particularly in Otorhinolaryngology and Respiratory medicine, demands primarily the assessment of potential risks and the prevention of any direct and future cell damages. Consequently, the application of a special intensity of CAP that is well tolerated by cells and tissues is of particular interest. Although improvement of wound healing by CAP treatment has been described, the underlying mechanisms and the molecular influences on human tissues are so far only partially characterized. In this study, human S9 bronchial epithelial cells were treated with cold plasma of atmospheric pressure plasma jet that was previously proven to accelerate the wound healing in a clinically relevant extent. We studied the detailed cellular adaptation reactions for a specified plasma intensity by time-resolved comparative proteome analyses of plasma treated vs. nontreated cells to elucidate the mechanisms of the observed improved wound healing and to define potential biomarkers and networks for the evaluation of plasma effects on human epithelial cells. K-means cluster analysis and time-related analysis of fold-change factors indicated concordantly clear differences between the short-term (up to 1 h) and long-term (24-72 h) adaptation reactions. Thus, the induction of Nrf2-mediated oxidative and endoplasmic reticulum stress response, PPAR-alpha/RXR activation as well as production of peroxisomes, and prevention of apoptosis already during the first hour after CAP treatment are important cell strategies to overcome oxidative stress and to protect and maintain cell integrity and especially microtubule dynamics. After resolving of stress, when stress adaptation was accomplished, the cells seem to start again with proliferation and cellular assembly and organization. The observed strategies and identification of marker proteins might explain the accelerated wound healing induced by CAP, and these indicators might be subsequently used for risk assessment and quality management of application of nonthermal plasma sources in clinical settings.
Collapse
Affiliation(s)
- Christian Scharf
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Christine Eymann
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Philipp Emicke
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Jörg Bernhardt
- 2Institute for Microbiology, University of Greifswald, Germany
| | - Martin Wilhelm
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Fabian Görries
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Jörn Winter
- 3Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
| | - Thomas von Woedtke
- 3Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- 4Department of Hygiene and Environmental Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Darm
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Georg Daeschlein
- 5Department of Dermatology, University Medicine Greifswald, Greifswald, Germany
| | - Leif Steil
- 6Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Germany
| | - Werner Hosemann
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
| | - Achim Beule
- 1Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Germany
- 7Department of Otorhinolaryngology, University Hospital Münster, Münster, Germany
| |
Collapse
|
76
|
Abstract
SIGNIFICANCE Nuclear factor E2-related factor 2 (Nrf2) is a transcription factor that coordinates the basal and stress-inducible activation of a vast array of cytoprotective genes. Understanding the regulation of Nrf2 activity and downstream pathways has major implications for human health. Recent Advances: Nrf2 regulates the transcription of components of the glutathione and thioredoxin antioxidant systems, as well as enzymes involved in phase I and phase II detoxification of exogenous and endogenous products, NADPH regeneration, and heme metabolism. It therefore represents a crucial regulator of the cellular defense mechanisms against xenobiotic and oxidative stress. In addition to antioxidant responses, Nrf2 is involved in other cellular processes, such as autophagy, intermediary metabolism, stem cell quiescence, and unfolded protein response. Given the wide range of processes that Nrf2 controls, its activity is tightly regulated at multiple levels. Here, we review the different modes of regulation of Nrf2 activity and the current knowledge of Nrf2-mediated transcriptional control. CRITICAL ISSUES It is now clear that Nrf2 lies at the center of a complex regulatory network. A full comprehension of the Nrf2 program will require an integrated consideration of all the different factors determining Nrf2 activity. FUTURE DIRECTIONS Additional computational and experimental studies are needed to obtain a more dynamic global view of Nrf2-mediated gene regulation. In particular, studies comparing how the Nrf2-dependent network changes from a physiological to a pathological condition can provide insight into mechanisms of disease and instruct new treatment strategies.
Collapse
Affiliation(s)
- Claudia Tonelli
- 1 Cold Spring Harbor Laboratory , Cold Spring Harbor, New York
| | | | - David A Tuveson
- 1 Cold Spring Harbor Laboratory , Cold Spring Harbor, New York.,2 Lustgarten Foundation Pancreatic Cancer Research Laboratory , Cold Spring Harbor, New York
| |
Collapse
|
77
|
Ma S, Paiboonrungruan C, Yan T, Williams KP, Major MB, Chen XL. Targeted therapy of esophageal squamous cell carcinoma: the NRF2 signaling pathway as target. Ann N Y Acad Sci 2018; 1434:164-172. [PMID: 29752726 PMCID: PMC6230513 DOI: 10.1111/nyas.13681] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/04/2018] [Accepted: 02/24/2018] [Indexed: 02/07/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly disease that requires extensive research. Here, we review the current understanding of the functions of the nuclear factor erythroid-derived 2-like 2 (NRF2) signaling pathway in the esophagus. Genomic data suggest that gene mutations and several other mechanisms result in NRF2 hyperactivation in human ESCC. As a consequence, NRF2high ESCC is more resistant to chemoradiotherapy and associated with poorer survival than NRF2low ESCC. Mechanistically, we believe NRF2, functioning as a transcription factor, causes an esophageal phenotype through regulation of gene transcription. We discuss metabolism, mitochondria, proteasomes, and several signaling pathways as downstream players that may contribute to an esophageal phenotype due to NRF2 hyperactivation. Finally, strategies are proposed to target the NRF2 signaling pathway for therapy of NRF2high ESCC.
