51
|
Role of Nrf2 and Its Activators in Cardiocerebral Vascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4683943. [PMID: 32831999 PMCID: PMC7428967 DOI: 10.1155/2020/4683943] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/16/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
Cardiocerebral vascular disease (CCVD) is a common disease with high morbidity, disability, and mortality. Oxidative stress (OS) is closely related to the progression of CCVD. Abnormal redox regulation leads to OS and overproduction of reactive oxygen species (ROS), which can cause biomolecular and cellular damage. The Nrf2/antioxidant response element (ARE) signaling pathway is one of the most important defense systems against exogenous and endogenous OS injury, and Nrf2 is regarded as a vital pharmacological target. The complexity of the CCVD pathological process and the current difficulties in conducting clinical trials have hindered the development of therapeutic drugs. Furthermore, little is known about the role of the Nrf2/ARE signaling pathway in CCVD. Clarifying the role of the Nrf2/ARE signaling pathway in CCVD can provide new ideas for drug design. This review details the recent advancements in the regulation of the Nrf2/ARE system and its role and activators in common CCVD development.
Collapse
|
52
|
Pomatto LCD, Dill T, Carboneau B, Levan S, Kato J, Mercken EM, Pearson KJ, Bernier M, de Cabo R. Deletion of Nrf2 shortens lifespan in C57BL6/J male mice but does not alter the health and survival benefits of caloric restriction. Free Radic Biol Med 2020; 152:650-658. [PMID: 31953150 PMCID: PMC7382945 DOI: 10.1016/j.freeradbiomed.2020.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/02/2020] [Accepted: 01/05/2020] [Indexed: 12/18/2022]
Abstract
Caloric restriction (CR) is the leading non-pharmaceutical dietary intervention to improve health- and lifespan in most model organisms. A wide array of cellular pathways is induced in response to CR and CR-mimetics, including the transcriptional activator Nuclear factor erythroid-2-related factor 2 (Nrf2), which is essential in the upregulation of multiple stress-responsive and mitochondrial enzymes. Nrf2 is necessary in tumor protection but is not essential for the lifespan extending properties of CR in outbred mice. Here, we sought to study Nrf2-knockout (KO) mice and littermate controls in male C57BL6/J, an inbred mouse strain. Deletion of Nrf2 resulted in shortened lifespan compared to littermate controls only under ad libitum conditions. CR-mediated lifespan extension and physical performance improvements did not require Nrf2. Metabolic and protein homeostasis and activation of tissue-specific cytoprotective proteins were dependent on Nrf2 expression. These results highlight an important contribution of Nrf2 for normal lifespan and stress response.
Collapse
Affiliation(s)
- Laura C D Pomatto
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA; National Institute on General Medical Sciences, National Institute of Health, Bethesda, MD, 20892, USA
| | - Theresa Dill
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA
| | - Bethany Carboneau
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA
| | - Sophia Levan
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA
| | - Jonathan Kato
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA
| | - Evi M Mercken
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA
| | - Kevin J Pearson
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
53
|
Moradifard S, Saghiri R, Ehsani P, Mirkhani F, Ebrahimi‐Rad M. A preliminary computational outputs versus experimental results: Application of sTRAP, a biophysical tool for the analysis of SNPs of transcription factor-binding sites. Mol Genet Genomic Med 2020; 8:e1219. [PMID: 32155318 PMCID: PMC7216802 DOI: 10.1002/mgg3.1219] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/25/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In the human genome, the transcription factors (TFs) and transcription factor-binding sites (TFBSs) network has a great regulatory function in the biological pathways. Such crosstalk might be affected by the single-nucleotide polymorphisms (SNPs), which could create or disrupt a TFBS, leading to either a disease or a phenotypic defect. Many computational resources have been introduced to predict the TFs binding variations due to SNPs inside TFBSs, sTRAP being one of them. METHODS A literature review was performed and the experimental data for 18 TFBSs located in 12 genes was provided. The sequences of TFBS motifs were extracted using two different strategies; in the size similar with synthetic target sites used in the experimental techniques, and with 60 bp upstream and downstream of the SNPs. The sTRAP (http://trap.molgen.mpg.de/cgi-bin/trap_two_seq_form.cgi) was applied to compute the binding affinity scores of their cognate TFs in the context of reference and mutant sequences of TFBSs. The alternative bioinformatics model used in this study was regulatory analysis of variation in enhancers (RAVEN; http://www.cisreg.ca/cgi-bin/RAVEN/a). The bioinformatics outputs of our study were compared with experimental data, electrophoretic mobility shift assay (EMSA). RESULTS In 6 out of 18 TFBSs in the following genes COL1A1, Hb ḉᴪ, TF, FIX, MBL2, NOS2A, the outputs of sTRAP were inconsistent with the results of EMSA. Furthermore, no p value of the difference between the two scores of binding affinity under the wild and mutant conditions of TFBSs was presented. Nor, were any criteria for preference or selection of any of the measurements of different matrices used for the same analysis. CONCLUSION Our preliminary study indicated some paradoxical results between sTRAP and experimental data. However, to link the data of sTRAP to the biological functions, its optimization via experimental procedures with the integration of expanded data and applying several other bioinformatics tools might be required.
Collapse
Affiliation(s)
| | - Reza Saghiri
- Biochemistry DepartmentPasteur Institute of IranTehranIran
| | - Parastoo Ehsani
- Molecular Biology DepartmentPasteur Institute of IranTehranIran
| | | | | |
Collapse
|
54
|
Beyond Heat Stress: Intestinal Integrity Disruption and Mechanism-Based Intervention Strategies. Nutrients 2020; 12:nu12030734. [PMID: 32168808 PMCID: PMC7146479 DOI: 10.3390/nu12030734] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
The current climate changes have increased the prevalence and intensity of heat stress (HS) conditions. One of the initial consequences of HS is the impairment of the intestinal epithelial barrier integrity due to hyperthermia and hypoxia following blood repartition, which often results in a leaky gut followed by penetration and transfer of luminal antigens, endotoxins, and pathogenic bacteria. Under extreme conditions, HS may culminate in the onset of “heat stroke”, a potential lethal condition if remaining untreated. HS-induced alterations of the gastrointestinal epithelium, which is associated with a leaky gut, are due to cellular oxidative stress, disruption of intestinal integrity, and increased production of pro-inflammatory cytokines. This review summarizes the possible resilience mechanisms based on in vitro and in vivo data and the potential interventions with a group of nutritional supplements, which may increase the resilience to HS-induced intestinal integrity disruption and maintain intestinal homeostasis.
