201
|
Quassinoid analogs with enhanced efficacy for treatment of hematologic malignancies target the PI3Kγ isoform. Commun Biol 2020; 3:267. [PMID: 32461675 PMCID: PMC7253423 DOI: 10.1038/s42003-020-0996-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
Development of novel PI3K inhibitors is an important strategy to overcome their resistance and poor tolerability in clinical trials. The quassinoid family member Brusatol shows specific inhibitory activity against hematologic malignancies. However, the mechanism of its anti-cancer activity is unknown. We investigated the anti-cancer activity of Brusatol on multiple hematologic malignancies derived cell lines. The results demonstrated that the PI3Kγ isoform was identified as a direct target of Brusatol, and inhibition was dramatically reduced on cells with lower PI3Kγ levels. Novel synthetic analogs were also developed and tested in vitro and in vivo. They shared comparable or superior potency in their ability to inhibit malignant hematologic cell lines, and in a xenograft transplant mouse model. One unique analog had minimal toxicity to normal human cells and in a mouse model. These new analogs have enhanced potential for development as a new class of PI3K inhibitors for treatment of hematologic malignancies. Pei et al. demonstrate that PI3Kγ isoform is a direct target of Brusatol, a natural compound with inhibitory activity against hematologic malignancies. They further develop several Brusatol analogs with superior in vitro and in vivo anti-cancer activity.
Collapse
|
202
|
Guo S, Zhang J, Wei C, Lu Z, Cai R, Pan D, Zhang H, Liang B, Zhang Z. Anticancer effects of brusatol in nasopharyngeal carcinoma through suppression of the Akt/mTOR signaling pathway. Cancer Chemother Pharmacol 2020; 85:1097-1108. [PMID: 32449143 DOI: 10.1007/s00280-020-04083-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 05/13/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Brusatol, a natural quassinoid that is isolated from a traditional Chinese herbal medicine known as Bruceae Fructus, possesses biological activity in various types of human cancers, but its effects in nasopharyngeal carcinoma (NPC) have not been reported. This study aimed to explore the effect and molecular mechanism of brusatol in NPC in vivo and in vitro. METHODS The antiproliferative effect of brusatol was assessed by MTT and colony formation assays. Apoptosis was determined by flow cytometry. The expression of mitochondrial apoptosis, cell cycle arrest, and Akt/mTOR pathway proteins were determined by western blot analysis. Further in vivo confirmation was performed in a nude mouse model. RESULTS Brusatol showed antiproliferative activity against four human NPC cell lines (CNE-1, CNE-2, 5-8F, and 6-10B) in a dose-dependent manner. This antiproliferative effect was accompanied by mitochondrial apoptosis and cell cycle arrest through the modulation of several key molecular targets, such as Bcl-xl, Bcl-2, Bad, Bax, PARP, Caspase-9, Caspase-7, Caspase-3, Cdc25c, Cyclin B1, Cdc2 p34, and Cyclin D1. In addition, we found that brusatol inhibited the activation of Akt, mTOR, 4EBP1, and S6K, suggesting that the Akt/mTOR pathway is a key underlying mechanism by which brusatol inhibits growth and promotes apoptosis. Further in vivo nude mouse models proved that brusatol significantly inhibited the growth of CNE-1 xenografts with no significant toxicity. CONCLUSIONS These observations indicate that brusatol is a promising antitumor drug candidate or a supplement to current chemotherapeutic therapies to treat NPC.
Collapse
Affiliation(s)
- Songbin Guo
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, Guangdong, People's Republic of China
| | - Jinling Zhang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Cairong Wei
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, Guangdong, People's Republic of China
| | - Zhiyong Lu
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, Guangdong, People's Republic of China
| | - Rulong Cai
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, Guangdong, People's Republic of China
| | - Danqi Pan
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, Guangdong, People's Republic of China
| | - Hanbin Zhang
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, Guangdong, People's Republic of China
| | - Baoxia Liang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Zhenfeng Zhang
- The Second Clinical Medical College, Guangzhou Medical University, Guangzhou, 511436, Guangdong, People's Republic of China. .,Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
203
|
Copper-imidazo[1,2-a]pyridines induce intrinsic apoptosis and modulate the expression of mutated p53, haem-oxygenase-1 and apoptotic inhibitory proteins in HT-29 colorectal cancer cells. Apoptosis 2020; 24:623-643. [PMID: 31073781 DOI: 10.1007/s10495-019-01547-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Metastatic colorectal cancer responds poorly to treatment and is a leading cause of cancer related deaths. Worldwide, chemotherapy of metastatic colorectal cancer remains plagued by poor efficacy, development of resistance and serious adverse effects. Copper-imidazo[1,2-a]pyridines were previously shown by our group to be selectively active against several cancer cell lines, with three complexes, JD46(27), JD47(29), and JD88(21), showing IC50 values between 0.8 and 1.8 μM against HT-29 cells. Here, we report that treatment with the copper complexes resulted in fragmented nuclei suggestive of apoptotic cell death, which was confirmed by increased annexin V binding and caspase-3/7 activity. The copper complexes caused a loss of mitochondrial membrane potential and increased caspase-9 activity. The absence of caspase-8 activity indicated activation of the intrinsic pathway. Proteomic analysis revealed that copper-imidazo[1,2-a]pyridines decreased the expression of phosphorylated forms of p53 [phospho-p53(S15), phospho-p53(S46) and phospho-p53(S392)]. The expression of inhibitor of apoptosis proteins, XIAP, cIAP1, livin, and the antiapoptotic proteins, Bcl-2 and Bcl-x, was decreased. HO/HMOX/HSP32, expression was notably increased, which suggested the accumulation of reactive oxygen species. Increased expression of TRAIL-R2/DR5 death receptor indicated the possible dual activation of both the extrinsic and intrinsic apoptotic pathways; however, caspase-8 activation could not be demonstrated. In conclusion, the copper-imidazo[1,2-a]pyridines were effective inducers of apoptotic cell death at low micromolar concentrations and changed the expression levels of proteins important for cell survival and cell death. These copper complexes may be useful tools to better understand the complexity of signalling networks in cancer cell death in response to cell stress.
Collapse
|
204
|
Tang KY, Du SL, Wang QL, Zhang YF, Song HY. Traditional Chinese medicine targeting cancer stem cells as an alternative treatment for hepatocellular carcinoma. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2020; 18:196-202. [DOI: 10.1016/j.joim.2020.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023]
|
205
|
Molecular mechanisms and systemic targeting of NRF2 dysregulation in cancer. Biochem Pharmacol 2020; 177:114002. [PMID: 32360363 DOI: 10.1016/j.bcp.2020.114002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
NF-E2-related factor 2 (NRF2) is a master regulator of redox homeostasis and provides cellular protection against oxidants and electrophiles by inducing the expression of a wide array of phase II cytoprotective genes. Until now, a number of NRF2 activators have been developed for treatment of chronic diseases and some are under evaluation in the clinical studies. On the other hand, accumulating evidence indicates that NRF2 confers chemoresistance and radioresistance, and its expression is correlated with poor prognosis in cancer patients. Studies in the last decade demonstrate that diverse mechanisms such as somatic mutations, accumulation of KEAP1 binding proteins, transcriptional dysregulation, oncogene activation, and accumulation of reactive metabolites contribute to NRF2 activation in cancer. In the present review, we illustrate the molecular mechanisms governing the function of NRF2 and explain how they are hijacked in cancer. We also provide some examples of NRF2 inhibitors together with a brief explanation of their mechanisms of action.
Collapse
|
206
|
Smith RE. The Effects of Dietary Supplements that Overactivate the Nrf2/ARE System. Curr Med Chem 2020; 27:2077-2094. [DOI: 10.2174/0929867326666190517113533] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 01/31/2019] [Accepted: 04/15/2019] [Indexed: 02/07/2023]
Abstract
Background:
Inflammation is one of the most misunderstood aspects of human
health. People have been encouraged to eat foods that have a high antioxidant capacity, and in
vitro tests for total antioxidant capacity emerged. They were based on measuring the destruction
of oxidized test compounds in direct reactions with the antioxidants in foods. Many dietary
supplements arrived in the market. They contained purified antioxidants, such as resveratrol
and EGCG that were and still are widely assumed by many to be quite healthy at any
dose.
Methods:
The literature on inflammation and the Nrf2/ARE antioxidant system was searched
systematically. Articles from prestigious, peer-reviewed journals were obtained and read. The
information obtained from them was used to write this review article.
Results:
Over 150 articles and books were read. The information obtained from them showed
that very few dietary antioxidants exert their effects by reacting directly with Reactive Oxygen
and Nitrogen Species (RONS). Instead, most of the effective antioxidants activate the endogenous
Nrf2/ARE antioxidant system. This helps prevent smoldering inflammation and the
diseases that it can cause. However, when overactivated or activated constitutively, the
Nrf2/ARE antioxidant system can cause some of these diseases, including many types of
multidrug resistant cancer, autoimmune, neurodegenerative and cardiovascular diseases.
Conclusion:
Even though green tea, as well as many fruits, vegetables and spices are quite
healthy, dietary supplements that deliver much higher doses of antioxidants may not be. People
who are diagnosed with cancer and plan to start chemotherapy and/or radiotherapy should
probably avoid such supplements. This is because multidrug resistant tumors can hijack and
overactivate the Nrf2/ARE antioxidant system.
Collapse
|
207
|
Triptolide suppresses IDH1-mutated malignancy via Nrf2-driven glutathione metabolism. Proc Natl Acad Sci U S A 2020; 117:9964-9972. [PMID: 32312817 DOI: 10.1073/pnas.1913633117] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Isocitrate dehydrogenase (IDH) mutation is a common genetic abnormality in human malignancies characterized by remarkable metabolic reprogramming. Our present study demonstrated that IDH1-mutated cells showed elevated levels of reactive oxygen species and higher demands on Nrf2-guided glutathione de novo synthesis. Our findings showed that triptolide, a diterpenoid epoxide from Tripterygium wilfordii, served as a potent Nrf2 inhibitor, which exhibited selective cytotoxicity to patient-derived IDH1-mutated glioma cells in vitro and in vivo. Mechanistically, triptolide compromised the expression of GCLC, GCLM, and SLC7A11, which disrupted glutathione metabolism and established synthetic lethality with reactive oxygen species derived from IDH1 mutant neomorphic activity. Our findings highlight triptolide as a valuable therapeutic approach for IDH1-mutated malignancies by targeting the Nrf2-driven glutathione synthesis pathway.