Collapse
Affiliation(s)
- Shaohua Ma
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing, China
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
| | - Chorlada Paiboonrungruan
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
| | - Tiansheng Yan
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing, China
| | - Kevin P Williams
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina
| | - M Ben Major
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Xiaoxin Luke Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
- Center for Esophageal Disease and Swallowing, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
78
|
The effects of Nrf2 knockout on regulation of benzene-induced mouse hematotoxicity. Toxicol Appl Pharmacol 2018; 358:56-67. [DOI: 10.1016/j.taap.2018.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/02/2018] [Accepted: 09/04/2018] [Indexed: 01/29/2023]
|
79
|
Paul S, Ghosh S, Mandal S, Sau S, Pal M. NRF2 transcriptionally activates the heat shock factor 1 promoter under oxidative stress and affects survival and migration potential of MCF7 cells. J Biol Chem 2018; 293:19303-19316. [PMID: 30309986 DOI: 10.1074/jbc.ra118.003376] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/02/2018] [Indexed: 01/07/2023] Open
Abstract
Functional up-regulation of heat shock factor 1 (HSF1) activity through different posttranslational modifications has been implicated in the survival and proliferation of various cancers. It is increasingly recognized that the HSF1 gene is also up-regulated at the transcriptional level, a phenomenon correlated with poor prognosis for patients with different cancers, including breast cancer. Here, we analyzed the transcriptional up-regulation of HSF1 in human cells upon arsenite- or peroxide-induced oxidative stress. Sequential promoter truncation coupled with bioinformatics analysis revealed that this activation is mediated by two antioxidant response elements (AREs) located between 1707 and 1530 bp upstream of the transcription start site of the HSF1 gene. Using shRNA-mediated down-regulation, ChIP of NRF2, site-directed mutagenesis of the AREs, and DNase I footprinting of the HSF1 promoter, we confirmed that nuclear factor erythroid-derived 2-like 2 (NRF2, also known as NFE2L2) interacts with these AREs and up-regulates HSF1 expression. We also found that BRM/SWI2-related gene 1 (BRG1), a catalytic subunit of SWI2/SNF2-like chromatin remodeler, is involved in this process. We further show that NRF2-dependent HSF1 gene regulation plays a crucial role in cancer cell biology, as interference with NRF2-mediated HSF1 activation compromised survival, migration potential, and the epithelial-to-mesenchymal transition and autophagy in MCF7 breast cancer cells exposed to oxidative stress. Taken together, our findings unravel the mechanistic basis of HSF1 gene regulation in cancer cells and provide molecular evidence supporting a direct interaction between HSF1 and NRF2, critical regulators of two cytoprotective mechanisms exploited by cancer cells.
Collapse
Affiliation(s)
| | | | - Sukhendu Mandal
- Department of Biochemistry, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata, India 700054
| | - Subrata Sau
- Department of Biochemistry, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata, India 700054
| | | |
Collapse
|
80
|
Yamada T, Murata D, Adachi Y, Itoh K, Kameoka S, Igarashi A, Kato T, Araki Y, Huganir RL, Dawson TM, Yanagawa T, Okamoto K, Iijima M, Sesaki H. Mitochondrial Stasis Reveals p62-Mediated Ubiquitination in Parkin-Independent Mitophagy and Mitigates Nonalcoholic Fatty Liver Disease. Cell Metab 2018; 28:588-604.e5. [PMID: 30017357 PMCID: PMC6170673 DOI: 10.1016/j.cmet.2018.06.014] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 03/07/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
Abstract
It is unknown what occurs if both mitochondrial division and fusion are completely blocked. Here, we introduced mitochondrial stasis by deleting two dynamin-related GTPases for division (Drp1) and fusion (Opa1) in livers. Mitochondrial stasis rescues liver damage and hypotrophy caused by the single knockout (KO). At the cellular level, mitochondrial stasis re-establishes mitochondrial size and rescues mitophagy defects caused by division deficiency. Using Drp1KO livers, we found that the autophagy adaptor protein p62/sequestosome-1-which is thought to function downstream of ubiquitination-promotes mitochondrial ubiquitination. p62 recruits two subunits of a cullin-RING ubiquitin E3 ligase complex, Keap1 and Rbx1, to mitochondria. Resembling Drp1KO, diet-induced nonalcoholic fatty livers enlarge mitochondria and accumulate mitophagy intermediates. Resembling Drp1Opa1KO, Opa1KO rescues liver damage in this disease model. Our data provide a new concept that mitochondrial stasis leads the spatial dimension of mitochondria to a stationary equilibrium and a new mechanism for mitochondrial ubiquitination in mitophagy.
Collapse
Affiliation(s)
- Tatsuya Yamada
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daisuke Murata
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoshihiro Adachi
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kie Itoh
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shoichiro Kameoka
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Atsushi Igarashi
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Takashi Kato
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoichi Araki
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Koji Okamoto
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
81
|
Abstract
The concept of cell signaling in the context of nonenzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. Our Review reflects on five aspects critical for understanding how nature harnesses these noncanonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation, (2) physicochemical properties, (3) mechanisms of action, (4) methods of interrogation, and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, our Review is anticipated to function as a primer. For the expert, we aim to stimulate thought and discussion about fundamentals of redox signaling mechanisms and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chemistry and biology.
Collapse
Affiliation(s)
- Saba Parvez
- Department of Pharmacology and Toxicology, College of
Pharmacy, University of Utah, Salt Lake City, Utah, 84112, USA
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Marcus J. C. Long
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Jesse R. Poganik
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Yimon Aye
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
- Department of Biochemistry, Weill Cornell Medicine, New
York, New York, 10065, USA
| |
Collapse
|
82
|
Prestigiacomo V, Suter-Dick L. Nrf2 protects stellate cells from Smad-dependent cell activation. PLoS One 2018; 13:e0201044. [PMID: 30028880 PMCID: PMC6054401 DOI: 10.1371/journal.pone.0201044] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Hepatic stellate cells (HSC) orchestrate the deposition of extracellular matrix (ECM) and are the primary effector of liver fibrosis. Several factors, including TGF-β1, PDGF and oxidative stress, have been shown to trigger HSC activation. However, the involvement of cellular defence mechanisms, such as the activation of antioxidant response by Nrf2/Keap1 in the modulation of HSC activation is not known. The aim of this work was to elucidate the role of Nrf2 pathway in HSC trans-differentiation involved in the development of fibrosis. To this end, we repressed Nrf2 and Keap1 expression in HSC with specific siRNAs. We then assessed activation markers, as well as proliferation and migration, in both primary and immortalised human HSCs exposed to Smad inhibitors (SB-431542 hydrate and SB-525334), TGF-β1 and/or PDGF. Our results indicate that knocking down Nrf2 induces HSC activation, as shown by an increase in αSMA-positive cells and by gene expression induction of ECM components (collagens and fibronectin). HSC with reduced Nrf2-levels also showed an increase in migration and a decrease in proliferation. We could also demonstrate that the activation of Nrf2-deficient HSC involves the TGF-β1/Smad pathway, as the activation was successfully inhibited with the two tested Smad inhibitors. Moreover, TGF-β1 elicited a stronger induction of HSC activation markers in Nrf2 deficient cells than in wild type cells. Thus, our data suggest that Nrf2 limits HSCs activation, through the inhibition of the TGF-β1/Smad pathway in HSCs.