Collapse
|
55
|
Zhao Y, Li MZ, Shen Y, Lin J, Wang HR, Talukder M, Li JL. Lycopene Prevents DEHP-Induced Leydig Cell Damage with the Nrf2 Antioxidant Signaling Pathway in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:2031-2040. [PMID: 31814398 DOI: 10.1021/acs.jafc.9b06882] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
As a plasticizer, di(2-ethylhexyl) phthalate (DEHP) is the most usually used phthalate. Leydig cell is a male-specific cell, which plays a principal role in spermatogenesis and masculinization by the androgens of synthesis and secretion. Numerous researchers have indicated that DEHP can result in testicular toxicity by inducing oxidative stress. Lycopene (LYC) is a possible treatment option for male infertility due to its natural antioxidant properties. Our study was aimed to investigate whether LYC could rescue DEHP-induced Leydig cell damage. The mice were treated with DEHP (500 mg/kg BW/day or 1000 mg/kg BW/day) and/or LYC (5 mg/kg BW/day) for 28 days. We found that LYC attenuated DEHP-induced Leydig cell damage. Moreover, the protective role of LYC was verified by the histopathological and ultrastructural analysis of the Leydig cell. LYC suppressed oxidative stress that was induced by DEHP. In the Leydig cell, the expressions of the nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target genes were improved through LYC-mediated protection in DEHP-induced Leydig cell damage. Our findings indicated that LYC could increase the antioxidant capacity via mediating Nrf2 signaling pathway, thereby attenuating DEHP-induced Leydig cell damage.
Collapse
Affiliation(s)
| | | | | | | | | | - Milton Talukder
- Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine , Patuakhali Science and Technology University , Barishal 8210 , Bangladesh
| | | |
Collapse
|
56
|
Singh D, Arora R, Bhatia A, Singh H, Singh B, Arora S. Molecular targets in cancer prevention by 4-(methylthio)butyl isothiocyanate - A comprehensive review. Life Sci 2020; 241:117061. [PMID: 31794774 DOI: 10.1016/j.lfs.2019.117061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/26/2022]
Abstract
The consumption of cruciferous vegetables rich in isothiocyanates has long been associated with a reduced risk of various types of cancer. 4-(methylthio)butyl isothiocyanate also called erucin is an isothiocyanate present in appreciable quantity in the seeds of Eruca sativa Mill. plant. Although the literature has revealed its protective effects via inducing phase II enzymes and inhibiting carcinogen activating phase I enzymes, recent studies also suggest that, it inhibits the proliferation of cancer cells by altering the telomerase activity, dynamics of microtubules, expression of histone deacetylases, and other molecular pathways. With this in mind, the emphasis has been made to review the molecular targets involved in cancer prevention by 4-(methylthio)butyl isothiocyanate.
Collapse
Affiliation(s)
- Davinder Singh
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar 143005, India.
| | - Rohit Arora
- Department of Biochemistry, Sri Guru Ram Das University of Health Science, Amritsar 143005, India.
| | - Astha Bhatia
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar 143005, India.
| | - Hasandeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, India.
| | - Balbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, India.
| | - Saroj Arora
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar 143005, India.
| |
Collapse
|
57
|
Qin JJ, Cheng XD, Zhang J, Zhang WD. Dual roles and therapeutic potential of Keap1-Nrf2 pathway in pancreatic cancer: a systematic review. Cell Commun Signal 2019; 17:121. [PMID: 31511020 PMCID: PMC6740038 DOI: 10.1186/s12964-019-0435-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 09/02/2019] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most fatal diseases with a very high rate of metastasis and low rate of survival. Despite the advances in understanding this devastating disease, PC still accounts for 3% of all cancers and causes almost 7% of death of cancer patients. Recent studies have demonstrated that the transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) and its key negative regulator Kelch-like ECH-associated protein 1 (Keap1) are dysregulated in PC and the Keap1-Nrf2 pathway is an emerging target for PC prevention and therapy. Indeed, Nrf2 plays an either tumor-suppressive or promoting function in PC, which depends on the developmental stages of the disease and the cellular context. Several natural-product Nrf2 activators have been developed to prevent pancreatic carcinogenesis, while the Nrf2 inhibitors have been examined for their efficacy in inhibiting PC growth and metastasis and reversing chemoresistance. However, further preclinical and clinical studies for determining the effectiveness and safety of targeting the Keap1-Nrf2 pathway for PC prevention and therapy are warranted. In this review, we comprehensively discuss the dual roles of the Keap1-Nrf2 signaling pathway in PC as well as the current targeting strategies and known activators and inhibitors of Nrf2. We also propose new strategies that may be used to address the current issues and develop more specific and more effective Nrf2 activator/inhibitors for PC prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China. .,Zhejiang Cancer Hospital, Hangzhou, 310022, China.
| | | | - Jia Zhang
- Shanxi Institute of Traditional Chinese Medicine, Taiyuan, 030012, China
| | - Wei-Dong Zhang
- School of Pharmacy, Naval Medical University, 325 Guohe Road, Yangpu District, Shanghai, 200433, China. .,Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
58
|
Alleva R, Tognù A, Tomasetti M, Benassi MS, Pazzaglia L, van Oven H, Viganò E, De Simone N, Pacini I, Giannone S, Gagic S, Borghi R, Picone S, Borghi B. Effect of different anaesthetic techniques on gene expression profiles in patients who underwent hip arthroplasty. PLoS One 2019; 14:e0219113. [PMID: 31344051 PMCID: PMC6657832 DOI: 10.1371/journal.pone.0219113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 06/15/2019] [Indexed: 11/18/2022] Open
Abstract
Objectives To investigate the modulation of genes whose expression level is indicative of stress and toxicity following exposure to three anaesthesia techniques, general anaesthesia (GA), regional anaesthesia (RA), or integrated anaesthesia (IA). Methods Patients scheduled for hip arthroplasty receiving GA, RA and IA were enrolled at Rizzoli Orthopaedic Institute of Bologna, Italy and the expression of genes involved in toxicology were evaluated in peripheral blood mononuclear cells (PBMCs) collected before (T0), immediately after surgery (T1), and on the third day (T2) after surgery in association with biochemical parameters. Results All three anaesthesia methods proved safe and reliable in terms of pain relief and patient recovery. Gene ontology analysis revealed that GA and mainly IA were associated with deregulation of DNA repair system and stress-responsive genes, which was observed even after 3-days from anaesthesia. Conversely, RA was not associated with substantial changes in gene expression. Conclusions Based on the gene expression analysis, RA technique showed the smallest toxicological effect in hip arthroplasty. Trial registration ClinicalTrials.gov number NCT03585647.