Collapse
|
208
|
Guan X, Yuan Y, Wang G, Zheng R, Zhang J, Dong B, Ran N, Hsu ACY, Wang C, Wang F. Ginsenoside Rg3 ameliorates acute exacerbation of COPD by suppressing neutrophil migration. Int Immunopharmacol 2020; 83:106449. [PMID: 32278128 DOI: 10.1016/j.intimp.2020.106449] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Acute Exacerbation of Chronic Obstructive Pulmonary Disease (AECOPD) is an irreversible inflammatory airways disease responsible for global health burden, involved with a complex condition of immunological change. Exacerbation-mediated neutrophilia is an important factor in the pathogenesis of cigarette smoke-induced AECOPD. Ginsenoside Rg3, a red-ginseng-derived compound, has multiple pharmacological properties such as anti-inflammatory and antitumor activities. Here, we investigated a protective role of Rg3 against AECOPD, focusing on neutrophilia. 14-week-cigarette smoke (CS) exposure and non-typeable Haemophilus inflenzae (NTHi) infection were used to establish the AECOPD murine model. Rg3 (10, 20, 40 mg/kg) was administered intragastrically from the 12th week of CS exposure before infection, and this led to improved lung function and lung morphology, and reduced neutrophilic inflammation, indicating a suppressive effect on neutrophil infiltration by Rg3. Further investigations on the mechanism of Rg3 on neutrophils were carried out using bronchial epithelial cell (BEAS-2B) and neutrophil co-culture and transepithelial migration model. Pre-treatment of neutrophils with Rg3 reduced neutrophil migration, which seemed to be the result of inhibition of phosphatidylinositol (PtdIns) 3-kinases (PI3K) activation within neutrophils. Thus, Rg3 could inhibit exacerbation-induced neutrophilia in COPD by negatively regulating PI3K activities in neutrophils. This study provides a potential natural drug against AECOPD neutrophil inflammation.
Collapse
Affiliation(s)
- Xuewa Guan
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yuze Yuan
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Guoqiang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ruipeng Zheng
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Invasive Technology, First Hospital of Jilin University, Changchun 130021, China
| | - Jing Zhang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Intensive Care Unit, First Hospital of Jilin University, Changchun 130021, China
| | - Bing Dong
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Nan Ran
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Alan Chen-Yu Hsu
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and the University of Newcastle, NSW, Australia
| | - Cuizhu Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Key laboratory of Zoonosis Research Ministry of Education, Jilin University, Changchun 130021, China.
| |
Collapse
|
209
|
Zhang ZW, Liang J, Yan JX, Ye YC, Wang JJ, Chen C, Sun HT, Chen F, Tu Y, Li XH. TBHQ improved neurological recovery after traumatic brain injury by inhibiting the overactivation of astrocytes. Brain Res 2020; 1739:146818. [PMID: 32275911 DOI: 10.1016/j.brainres.2020.146818] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is a major leading cause of death and long-term disability. Although astrocytes play a key role in neuroprotection after TBI in the early stage, the overactivation of astrocytes can lead to long-term functional deficits, and the underlying pathophysiological mechanisms remain unclear. In addition, it is unknown whether the nuclear factor erythroid 2-related factor2/haem oxygenase-1 (Nrf-2/HO-1) pathway could elicit a neuroprotective effect by decreasing astrocyte overactivation after TBI. We aimed to study the effects of tert-butylhydroquinone (TBHQ) in reducing astrocyte overactivation after TBI and explored the underlying mechanisms. We first established a controlled cortical impact (CCI) model in rats and performed Haematoxylin and eosin (H&E) staining to observe brain tissue damage. The cognitive function of rats was assessed by modified neurological severity scoring (mNSS) and Morris water maze (MWM) test. Astrocyte and microglia activation was detected by immunofluorescence staining. Oxidative stress conditions were investigated using Western blotting. An enzyme-linked immunosorbent assay (ELISA) was designed to assess the level of the proinflammatory factor tumour necrosis factor-alpha (TNF-α). Dihydroethidium (DHE) staining was used to detect reactive oxygen species (ROS). Apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The results showed that the administration of TBHQ ameliorated motor function and cognitive deficits and decreased the lesion volume. In addition, TBHQ significantly decreased astrocyte overactivation, diminished the pro-inflammatory phenotype M1 and inflammatory cytokines production after TBI, increased Nrf-2 nuclear accumulation, and enhanced the levels of the Nrf-2 downstream antioxidative genes HO-1 and NADPH-quinone oxidoreductase-1 (NQO-1). Furthermore, TBHQ treatment alleviated apoptosis and neuronal death in the cerebral cortex. Overall, our data indicated that the upregulation of Nrf-2 expression could enhance neuroprotection and decrease astrocyte overactivation and might represent a new theoretical basis for treating TBI.
Collapse
Affiliation(s)
- Zhen-Wen Zhang
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jun Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Xing Yan
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Yi-Chao Ye
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Chong Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Feng Chen
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yue Tu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China.
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
210
|
Progressive Rotavirus Infection Downregulates Redox-Sensitive Transcription Factor Nrf2 and Nrf2-Driven Transcription Units. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7289120. [PMID: 32322337 PMCID: PMC7165344 DOI: 10.1155/2020/7289120] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/31/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022]
Abstract
Eukaryotic cells adopt highly tuned stress response physiology under threats of exogenous stressors including viruses to maintain cellular homeostasis. Not surprisingly, avoidance of cellular stress response pathways is an essential facet of virus-induced obligatory host reprogramming to invoke a cellular environment conducive to viral perpetuation. Adaptive cellular responses to oxidative and electrophilic stress are usually taken care of by an antioxidant defense system, core to which lies the redox-responsive transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and Nrf2-driven transcriptional cascade. Deregulation of host redox balance and redox stress-sensitive Nrf2 antioxidant defense have been reported for many viruses. In the current study, we aimed to study the modulation of the Nrf2-based host cellular redox defense system in response to Rotavirus (RV) infection in vitro. Interestingly, we found that Nrf2 protein levels decline sharply with progression of RV infection beyond an initial upsurge. Moreover, Nrf2 decrease as a whole was found to be accompanied by active nuclear vacuity of Nrf2, resulting in lowered expression of stress-responsive Nrf2 target genes heme oxygenase-1 (HO-1), NAD(P)H quinone dehydrogenase 1, and superoxide dismutase 1 both in the presence and absence of Nrf2-driven transcriptional inducers. Initial induction of Nrf2 concurred with RV-induced early burst of oxidative stress and therefore was sensitive to treatments with antioxidants. Reduction of Nrf2 levels beyond initial hours, however, was found to be independent of the cellular redox status. Furthermore, increasing the half-life of Nrf2 through inhibition of the Kelch-like erythroid cell-derived protein with CNC homology- (ECH-) associated protein 1/Cullin3-RING Box1-based canonical Nrf2 turnover pathway could not restore Nrf2 levels post RV-SA11 infection. Depletion of the Nrf2/HO-1 axis was subsequently found to be sensitive to proteasome inhibition with concurrent observation of increased K48-linked ubiquitination associated with Nrf2. Together, the present study describes robust downregulation of Nrf2-dependent cellular redox defense beyond initial hours of RV infection, justifying our previous observation of potent antirotaviral implications of Nrf2 agonists.
Collapse
|
211
|
Vernier M, Dufour CR, McGuirk S, Scholtes C, Li X, Bourmeau G, Kuasne H, Park M, St-Pierre J, Audet-Walsh E, Giguère V. Estrogen-related receptors are targetable ROS sensors. Genes Dev 2020; 34:544-559. [PMID: 32079653 PMCID: PMC7111261 DOI: 10.1101/gad.330746.119] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/21/2020] [Indexed: 12/17/2022]
Abstract
Excessive reactive oxygen species (ROS) can cause oxidative stress and consequently cell injury contributing to a wide range of diseases. Addressing the critical gaps in our understanding of the adaptive molecular events downstream ROS provocation holds promise for the identification of druggable metabolic vulnerabilities. Here, we unveil a direct molecular link between the activity of two estrogen-related receptor (ERR) isoforms and the control of glutamine utilization and glutathione antioxidant production. ERRα down-regulation restricts glutamine entry into the TCA cycle, while ERRγ up-regulation promotes glutamine-driven glutathione production. Notably, we identify increased ERRγ expression/activation as a hallmark of oxidative stress triggered by mitochondrial disruption or chemotherapy. Enhanced tumor antioxidant capacity is an underlying feature of human breast cancer (BCa) patients that respond poorly to treatment. We demonstrate that pharmacological inhibition of ERRγ with the selective inverse agonist GSK5182 increases antitumor efficacy of the chemotherapeutic paclitaxel on poor outcome BCa tumor organoids. Our findings thus underscore the ERRs as novel redox sensors and effectors of a ROS defense program and highlight the potential therapeutic advantage of exploiting ERRγ inhibitors for the treatment of BCa and other diseases where oxidative stress plays a central role.
Collapse
Affiliation(s)
- Mathieu Vernier
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Catherine R Dufour
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Shawn McGuirk
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Charlotte Scholtes
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Xiaojing Li
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Guillaume Bourmeau
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Hellen Kuasne
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec H3G 1Y6, Canada
- Department of Medicine, McGill University, Montréal, Quebec H3G 1Y6, Canada
- Department of Oncology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Julie St-Pierre
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec H3G 1Y6, Canada
| | - Etienne Audet-Walsh
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec H3G 1Y6, Canada
- Department of Medicine, McGill University, Montréal, Quebec H3G 1Y6, Canada
- Department of Oncology, McGill University, Montréal, Quebec H3G 1Y6, Canada
| |
Collapse
|
212
|
Noman ASM, Parag RR, Rashid MI, Rahman MZ, Chowdhury AA, Sultana A, Jerin C, Siddiqua A, Rahman L, Shirin A, Nayeem J, Mahmud R, Akther S, Shil RK, Hossain I, Alam S, Chowdhury A, Basher SB, Hasan A, Bithy S, Aklima J, Rahman M, Chowdhury N, Banu T, Karakas B, Yeger H, Farhat WA, Islam SS. Widespread expression of Sonic hedgehog (Shh) and Nrf2 in patients treated with cisplatin predicts outcome in resected tumors and are potential therapeutic targets for HPV-negative head and neck cancer. Ther Adv Med Oncol 2020; 12:1758835920911229. [PMID: 32206093 PMCID: PMC7074584 DOI: 10.1177/1758835920911229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/29/2020] [Indexed: 01/31/2023] Open
Abstract
Background: Sonic hedgehog (Shh) and Nrf2 play a critical role in chemotherapeutic resistance. These two genes have been found to be dysregulated in head and neck squamous cell carcinomas (HNSCC). The purpose of this study was to analyze the expression, function and clinical prognostic relationship of Shh and Nrf2 in HNSCC in the context of therapeutic resistance and cancer stem cells (CSCs). Methods: We analyzed a cohort of patients with HNSCC to identify potential therapeutic biomarkers correlating with overall survival (OS) as well as disease-free survival (DFS) from our own data and validated these results using The Cancer Genome Atlas dataset. Expression of Shh and Nrf2 was knocked down by siRNA and cell growth, sphere growth and chemotherapeutic resistance were evaluated. Results: Widespread abundant expression of Shh and Nrf2 proteins were associated with shorter OS and DFS. The combination of Shh and Nrf2 expression levels was found to be a significant predictor of patient DFS. The tumor stromal index was correlated with Shh expression and inversely associated with shorter OS and DFS. Inhibition of Shh by siRNA or cyclopamine resulted in the attenuation of resistant CSC self-renewal, invasion, clonogenic growth and re-sensitization to the chemotherapeutic agents. Concomitant upregulation of Shh and Nrf2 proved to be an independent predictor of poor OS and DFS in patients with HNSCC. Conclusions: These findings suggest that Shh and Nrf2 could serve as therapeutic targets as well as promising dual prognostic therapeutic biomarkers for HNSCC.