Collapse
Affiliation(s)
- Vincenzo Prestigiacomo
- University of Applied Sciences Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
- University of Basel, Department of Pharmaceutical Sciences, Basel, Switzerland
- * E-mail:
| | - Laura Suter-Dick
- University of Applied Sciences Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| |
Collapse
|
83
|
Rojo de la Vega M, Chapman E, Zhang DD. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018; 34:21-43. [PMID: 29731393 PMCID: PMC6039250 DOI: 10.1016/j.ccell.2018.03.022] [Citation(s) in RCA: 1155] [Impact Index Per Article: 165.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/11/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022]
Abstract
The transcription factor NRF2 is the master regulator of the cellular antioxidant response. Though recognized originally as a target of chemopreventive compounds that help prevent cancer and other maladies, accumulating evidence has established the NRF2 pathway as a driver of cancer progression, metastasis, and resistance to therapy. Recent studies have identified new functions for NRF2 in the regulation of metabolism and other essential cellular functions, establishing NRF2 as a truly pleiotropic transcription factor. In this review, we explore the roles of NRF2 in the hallmarks of cancer, indicating both tumor suppressive and tumor-promoting effects.
Collapse
Affiliation(s)
- Montserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
84
|
Yamamoto M, Kensler TW, Motohashi H. The KEAP1-NRF2 System: a Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol Rev 2018; 98:1169-1203. [PMID: 29717933 PMCID: PMC9762786 DOI: 10.1152/physrev.00023.2017] [Citation(s) in RCA: 1264] [Impact Index Per Article: 180.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The Kelch-like ECH-associated protein 1-NF-E2-related factor 2 (KEAP1-NRF2) system forms the major node of cellular and organismal defense against oxidative and electrophilic stresses of both exogenous and endogenous origins. KEAP1 acts as a cysteine thiol-rich sensor of redox insults, whereas NRF2 is a transcription factor that robustly transduces chemical signals to regulate a battery of cytoprotective genes. KEAP1 represses NRF2 activity under quiescent conditions, whereas NRF2 is liberated from KEAP1-mediated repression on exposure to stresses. The rapid inducibility of a response based on a derepression mechanism is an important feature of the KEAP1-NRF2 system. Recent studies have unveiled the complexities of the functional contributions of the KEAP1-NRF2 system and defined its broader involvement in biological processes, including cell proliferation and differentiation, as well as cytoprotection. In this review, we describe historical milestones in the initial characterization of the KEAP1-NRF2 system and provide a comprehensive overview of the molecular mechanisms governing the functions of KEAP1 and NRF2, as well as their roles in physiology and pathology. We also refer to the clinical significance of the KEAP1-NRF2 system as an important prophylactic and therapeutic target for various diseases, particularly aging-related disorders. We believe that controlled harnessing of the KEAP1-NRF2 system is a key to healthy aging and well-being in humans.
Collapse
|
85
|
Ziros PG, Habeos IG, Chartoumpekis DV, Ntalampyra E, Somm E, Renaud CO, Bongiovanni M, Trougakos IP, Yamamoto M, Kensler TW, Santisteban P, Carrasco N, Ris-Stalpers C, Amendola E, Liao XH, Rossich L, Thomasz L, Juvenal GJ, Refetoff S, Sykiotis GP. NFE2-Related Transcription Factor 2 Coordinates Antioxidant Defense with Thyroglobulin Production and Iodination in the Thyroid Gland. Thyroid 2018; 28:780-798. [PMID: 29742982 PMCID: PMC5994681 DOI: 10.1089/thy.2018.0018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND The thyroid gland has a special relationship with oxidative stress. While generation of oxidative substances is part of normal iodide metabolism during thyroid hormone synthesis, the gland must also defend itself against excessive oxidation in order to maintain normal function. Antioxidant and detoxification enzymes aid thyroid cells to maintain homeostasis by ameliorating oxidative insults, including during exposure to excess iodide, but the factors that coordinate their expression with the cellular redox status are not known. The antioxidant response system comprising the ubiquitously expressed NFE2-related transcription factor 2 (Nrf2) and its redox-sensitive cytoplasmic inhibitor Kelch-like ECH-associated protein 1 (Keap1) defends tissues against oxidative stress, thereby protecting against pathologies that relate to DNA, protein, and/or lipid oxidative damage. Thus, it was hypothesized that Nrf2 should also have important roles in maintaining thyroid homeostasis. METHODS Ubiquitous and thyroid-specific male C57BL6J Nrf2 knockout (Nrf2-KO) mice were studied. Plasma and thyroids were harvested for evaluation of thyroid function tests by radioimmunoassays and of gene and protein expression by real-time polymerase chain reaction and immunoblotting, respectively. Nrf2-KO and Keap1-KO clones of the PCCL3 rat thyroid follicular cell line were generated using CRISPR/Cas9 technology and were used for gene and protein expression studies. Software-predicted Nrf2 binding sites on the thyroglobulin enhancer were validated by site-directed in vitro mutagenesis and chromatin immunoprecipitation. RESULTS The study shows that Nrf2 mediates antioxidant transcriptional responses in thyroid cells and protects the thyroid from oxidation induced by iodide overload. Surprisingly, it was also found that Nrf2 has a dramatic impact on both the basal abundance and the thyrotropin-inducible intrathyroidal abundance of thyroglobulin (Tg), the precursor protein of thyroid hormones. This effect is mediated by cell-autonomous regulation of Tg gene expression by Nrf2 via its direct binding to two evolutionarily conserved antioxidant response elements in an upstream enhancer. Yet, despite upregulating Tg levels, Nrf2 limits Tg iodination both under basal conditions and in response to excess iodide. CONCLUSIONS Nrf2 exerts pleiotropic roles in the thyroid gland to couple cell stress defense mechanisms to iodide metabolism and the thyroid hormone synthesis machinery, both under basal conditions and in response to excess iodide.