Collapse
Affiliation(s)
- Renata Alleva
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- * E-mail:
| | - Andrea Tognù
- Department of Anaesthesia and Postoperative Intensive Care, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Marco Tomasetti
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Maria Serena Benassi
- Laboratory of Experimental Oncology, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Laura Pazzaglia
- Laboratory of Experimental Oncology, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Hanna van Oven
- Department of Anaesthesia and Postoperative Intensive Care, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Ettore Viganò
- Department of Anaesthesia and Postoperative Intensive Care, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Nicola De Simone
- Department of Anaesthesia and Postoperative Intensive Care, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Ilaria Pacini
- Department of Anaesthesia and Postoperative Intensive Care, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Sandra Giannone
- Department of Anaesthesia and Postoperative Intensive Care, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Sanjin Gagic
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Research Unit of Anaesthesia and Pain Therapy, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Raffaele Borghi
- Department of Anaesthesia and Postoperative Intensive Care, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Sara Picone
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Research Unit of Anaesthesia and Pain Therapy, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Battista Borghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Research Unit of Anaesthesia and Pain Therapy, Rizzoli Orthopaedic Institute, Bologna, Italy
| |
Collapse
|
59
|
Antioxidant Defence Systems and Oxidative Stress in Poultry Biology: An Update. Antioxidants (Basel) 2019; 8:antiox8070235. [PMID: 31336672 PMCID: PMC6680731 DOI: 10.3390/antiox8070235] [Citation(s) in RCA: 292] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Poultry in commercial settings are exposed to a range of stressors. A growing body of information clearly indicates that excess ROS/RNS production and oxidative stress are major detrimental consequences of the most common commercial stressors in poultry production. During evolution, antioxidant defence systems were developed in poultry to survive in an oxygenated atmosphere. They include a complex network of internally synthesised (e.g., antioxidant enzymes, (glutathione) GSH, (coenzyme Q) CoQ) and externally supplied (vitamin E, carotenoids, etc.) antioxidants. In fact, all antioxidants in the body work cooperatively as a team to maintain optimal redox balance in the cell/body. This balance is a key element in providing the necessary conditions for cell signalling, a vital process for regulation of the expression of various genes, stress adaptation and homeostasis maintenance in the body. Since ROS/RNS are considered to be important signalling molecules, their concentration is strictly regulated by the antioxidant defence network in conjunction with various transcription factors and vitagenes. In fact, activation of vitagenes via such transcription factors as Nrf2 leads to an additional synthesis of an array of protective molecules which can deal with increased ROS/RNS production. Therefore, it is a challenging task to develop a system of optimal antioxidant supplementation to help growing/productive birds maintain effective antioxidant defences and redox balance in the body. On the one hand, antioxidants, such as vitamin E, or minerals (e.g., Se, Mn, Cu and Zn) are a compulsory part of the commercial pre-mixes for poultry, and, in most cases, are adequate to meet the physiological requirements in these elements. On the other hand, due to the aforementioned commercially relevant stressors, there is a need for additional support for the antioxidant system in poultry. This new direction in improving antioxidant defences for poultry in stress conditions is related to an opportunity to activate a range of vitagenes (via Nrf2-related mechanisms: superoxide dismutase, SOD; heme oxygenase-1, HO-1; GSH and thioredoxin, or other mechanisms: Heat shock protein (HSP)/heat shock factor (HSP), sirtuins, etc.) to maximise internal AO protection and redox balance maintenance. Therefore, the development of vitagene-regulating nutritional supplements is on the agenda of many commercial companies worldwide.
Collapse
|
60
|
Cirone M, Gilardini Montani MS, Granato M, Garufi A, Faggioni A, D'Orazi G. Autophagy manipulation as a strategy for efficient anticancer therapies: possible consequences. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:262. [PMID: 31200739 PMCID: PMC6570888 DOI: 10.1186/s13046-019-1275-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022]
Abstract
Autophagy is a catabolic process whose activation may help cancer cells to adapt to cellular stress although, in some instances, it can induce cell death. Autophagy stimulation or inhibition has been considered an opportunity to treat cancer, especially in combination with anticancer therapies, although autophagy manipulation may be viewed as controversial. Thus, whether to induce or to inhibit autophagy may be the best option in the different cancer patients is still matter of debate. Her we will recapitulate the possible advantages or disadvantages of manipulating autophagy in cancer, not only with the aim to obtain cancer cell death and disable oncogenes, but also to evaluate its interplay with the immune response which is fundamental for the success of anticancer therapies.
Collapse
Affiliation(s)
- Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy. .,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy.
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Marisa Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessia Garufi
- Department of Medical Science, University 'G. D'Annunzio', 66013, Chieti, Italy.,Department of Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Alberto Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Gabriella D'Orazi
- Department of Medical Science, University 'G. D'Annunzio', 66013, Chieti, Italy. .,Department of Research, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy.
| |
Collapse
|
61
|
Gilardini Montani MS, Cecere N, Granato M, Romeo MA, Falcinelli L, Ciciarelli U, D'Orazi G, Faggioni A, Cirone M. Mutant p53, Stabilized by Its Interplay with HSP90, Activates a Positive Feed-Back Loop Between NRF2 and p62 that Induces Chemo-Resistance to Apigenin in Pancreatic Cancer Cells. Cancers (Basel) 2019; 11:cancers11050703. [PMID: 31121848 PMCID: PMC6562395 DOI: 10.3390/cancers11050703] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is one of the most aggressive cancers whose prognosis is worsened by the poor response to the current chemotherapies. In this study, we investigated the cytotoxic effect of Apigenin, against two pancreatic cell lines, namely Panc1 and PaCa44, harboring different p53 mutations. Apigenin is a flavonoid widely distributed in nature that displays anti-inflammatory and anticancer properties against a variety of cancers. Here we observed that Apigenin exerted a stronger cytotoxic effect against Panc1 cell line in comparison to PaCa44. Searching for mechanisms responsible for such different effect, we found that the higher cytotoxicity of Apigenin correlated with induction of higher level of intracellular ROS, reduction of mutant (mut) p53 and HSP90 expression and mTORC1 inhibition. Interestingly, we found that mutp53 was stabilized by its interplay with HSP90 and activates a positive feed-back loop between NRF2 and p62, up-regulating the antioxidant response and reducing the cytotoxicity of Apigenin. These results suggest that targeting the molecules involved in the mTOR-HSP90-mutp53-p62-NRF2-antioxidant response axis could help to overcome the chemo-resistance of pancreatic cancer to Apigenin.
Collapse
Affiliation(s)
- Maria Saveria Gilardini Montani
- Department of Experimental Medicine, Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| | - Nives Cecere
- Department of Experimental Medicine, Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| | - Marisa Granato
- Department of Experimental Medicine, Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| | - Maria Anele Romeo
- Department of Experimental Medicine, Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| | - Luca Falcinelli
- Department of Experimental Medicine, Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| | - Umberto Ciciarelli
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio", 66100 Chieti, Italy.
| | - Alberto Faggioni
- Department of Experimental Medicine, Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy.
| |
Collapse
|
62
|
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: 53] [Impact Index Per Article: 8.8] [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
|
63
|
Patinen T, Adinolfi S, Cortés CC, Härkönen J, Jawahar Deen A, Levonen AL. Regulation of stress signaling pathways by protein lipoxidation. Redox Biol 2019; 23:101114. [PMID: 30709792 PMCID: PMC6859545 DOI: 10.1016/j.redox.2019.101114] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/12/2019] [Accepted: 01/15/2019] [Indexed: 12/30/2022] Open
Abstract
Enzymatic and non-enzymatic oxidation of unsaturated fatty acids gives rise to reactive species that covalently modify nucleophilic residues within redox sensitive protein sensors in a process called lipoxidation. This triggers adaptive signaling pathways that ultimately lead to increased resistance to stress. In this graphical review, we will provide an overview of pathways affected by protein lipoxidation and the key signaling proteins being altered, focusing on the KEAP1-NRF2 and heat shock response pathways. We review the mechanisms by which lipid peroxidation products can serve as second messengers and evoke cellular responses via covalent modification of key sensors of altered cellular environment, ultimately leading to adaptation to stress.