Collapse
Affiliation(s)
- Abu Shadat M Noman
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Rashed R Parag
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Muhammad I Rashid
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Mohammad Z Rahman
- Department of Pathology, Chittagong Medical College and Hospital, Chittagong, Bangladesh
| | - Ali A Chowdhury
- Department of Radiotherapy, Chittagong Medical College and Hospital, Chittagong, Bangladesh
| | - Afrin Sultana
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Chandsultana Jerin
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Ayesha Siddiqua
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Lutfur Rahman
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Afsana Shirin
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Junayed Nayeem
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Reaz Mahmud
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Sonam Akther
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Rajib K Shil
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Ikram Hossain
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Sharmin Alam
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Arfina Chowdhury
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Shabnam B Basher
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Abul Hasan
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Shammy Bithy
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Jannatul Aklima
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Mizanur Rahman
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Nabila Chowdhury
- Department of Biochemistry and Molecular Biology, The University of Chittagong, Chittagong, Bangladesh
| | - Tahmina Banu
- Chittagong Research Institute of Children Surgery, Chittagong, Bangladesh
| | - Bedri Karakas
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Herman Yeger
- Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Walid A Farhat
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Syed S Islam
- Department of Molecular Oncology, Cancer Biology and Experimental Therapeutics, King Faisal Specialist Hospital and Research Centre, School of Medicine, Alfaisal University, Thakassussi Street, Riyadh, 11211, Saudi Arabia
| |
Collapse
|
213
|
Wu WL, Papagiannakopoulos T. The Pleiotropic Role of the KEAP1/NRF2 Pathway in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2020. [DOI: 10.1146/annurev-cancerbio-030518-055627] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The unregulated proliferative capacity of many tumors is dependent on dysfunctional nutrient utilization and ROS (reactive oxygen species) signaling to sustain a deranged metabolic state. Although it is clear that cancers broadly rely on these survival and signaling pathways, how they achieve these aims varies dramatically. Mutations in the KEAP1/NRF2 pathway represent a potent cancer adaptation to exploit native cytoprotective pathways that involve both nutrient metabolism and ROS regulation. Despite activating these advantageous processes, mutations within KEAP1/ NRF2 are not universally selected for across cancers and instead appear to interact with particular tumor driver mutations and tissues of origin. Here, we highlight the relationship between the KEAP1/NRF2 signaling axis and tumor biology with a focus on genetic mutation, metabolism, immune regulation, and treatment implications and opportunities. Understanding the dysregulation of KEAP1 and NRF2 provides not only insight into a commonly mutated tumor suppressor pathway but also a window into the factors dictating the development and evolution of many cancers.
Collapse
Affiliation(s)
- Warren L. Wu
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
214
|
Schmidlin CJ, Dodson MB, Zhang DD. Filtering through the role of NRF2 in kidney disease. Arch Pharm Res 2020; 43:361-369. [PMID: 31372933 PMCID: PMC6994339 DOI: 10.1007/s12272-019-01177-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023]
Abstract
Kidney disease affects ~ 10% of the population worldwide, resulting in millions of deaths each year. Mechanistically, oxidative stress is a major driver of various kidney diseases, and promotes the progression from acute to chronic injury, as well as renal cancer development. NRF2, the master regulator of redox balance, has been shown to protect against kidney disease through its negation of reactive oxygen species (ROS). However, many kidney diseases exhibit high levels of ROS as a result of decreased NRF2 protein levels and transcriptional activity. Many studies have tested the strategy of using NRF2 inducing compounds to alleviate ROS to prevent or slow down the progression of kidney diseases. Oppositely, in specific subsets of renal cancer, NRF2 is constitutively activated and contributes to tumor burden and overall poor prognosis; therefore, there has been a recent interest in studies investigating the benefits of NRF2 inhibition. In this review, we summarize recent literature investigating the role of NRF2 and oxidative stress in various kidney diseases, and how pharmacological modification of NRF2 signaling could play a protective role.
Collapse
Affiliation(s)
- Cody J Schmidlin
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Matthew B Dodson
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
215
|
Potential Applications of NRF2 Modulators in Cancer Therapy. Antioxidants (Basel) 2020; 9:antiox9030193. [PMID: 32106613 PMCID: PMC7139512 DOI: 10.3390/antiox9030193] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/17/2023] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (NRF2)-Kelch-like ECH-associated protein 1 (KEAP1) regulatory pathway plays an essential role in protecting cells and tissues from oxidative, electrophilic, and xenobiotic stress. By controlling the transactivation of over 500 cytoprotective genes, the NRF2 transcription factor has been implicated in the physiopathology of several human diseases, including cancer. In this respect, accumulating evidence indicates that NRF2 can act as a double-edged sword, being able to mediate tumor suppressive or pro-oncogenic functions, depending on the specific biological context of its activation. Thus, a better understanding of the mechanisms that control NRF2 functions and the most appropriate context of its activation is a prerequisite for the development of effective therapeutic strategies based on NRF2 modulation. In line of principle, the controlled activation of NRF2 might reduce the risk of cancer initiation and development in normal cells by scavenging reactive-oxygen species (ROS) and by preventing genomic instability through decreased DNA damage. In contrast however, already transformed cells with constitutive or prolonged activation of NRF2 signaling might represent a major clinical hurdle and exhibit an aggressive phenotype characterized by therapy resistance and unfavorable prognosis, requiring the use of NRF2 inhibitors. In this review, we will focus on the dual roles of the NRF2-KEAP1 pathway in cancer promotion and inhibition, describing the mechanisms of its activation and potential therapeutic strategies based on the use of context-specific modulation of NRF2.
Collapse
|
216
|
Arefin S, Buchanan S, Hobson S, Steinmetz J, Alsalhi S, Shiels PG, Kublickiene K, Stenvinkel P. Nrf2 in early vascular ageing: Calcification, senescence and therapy. Clin Chim Acta 2020; 505:108-118. [PMID: 32097628 DOI: 10.1016/j.cca.2020.02.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022]
Abstract
Under normal physiological conditions, free radical generation and antioxidant defences are balanced, and reactive oxygen species (ROS) usually act as secondary messengers in a plethora of biological processes. However, when this balance is impaired, oxidative stress develops due to imbalanced redox homeostasis resulting in cellular damage. Oxidative stress is now recognized as a trigger of cellular senescence, which is associated with multiple chronic 'burden of lifestyle' diseases, including atherosclerosis, type-2 diabetes, chronic kidney disease and vascular calcification; all of which possess signs of early vascular ageing. Nuclear factor erythroid 2-related factor 2 (Nrf2), termed the master regulator of antioxidant responses, is a transcription factor found to be frequently dysregulated in conditions characterized by oxidative stress and inflammation. Recent evidence suggests that activation of Nrf2 may be beneficial in protecting against vascular senescence and calcification. Both natural and synthetic Nrf2 agonists have been introduced as promising drug classes in different phases of clinical trials. However, overexpression of the Nrf2 pathway has also been linked to tumorigenesis, which highlights the requirement for further understanding of pathways involving Nrf2 activity, especially in the context of cellular senescence and vascular calcification. Therefore, comprehensive translational pre-clinical and clinical studies addressing the targeting capabilities of Nrf2 agonists are urgently required. The present review discusses the impact of Nrf2 in senescence and calcification in early vascular ageing, with focus on the potential clinical implications of Nrf2 agonists and non-pharmacological Nrf2 therapeutics.
Collapse
Affiliation(s)
- Samsul Arefin
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Sarah Buchanan
- Institute of Cancer Sciences, Wolfson Wohl CRC, ICS, MVLS, University of Glasgow, Glasgow, UK
| | - Sam Hobson
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Julia Steinmetz
- Rheumatology Unit, Dep. of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Shno Alsalhi
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden; Research Center, Salahaddin University-Erbil, 44001 Erbil, Kurdistan-Region, Iraq
| | - Paul G Shiels
- Institute of Cancer Sciences, Wolfson Wohl CRC, ICS, MVLS, University of Glasgow, Glasgow, UK
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Karolinska University Hospital, 14186 Stockholm, Sweden.
| |
Collapse
|
217
|
Zhang X, Lai W, Ying X, Xu L, Chu K, Brown J, Chen L, Hong G. Salidroside Reduces Inflammation and Brain Injury After Permanent Middle Cerebral Artery Occlusion in Rats by Regulating PI3K/PKB/Nrf2/NFκB Signaling Rather than Complement C3 Activity. Inflammation 2020; 42:1830-1842. [PMID: 31230155 DOI: 10.1007/s10753-019-01045-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Salidroside, an active constituent of Rhodiola rosea, is neuroprotective after transient middle cerebral artery occlusion (tMCAO). However, its effects in other experimental stroke models are less understood. Here, we investigated the effect of daily intraperitoneal injections of salidroside in rats after permanent MCAO (pMCAO). Cerebral infarct volumes at 1 day after pMCAO were significantly reduced by treatment with 100 mg/kg/day salidroside, but not by 25 or 50 mg/kg/day, and this benefit of salidroside increased significantly over at least 7 days of treatment, when it was also accompanied by decreased neurological deficit scores. These observations led us to investigate the underlying mechanism of action of salidroside. 100 mg/kg salidroside for 1 day increased NeuN, Nrf2, and its downstream mediator HO-1, while it reduced nuclear NFκB p50, IL-6, and TNFα. Brusatol, a Nrf2 inhibitor, blocked the actions of salidroside on Nrf2, NFκB p50, IL-6, and TNFα. Salidroside also increased the ratio of p-PKB/PKB at 1 day after pMCAO even in the presence of brusatol. LY294002, a PI3K inhibitor, prevented all these effects of salidroside, including those on NeuN, p-PKB/PKB, Nrf2, HO-1, and pro-inflammatory mediators. In contrast, salidroside had no significant effect on the level of cerebral complement C3 after pMCAO, or on the activity of C3 as measured by the expression of cerebral Egr1. Our findings therefore suggest that salidroside reduces neuroinflammation and neural damage by regulating the PI3K/PKB/Nrf2/NFκB signaling pathway after pMCAO, and that this neuroprotective effect does not involve modulation of complement C3 activity.