Collapse
Affiliation(s)
- Panos G. Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ioannis G. Habeos
- Department of Internal Medicine, Division of Endocrinology, School of Medicine, University of Patras, Patras, Greece
| | | | - Eleni Ntalampyra
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Emmanuel Somm
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Cédric O. Renaud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Massimo Bongiovanni
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Thomas W. Kensler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid, CIBERONC (ISCIII), Madrid, Spain
| | - Nancy Carrasco
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Carrie Ris-Stalpers
- Women's and Children's Clinic, Department of Obstetrics and Gynaecology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Elena Amendola
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli, Federico II, Naples, Italy
| | - Xiao-Hui Liao
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Luciano Rossich
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - Lisa Thomasz
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - Guillermo J. Juvenal
- Nuclear Biochemistry Division, Argentine National Atomic Energy Commission, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - Samuel Refetoff
- Department of Medicine, The University of Chicago, Chicago, Illinois
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
- Department of Committee on Genetics, The University of Chicago, Chicago, Illinois
| | - Gerasimos P. Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
86
|
Zhao Q, Mao A, Guo R, Zhang L, Yan J, Sun C, Tang J, Ye Y, Zhang Y, Zhang H. Suppression of radiation-induced migration of non-small cell lung cancer through inhibition of Nrf2-Notch Axis. Oncotarget 2018; 8:36603-36613. [PMID: 28402268 PMCID: PMC5482680 DOI: 10.18632/oncotarget.16622] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/09/2017] [Indexed: 01/23/2023] Open
Abstract
Nuclear factor E2 related factor 2 (Nrf2) is a transcription factor that is associated with tumor growth and resistance to radiation. The canonical Notch signaling pathway is also crucial for maintaining non-small cell lung cancer (NSCLC). Aberrant Nrf2 and Notch signaling has repeatedly been showed to facilitate metastasis of NSCLC. Here, we show that radiation induce Nrf2 and Notch1 expression in NSCLC. Knockdown of Nrf2 enhanced radiosensitivity of NSCLC and reduced epithelial-to-mesenchymal transition. Importantly, we found that knockdown of Nrf2 dramatically decreased radiation-induced NSCLC invasion and significantly increased E-cadherin, but reduced N-cadherin and matrix metalloproteinase (MMP)-2/9 expression. We found that Notch1 knockdown also upregulated E-cadherin and suppressed N-cadherin expression. Nrf2 contributes to NSCLC cell metastatic properties and this inhibition correlated with reduced Notch1 expression. These results establish that Nrf2 and Notch1 downregulation synergistically inhibit radiation-induced migratory and invasive properties of NSCLC cells.
Collapse
Affiliation(s)
- Qiuyue Zhao
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China.,University of Chinese Academy of Sciences, Beijing 100039, China
| | - Aihong Mao
- Institute of Gansu Medical Science Research, Lanzhou 730000, China
| | - Ruoshui Guo
- South China Normal University, Guangzhou 510642, China
| | - Liping Zhang
- Northwest Normal University, Lanzhou 730000, China
| | - Jiawei Yan
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China.,University of Chinese Academy of Sciences, Beijing 100039, China
| | - Chao Sun
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | | | - Yancheng Ye
- Gansu Wuwei Institute of Medical Sciences, Gansu Province, Wuwei 733000, China
| | - Yanshan Zhang
- Gansu Wuwei Institute of Medical Sciences, Gansu Province, Wuwei 733000, China
| | - Hong Zhang
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China.,Key Laboratory of Heavy Ion Radiation and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China.,Gansu Wuwei Institute of Medical Sciences, Gansu Province, Wuwei 733000, China.,Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| |
Collapse
|
87
|
Chartoumpekis DV, Yagishita Y, Fazzari M, Palliyaguru DL, Rao UN, Zaravinos A, Khoo NK, Schopfer FJ, Weiss KR, Michalopoulos GK, Sipula I, O’Doherty RM, Kensler TW, Wakabayashi N. Nrf2 prevents Notch-induced insulin resistance and tumorigenesis in mice. JCI Insight 2018; 3:e97735. [PMID: 29515034 PMCID: PMC5922294 DOI: 10.1172/jci.insight.97735] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/30/2018] [Indexed: 12/30/2022] Open
Abstract
Insulin resistance is associated with increased incidence and enhanced progression of cancers. However, little is known about strategies that can effectively ameliorate insulin resistance and consequently halt cancer progression. Herein, we propose that the transcription factor Nrf2 (also known as Nfe2l2) may be such a target, given its central role in disease prevention. To this end, we developed a mouse that overexpresses the Notch intracellular domain in adipocytes (AdNICD), leading to lipodystrophy-induced severe insulin resistance and subsequent development of sarcomas, as a model reflecting that Notch signaling is deregulated in cancers and shows positive associations with insulin resistance and fatty liver disease in humans. Nrf2 pathway activation was achieved by knocking down Keap1, a repressor of Nrf2, in the AdNICD background. Constitutively enhanced Nrf2 signaling in this setting led to prevention of hepatic steatosis, dyslipidemia, and insulin resistance by repressing hepatic lipogenic pathways and restoration of the hepatic fatty acid profile to control levels. This protective effect of Nrf2 against diabetes extended to significant reduction and delay in sarcoma incidence and latency. Our study highlights that the Nrf2 pathway, which has been induced by small molecules in clinical trials, is a potential therapeutic target against insulin resistance and subsequent risk of cancer.