Collapse
Affiliation(s)
- Tommi Patinen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Simone Adinolfi
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Carlos Cruz Cortés
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland; Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City MX-07360, Mexico
| | - Jouni Härkönen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Ashik Jawahar Deen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland
| | - Anna-Liisa Levonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FIN-70211, Finland.
| |
Collapse
|
64
|
Identification of a potential transcriptional regulator encoded by grass carp reovirus. Arch Virol 2019; 164:1393-1404. [DOI: 10.1007/s00705-019-04204-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/09/2019] [Indexed: 01/26/2023]
|
65
|
Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med 2019; 133:162-168. [PMID: 29800655 PMCID: PMC6251771 DOI: 10.1016/j.freeradbiomed.2018.05.074] [Citation(s) in RCA: 482] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/16/2018] [Accepted: 05/19/2018] [Indexed: 12/14/2022]
Abstract
Ferroptosis is a form of lipid peroxidation-induced cell death that can be regulated in many ways, from altering the activity of antioxidant enzymes to the level of transcription factors. The p53 tumor suppressor is 'the guardian of the genome' that participates in the control of cell survival and division under various stresses. Beyond its effects on apoptosis, autophagy, and cell cycle, p53 also regulates ferroptosis either through a transcriptional or posttranslational mechanism. On one hand, p53 can enhance ferroptosis by inhibiting the expression of SLC7A11 (solute carrier family 7 member 11) or by enhancing that of SAT1 (spermidine/spermine N1-acetyltransferase 1) and GLS2 (glutaminase 2). On the other hand, p53 suppresses ferroptosis through the direct inhibition of DPP4 (dipeptidyl peptidase 4) activity or by the induction of CDKN1A/p21 (cyclin dependent kinase inhibitor 1 A) expression. Here, we review recent discoveries and emerging trends in the study of the ferroptosis network and highlight the context-dependent impact of p53 on ferroptosis and oxidative stress.
Collapse
Affiliation(s)
- Rui Kang
- The Third Affiliated Hospital, Central of DAMP Biology, Guangzhou Medical University, Guangzhou, Guangdong 510150, China; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Guido Kroemer
- Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France; Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France; Université Pierre et Marie Curie, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France; Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Daolin Tang
- The Third Affiliated Hospital, Central of DAMP Biology, Guangzhou Medical University, Guangzhou, Guangdong 510150, China; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
66
|
Mao L, Yang T, Li X, Lei X, Sun Y, Zhao Y, Zhang W, Gao Y, Sun B, Zhang F. Protective effects of sulforaphane in experimental vascular cognitive impairment: Contribution of the Nrf2 pathway. J Cereb Blood Flow Metab 2019; 39. [PMID: 29533123 PMCID: PMC6365596 DOI: 10.1177/0271678x18764083] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The major pathophysiological process of vascular cognitive impairment (VCI) is chronic cerebral ischemia, which causes disintegration of the blood-brain barrier (BBB), neuronal death, and white matter injury. This study aims to test whether sulforaphane (Sfn), a natural activator of nuclear factor erythroid 2-related factor 2 (Nrf2), reduces the chronic ischemic injury and cognitive dysfunction after VCI. Experimental VCI was induced in rats by permanent occlusion of both common carotid arteries for six weeks. This procedure caused notable neuronal death in the cortex and hippocampal CA1, myelin loss in the corpus callosum and hippocampal fimbria, accumulation of myelin debris in the corpus callosum, and remarkable cognitive impairment. Sfn treatment alleviated these ischemic injuries and the cognitive dysfunction. Sfn-mediated neuroprotection was associated with enhanced activation of Nrf2 and upregulation of heme oxygenase 1. Sfn also reduced neuronal and endothelial death and maintained the integrity of BBB after oxygen-glucose deprivation in vitro in an Nrf2 dependent manner. Furthermore, Nrf2 knockdown in endothelial cells decreased claudin-5 protein expression with downregulated claudin-5 promoter activity, suggesting that claudin-5 might be a target gene of Nrf2. Our results demonstrate that Sfn provides robust neuroprotection against chronic brain ischemic injury and may be a promising agent for VCI treatment.
Collapse
Affiliation(s)
- Leilei Mao
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Tuo Yang
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xin Li
- 2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Xia Lei
- 2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Yang Sun
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yongfang Zhao
- 3 State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China
| | - Wenting Zhang
- 3 State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China
| | - Yanqin Gao
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,3 State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China
| | - Baoliang Sun
- 2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Feng Zhang
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| |
Collapse
|
67
|
Kamikubo Y, Yamana T, Hashimoto Y, Sakurai T. Induction of Oxidative Stress and Cell Death in Neural Cells by Silica Nanoparticles. ACS Chem Neurosci 2019; 10:304-312. [PMID: 30230808 DOI: 10.1021/acschemneuro.8b00248] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Silica nanoparticles (SiNPs) are produced on an industrial scale and used in various fields including health care, because silica is stable, inexpensive, and easy to handle. Despite these benefits, there is concern that exposure to SiNPs may lead to adverse effects in certain types of cells or tissues, such as hemolysis, immune responses, and developmental abnormalities in the brain and developing embryos. Although investigations on the toxicity of SiNPs against neurons are essential for medicinal use, only a few studies have assessed the direct effects of SiNPs on cells derived from the central nervous system. In this study, we investigated the toxic effects of SiNPs on primary cultures of hippocampal cells, using SiNPs with diameters of 10-1500 nm. We showed that treatment with SiNPs caused oxidative stress and cell death. Furthermore, we found that these cytotoxicities were dependent on the particle size, concentration, and surface charge of SiNPs, as well as the treatment temperature. The toxicity was reduced by SiNP surface functionalization or protein coating and by pretreating cells with an antioxidant, suggesting that contact-induced oxidative stress may be partially responsible for SiNP-induced cell death. These data will be valuable for utilizing SiNPs in biomedical applications.