Collapse
Affiliation(s)
- X Zhang
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China
| | - W Lai
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China
| | - X Ying
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China
| | - L Xu
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China
| | - K Chu
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China
| | - J Brown
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China
| | - L Chen
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China
| | - G Hong
- Centre of Biomedical Research & Development, Fujian University of Traditional Chinese Medicine, No. 1 Huatou Road, Minhou Shangjie, Fuzhou, China.
| |
Collapse
|
218
|
Iuchi K, Tasaki Y, Shirai S, Hisatomi H. Upregulation of nuclear factor (erythroid-derived 2)-like 2 protein level in the human colorectal adenocarcinoma cell line DLD-1 by a heterocyclic organobismuth(III) compound: Effect of organobismuth(III) compound on NRF2 signaling. Biomed Pharmacother 2020; 125:109928. [PMID: 32004978 DOI: 10.1016/j.biopha.2020.109928] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/09/2019] [Accepted: 12/18/2019] [Indexed: 02/09/2023] Open
Abstract
An increasing number of metal-based compounds, including arsenic trioxide, auranofin, and cisplatin, have been reported to have antitumor activity. Their beneficial effects are controlled by a transcription factor, nuclear factor (erythroid-derived 2)-like 2 (NRF2). In response to oxidative stress, NRF2 induces the expression of cytoprotective genes. NRF2 protein levels are regulated by Kelch-like ECH-associated protein 1 (KEAP1) via ubiquitination. Bi-chlorodibenzo[c,f][1,5]thiabismocine (compound 3), a bismuth compound, is known for its potent anti-proliferative activity against various cancer cell lines. In the present study, we investigated the effect of compound 3 on NRF2 signaling in the human colorectal adenocarcinoma cell line DLD-1 in terms of cell viability as well as mRNA and protein expression levels of NRF2. Compound 3 upregulated NRF2 protein levels in a time- and concentration-dependent manner, accompanied by a marked increase in heme-oxygenase-1 (HO-1) mRNA and protein levels. We observed that brusatol, an NRF2 inhibitor, as well as small interfering RNA (siRNA)-mediated knockdown of NRF2 in DLD-1 cells suppressed compound 3-induced HO-1 expression. The anticancer activity of compound 3 was enhanced by compounds that downregulate NRF2. These results suggest that compound 3 upregulates HO-1 via NRF2 activation and that the NRF2-HO-1 pathway is the cellular response to compound 3. We also discovered that compound 3 slightly downregulated KEAP1; thus, NRF2 activation may be associated with KEAP1 modification. Collectively, our results indicate that compound 3 simultaneously activates an anti-oxidative stress pathway, such as NRF2 and HO-1, and a pro-cell death signal in DLD-1 cells. Our findings may provide useful information for the development of a potent anticancer organobismuth(III) compound.
Collapse
Affiliation(s)
- Katsuya Iuchi
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, 3-3-1 Kichijojikitamachi, Musashino-shi, Tokyo, 180-8633, Japan.
| | - Yuji Tasaki
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, 3-3-1 Kichijojikitamachi, Musashino-shi, Tokyo, 180-8633, Japan
| | - Sayo Shirai
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, 3-3-1 Kichijojikitamachi, Musashino-shi, Tokyo, 180-8633, Japan
| | - Hisashi Hisatomi
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, 3-3-1 Kichijojikitamachi, Musashino-shi, Tokyo, 180-8633, Japan
| |
Collapse
|
219
|
Targeting NRF2-Governed Glutathione Synthesis for SDHB-Mutated Pheochromocytoma and Paraganglioma. Cancers (Basel) 2020; 12:cancers12020280. [PMID: 31979226 PMCID: PMC7072390 DOI: 10.3390/cancers12020280] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Succinate dehydrogenase subunit B (SDHB) deficiency frequently occurs in cluster I pheochromocytomas and paragangliomas (PCPGs). SDHB-mutated PCPGs are characterized by alterations in the electron transport chain, metabolic reprogramming of the tricarboxylic cycle, and elevated levels of reactive oxygen species (ROS). We discovered that SDHB-deficient PCPG cells exhibit increased oxidative stress burden, which leads to elevated demands for glutathione metabolism. Mechanistically, nuclear factor erythroid 2-related factor 2 (NRF2)-guided glutathione de novo synthesis plays a key role in supporting cellular survival and the proliferation of SDHB-knockdown (SDHBKD) cells. NRF2 blockade not only disrupted ROS homeostasis in SDHB-deficient cells but also caused severe cytotoxicity by the accumulation of DNA oxidative damage. Brusatol, a potent NRF2 inhibitor, showed a promising effect in suppressing SDHBKD metastatic lesions in vivo, with prolonged overall survival in mice bearing PCPG allografts. Our findings highlight a novel therapeutic strategy of targeting the NRF2-driven glutathione metabolic pathway against SDHB-mutated PCPG.
Collapse
|
220
|
Abstract
The basic leucine zipper transcription factor Nrf2 is the primary regulator of cellular oxidative stress. Activation of Nrf2 is regarded as a potential preventive and therapeutic strategy. However, aberrant hyperactivation of Nrf2 is found in a variety of cancers and promotes cancer progression and metastasis. Moreover, constitutive activation of Nrf2 confers cancer cells resistance to chemo- and radio-therapy. Thus, inhibiting Nrf2 could be a new therapeutic strategy for cancer. With the aim of accelerating the discovery and development of novel Nrf2 inhibitors, we summarize the biological and pathological functions of Nrf2 in cancer. Furthermore, the recent studies of small molecular Nrf2 inhibitors and potential Nrf2 inhibitory mechanisms are also summarized in this review.
Collapse
|
221
|
Zhou Y, Zhou Y, Wang K, Li T, Yang M, Wang R, Chen Y, Cao M, Hu R. Flumethasone enhances the efficacy of chemotherapeutic drugs in lung cancer by inhibiting Nrf2 signaling pathway. Cancer Lett 2020; 474:94-105. [PMID: 31954771 DOI: 10.1016/j.canlet.2020.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/16/2022]
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2), a transcription factor, participates in protecting cells from electrophilic or oxidative stresses through regulating expression of cytoprotective and antioxidant genes. It has become one of the emerging targets for cancer chemosensitization, and small molecule inhibitors of Nrf2 can enhance the efficacy of chemotherapeutic drugs. Here, we found that flumethasone, a glucocorticoid, inhibited Nrf2 signaling in A549 and H460 cells by promoting Nrf2 protein degradation. Moreover, flumethasone significantly increased the sensitivity of A549 and H460 cells to chemotherapeutic drugs including cisplatin, doxorubicin and 5-FU. In mice bearing A549-shControl cells-derived xenografts, the size and weight of xenografts in the flumethasone and cisplatin combination group had a significant reduction compared with those in the cisplatin group, while in mice bearing A549-shNrf2 cells-derived xenografts, the size and weight of the xenografts in the combination group had no significant difference compared with those in the cisplatin group, demonstrating that chemosensitization effect of flumethasone is Nrf2-dependent. This work suggests that flumethasone can potentially be used as an adjuvant sensitizer to enhance the efficacy of chemotherapeutic drugs in lung cancer.
Collapse
Affiliation(s)
- Yunjiang Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yang Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Keke Wang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tao Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mengdi Yang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Rui Wang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yaxin Chen
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mengran Cao
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Rong Hu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
222
|
Mendez B, Reyes J, Conde I, Ramos Z, Lozada E, Cruz AM, Asencio G, Carvajal A, Dharmawardhane S, Piñero-Cruz DM, Hernández E, Vivas P, Ospina CA. Simalikalactone D, a Potential Anticancer Compound from Simarouba tulae, an Endemic Plant of Puerto Rico. PLANTS (BASEL, SWITZERLAND) 2020; 9:E93. [PMID: 31940804 PMCID: PMC7020415 DOI: 10.3390/plants9010093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 11/16/2022]
Abstract
Species of the genus Simarouba have been studied because of their antimalarial and antileukemic activities. A group of oxygenated terpenes called quassinoids have been isolated from species of the Simarouba genus, and are responsible for its therapeutic properties. We hypothesized that Simarouba tulae, an endemic plant from Puerto Rico, is a natural source rich in quassinoid compounds with anticancer activity. The leaves were processed and extracted with solvents of different polarities. The extracts were screened for their antiproliferative activity, and it was shown that the chloroform extract was the most active extract. This extract was purified using different chromatographic techniques to afford the quassinoid simalikalactone D (SKD). This compound was further characterized using NMR and X-ray diffraction analysis. A reassessment of original structural assignments for SKD is proposed. SKD showed high cytotoxicity activity, with an IC50 of 55, 58, and 65 nM in A2780CP20 (ovarian), MDA-MB-435 (breast), and MDA-MB-231 (breast) cell lines, respectively. Exposure to SKD led to 15% inhibition of the migration of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Belmari Mendez
- Natural Sciences Program, University of Puerto Rico at Cayey, Cayey 00736, Puerto Rico; (B.M.); (A.C.)
| | - Jeyshka Reyes
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.R.); (I.C.); (A.M.C.); (S.D.); (P.V.)
| | - Isabel Conde
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.R.); (I.C.); (A.M.C.); (S.D.); (P.V.)
| | - Zulma Ramos
- Department of Pharmaceutical Sciences, University of Puerto Rico, School of Pharmacy, San Juan 00936, Puerto Rico; (Z.R.); (G.A.); (E.H.)
| | - Eunice Lozada
- Department of Biology, University of Puerto Rico, Río Piedras Campus, San Juan 00936, Puerto Rico;
| | - Ailed M. Cruz
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.R.); (I.C.); (A.M.C.); (S.D.); (P.V.)
| | - Gabriela Asencio
- Department of Pharmaceutical Sciences, University of Puerto Rico, School of Pharmacy, San Juan 00936, Puerto Rico; (Z.R.); (G.A.); (E.H.)
| | - Augusto Carvajal
- Natural Sciences Program, University of Puerto Rico at Cayey, Cayey 00736, Puerto Rico; (B.M.); (A.C.)
| | - Suranganie Dharmawardhane
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.R.); (I.C.); (A.M.C.); (S.D.); (P.V.)
| | - Dalice M. Piñero-Cruz
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan 00936, Puerto Rico;
| | - Eliud Hernández
- Department of Pharmaceutical Sciences, University of Puerto Rico, School of Pharmacy, San Juan 00936, Puerto Rico; (Z.R.); (G.A.); (E.H.)
| | - Pablo Vivas
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan 00936, Puerto Rico; (J.R.); (I.C.); (A.M.C.); (S.D.); (P.V.)
| | - Claudia A. Ospina
- Department of Chemistry and Physics, Universidad Ana G Mendez, Gurabo 00778, Puerto Rico
| |
Collapse
|
223
|
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
|
224
|
Spotlight on ROS and β3-Adrenoreceptors Fighting in Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6346529. [PMID: 31934266 PMCID: PMC6942895 DOI: 10.1155/2019/6346529] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023]
Abstract
The role of ROS and RNS is a long-standing debate in cancer. Increasing the concentration of ROS reaching the toxic threshold can be an effective strategy for the reduction of tumor cell viability. On the other hand, cancer cells, by maintaining intracellular ROS concentration at an intermediate level called “mild oxidative stress,” promote the activation of signaling that favors tumor progression by increasing cell viability and dangerous tumor phenotype. Many chemotherapeutic treatments induce cell death by rising intracellular ROS concentration. The persistent drug stimulation leads tumor cells to simulate a process called hormesis by which cancer cells exhibit a biphasic response to exposure to drugs used. After a first strong response to a low dose of chemotherapeutic agent, cancer cells start to decrease the response even if high doses of drugs were used. In this framework, β3-adrenoreceptors (β3-ARs) fit with an emerging antioxidant role in cancer. β3-ARs are involved in tumor proliferation, angiogenesis, metastasis, and immune tolerance. Its inhibition, by the selective β3-ARs antagonist (SR59230A), leads cancer cells to increase ROS concentration thus inducing cell death and to decrease NO levels thus inhibiting angiogenesis. In this review, we report an overview on reactive oxygen biology in cancer cells focusing on β3-ARs as new players in the antioxidant pathway.