Collapse
Affiliation(s)
- Dionysios V. Chartoumpekis
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yoko Yagishita
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Fondazione Ri.MED, Palermo, Italy
| | - Dushani L. Palliyaguru
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Uma N.M. Rao
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Nicholas K.H. Khoo
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Francisco J. Schopfer
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Ian Sipula
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert M. O’Doherty
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Thomas W. Kensler
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nobunao Wakabayashi
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
88
|
Luo L, Chen Y, Wang H, Wang S, Liu K, Li X, Wang XJ, Tang X. Mkp-1 protects mice against toxin-induced liver damage by promoting the Nrf2 cytoprotective response. Free Radic Biol Med 2018; 115:361-370. [PMID: 29241671 DOI: 10.1016/j.freeradbiomed.2017.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/14/2017] [Accepted: 12/07/2017] [Indexed: 12/30/2022]
Abstract
The present study was undertaken to investigate the possible protective effect of mitogen-activated protein kinase phosphatase 1 (Mkp-1) on toxin-induced hepatic injury. Here, we uncovered a positive feedback loop between Mkp-1, a dual threonine/tyrosine phosphatase, and nuclear factor erythroid 2-related factor 2 (Nrf2), a crucial regulator of the defense system in the liver. Mkp-1-/- mice exhibited decreased protein levels of Nrf2, phase II gene products, and reduced glutathione (GSH) in the liver. Induction of detoxifying enzymes by the Nrf2 activator butylated hydroxyanisole (BHA) or sulforaphane, was attenuated in the liver and small intestines of Mkp-1-/- mice, indicating that the Nrf2 signaling pathway is impaired as a result of Mkp-1 deficiency. Mkp-1-/- mice suffered more severe liver injury after a single exposure to hepatotoxin carbon tetrachloride (CCl4) than their wild-type (WT) counterparts. BHA partially rescued the CCl4-induced liver damage in WT mice, but not in Mkp-1-/- mice, suggesting the requirement of Mkp-1 in the activation of Nrf2 signaling against the liver injury. Mechanistically, Mkp-1 upregulated Nrf2 through a direct interaction with the Neh2 domain in the transcription factor, while Nrf2 enhanced the expression of Mkp-1 mRNA by binding to the ARE site at -1719 to -1710bp in the Mkp-1 promoter. Our results reveal novel role of Mkp-1 in the maintenance of redox homeostasis in the liver. Thus, strategies aimed at augmenting Mkp-1 expression may be beneficial in protecting the liver and may provide novel therapeutic approaches to toxin-induced liver injury.
Collapse
Affiliation(s)
- Lin Luo
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Yeru Chen
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Hongyan Wang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China; Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Shengcun Wang
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Kaihua Liu
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xin Li
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Xiu Jun Wang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, PR China.
| | - Xiuwen Tang
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, PR China.
| |
Collapse
|
89
|
Fan H, Paiboonrungruan C, Zhang X, Prigge JR, Schmidt EE, Sun Z, Chen X. Nrf2 regulates cellular behaviors and Notch signaling in oral squamous cell carcinoma cells. Biochem Biophys Res Commun 2017; 493:833-839. [PMID: 28821430 PMCID: PMC6037169 DOI: 10.1016/j.bbrc.2017.08.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/13/2017] [Indexed: 11/18/2022]
Abstract
Oxidative stress is known to play a pivotal role in the development of oral squamous cell carcinoma (OSCC). We have demonstrated that activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway has chemopreventive effects against oxidative stress-associated OSCC. However, Nrf2 have dual roles in cancer development; while it prevents carcinogenesis of normal cells, hyperactive Nrf2 also promotes the survival of cancer cells. This study is aimed to understand the function of Nrf2 in regulating cellular behaviors of OSCC cells, and the potential mechanisms through which Nrf2 facilitates OSCC. We established the Nrf2-overexpressing and Nrf2-knockdown OSCC cell lines, and examined the function of Nrf2 in regulating cell proliferation, migration, invasion, cell cycle and colony formation. Our data showed that Nrf2 overexpression promoted cancer phenotypes in OSCC cells, whereas Nrf2 silencing inhibited these phenotypes. In addition, Nrf2 positively regulated Notch signaling pathway in OSCC cells in vitro. Consistent with this observation, Nrf2 activation in Keap1-/- mice resulted in not only hyperproliferation of squamous epithelial cells in mouse tongue as evidenced by increased expression of PCNA, but also activation of Notch signaling in these cells as evidenced by increased expression of NICD1 and Hes1. In conclusion, Nrf2 regulates cancer behaviors and Notch signaling in OSCC cells.
Collapse
Affiliation(s)
- Hong Fan
- Department of Oral Medicine, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | | | - Xinyan Zhang
- Department of Oral Medicine, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Justin R Prigge
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59717, USA
| | - Edward E Schmidt
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59717, USA
| | - Zheng Sun
- Department of Oral Medicine, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing 100050, China.
| | - Xiaoxin Chen
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
90
|
Dinkova-Kostova AT, Fahey JW, Kostov RV, Kensler TW. KEAP1 and Done? Targeting the NRF2 Pathway with Sulforaphane. Trends Food Sci Technol 2017; 69:257-269. [PMID: 29242678 PMCID: PMC5725197 DOI: 10.1016/j.tifs.2017.02.002] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/12/2017] [Accepted: 02/14/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Since the re-discovery of sulforaphane in 1992 and the recognition of the bioactivity of this phytochemical, many studies have examined its mode of action in cells, animals and humans. Broccoli, especially as young sprouts, is a rich source of sulforaphane and broccoli-based preparations are now used in clinical studies probing efficacy in health preservation and disease mitigation. Many putative cellular targets are affected by sulforaphane although only one, KEAP1-NRF2 signaling, can be considered a validated target at this time. The transcription factor NRF2 is a master regulator of cell survival responses to endogenous and exogenous stressors. SCOPE AND APPROACH This review summarizes the chemical biology of sulforaphane as an inducer of NRF2 signaling and efficacy as an inhibitor of carcinogenesis. It also provides a summary of the current findings from clinical trials using a suite of broccoli sprout preparations on a series of short-term endpoints reflecting a diversity of molecular actions. KEY FINDINGS AND CONCLUSIONS Sulforaphane, as a pure chemical, protects against chemical-induced skin, oral, stomach, colon, lung and bladder carcinogenesis and in genetic models of colon and prostate carcinogenesis. In many of these settings the antitumorigenic efficacy of sulforaphane is dampened in Nrf2-disrupted animals. Broccoli preparations rich in glucoraphanin or sulforaphane exert demonstrable pharmacodynamic action in over a score of clinical trials. Measures of NRF2 pathway response and function are serving as guideposts for the optimization of dose, schedule and formulation as clinical trials with broccoli-based preparations become more commonplace and more rigorous in design and implementation.