Collapse
Affiliation(s)
- Yuji Kamikubo
- Department of Pharmacology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Tomohito Yamana
- Department of Pharmacology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoshie Hashimoto
- Department of Pharmacology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Takashi Sakurai
- Department of Pharmacology, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
68
|
Samuelsen JT, Michelsen VB, Bruun JA, Dahl JE, Jensen E, Örtengren U. The dental monomer HEMA causes proteome changes in human THP-1 monocytes. J Biomed Mater Res A 2019; 107:851-859. [DOI: 10.1002/jbm.a.36601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/16/2018] [Accepted: 12/26/2018] [Indexed: 12/15/2022]
Affiliation(s)
| | | | - Jack-Ansgar Bruun
- Department of Medical Biology, Faculty of Health Sciences; University of Tromsø; Tromsø Norway
| | - Jon E. Dahl
- Nordic Institute of Dental Materials (NIOM); Oslo Norway
| | - Einar Jensen
- Department of Pharmacy, Faculty of Health Sciences; University of Tromsø; Tromsø Norway
| | - Ulf Örtengren
- Department of Clinical Dentistry, Faculty of Health Sciences; University of Tromsø; Tromsø Norway
- Department of Cariology; Institute for Odontology, Sahlgrenska Academy, Göteborg University; Göteborg Sweden
| |
Collapse
|
69
|
Lee EH, Kim S, Choi MS, Yang H, Park SM, Oh HA, Moon KS, Han JS, Kim YB, Yoon S, Oh JH. Gene networking in colistin-induced nephrotoxicity reveals an adverse outcome pathway triggered by proteotoxic stress. Int J Mol Med 2019; 43:1343-1355. [PMID: 30628653 PMCID: PMC6365082 DOI: 10.3892/ijmm.2019.4052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/17/2018] [Indexed: 01/18/2023] Open
Abstract
Colistin has been widely used for the treatment of infections of multidrug-resistant Gram-negative bacteria, despite the fact that it induces serious kidney injury as a side effect. To investigate the mechanism underlying its nephrotoxicity, colistin methanesulfonate sodium (CMS; 25 or 50 mg/kg) was administered via intraperitoneal injection to Sprague-Dawley rats daily over 7 days. Serum biochemistry and histopathology indicated that nephrotoxicity occurred in the rats administered with CMS. Whole-genome microarrays indicated 894 differentially expressed genes in the group treated with CMS (analysis of variance, false discovery rate <0.05, fold-change ≥1.3). Gene pathway and networking analyses revealed that genes associated with proteotoxic stress, including ribosome synthesis, protein translation, and protein folding, were significantly associated with the nephrotoxicity induced by CMS. It was found that colistin inhibited the expression of the target genes heat shock factor 1 and nuclear factor erythroid-2-related factor-2, which are associated with proteostasis, and that nephrotoxicity of CMS may be initiated by proteotoxic stress due to heat shock response inhibition, leading to oxidative stress, endoplasmic reticulum stress, cell cycle arrest and apoptosis, eventually leading to cell death. A putative adverse outcome pathway was constructed based on the integrated gene networking data, which may clarify the mode of action of colistin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Eun Hee Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Soojin Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Mi-Sun Choi
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Heeyoung Yang
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Se-Myo Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Hyun-A Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Kyoung-Sik Moon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Ji-Seok Han
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Yong-Bum Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| |
Collapse
|
70
|
Granato M, Gilardini Montani MS, Angiolillo C, D'Orazi G, Faggioni A, Cirone M. Cytotoxic Drugs Activate KSHV Lytic Cycle in Latently Infected PEL Cells by Inducing a Moderate ROS Increase Controlled by HSF1, NRF2 and p62/SQSTM1. Viruses 2018; 11:v11010008. [PMID: 30586869 PMCID: PMC6356381 DOI: 10.3390/v11010008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/17/2018] [Accepted: 12/21/2018] [Indexed: 12/26/2022] Open
Abstract
Previous studies have indicated that cytotoxic treatments may induce or not activate viral lytic cycle activation in cancer cells latently infected by Kaposi’s sarcoma-associated herpesvirus (KSHV). To investigate the molecular mechanisms responsible for such an effect, we compared two cytotoxic treatments able to induce the viral lytic cycle, named 12-O-tetradecanoylphorbol 13-acetate (TPA) (T) in combination with sodium butyrate (B) and bortezomib (BZ), with two cytotoxic treatments that did not activate this process, named metformin (MET) and quercetin (Q). Our results indicated that TB and bortezomib increased levels of oxygen reactive species (ROS) while metformin and quercetin reduced them. The finding that N-acetylcysteine (NAC), a reactive oxigen species (ROS) scavenger, counteracted K-bZIP expression induced by TB or bortezomib, confirmed that an ROS increase played a role in KSHV lytic cycle activation. Moreover, we found that TB and bortezomib up-regulated p62/Sequestosome1(p62/SQSTM1) protein, while metformin and quercetin down-regulated it. p62/SQSTM1 silencing or the inhibition of NF-E2-related factor 2 (NRF2) or Heat Shock Factor 1 (HSF1), that mediate p62/SQSTM1 transcription, also reduced KSHV lytic antigen expression induced by TB or bortezomib. Interestingly, such combination treatments further increased intracellular ROS and cytotoxicity induced by the single TB or bortezomib treatment, suggesting that NRF2, HSF1 and p62/SQSTM1 keep the ROS level under control, allowing primary effusion lymphoma (PEL) cells to continue to survive and KSHV to replicate.
Collapse
Affiliation(s)
- Marisa Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Camilla Angiolillo
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Gabriella D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, 00144 Rome, Italy.
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", 66013 Chieti, Italy.
| | - Alberto Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 324, 00161 Rome, Italy.
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Viale Regina Elena 324, 00161 Rome, Italy.
| |
Collapse
|
71
|
Matzinger M, Fischhuber K, Heiss EH. Activation of Nrf2 signaling by natural products-can it alleviate diabetes? Biotechnol Adv 2018; 36:1738-1767. [PMID: 29289692 PMCID: PMC5967606 DOI: 10.1016/j.biotechadv.2017.12.015] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/19/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (DM) has reached pandemic proportions and effective prevention strategies are wanted. Its onset is accompanied by cellular distress, the nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor boosting cytoprotective responses, and many phytochemicals activate Nrf2 signaling. Thus, Nrf2 activation by natural products could presumably alleviate DM. We summarize function, regulation and exogenous activation of Nrf2, as well as diabetes-linked and Nrf2-susceptible forms of cellular stress. The reported amelioration of insulin resistance, β-cell dysfunction and diabetic complications by activated Nrf2 as well as the status quo of Nrf2 in precision medicine for DM are reviewed.