Collapse
|
225
|
Li X, Zhan J, Hou Y, Hou Y, Chen S, Luo D, Luan J, Wang L, Lin D. Coenzyme Q10 Regulation of Apoptosis and Oxidative Stress in H 2O 2 Induced BMSC Death by Modulating the Nrf-2/NQO-1 Signaling Pathway and Its Application in a Model of Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6493081. [PMID: 31915512 PMCID: PMC6930770 DOI: 10.1155/2019/6493081] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/16/2019] [Indexed: 12/23/2022]
Abstract
Spinal cord injury (SCI) has always been considered to be a devastating problem that results in catastrophic dysfunction, high disability rate, low mortality rate, and huge cost for the patient. Stem cell-based therapy, especially using bone marrow mesenchymal stem cells (BMSCs), is a promising strategy for the treatment of SCI. However, SCI results in low rates of cell survival and a poor microenvironment, which limits the therapeutic efficiency of BMSC transplantation. Coenzyme Q10 (CoQ10) is known as a powerful antioxidant, which inhibits lipid peroxidation and scavenges free radicals, and its combined effect with BMSC transplantation has been shown to have a powerful impact on protecting the vitality of cells, as well as antioxidant and antiapoptotic compounds in SCI. Therefore, we aimed to evaluate whether CoQ10 could decrease oxidative stress against the apoptosis of BMSCs in vitro and explored its molecular mechanisms. Furthermore, we investigated the protective effect of CoQ10 combined with BMSCs transplanted into a SCI model to verify its ability. Our results demonstrate that CoQ10 treatment significantly decreases the expression of the proapoptotic proteins Bax and Caspase-3, as shown through TUNEL-positive staining and the products of oxidative stress (ROS), while increasing the expression of the antiapoptotic protein Bcl-2 and the products of antioxidation, such as glutathione (GSH), against apoptosis and oxidative stress, in a H2O2-induced model. We also identified consistent results from the CoQ10 treatment of BMSCs transplanted into SCI rats in vivo. Moreover, the Nrf-2 signaling pathway was also investigated in order to detail its molecular mechanism, and the results show that it plays an important role, both in vitro and in vivo. Thus, CoQ10 exerts an antiapoptotic and antioxidant effect, as well as improves the microenvironment in vitro and in vivo. It may also protect BMSCs from oxidative stress and enhance their therapeutic efficiency when transplanted for SCI treatment.
Collapse
Affiliation(s)
- Xing Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiheng Zhan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu Hou
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yonghui Hou
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shudong Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Dan Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiyao Luan
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Le Wang
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dingkun Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, Guangdong 510120, China
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou 510405, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
226
|
Ștefanache T, Forna N, Bădescu M, Jitaru D, Dragos ML, Rezuș C, Diaconescu BM, Bădulescu O, Rezuș E, Ciocoiu M, Bădescu C. Modulation of the activity of certain genes involved in tumor cell metabolism in the presence of the cytotoxic peptides defensin and cathelicidin LL37. Exp Ther Med 2019; 18:5033-5040. [PMID: 31819768 PMCID: PMC6895780 DOI: 10.3892/etm.2019.8117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/30/2019] [Indexed: 11/06/2022] Open
Abstract
It is common knowledge that some natural antimicrobial peptides also have a tumoricidal effect. We have shown that the peptides defensin and cathelicidin LL37 have cytostatic effects on human tumor cell lines HT29 (colorectal carcinoma) and A549 (alveolar carcinoma). In order to determine the modulating mechanism of these peptides we assessed the gene expression of the AKT, HIF-1α, XBP, NRF2, PERK, CHOP, BCL2, IRE1α and PI3K molecular targets involved in the survival, growth, proliferation and apoptosis pathways of tumor cells in the presence or absence of the studied peptides. Thus, this research enabled us to determine molecular markers and methods of assessment and monitoring of tumor cell cytotoxicity by high-performance molecular biology techniques. Defensin and cathelicidin LL37 activated tumor cell apoptosis, especially for the HT29, but also for A549 line, by increasing gene expression of CHOP and by lowering BCL2 gene expression. Oxidative stress determined the increase in gene expression of XBP, which directly influenced CHOP. The decrease in NRF2 gene expression highlighted the inhibition of cell proliferation, while the decrease in HIF1α gene expression evidenced the decrease in cell survival.
Collapse
Affiliation(s)
- Teodor Ștefanache
- Department of Pathophysiology, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| | - Norina Forna
- Department of Implantology, Dental Medicine, 700115 Iaşi, Romania
| | - Magda Bădescu
- Department of Pathophysiology, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| | - Daniela Jitaru
- Regional Institute of Oncology Iasi, 700115 Iaşi, Romania
| | | | - Ciprian Rezuș
- Department of Internal Medicine, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| | - Bogdan Mihail Diaconescu
- Department of Pathophysiology, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| | - Oana Bădulescu
- Department of Pathophysiology, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| | - Elena Rezuș
- Rehabilitation Hospital of Iasi, Rheumatology Clinic, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| | - Codruta Bădescu
- Department of Internal Medicine, University of Medicine and Pharmacy 'Grigore T. Popa', 700115 Iaşi, Romania
| |
Collapse
|
227
|
Gagliardi M, Cotella D, Santoro C, Corà D, Barlev NA, Piacentini M, Corazzari M. Aldo-keto reductases protect metastatic melanoma from ER stress-independent ferroptosis. Cell Death Dis 2019; 10:902. [PMID: 31780644 PMCID: PMC6883066 DOI: 10.1038/s41419-019-2143-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 12/22/2022]
Abstract
The incidence of melanoma is increasing over the years with a still poor prognosis and the lack of a cure able to guarantee an adequate survival of patients. Although the new immuno-based coupled to target therapeutic strategy is encouraging, the appearance of targeted/cross-resistance and/or side effects such as autoimmune disorders could limit its clinical use. Alternative therapeutic strategies are therefore urgently needed to efficiently kill melanoma cells. Ferroptosis induction and execution were evaluated in metastasis-derived wild-type and oncogenic BRAF melanoma cells, and the process responsible for the resistance has been dissected at molecular level. Although efficiently induced in all cells, in an oncogenic BRAF- and ER stress-independent way, most cells were resistant to ferroptosis execution. At molecular level we found that: resistant cells efficiently activate NRF2 which in turn upregulates the early ferroptotic marker CHAC1, in an ER stress-independent manner, and the aldo-keto reductases AKR1C1 ÷ 3 which degrades the 12/15-LOX-generated lipid peroxides thus resulting in ferroptotic cell death resistance. However, inhibiting AKRs activity/expression completely resensitizes resistant melanoma cells to ferroptosis execution. Finally, we found that the ferroptotic susceptibility associated with the differentiation of melanoma cells cannot be applied to metastatic-derived cells, due to the EMT-associated gene expression reprogramming process. However, we identified SCL7A11 as a valuable marker to predict the susceptibility of metastatic melanoma cells to ferroptosis. Our results identify the use of pro-ferroptotic drugs coupled to AKRs inhibitors as a new valuable strategy to efficiently kill human skin melanoma cells.
Collapse
Affiliation(s)
- Mara Gagliardi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.,Department of Health Sciences, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara, Italy
| | - Diego Cotella
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Claudio Santoro
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara, Italy.,Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Davide Corà
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara, Italy.,Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Nickolai A Barlev
- Laboratory of Molecular Medicine, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia.,Laboratory of Intracellular Signaling, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russian Federation
| | - Mauro Piacentini
- Department of Biology, University of Rome Tor Vergata, Rome, Italy. .,Laboratory of Molecular Medicine, Institute of Cytology of the Russian Academy of Sciences, Saint Petersburg, Russia.
| | - Marco Corazzari
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy. .,Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara, Italy. .,Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy.
| |
Collapse
|
228
|
Zhou Y, Wang K, Zhou Y, Li T, Yang M, Wang R, Chen Y, Cao M, Hu R. HEATR1 deficiency promotes pancreatic cancer proliferation and gemcitabine resistance by up-regulating Nrf2 signaling. Redox Biol 2019; 29:101390. [PMID: 31785531 PMCID: PMC6888747 DOI: 10.1016/j.redox.2019.101390] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/17/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022] Open
Abstract
The human HEAT repeat-containing protein 1 (HEATR1), consisting of 2144 amino acids, is a member of the UTP10 family and contains one HEAT repeat at its C-terminal. HEATR1 has been reported to regulate cytotoxic T lymphocytes and rRNA synthesis, while its functions in tumors are poorly understood. Here, we found that HEATR1 competed with Keap1 for binding to p62/sequestosome 1 (SQSTM1), resulted in up-regulation of Keap1, which then inhibited Nrf2 signaling in pancreatic cancer cells. HEATR1 knockdown enhanced proliferation and gemcitabine resistance of pancreatic cancer cells. Moreover, HEATR1 deficiency significantly improved xenografts growth and led to gemcitabine resistance in pancreatic cancer cell-derived xenografts through up-regulating Nrf2 signaling. By analyzing tumor tissue samples from pancreatic cancer patients, we found that low expression of HEATR1 was closely correlated with poor prognosis and clinicopathological features. Collectively, we suggest that HEATR1 deficiency promotes proliferation and gemcitabine resistance of pancreatic cancer through up-regulating Nrf2 signaling, indicating that HEATR1 may be a promising therapeutic target for pancreatic cancer. HEATR1 inhibited Nrf2 signaling in pancreatic cancer cells. HEATR1 inhibited Nrf2 signaling through competing with Keap1 for p62 binding in pancreatic cancer cells. HEATR1 deficiency promoted pancreatic cancer proliferation and gemcitabine resistance by up-regulating Nrf2 signaling.
Collapse
Affiliation(s)
- Yunjiang Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Keke Wang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yang Zhou
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Tao Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mengdi Yang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Rui Wang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yaxin Chen
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mengran Cao
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Rong Hu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
229
|
Brusatol, a Nrf2 Inhibitor Targets STAT3 Signaling Cascade in Head and Neck Squamous Cell Carcinoma. Biomolecules 2019; 9:biom9100550. [PMID: 31575007 PMCID: PMC6843503 DOI: 10.3390/biom9100550] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
STAT3 is a latent transcription factor that plays a vital role in the transmission of extracellular signal from receptors to the nucleus. It has been regarded as a master transcription factor due to its role in the regulation of a broad spectrum of genes, which can contribute to oncogenesis. Persistent activation of STAT3 and deregulation of its signaling has been observed in various human cancers including head and neck squamous cell carcinoma (HNSCC). In the present work, we identified brusatol (BT) as a potential blocker of STAT3 signaling pathway in diverse HNSCC cells. The data from the cell-based experiments suggested that BT-induced cytotoxicity and abrogated the activation of STAT3 and that of upstream kinases such as JAK1, JAK2, and Src. It reduced the levels of nuclear STAT3 and its DNA binding ability. BT treatment increased annexin-V-positive cells, promoted procaspase-3 and PARP cleavage, and downregulated the mRNA and protein expression of diverse proteins (Bcl-2, Bcl-xl, survivin) in HNSCC cells. Taken together, brusatol can function as a promising inhibitor targeting STAT3 signaling pathway in HNSCC.