Collapse
Affiliation(s)
- Albena T. Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jed W. Fahey
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Human Nutrition, Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rumen V. Kostov
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W. Kensler
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
91
|
Guéraud F. 4-Hydroxynonenal metabolites and adducts in pre-carcinogenic conditions and cancer. Free Radic Biol Med 2017; 111:196-208. [PMID: 28065782 DOI: 10.1016/j.freeradbiomed.2016.12.025] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/14/2016] [Accepted: 12/17/2016] [Indexed: 12/22/2022]
Abstract
4-hydroxy-2-nonenal (HNE) is an amazing reactive compound, originating from lipid peroxidation within cells but also in food and considered as a "second messenger" of oxidative stress. Due to its chemical features, HNE is able to make covalent links with DNA, proteins and lipids. The aim of this review is to give a comprehensive summary of the chemical properties of HNE and of the consequences of its reactivity in relation to cancer development. The formation of exocyclic etheno-and propano-adducts and genotoxic effects are addressed. The adduction to cellular proteins and the repercussions on the regulation of cell signaling pathways involved in cancer development are reviewed, notably on the Nrf2/Keap1/ARE pathway. The metabolic pathways leading to the inactivation/elimination or, on the contrary, to the bioactivation of HNE are considered. A special focus is given on the link between HNE and colorectal cancer development, due to its occurrence in foodstuffs and in the digestive lumen, during digestion.
Collapse
Affiliation(s)
- Françoise Guéraud
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France.
| |
Collapse
|
92
|
NRF2 Activation Impairs Quiescence and Bone Marrow Reconstitution Capacity of Hematopoietic Stem Cells. Mol Cell Biol 2017; 37:MCB.00086-17. [PMID: 28674188 DOI: 10.1128/mcb.00086-17] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 06/27/2017] [Indexed: 12/12/2022] Open
Abstract
Tissue stem cells are maintained in quiescence under physiological conditions but proliferate and differentiate to replenish mature cells under stressed conditions. The KEAP1-NRF2 system plays an essential role in stress response and cytoprotection against redox disturbance. To clarify the role of the KEAP1-NRF2 system in tissue stem cells, we focused on hematopoiesis in this study and used Keap1-deficient mice to examine the effects of persistent activation of NRF2 on long-term hematopoietic stem cells (LT-HSCs). We found that persistent activation of NRF2 due to Keap1 deficiency did not change the number of LT-HSCs but reduced their quiescence in steady-state hematopoiesis. During hematopoietic regeneration after bone marrow (BM) transplantation, persistent activation of NRF2 reduced the BM reconstitution capacity of LT-HSCs, suggesting that NRF2 reduces the quiescence of LT-HSCs and promotes their differentiation, leading to eventual exhaustion. Transient activation of NRF2 by an electrophilic reagent also promotes the entry of LT-HSCs into the cell cycle. Taken together, our findings show that NRF2 drives the cell cycle entry and differentiation of LT-HSCs at the expense of their quiescence and maintenance, an effect that appears to be beneficial for prompt recovery from blood loss. We propose that the appropriate control of NRF2 activity by KEAP1 is essential for maintaining HSCs and guarantees their stress-induced regenerative response.
Collapse
|
93
|
Bachofner M, Speicher T, Bogorad RL, Muzumdar S, Derrer CP, Hürlimann F, Böhm F, Nanni P, Kockmann T, Kachaylo E, Meyer M, Padrissa-Altés S, Graf R, Anderson DG, Koteliansky V, Auf dem Keller U, Werner S. Large-Scale Quantitative Proteomics Identifies the Ubiquitin Ligase Nedd4-1 as an Essential Regulator of Liver Regeneration. Dev Cell 2017; 42:616-625.e8. [PMID: 28890072 DOI: 10.1016/j.devcel.2017.07.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 04/16/2017] [Accepted: 07/26/2017] [Indexed: 01/20/2023]
Abstract
The liver is the only organ in mammals that fully regenerates even after major injury. To identify orchestrators of this regenerative response, we performed quantitative large-scale proteomics analysis of cytoplasmic and nuclear fractions from normal versus regenerating mouse liver. Proteins of the ubiquitin-proteasome pathway were rapidly upregulated after two-third hepatectomy, with the ubiquitin ligase Nedd4-1 being a top hit. In vivo knockdown of Nedd4-1 in hepatocytes through nanoparticle-mediated delivery of small interfering RNA caused severe liver damage and inhibition of cell proliferation after hepatectomy, resulting in liver failure. Mechanistically, we demonstrate that Nedd4-1 is required for efficient internalization of major growth factor receptors involved in liver regeneration and their downstream mitogenic signaling. These results highlight the power of large-scale proteomics to identify key players in liver regeneration and the importance of posttranslational regulation of growth factor signaling in this process. Finally, they identify an essential function of Nedd4-1 in tissue repair.