Collapse
Affiliation(s)
- Manuel Matzinger
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Katrin Fischhuber
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Elke H Heiss
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
72
|
NRF2 and HSF1 coordinately regulate heme oxygenase-1 expression. Biochem Biophys Res Commun 2018; 506:7-11. [DOI: 10.1016/j.bbrc.2018.10.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/05/2018] [Indexed: 01/13/2023]
|
73
|
Fernandes FA, Dutra BK, Mosele F, Araujo ASR, Ferreira GD, Belló-Klein A, Kucharski LC, Vinagre AS, Da Silva RSM. Redox and metabolic strategies developed by anterior and posterior gills of the crab Neohelice granulata after short periods of hypo- or hyper-osmotic stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 639:457-464. [PMID: 29800839 DOI: 10.1016/j.scitotenv.2018.05.162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/11/2018] [Accepted: 05/13/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study was to identify the response pattern of redox balance, Na+/K+ATPase activity and HSP70 expression in the posterior and anterior gills of the crab Neohelice granulata submitted to hypo- or hyper-osmotic stress for 1 h and 6 h. After 1 h of either type of osmotic stress, there was an increase in catalase activity, but a decrease in GSSG/GSH ratio (oxidized to reduced glutathione ratio) and Na+/K+ATPase activity in both gill sets. H2O2 levels decreased only in the posterior gills. H2O2 levels and Na+/K+ATPase activity remained reduced after 6 h of exposure to either type of osmotic stress in both gill sets. The GSSG/GSH ratio returned to initial levels after 6 h of hyper-osmotic stress, whereas it increased 10 times in both gill sets after hypo-osmotic stress. Furthermore, HSP70 protein expression increased in posterior gills after 6 h of hypo-osmotic stress. H2O2 levels in tank water decreased after hypo-osmotic challenge and increased after 6 h of hyper-osmotic stress, indicating increased H2O2 excretion. Therefore, N. granulata gills have redox, metabolic and molecular strategies to deal with rapid osmotic challenges, an important environmental parameter that influences juvenile and adult crab distribution and abundance within different populations.
Collapse
Affiliation(s)
- F A Fernandes
- Laboratório de Metabolismo e Endocrinologia Comparada (LAMEC), Departamento de Fisiologia, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Brazil; Laboratório de Ictiologia, Instituto Federal de Educação, Ciência e Tecnologia Farroupilha - Campus São Vicente do Sul (IFFAR), Brazil
| | - B K Dutra
- Laboratório de Ictiologia, Instituto Federal de Educação, Ciência e Tecnologia Farroupilha - Campus São Vicente do Sul (IFFAR), Brazil
| | - F Mosele
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, ICBS, UFRGS, Brazil
| | - A S R Araujo
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, ICBS, UFRGS, Brazil
| | - G D Ferreira
- Departamento de Fisiologia, Federal University of Pelotas (UFPEL), Brazil
| | - A Belló-Klein
- Laboratório de Fisiologia Cardiovascular, Departamento de Fisiologia, ICBS, UFRGS, Brazil
| | - L C Kucharski
- Laboratório de Metabolismo e Endocrinologia Comparada (LAMEC), Departamento de Fisiologia, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Brazil
| | - A S Vinagre
- Laboratório de Metabolismo e Endocrinologia Comparada (LAMEC), Departamento de Fisiologia, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Brazil.
| | - R S M Da Silva
- Laboratório de Metabolismo e Endocrinologia Comparada (LAMEC), Departamento de Fisiologia, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Brazil
| |
Collapse
|
74
|
Paul S, Ghosh S, Mandal S, Sau S, Pal M. NRF2 transcriptionally activates the heat shock factor 1 promoter under oxidative stress and affects survival and migration potential of MCF7 cells. J Biol Chem 2018; 293:19303-19316. [PMID: 30309986 DOI: 10.1074/jbc.ra118.003376] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/02/2018] [Indexed: 01/07/2023] Open
Abstract
Functional up-regulation of heat shock factor 1 (HSF1) activity through different posttranslational modifications has been implicated in the survival and proliferation of various cancers. It is increasingly recognized that the HSF1 gene is also up-regulated at the transcriptional level, a phenomenon correlated with poor prognosis for patients with different cancers, including breast cancer. Here, we analyzed the transcriptional up-regulation of HSF1 in human cells upon arsenite- or peroxide-induced oxidative stress. Sequential promoter truncation coupled with bioinformatics analysis revealed that this activation is mediated by two antioxidant response elements (AREs) located between 1707 and 1530 bp upstream of the transcription start site of the HSF1 gene. Using shRNA-mediated down-regulation, ChIP of NRF2, site-directed mutagenesis of the AREs, and DNase I footprinting of the HSF1 promoter, we confirmed that nuclear factor erythroid-derived 2-like 2 (NRF2, also known as NFE2L2) interacts with these AREs and up-regulates HSF1 expression. We also found that BRM/SWI2-related gene 1 (BRG1), a catalytic subunit of SWI2/SNF2-like chromatin remodeler, is involved in this process. We further show that NRF2-dependent HSF1 gene regulation plays a crucial role in cancer cell biology, as interference with NRF2-mediated HSF1 activation compromised survival, migration potential, and the epithelial-to-mesenchymal transition and autophagy in MCF7 breast cancer cells exposed to oxidative stress. Taken together, our findings unravel the mechanistic basis of HSF1 gene regulation in cancer cells and provide molecular evidence supporting a direct interaction between HSF1 and NRF2, critical regulators of two cytoprotective mechanisms exploited by cancer cells.
Collapse
Affiliation(s)
| | | | - Sukhendu Mandal
- Department of Biochemistry, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata, India 700054
| | - Subrata Sau
- Department of Biochemistry, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata, India 700054
| | | |
Collapse
|
75
|
Lellahi SM, Rosenlund IA, Hedberg A, Kiær LT, Mikkola I, Knutsen E, Perander M. The long noncoding RNA NEAT1 and nuclear paraspeckles are up-regulated by the transcription factor HSF1 in the heat shock response. J Biol Chem 2018; 293:18965-18976. [PMID: 30305397 DOI: 10.1074/jbc.ra118.004473] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/02/2018] [Indexed: 01/08/2023] Open
Abstract
The long noncoding RNA (lncRNA) NEAT1 (nuclear enriched abundant transcript 1) is the architectural component of nuclear paraspeckles, and it has recently gained considerable attention as it is abnormally expressed in pathological conditions such as cancer and neurodegenerative diseases. NEAT1 and paraspeckle formation are increased in cells upon exposure to a variety of environmental stressors and believed to play an important role in cell survival. The present study was undertaken to further investigate the role of NEAT1 in cellular stress response pathways. We show that NEAT1 is a novel target gene of heat shock transcription factor 1 (HSF1) and is up-regulated when the heat shock response pathway is activated by sulforaphane (SFN) or elevated temperature. HSF1 binds specifically to a newly identified conserved heat shock element in the NEAT1 promoter. In line with this, SFN induced the formation of NEAT1-containing paraspeckles via an HSF1-dependent mechanism. HSF1 plays a key role in the cellular response to proteotoxic stress by promoting the expression of a series of genes, including those encoding molecular chaperones. We have found that the expression of HSP70, HSP90, and HSP27 is amplified and sustained during heat shock in NEAT1-depleted cells compared with control cells, indicating that NEAT1 feeds back via an unknown mechanism to regulate HSF1 activity. This interrelationship is potentially significant in human diseases such as cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | | | - Ingvild Mikkola