Collapse
|
230
|
Hammad A, Namani A, Elshaer M, Wang XJ, Tang X. "NRF2 addiction" in lung cancer cells and its impact on cancer therapy. Cancer Lett 2019; 467:40-49. [PMID: 31574294 DOI: 10.1016/j.canlet.2019.09.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022]
Abstract
Nuclear factor erythroid 2-like factor 2 (NRF2) is a master regulator of the antioxidant enzymes and the detoxification proteins that play major roles in redox homeostasis. Although it plays a protective role against tumorigenesis, emerging evidence has shown that the NRF2 pathway is frequently altered in different types of cancer, including lung cancer. NRF2 activation influences many of the hallmarks of cancer and their signaling pathways, mainly apoptosis, proliferation, angiogenesis, metastasis, and metabolic reprogramming to establish cellular metabolic processes leading to "NRF2 addiction" in lung cancer cells. Intriguingly, constitutive activation of NRF2 promotes cancer development as well as resistance to chemotherapy and radiotherapy, and these malignant phenotypes lead to a poor prognosis in lung cancer patients. Therefore, targeted inhibition of the NRF2 together with traditional chemotherapy, radiotherapy, and immunotherapy, may be a promising approach to improving the survival rates of the NRF2-addicted lung cancer cases. Here we summarize the recent advances in NRF2-addicted lung cancer.
Collapse
Affiliation(s)
- Ahmed Hammad
- Department of Biochemistry and Department of Thoracic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China
| | - Akhileshwar Namani
- Department of Biochemistry and Department of Thoracic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China
| | - Mohamed Elshaer
- Department of Biochemistry and Department of Thoracic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China
| | - Xiu Jun Wang
- Department of Pharmacology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, PR China
| | - Xiuwen Tang
- Department of Biochemistry and Department of Thoracic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China.
| |
Collapse
|
231
|
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: 64] [Impact Index Per Article: 12.8] [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
|
232
|
Tang X, Fu X, Liu Y, Yu D, Cai SJ, Yang C. Blockade of Glutathione Metabolism in IDH1-Mutated Glioma. Mol Cancer Ther 2019; 19:221-230. [PMID: 31548295 DOI: 10.1158/1535-7163.mct-19-0103] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/20/2019] [Accepted: 09/17/2019] [Indexed: 12/21/2022]
Abstract
Mutations in genes encoding isocitrate dehydrogenases (IDH) 1 and 2 are common cancer-related genetic abnormalities. Malignancies with mutated IDHs exhibit similar pathogenesis, metabolic pattern, and resistance signature. However, an effective therapy against IDH1-mutated solid tumor remains unavailable. In this study, we showed that acquisition of IDH1 mutation results in the disruption of NADP+/NADPH balance and an increased demand for glutathione (GSH) metabolism. Moreover, the nuclear factor erythroid 2-related factor 2 (Nrf2) plays a key protective role in IDH1-mutated cells by prompting GSH synthesis and reactive oxygen species scavenging. Pharmacologic inhibition of the Nrf2/GSH pathway via brusatol administration exhibited a potent tumor suppressive effect on IDH1-mutated cancer in vitro and in vivo Our findings highlight a possible therapeutic strategy that could be valuable for IDH1-mutated cancer treatment.
Collapse
Affiliation(s)
- Xiaoying Tang
- School of Life Science and Technology, Beijing Institute of Technology, Beijing, China
| | - Xiao Fu
- School of Life Science and Technology, Beijing Institute of Technology, Beijing, China.,Neuro-Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Yang Liu
- Neuro-Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Di Yu
- Neuro-Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Sabrina J Cai
- Neuro-Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, NCI, Bethesda, Maryland.
| |
Collapse
|
233
|
Targeting Reactive Oxygen Species in Cancer via Chinese Herbal Medicine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9240426. [PMID: 31583051 PMCID: PMC6754955 DOI: 10.1155/2019/9240426] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023]
Abstract
Recently, reactive oxygen species (ROS), a class of highly bioactive molecules, have been extensively studied in cancers. Cancer cells typically exhibit higher levels of basal ROS than normal cells, primarily due to their increased metabolism, oncogene activation, and mitochondrial dysfunction. This moderate increase in ROS levels facilitates cancer initiation, development, and progression; however, excessive ROS concentrations can lead to various types of cell death. Therefore, therapeutic strategies that either increase intracellular ROS to toxic levels or, conversely, decrease the levels of ROS may be effective in treating cancers via ROS regulation. Chinese herbal medicine (CHM) is a major type of natural medicine and has greatly contributed to human health. CHMs have been increasingly used for adjuvant clinical treatment of tumors. Although their mechanism of action is unclear, CHMs can execute a variety of anticancer effects by regulating intracellular ROS. In this review, we summarize the dual roles of ROS in cancers, present a comprehensive analysis of and update the role of CHM—especially its active compounds and ingredients—in the prevention and treatment of cancers via ROS regulation and emphasize precautions and strategies for the use of CHM in future research and clinical trials.
Collapse
|
234
|
Paunkov A, Chartoumpekis DV, Ziros PG, Sykiotis GP. A Bibliometric Review of the Keap1/Nrf2 Pathway and its Related Antioxidant Compounds. Antioxidants (Basel) 2019; 8:antiox8090353. [PMID: 31480567 PMCID: PMC6769514 DOI: 10.3390/antiox8090353] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
Nrf2 is a master transcriptional regulator of antioxidant and cytoprotective pathways. Currently in its third decade, research on Nrf2 has expanded to encompass not only basic but also clinical studies. In the present bibliometric review, we employed the VOSviewer tool to describe the existing Nrf2 literature landscape. As of July 2019, 11,931 papers on Nrf2 were listed in the “Web of Science” database, with more than 1000 new papers published each year. As expected, terms related to oxidative stress and antioxidant molecules occur very often in the Nrf2 literature throughout the years. Interestingly, there is also a gradual increase in the occurrence of terms related to diseases or to natural compounds, the most prominent being sulforaphane, curcumin, and resveratrol that modulate the Nrf2 pathway. Going beyond molecular biology/biochemistry and related fields, Nrf2 research has begun to spread into more clinical areas like endocrinology/metabolism, cardiology, and nephrology, likely reflecting an increased interest in clinical applications of Nrf2 pathway activators. China has become the most prolific producer of Nrf2 papers the last five years followed by the USA and Japan, a reverse pattern compared to the past. In conclusion, Nrf2 is the subject of a globally active research field that keeps growing and extends from bench to bedside.
Collapse
Affiliation(s)
- Ana Paunkov
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland
| | - Dionysios V Chartoumpekis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland
- Division of Endocrinology, Department of Internal Medicine, School of Medicine, University of Patras, 265 04 Patras, Greece
| | - Panos G Ziros
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland
| | - Gerasimos P Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, SA08/02/250, Ave de la Sallaz 8, CH-1011 Lausanne, Switzerland.
| |
Collapse
|
235
|
Ji L, Zhang R, Chen J, Xue Q, Moghal N, Tsao MS. PIDD interaction with KEAP1 as a new mutation-independent mechanism to promote NRF2 stabilization and chemoresistance in NSCLC. Sci Rep 2019; 9:12437. [PMID: 31455821 PMCID: PMC6712044 DOI: 10.1038/s41598-019-48763-4] [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: 10/25/2018] [Accepted: 08/12/2019] [Indexed: 01/15/2023] Open
Abstract
Chemotherapy resistance is a major problem in non-small cell lung cancer (NSCLC) treatment. A major mechanism of chemoresistance involves stabilization of the NRF2 transcription factor. NRF2 levels are normally tightly regulated through interaction with KEAP1, an adaptor that targets NRF2 to the CUL3 E3 ubiquitin ligase for proteolysis. In NSCLC, aberrant NRF2 stabilization is best understood through mutations in NRF2, KEAP1, or CUL3 that disrupt their interaction. Biochemical studies, however, have revealed that NRF2 can also be stabilized through expression of KEAP1-interacting proteins that competitively sequester KEAP1 away from NRF2. Here, we have identified PIDD, as a novel KEAP1-interactor in NSCLC that regulates NRF2. We show that this interaction allows PIDD to reduce NRF2 ubiquitination and increase its stability. We also demonstrate that PIDD promotes chemoresistance in NSCLC cells both in vitro and in vivo, and that this effect is dependent on NRF2. Finally, we report that NRF2 protein expression in a NSCLC cohort exceeds the typical incidence of combined NRF2, KEAP1, and CUL3 mutations, and that NRF2 expression in this cohort is correlated with PIDD levels. Our data identify PIDD as a new NRF2 regulator, and suggest that variations in PIDD levels contribute to differential chemosensitivities among NSCLC patients.
Collapse
Affiliation(s)
- Lili Ji
- Department of Pathology, Medical College of Nantong University, Nantong, Jiangsu, 226001, China. .,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 1L7, Canada.
| | - Rui Zhang
- Department of Tuberculosis, the Sixth Hospital of Nantong, Nantong, Jiangsu, 226000, China
| | - Jie Chen
- Department of Oncology, Jiangyin People's Hospital, Jiangyin, China
| | - Qun Xue
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Nadeem Moghal
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 1L7, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| |
Collapse
|
236
|
Zhang Y, Xu M, Hu C, Liu A, Chen J, Gu C, Zhang X, You C, Tong H, Wu M, Chen P. Sargassum fusiforme Fucoidan SP2 Extends the Lifespan of Drosophila melanogaster by Upregulating the Nrf2-Mediated Antioxidant Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8918914. [PMID: 31485301 PMCID: PMC6710776 DOI: 10.1155/2019/8918914] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/31/2019] [Accepted: 07/02/2019] [Indexed: 01/19/2023]
Abstract
Damage accumulated in the genome and macromolecules is largely attributed to increased oxidative damage and a lack of damage repair in a cell, and this can eventually trigger the process of aging. Alleviating the extent of oxidative damage is therefore considered as a potential way to promote longevity. SFPS, a heteropolysaccharide extracted from the brown alga Sargassum fusiforme, has previously been shown to alleviate oxidative damage during the aging process in mice, but whether SFPS could extend the lifespan of an organism was not demonstrated. Furthermore, the precise component within SFPS that is responsible for the antioxidant activity and the underlying mechanism of such activity was also not resolved. In this study, SP2, a fucoidan derived from SFPS, was shown to exhibit strong antioxidant activity as measured by in vitro radical-scavenging assays. SP2 also improved the survival rate of D. melanogaster subjected to oxidative stress. The flies that were fed with a diet containing SP2 from the time of eclosion displayed significant enhancement in lifespan and reduced accumulation of triglyceride at the old-age stage. In addition, SP2 markedly improved the activities of the antioxidant enzymes, superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) and reduced the contents of the malondialdehyde (MDA) and oxidized glutathione (GSSG) in old flies. Furthermore, SP2 also upregulated the expression levels of the nuclear factor-erythroid-2-like 2 (nfe2l2 or nrf2) and its downstream target genes, accompanied by a dramatic reduction in the expression of kelch-like ECH-associated protein 1 (keap1, a canonical inhibitor of the Nrf2) in old flies. Additional support linking the crucial role of the Nrf2/ARE pathway to the antioxidant effect of SP2 was the relatively high survival rate under heat stress for D. melanogaster individuals receiving SP2 supplement, an effect that was abolished by the inclusion of inhibitors specific for the Nrf2/ARE pathway. Collectively, the results indicated that SP2, a S. fusiforme fucoidan, could promote longevity in D. melanogaster by enhancing the Nrf2-mediated antioxidant signaling pathway during the aging process.