Collapse
Affiliation(s)
- Marc Bachofner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Tobias Speicher
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Roman L Bogorad
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sukalp Muzumdar
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Carina P Derrer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Fabrizio Hürlimann
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Friederike Böhm
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Paolo Nanni
- Functional Genomics Center Zürich, University of Zürich/ETH Zürich, 8057 Zürich, Switzerland
| | - Tobias Kockmann
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland; Functional Genomics Center Zürich, University of Zürich/ETH Zürich, 8057 Zürich, Switzerland
| | - Ekaterina Kachaylo
- Swiss HPB Center, Division of Visceral and Transplantation Surgery, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Michael Meyer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Susagna Padrissa-Altés
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Rolf Graf
- Swiss HPB Center, Division of Visceral and Transplantation Surgery, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Health Science Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Victor Koteliansky
- Skolkovo Institute of Science and Technology, ul. Novaya, d.100, Skolkovo 143025, Russian Federation
| | - Ulrich Auf dem Keller
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland.
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland.
| |
Collapse
|
94
|
Cap-n-Collar Promotes Tissue Regeneration by Regulating ROS and JNK Signaling in the Drosophila melanogaster Wing Imaginal Disc. Genetics 2017; 206:1505-1520. [PMID: 28512185 DOI: 10.1534/genetics.116.196832] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/10/2017] [Indexed: 01/03/2023] Open
Abstract
Regeneration is a complex process that requires an organism to recognize and repair tissue damage, as well as grow and pattern new tissue. Here, we describe a genetic screen to identify novel regulators of regeneration. We ablated the Drosophila melanogaster larval wing primordium by inducing apoptosis in a spatially and temporally controlled manner and allowed the tissue to regenerate and repattern. To identify genes that regulate regeneration, we carried out a dominant-modifier screen by assessing the amount and quality of regeneration in adult wings heterozygous for isogenic deficiencies. We have identified 31 regions on the right arm of the third chromosome that modify the regenerative response. Interestingly, we observed several distinct phenotypes: mutants that regenerated poorly, mutants that regenerated faster or better than wild-type, and mutants that regenerated imperfectly and had patterning defects. We mapped one deficiency region to cap-n-collar (cnc), the Drosophila Nrf2 ortholog, which is required for regeneration. Cnc regulates reactive oxygen species levels in the regenerating epithelium, and affects c-Jun N-terminal protein kinase (JNK) signaling, growth, debris localization, and pupariation timing. Here, we present the results of our screen and propose a model wherein Cnc regulates regeneration by maintaining an optimal level of reactive oxygen species to promote JNK signaling.
Collapse
|
95
|
Alam MM, Okazaki K, Nguyen LTT, Ota N, Kitamura H, Murakami S, Shima H, Igarashi K, Sekine H, Motohashi H. Glucocorticoid receptor signaling represses the antioxidant response by inhibiting histone acetylation mediated by the transcriptional activator NRF2. J Biol Chem 2017; 292:7519-7530. [PMID: 28314773 DOI: 10.1074/jbc.m116.773960] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/17/2017] [Indexed: 12/30/2022] Open
Abstract
NRF2 (nuclear factor erythroid 2-related factor 2) is a key transcriptional activator that mediates the inducible expression of antioxidant genes. NRF2 is normally ubiquitinated by KEAP1 (Kelch-like ECH-associated protein 1) and subsequently degraded by proteasomes. Inactivation of KEAP1 by oxidative stress or electrophilic chemicals allows NRF2 to activate transcription through binding to antioxidant response elements (AREs) and recruiting histone acetyltransferase CBP (CREB-binding protein). Whereas KEAP1-dependent regulation is a major determinant of NRF2 activity, NRF2-mediated transcriptional activation varies from context to context, suggesting that other intracellular signaling cascades may impact NRF2 function. To identify a signaling pathway that modifies NRF2 activity, we immunoprecipitated endogenous NRF2 and its interacting proteins from mouse liver and identified glucocorticoid receptor (GR) as a novel NRF2-binding partner. We found that glucocorticoids, dexamethasone and betamethasone, antagonize diethyl maleate-induced activation of NRF2 target genes in a GR-dependent manner. Dexamethasone treatment enhanced GR recruitment to AREs without affecting chromatin binding of NRF2, resulting in the inhibition of CBP recruitment and histone acetylation at AREs. This repressive effect was canceled by the addition of histone deacetylase inhibitors. Thus, GR signaling decreases NRF2 transcriptional activation through reducing the NRF2-dependent histone acetylation. Consistent with these observations, GR signaling blocked NRF2-mediated cytoprotection from oxidative stress. This study suggests that an impaired antioxidant response by NRF2 and a resulting decrease in cellular antioxidant capacity account for the side effects of glucocorticoids, providing a novel viewpoint for the pathogenesis of hypercorticosteroidism.
Collapse
Affiliation(s)
- Md Morshedul Alam
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575
| | - Keito Okazaki
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575
| | - Linh Thi Thao Nguyen
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575
| | - Nao Ota
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575
| | - Hiroshi Kitamura
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575
| | - Shohei Murakami
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575
| | - Hiroki Shima
- the Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, and.,CREST, AMED, Sendai, Miyagi 980-8575, Japan
| | - Kazuhiko Igarashi
- the Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, and.,CREST, AMED, Sendai, Miyagi 980-8575, Japan
| | - Hiroki Sekine
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575,
| | - Hozumi Motohashi
- From the Department of Gene Expression Regulation, Institute of Development, Aging, and Cancer, Tohoku University, 4-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575,
| |
Collapse
|
96
|
Li W, Guo Y, Zhang C, Wu R, Yang AY, Gaspar J, Kong ANT. Dietary Phytochemicals and Cancer Chemoprevention: A Perspective on Oxidative Stress, Inflammation, and Epigenetics. Chem Res Toxicol 2016; 29:2071-2095. [PMID: 27989132 DOI: 10.1021/acs.chemrestox.6b00413] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxidative stress occurs when cellular reactive oxygen species levels exceed the self-antioxidant capacity of the body. Oxidative stress induces many pathological changes, including inflammation and cancer. Chronic inflammation is believed to be strongly associated with the major stages of carcinogenesis. The nuclear factor erythroid 2-related factor 2 (Nrf2) pathway plays a crucial role in regulating oxidative stress and inflammation by manipulating key antioxidant and detoxification enzyme genes via the antioxidant response element. Many dietary phytochemicals with cancer chemopreventive properties, such as polyphenols, isothiocyanates, and triterpenoids, exert antioxidant and anti-inflammatory functions by activating the Nrf2 pathway. Furthermore, epigenetic changes, including DNA methylation, histone post-translational modifications, and miRNA-mediated post-transcriptional alterations, also lead to various carcinogenesis processes by suppressing cancer repressor gene transcription. Using epigenetic research tools, including next-generation sequencing technologies, many dietary phytochemicals are shown to modify and reverse aberrant epigenetic/epigenome changes, potentially leading to cancer prevention/treatment. Thus, the beneficial effects of dietary phytochemicals on cancer development warrant further investigation to provide additional impetus for clinical translational studies.