- Pharmacy, Faculty of Health Sciences, UiT-The Arctic University of Norway, N-9037 Tromsø, Norway
| | | | | |
Collapse
|
76
|
Moyano P, García JM, Lobo M, Anadón MJ, Sola E, Pelayo A, García J, Frejo MT, Pino JD. Cadmium alters heat shock protein pathways in SN56 cholinergic neurons, leading to Aβ and phosphorylated Tau protein generation and cell death. Food Chem Toxicol 2018; 121:297-308. [PMID: 30213552 DOI: 10.1016/j.fct.2018.09.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 11/28/2022]
Abstract
Cadmium, a neurotoxic environmental compound, produces cognitive disorders, although the mechanism remains unknown. Cadmium induces a more pronounced cell death on cholinergic neurons from basal forebrain (BF), mediated, in part, by increase in Aβ and total and phosphorylated Tau protein levels, which may explain cadmium effects on learning and memory processes. Cadmium downregulates the expression of heat shock proteins (HSPs) HSP 90, HSP70 and HSP27, and of HSF1, the master regulator of the HSP pathway. HSPs proteins reduce the production of Aβ and phosphorylated Tau proteins and avoid cell death pathways induction. Thus, we hypothesized that cadmium induced the production of Aβ and Tau proteins by HSP pathway disruption through HSF1 expression alteration, leading to BF cholinergic neurons cell death. Our results show that cadmium downregulates HSF1, leading to HSP90, HSP70 and HSP27 gene expression downregulation in BF SN56 cholinergic neurons. In addition, cadmium induced Aβ and total and phosphorylated Tau proteins generation, mediated partially by HSP90, HSP70 and HSP27 disruption, leading to cell death. These results provide new understanding of the mechanisms contributing to cadmium harmful effects on cholinergic neurons.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Toxicology and Legal Medicine, Medical School, Complutense University of Madrid, 28041, Madrid, Spain
| | - José Manuel García
- Department of Toxicology and Legal Medicine, Medical School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Margarita Lobo
- Department of Toxicology and Pharmacology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - María José Anadón
- Department of Toxicology and Legal Medicine, Medical School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Emma Sola
- Department of Pathological Anatomy, Medical School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Adela Pelayo
- Department of Pathological Anatomy, Medical School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Jimena García
- Department of Toxicology and Legal Medicine, Medical School, Complutense University of Madrid, 28041, Madrid, Spain
| | - María Teresa Frejo
- Department of Toxicology and Pharmacology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Javier Del Pino
- Department of Toxicology and Pharmacology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
77
|
Bianchi M, Crinelli R, Arbore V, Magnani M. Induction of ubiquitin C ( UBC) gene transcription is mediated by HSF1: role of proteotoxic and oxidative stress. FEBS Open Bio 2018; 8:1471-1485. [PMID: 30186748 PMCID: PMC6120222 DOI: 10.1002/2211-5463.12484] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/16/2018] [Accepted: 06/26/2018] [Indexed: 12/14/2022] Open
Abstract
The polyubiquitin gene ubiquitin C (UBC) is considered a stress protective gene and is upregulated under various stressful conditions, which is probably a consequence of an increased demand for ubiquitin in order to remove toxic misfolded proteins. We previously identified heat shock elements (HSEs) within the UBC promoter, which are responsible for heat shock factor (HSF)1‐driven induction of the UBC gene and are activated by proteotoxic stress. Here, we determined the molecular players driving the UBC gene transcriptional response to arsenite treatment, mainly addressing the role of the nuclear factor‐erythroid 2‐related factor 2 (Nrf2)‐mediated antioxidant pathway. Exposure of HeLa cells to arsenite caused a time‐dependent increase of UBCmRNA, while cell viability and proteasome activity were not affected. Nuclear accumulation of HSF1 and Nrf2 transcription factors was detected upon both arsenite and MG132 treatment, while HSF2 nuclear levels increased in MG132‐treated cells. Notably, siRNA‐mediated knockdown of Nrf2 did not reduce UBC transcription under either basal or stressful conditions, but significantly impaired the constitutive and inducible expression of well‐known antioxidant response element‐dependent genes. A chromatin immunoprecipitation assay consistently failed to detect Nrf2 binding to the UBC promoter sequence. By contrast, depletion of HSF1, but not HSF2, significantly compromised stress‐induced UBC expression. Critically, HSF1‐mediated UBC trans‐activation upon arsenite exposure relies on transcription factor binding to previously mapped distal HSEs, as demonstrated to occur under proteasome inhibition. These data highlight HSF1 as the pivotal transcription factor that translates different stress signals into UBC gene transcriptional induction.
Collapse
Affiliation(s)
- Marzia Bianchi
- Department of Biomolecular Sciences, Biochemistry and Molecular Biology Section University of Urbino 'Carlo Bo' Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, Biochemistry and Molecular Biology Section University of Urbino 'Carlo Bo' Italy
| | - Vanessa Arbore
- Department of Biomolecular Sciences, Biochemistry and Molecular Biology Section University of Urbino 'Carlo Bo' Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, Biochemistry and Molecular Biology Section University of Urbino 'Carlo Bo' Italy
| |
Collapse
|
78
|
Rojo de la Vega M, Chapman E, Zhang DD. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018; 34:21-43. [PMID: 29731393 PMCID: PMC6039250 DOI: 10.1016/j.ccell.2018.03.022] [Citation(s) in RCA: 1153] [Impact Index Per Article: 164.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/11/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022]
Abstract
The transcription factor NRF2 is the master regulator of the cellular antioxidant response. Though recognized originally as a target of chemopreventive compounds that help prevent cancer and other maladies, accumulating evidence has established the NRF2 pathway as a driver of cancer progression, metastasis, and resistance to therapy. Recent studies have identified new functions for NRF2 in the regulation of metabolism and other essential cellular functions, establishing NRF2 as a truly pleiotropic transcription factor. In this review, we explore the roles of NRF2 in the hallmarks of cancer, indicating both tumor suppressive and tumor-promoting effects.
Collapse
Affiliation(s)
- Montserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
79
|
Deen AJ, Sihvola V, Härkönen J, Patinen T, Adinolfi S, Levonen AL. Regulation of stress signaling pathways by nitro-fatty acids. Nitric Oxide 2018; 78:S1089-8603(17)30323-3. [PMID: 29567143 DOI: 10.1016/j.niox.2018.03.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/09/2018] [Accepted: 03/16/2018] [Indexed: 12/31/2022]
Abstract
Electrophilic nitrated-fatty acids (NO2-FA, nitroalkenes) are formed during reactions of NO-derived oxidized species (•NO, •NO2) with either free or esterified polyunsaturated fatty acids. Due to their electrophilic character, they react with nucleophiles such as cysteine thiols in signaling proteins, thereby triggering downstream signaling cascades. Herein, we review two stress-signaling pathways activated by nitroalkenes, the KEAP1-NRF2 signaling pathway and the heat shock response (HSR) pathway. In addition, their biological and pharmacological relevance are discussed. Given that perturbations in both proteostasis and redox balance are common in many disease processes, dual activation of both pathways by nitroalkenes is a promising pharmacological approach for their treatment.