Collapse
Affiliation(s)
- Ya Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
- Department of Natural Resources and Environmental Studies, University of Northern British Columbia, Prince George, BC, Canada
| | - Man Xu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Chenxi Hu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
- Department of Natural Resources and Environmental Studies, University of Northern British Columbia, Prince George, BC, Canada
| | - Amei Liu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Junjie Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Chenfei Gu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Xu Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Cuiping You
- Department of Central Laboratory, Linyi People's Hospital, Shandong University, Linyi, Shandong Province, China
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Mingjiang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Peichao Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| |
Collapse
|
237
|
Honokiol alleviates acetaminophen-induced hepatotoxicity via decreasing generation of acetaminophen-protein adducts in liver. Life Sci 2019; 230:97-103. [DOI: 10.1016/j.lfs.2019.05.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023]
|
238
|
Garufi A, Traversi G, Gilardini Montani MS, D'Orazi V, Pistritto G, Cirone M, D'Orazi G. Reduced chemotherapeutic sensitivity in high glucose condition: implication of antioxidant response. Oncotarget 2019; 10:4691-4702. [PMID: 31384396 PMCID: PMC6659798 DOI: 10.18632/oncotarget.27087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/05/2019] [Indexed: 12/19/2022] Open
Abstract
Resistance to chemotherapy represents a major obstacle to successful treatment. The generation of reactive oxygen species (ROS) has been directly linked to the cytotoxic effects of several antitumor agents, including Adriamycin (ADR), and modulation of the oxidative balance has been implicated in the development and/or regulation of resistance to chemotherapeutic drugs. We recently showed that high glucose (HG) markedly diminished the cancer cell death induced by anticancer agents such as ADR. In the present study we attempted to evaluate the mechanism that impaired the cytotoxic effect of ADR in HG. We found that, in colon cancer cells, HG attenuated ADR-induced ROS production that consequently diminished ADR-induced H2AX phosphorylation and micronuclei (MN) formation. Mechanistically, HG attenuation of ADR-induced ROS production correlated with increased antioxidant response promoted by NRF2 activity. Thus, pharmacologic inhibition of NRF2 pathway by brusatol re-established the ADR cytotoxic effect impaired by HG. Together, the data provide new insights into chemotherapeutic-resistance mechanisms in HG condition dictated by increased NRF2-induced antioxidant response and how they may be overcome in order to restore chemosensitivity and ADR-induced cell death.
Collapse
Affiliation(s)
- Alessia Garufi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| | - Gianandrea Traversi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| | | | | | - Giuseppa Pistritto
- University Tor Vergata, Department of Systems Medicine, Rome 00133, Italy
| | - Mara Cirone
- Sapienza University, Department of Experimental Medicine, Rome 00161, Italy
| | - Gabriella D'Orazi
- IRCCS Regina Elena National Cancer Institute, Department of Research, Rome 00144, Italy.,University 'G. d'Annunzio', Department of Medical and Biotechnological Sciences, Chieti 66013, Italy
| |
Collapse
|
239
|
Norwood VM, Huigens RW. Harnessing the Chemistry of the Indole Heterocycle to Drive Discoveries in Biology and Medicine. Chembiochem 2019; 20:2273-2297. [DOI: 10.1002/cbic.201800768] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Verrill M. Norwood
- Department of Medicinal ChemistryCenter for Natural Products Drug Discovery and Development (CNPD3)University of Florida 1345 Center Drive Gainesville FL 32610 USA
| | - Robert W. Huigens
- Department of Medicinal ChemistryCenter for Natural Products Drug Discovery and Development (CNPD3)University of Florida 1345 Center Drive Gainesville FL 32610 USA
| |
Collapse
|
240
|
Activators and Inhibitors of NRF2: A Review of Their Potential for Clinical Development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9372182. [PMID: 31396308 PMCID: PMC6664516 DOI: 10.1155/2019/9372182] [Citation(s) in RCA: 344] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/26/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023]
Abstract
The transcription factor NRF2 (nuclear factor erythroid 2-related factor 2) triggers the first line of homeostatic responses against a plethora of environmental or endogenous deviations in redox metabolism, proteostasis, inflammation, etc. Therefore, pharmacological activation of NRF2 is a promising therapeutic approach for several chronic diseases that are underlined by oxidative stress and inflammation, such as neurodegenerative, cardiovascular, and metabolic diseases. A particular case is cancer, where NRF2 confers a survival advantage to constituted tumors, and therefore, NRF2 inhibition is desired. This review describes the electrophilic and nonelectrophilic NRF2 activators with clinical projection in various chronic diseases. We also analyze the status of NRF2 inhibitors, which at this time provide proof of concept for blocking NRF2 activity in cancer therapy.
Collapse
|
241
|
Song Y, Yang H, Lin R, Jiang K, Wang BM. The role of ferroptosis in digestive system cancer. Oncol Lett 2019; 18:2159-2164. [PMID: 31402933 DOI: 10.3892/ol.2019.10568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
Ferroptosis is a type of regulated cell death dependent on iron and reactive oxygen species. Ferroptosis is distinct from other cell death modalities, including apoptosis, autophagy and necrosis. Dysregulated ferroptosis has been implicated in a number of diseases, including neuropathy, ischemia reperfusion injury, acute kidney failure and cancer. The digestive system consists of several organs. The morbidity and mortality rates of digestive system cancer are high. The current review summarizes the role of ferroptosis in digestive system cancer. A large number of molecules, including tumor protein p53, retinoblastoma protein, nuclear factor E2-related factor 2, KH RNA binding domain containing signal transduction associated 1, cysteine dioxygenase type 1, metallothionein-1G, nuclear receptor coactivator 4, CDGSH iron sulfur domain 1, heat shock protein family A (Hsp70) member 5 and acyl-CoA synthetase long chain family member 4, regulate ferroptosis in digestive system cancer. Drugs such as cisplatin, baicalein, haloperidol, artesunate, piperlongumine, saponin and bromelain may cause cancer cell death by inducing ferroptosis. An improved understanding of ferroptosis in digestive system cancer may give rise to novel diagnostic and making therapeutic strategies.
Collapse
Affiliation(s)
- Yan Song
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hu Yang
- Department of Nephrology, Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Rui Lin
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Kui Jiang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Bang-Mao Wang
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
242
|
Homoharringtonine stabilizes secondary structure of guanine-rich sequence existing in the 5'-untranslated region of Nrf2. Bioorg Med Chem Lett 2019; 29:2189-2196. [PMID: 31270017 DOI: 10.1016/j.bmcl.2019.06.049] [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] [Received: 02/08/2019] [Revised: 06/20/2019] [Accepted: 06/25/2019] [Indexed: 11/20/2022]
Abstract
Homoharringtonine, known as omacetaxine mepesuccinate, is a pharmaceutical drug substance approved for treatment of chronic myeloid leukemia. Here, we report that homoharringtonine (HHT) is a novel chemical inhibitor of NRF2. HHT significantly suppressed NRF2 and ARE-dependent gene expression in human lung carcinoma A549 cells. HHT stabilized secondary structure of guanine-rich sequence existing in the 5'-untranslated region (5'-UTR) of Nrf2 and sensitized A549 cells to etoposide-induced apoptosis. To the best of our knowledge, HHT is the first type of transcriptional inhibitor of Nrf2 that stabilizes guanine-rich sequence existing in the 5'-UTR. Our study also provides a novel mechanism of action underlying how HHT exerts anti-carcinogenic effects in cancer cells.
Collapse
|
243
|
Yan J, Pang Y, Zhuang J, Lin H, Zhang Q, Han L, Ke P, Zhuang J, Huang X. Selenepezil, a Selenium-Containing Compound, Exerts Neuroprotective Effect via Modulation of the Keap1-Nrf2-ARE Pathway and Attenuates Aβ-Induced Cognitive Impairment in Vivo. ACS Chem Neurosci 2019; 10:2903-2914. [PMID: 31035749 DOI: 10.1021/acschemneuro.9b00106] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is a major risk factor for neurodegenerative disease. The Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2 related factor 2 (Nrf2)-antioxidant response element (ARE) pathway is one of the most potent defensive systems against oxidative stress. Selenepezil, a selenium-based compound, was previously found to exhibit excellent acetylcholinesterase (AChE) inhibition, to mimic endogenous glutathione peroxidase (GPx) activity, and to exhibit scavenging activity for hydrogen peroxide in vitro. However, none of these activities have been evaluated in a cellular model, and detailed molecular mechanisms are not elucidated. Moreover, whether selenepezil ameliorates memory deficits in vivo remains unknown. This study validated the cytoprotective effect of selenepezil against 6-hydroxydopamine (6-OHDA)- or H2O2-induced SH-SY5Y cell damage via alleviation or neutralization of intracellular microtubule disorder, reactive oxygen species (ROS) accumulation, mitochondrial dysfunction, and cell apoptosis. Our study clearly demonstrated that selenepezil pretreatment exhibited remarkable cytoprotective effect in a Nrf2-dependent manner via activating the Keap1-Nrf2-ARE pathway and stimulating the transcription of Nrf2-ARE-regulated cytoprotective genes. Moreover, selenepezil·HCl exerts neuroprotective effect via attenuating Aβ-induced cognitive impairment in Alzheimer's disease (AD) rat and was more active than the reference drug donepezil. In summary, selenepezil deserves further consideration for AD therapy.