Collapse
Affiliation(s)
- Wenji Li
- Center for Cancer Prevention Research, ‡Department of Pharmaceutics, §Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey 08854, United States
| | - Yue Guo
- Center for Cancer Prevention Research, ‡Department of Pharmaceutics, §Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey 08854, United States
| | - Chengyue Zhang
- Center for Cancer Prevention Research, ‡Department of Pharmaceutics, §Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey 08854, United States
| | - Renyi Wu
- Center for Cancer Prevention Research, ‡Department of Pharmaceutics, §Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey 08854, United States
| | - Anne Yuqing Yang
- Center for Cancer Prevention Research, ‡Department of Pharmaceutics, §Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey 08854, United States
| | - John Gaspar
- Center for Cancer Prevention Research, ‡Department of Pharmaceutics, §Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey 08854, United States
| | - Ah-Ng Tony Kong
- Center for Cancer Prevention Research, ‡Department of Pharmaceutics, §Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey 08854, United States
| |
Collapse
|
97
|
|
98
|
Alizai PH, Bertram L, Fragoulis A, Wruck CJ, Kroy DC, Klinge U, Neumann UP, Schmeding M. In vivo imaging of antioxidant response element activity during liver regeneration after partial hepatectomy. J Surg Res 2016; 206:525-535. [DOI: 10.1016/j.jss.2016.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/03/2016] [Accepted: 08/02/2016] [Indexed: 02/07/2023]
|
99
|
Palliyaguru DL, Chartoumpekis DV, Wakabayashi N, Skoko JJ, Yagishita Y, Singh SV, Kensler TW. Withaferin A induces Nrf2-dependent protection against liver injury: Role of Keap1-independent mechanisms. Free Radic Biol Med 2016; 101:116-128. [PMID: 27717869 PMCID: PMC5154810 DOI: 10.1016/j.freeradbiomed.2016.10.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/28/2016] [Accepted: 10/02/2016] [Indexed: 12/30/2022]
Abstract
Small molecules of plant origin offer presumptively safe opportunities to prevent carcinogenesis, mutagenesis and other forms of toxicity in humans. However, the mechanisms of action of such plant-based agents remain largely unknown. In recent years the stress responsive transcription factor Nrf2 has been validated as a target for disease chemoprevention. Withania somnifera (WS) is a herb used in Ayurveda (an ancient form of medicine in South Asia). In the recent past, withanolides isolated from WS, such as Withaferin A (WA) have been demonstrated to be preventive and therapeutic against multiple diseases in experimental models. The goals of this study are to evaluate withanolides such as WA as well as Withania somnifera root extract as inducers of Nrf2 signaling, to probe the underlying signaling mechanism of WA and to determine whether prevention of acetaminophen (APAP)-induced hepatic toxicity in mice by WA occurs in an Nrf2-dependent manner. We observed that WA profoundly protects wild-type mice but not Nrf2-disrupted mice against APAP hepatotoxicity. WA is a potent inducer of Nrf2-dependent cytoprotective enzyme expression both in vivo and in vitro. Unexpectedly, WA induces Nrf2 signaling at least in part, in a Keap1-independent, Pten/Pi3k/Akt-dependent manner in comparison to prototypical Nrf2 inducers, sulforaphane and CDDO-Im. The identification of WA as an Nrf2 inducer that can signal through a non-canonical, Keap1-independent pathway provides an opportunity to evaluate the role of other regulatory partners of Nrf2 in the dietary and pharmacological induction of Nrf2-mediated cytoprotection.
Collapse
Affiliation(s)
- Dushani L Palliyaguru
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dionysios V Chartoumpekis
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nobunao Wakabayashi
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John J Skoko
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yoko Yagishita
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shivendra V Singh
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas W Kensler
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
100
|
Nrf2 and Notch Signaling in Lung Cancer: Near the Crossroad. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7316492. [PMID: 27847554 PMCID: PMC5099458 DOI: 10.1155/2016/7316492] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 07/08/2016] [Accepted: 09/20/2016] [Indexed: 01/01/2023]
Abstract
The transcription factor Nrf2 (NF-E2 related factor 2) is a master regulator of the cell antioxidant response associated with tumor growth and resistance to cytotoxic treatments. In particular, Nrf2 induces upregulation of cytoprotective genes by interacting with the closely situated AREs (Antioxidant Response Elements) in response to endogenous or exogenous stress stimuli and takes part to several oncogenic signaling pathways. Among these, the crosstalk with Notch pathway has been shown to enhance cytoprotection and maintenance of cellular homeostasis, tissue organization by modulating cell proliferation kinetics, and stem cell self-renewal in several organs. The role of Notch and Nrf2 related pathways in tumorigenesis is highly variable and when they are both abnormally activated they can synergistically cause neoplastic proliferation by promoting cell survival, differentiation, invasion, and metastases. NFE2L2, KEAP1, and NOTCH genes family appear in the list of significantly mutated genes in tumors in both combined and individual sets, supporting the crucial role that the aberrant Nrf2-Notch crosstalk might have in cancerogenesis. In this review, we summarize current knowledge about the alterations of Nrf2 and Notch pathways and their reciprocal transcriptional regulation throughout tumorigenesis and progression of lung tumors, supporting the potentiality of putative biomarkers and therapeutic targets.
Collapse
|