Collapse
Affiliation(s)
- Jun Yan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Yanqing Pang
- Department of Integrated Chinese medicine immunization, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Jialing Zhuang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Haibiao Lin
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Qiaoxuan Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Liqiao Han
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Peifeng Ke
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Junhua Zhuang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Xianzhang Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| |
Collapse
|
244
|
Cai SJ, Liu Y, Han S, Yang C. Brusatol, an NRF2 inhibitor for future cancer therapeutic. Cell Biosci 2019; 9:45. [PMID: 31183074 PMCID: PMC6554866 DOI: 10.1186/s13578-019-0309-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 05/30/2019] [Indexed: 11/10/2022] Open
Abstract
Background Natural products from herbal medicines have long been investigated for their potentials as cancer therapeutics. Besides the development of several herbal medicine-derived anti-cancer agents, such as paclitaxel, vincristine and podophyllotoxin, many recent laboratory findings demonstrated that brusatol, a quassinoid from the seeds of Brucea sumatrana, exhibits potent tumor suppressing effect with improved disease outcome. Our recent finding further demonstrated that brusatol synergizes with the intrinsic metabolic burden in cancer cells. Main body Here, we summarized the recent investigations of brusatol as an experimental therapeutic for human malignancies, such as leukemia, lung cancer, pancreatic cancer and brain tumor. We also discussed the molecular target brusatol, with a focus on the Nuclear factor erythroid 2-related factor 2 (NRF2)-guided gene transcription, as well as glutathione de novo synthesis. Further, we discussed the challenges and future applications of brusatol for cancer therapy. Conclusion In conclusion, we believe increasing evidences have shown the value of brusatol as a novel strategy for cancer treatment, which may indicate future drug development and clinical translation.
Collapse
Affiliation(s)
- Sabrina J Cai
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Room 1142E, Bethesda, MD 20892 USA
| | - Yang Liu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Room 1142E, Bethesda, MD 20892 USA
| | - Sue Han
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Room 1142E, Bethesda, MD 20892 USA
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 37, Room 1142E, Bethesda, MD 20892 USA
| |
Collapse
|
245
|
Pharmacokinetic study on bruceoside A revealed the potential role of quassinoid glycosides for the anticancer properties of Fructus Bruceae. J Pharm Biomed Anal 2019; 170:264-272. [DOI: 10.1016/j.jpba.2019.03.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/11/2019] [Accepted: 03/22/2019] [Indexed: 11/24/2022]
|
246
|
Effects of KEAP1 Silencing on the Regulation of NRF2 Activity in Neuroendocrine Lung Tumors. Int J Mol Sci 2019; 20:ijms20102531. [PMID: 31126053 PMCID: PMC6566555 DOI: 10.3390/ijms20102531] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 01/09/2023] Open
Abstract
Background. The KEAP1/NRF2 pathway has been widely investigated in tumors since it was implicated in cancer cells survival and therapies resistance. In lung tumors the deregulation of this pathway is mainly related to point mutations of KEAP1 and NFE2L2 genes and KEAP1 promoter hypermethylation, but these two genes have been rarely investigated in low/intermediate grade neuroendocrine tumors of the lung. Methods. The effects of KEAP1 silencing on NRF2 activity was investigated in H720 and H727 carcinoid cell lines and results were compared with those obtained by molecular profiling of KEAP1 and NFE2L2 in a collection of 47 lung carcinoids. The correlation between methylation and transcript levels was assessed by 5-aza-dC treatment. Results. We demonstrated that in carcinoid cell lines, the KEAP1 silencing induces an upregulation of NRF2 and some of its targets and that there is a direct correlation between KEAP1 methylation and its mRNA levels. A KEAP1 hypermethylation and Loss of Heterozygosity at KEAP1 gene locus was also observed in nearly half of lung carcinoids. Conclusions. This is the first study that has described the effects of KEAP1 silencing on the regulation of NRF2 activity in lung carcinoids cells. The epigenetic deregulation of the KEAP1/NRF2 by a KEAP1 promoter hypermethylation system appears to be a frequent event in lung carcinoids.
Collapse
|
247
|
Kapoor R, Sirohi VK, Gupta K, Dwivedi A. Naringenin ameliorates progression of endometriosis by modulating Nrf2/Keap1/HO1 axis and inducing apoptosis in rats. J Nutr Biochem 2019; 70:215-226. [PMID: 31252288 DOI: 10.1016/j.jnutbio.2019.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/02/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022]
Abstract
Endometriosis is mainly characterized by the presence of endometrial tissue exterior to the uterus, however, the exact pathophysiology of this disease still remains uncertain. Moreover, the incidence significantly contributes to infertility among women and hence, a novel treatment for endometriosis is widely investigated. Naringenin is a plant-derived flavonoid having anti-proliferative, anti-inflammatory, and anti-angiogenic properties in chronic and metabolic diseases. The current study was planned with an objective to demonstrate the anti-endometriotic therapeutic potential of naringenin in rats and to examine its impact on various cellular aspects with a view to define the mechanism involved. The endometrial lesion volumes, weight, serum TNF-α level and the histopathologic scores were significantly reduced in the naringenin- treated group as compared to the endometriotic control group. Naringenin ameliorated the expression of prognostic markers (TAK1, PAK1, VEGF and PCNA) involved in development and progression of endometriotic cells. Naringenin caused dose-dependent loss of mitochondrial membrane potential, induced apoptosis and inhibited proliferation in these cells. Further, a significant increase in level of Nrf2 and its downstream molecules (NQO1, HO-1) was found in endometriotic lesion, with a subsequent decrease in its repressor molecule Keap-1. Naringenin significantly modulated the expression of Nrf2 and its effector molecules downstream. It also inhibited the invasion of endometrial cells by reducing the expression of MMP-2 and MMP-9 in in-vitro primary culture. We conclude that naringenin may have a therapeutic potential in the treatment of endometriosis via induction of ROS-mediated apoptosis and its anti-invasive effects.
Collapse
Affiliation(s)
- Radhika Kapoor
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vijay Kumar Sirohi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kanchan Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Anila Dwivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
248
|
Mutant p53 and Cellular Stress Pathways: A Criminal Alliance That Promotes Cancer Progression. Cancers (Basel) 2019; 11:cancers11050614. [PMID: 31052524 PMCID: PMC6563084 DOI: 10.3390/cancers11050614] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 02/07/2023] Open
Abstract
The capability of cancer cells to manage stress induced by hypoxia, nutrient shortage, acidosis, redox imbalance, loss of calcium homeostasis and exposure to drugs is a key factor to ensure cancer survival and chemoresistance. Among the protective mechanisms utilized by cancer cells to cope with stress a pivotal role is played by the activation of heat shock proteins (HSP) response, anti-oxidant response induced by nuclear factor erythroid 2-related factor 2 (NRF2), the hypoxia-inducible factor-1 (HIF-1), the unfolded protein response (UPR) and autophagy, cellular processes strictly interconnected. However, depending on the type, intensity or duration of cellular stress, the balance between pro-survival and pro-death pathways may change, and cell survival may be shifted into cell death. Mutations of p53 (mutp53), occurring in more than 50% of human cancers, may confer oncogenic gain-of-function (GOF) to the protein, mainly due to its stabilization and interaction with the above reported cellular pathways that help cancer cells to adapt to stress. This review will focus on the interplay of mutp53 with HSPs, NRF2, UPR, and autophagy and discuss how the manipulation of these interconnected processes may tip the balance towards cell death or survival, particularly in response to therapies.
Collapse
|
249
|
HACE1, an E3 Ubiquitin Protein Ligase, Mitigates Kaposi's Sarcoma-Associated Herpesvirus Infection-Induced Oxidative Stress by Promoting Nrf2 Activity. J Virol 2019; 93:JVI.01812-18. [PMID: 30787155 DOI: 10.1128/jvi.01812-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/12/2019] [Indexed: 12/14/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV)-induced activation of nuclear factor erythroid 2-related factor 2 (Nrf2) is essential for both the expression of viral genes (latency) and modulation of the host antioxidant machinery. Reactive oxygen species (ROS) are also regulated by the ubiquitously expressed HACE1 protein (HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1), which targets the Rac1 protein for proteasomal degradation, and this blocks the generation of ROS by Rac1-dependent NADPH oxidases. In this study, we examined the role of HACE1 in KSHV infection. Elevated levels of HACE1 expression were observed in de novo KSHV-infected endothelial cells, KSHV latently infected TIVE-LTC and PEL cells, and Kaposi's sarcoma skin lesion cells. The increased HACE1 expression in the infected cells was mediated by KSHV latent protein kaposin A. HACE1 knockdown resulted in high Rac1 and Nox 1 (NADPH oxidase 1) activity, increased ROS (oxidative stress), increased cell death, and decreased KSHV gene expression. Loss of HACE1 impaired KSHV infection-induced phosphoinositide 3-kinase (PI3-K), protein kinase C-ζ (PKC-ζ), extracellular signal-regulated kinase 1/2 (ERK1/2), NF-κB, and Nrf2 activation and nuclear translocation of Nrf2, and it reduced the expression of Nrf2 target genes responsible for balancing the oxidative stress. In the absence of HACE1, glutamine uptake increased in the cells to cope with the KSHV-induced oxidative stress. These findings reveal for the first time that HACE1 plays roles during viral infection-induced oxidative stress and demonstrate that HACE1 facilitates resistance to KSHV infection-induced oxidative stress by promoting Nrf2 activity. Our studies suggest that HACE1 could be a potential target to induce cell death in KSHV-infected cells and to manage KSHV infections.IMPORTANCE ROS play important roles in several cellular processes, and increased ROS cause several adverse effects. KSHV infection of endothelial cells induces ROS, which facilitate virus entry by amplifying the infection-induced host cell signaling cascade, which, in turn, induces the nuclear translocation of phospho-Nrf2 protein to regulate the expression of antioxidative genes and viral genes. The present study demonstrates that KSHV infection induces the E3 ligase HACE1 protein to regulate KSHV-induced oxidative stress by promoting the activation of Nrf2 and nuclear translocation. Absence of HACE1 results in increased ROS and cellular death and reduced nuclear Nrf2, antioxidant, and viral gene expression. Together, these studies suggest that HACE1 can be a potential target to induce cell death in KSHV-infected cells.
Collapse
|
250
|
Potential Applications of NRF2 Inhibitors in Cancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8592348. [PMID: 31097977 PMCID: PMC6487091 DOI: 10.1155/2019/8592348] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/10/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
The NRF2/KEAP1 pathway represents one of the most important cell defense mechanisms against exogenous or endogenous stressors. Indeed, by increasing the expression of several cytoprotective genes, the transcription factor NRF2 can shelter cells and tissues from multiple sources of damage including xenobiotic, electrophilic, metabolic, and oxidative stress. Importantly, the aberrant activation or accumulation of NRF2, a common event in many tumors, confers a selective advantage to cancer cells and is associated to malignant progression, therapy resistance, and poor prognosis. Hence, in the last years, NRF2 has emerged as a promising target in cancer treatment and many efforts have been made to identify therapeutic strategies aimed at disrupting its prooncogenic role. By summarizing the results from past and recent studies, in this review, we provide an overview concerning the NRF2/KEAP1 pathway, its biological impact in solid and hematologic malignancies, and the molecular mechanisms causing NRF2 hyperactivation in cancer cells. Finally, we also describe some of the most promising therapeutic approaches that have been successfully employed to counteract NRF2 activity in tumors, with a particular emphasis on the development of natural compounds and the adoption of drug repurposing strategies.
Collapse
|