Collapse
Affiliation(s)
- Ashik Jawahar Deen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FI-70211, Finland
| | - Virve Sihvola
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FI-70211, Finland
| | - Jouni Härkönen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FI-70211, Finland
| | - Tommi Patinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FI-70211, Finland
| | - Simone Adinolfi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FI-70211, Finland
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, Kuopio FI-70211, Finland.
| |
Collapse
|
80
|
Murakami A. Non-specific protein modifications may be novel mechanism underlying bioactive phytochemicals. J Clin Biochem Nutr 2018; 62:115-123. [PMID: 29610550 PMCID: PMC5874230 DOI: 10.3164/jcbn.17-113] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/27/2017] [Indexed: 12/27/2022] Open
Abstract
In a variety of experimental models, dietary phytochemicals have been demonstrated to exhibit pronounced and versatile bioactivities. Importantly, the possibility of such phytochemicals for human application has been supported in part by epidemiological surveys, which have demonstrated that frequent ingestion of vegetables and fruits containing abundant phytochemicals lowers the risk of onset of various diseases. However, the action mechanisms underlying those dietary phytochemical activities remain to be fully elucidated. For example, even though the anti-oxidant effects of natural polyphenols have long received widespread attention from food scientists, their roles in and contribution to those bioactivities remain controversial because of their poor bioavailability, resulting in extremely low concentrations in the bloodstream. Meanwhile, another important question is why phytochemicals have beneficial effects for animals, including humans, since they are biosynthesized by plants as compounds necessary for adaptation to environmental stress. In regard to that fundamental question, we recently reported novel and unique mechanisms of action of zerumbone, a sesquiterpene with anti-inflammatory and chemopreventive properties. This agent was found to partially exhibit bioactivity through its non-specific interactions with cellular proteins. More strikingly, a non-specific protein binding action of zerumbone was revealed to partially contribute to its anti-inflammatory functions via activation of heat shock factor 1. The present review article highlights and introduces our recent findings regarding the proteo-stress-mediated mechanisms of this phytochemical, along with the concept of hormesis.
Collapse
Affiliation(s)
- Akira Murakami
- Food Hormesis Laboratory, Department of Food Science & Nutrition, School of Human Science & Environment, Research Institute for Food and Nutritional Sciences, University of Hyogo, 1-1-12 Shinzaike-Honcho, Himeji, Hyogo 670-0092, Japan
| |
Collapse
|
81
|
Lu Z, Casalino-Matsuda SM, Nair A, Buchbinder A, Budinger GRS, Sporn PHS, Gates KL. A role for heat shock factor 1 in hypercapnia-induced inhibition of inflammatory cytokine expression. FASEB J 2018; 32:3614-3622. [PMID: 29405096 DOI: 10.1096/fj.201701164r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Hypercapnia, elevated levels of CO2 in the blood, is a known marker for poor clinical prognosis and is associated with increased mortality in patients hospitalized with both bacterial and viral pneumonias. Although studies have established a connection between elevated CO2 levels and poor pneumonia outcomes, a mechanistic basis of this association has not yet been established. We previously reported that hypercapnia inhibits expression of key NF-κB-regulated, innate immune cytokines, TNF-α, and IL-6, in LPS-stimulated macrophages in vitro and in mice during Pseudomonas pneumonia. The transcription factor heat shock factor 1 (HSF1) is important in maintaining proteostasis during stress and has been shown to negatively regulate NF-κB activity. In this study, we tested the hypothesis that HSF1 activation in response to hypercapnia results in attenuated NF-κB-regulated gene expression. We found that hypercapnia induced the protein expression and nuclear accumulation of HSF1 in primary murine alveolar macrophages and in an alveolar macrophage cell line (MH-S). In MH-S cells treated with short interfering RNA targeting Hsf1, LPS-induced IL-6 and TNF-α release were elevated during exposure to hypercapnia. Pseudomonas-infected Hsf1+/+ (wild-type) mice, maintained in a hypercapnic environment, showed lower levels of IL-6 and TNF-α in bronchoalveolar lavage fluid and IL-1β in lung tissue than did infected mice maintained in room air. In contrast, infected Hsf1+/- mice exposed to either hypercapnia or room air had similarly elevated levels of those cytokines. These results suggest that hypercapnia-mediated inhibition of NF-κB cytokine production is dependent on HSF1 expression and/or activation.-Lu, Z., Casalino-Matsuda, S. M., Nair, A., Buchbinder, A., Budinger, G. R. S., Sporn, P. H. S., Gates, K. L. A role for heat shock factor 1 in hypercapnia-induced inhibition of inflammatory cytokine expression.
Collapse
Affiliation(s)
- Ziyan Lu
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - S Marina Casalino-Matsuda
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Aisha Nair
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Jesse Brown Veteran Affairs Medical Center, Chicago, Illinois, USA
| | - Anja Buchbinder
- Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - G R Scott Budinger
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Jesse Brown Veteran Affairs Medical Center, Chicago, Illinois, USA
| | - Peter H S Sporn
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Jesse Brown Veteran Affairs Medical Center, Chicago, Illinois, USA
| | - Khalilah L Gates
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
82
|
Abstract
The NFE2L2 gene encodes the transcription factor Nrf2 best known for regulating the expression of antioxidant and detoxification genes. Gene knockout approaches have demonstrated its universal cytoprotective features. While Nrf2 has been the topic of intensive research in cancer biology since its discovery in 1994, understanding the role of Nrf2 in cardiovascular disease has just begun. The literature concerning Nrf2 in experimental models of atherosclerosis, ischemia, reperfusion, cardiac hypertrophy, heart failure, and diabetes supports its cardiac protective character. In addition to antioxidant and detoxification genes, Nrf2 has been found to regulate genes participating in cell signaling, transcription, anabolic metabolism, autophagy, cell proliferation, extracellular matrix remodeling, and organ development, suggesting that Nrf2 governs damage resistance as well as wound repair and tissue remodeling. A long list of small molecules, most derived from natural products, have been characterized as Nrf2 inducers. These compounds disrupt Keap1-mediated Nrf2 ubquitination, thereby prohibiting proteasomal degradation and allowing Nrf2 protein to accumulate and translocate to the nucleus, where Nrf2 interacts with sMaf to bind to ARE in the promoter of genes. Recently alternative mechanisms driving Nrf2 protein increase have been revealed, including removal of Keap1 by autophagy due to p62/SQSTM1 binding, inhibition of βTrCP or Synoviolin/Hrd1-mediated ubiquitination of Nrf2, and de novo Nrf2 protein translation. We review here a large volume of literature reporting historical and recent discoveries about the function and regulation of Nrf2 gene. Multiple lines of evidence presented here support the potential of dialing up the Nrf2 pathway for cardiac protection in the clinic.
Collapse
Affiliation(s)
- Qin M Chen
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Anthony J Maltagliati
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
83
|
Frigola J, Iturbide A, Lopez-Bigas N, Peiro S, Gonzalez-Perez A. Altered oncomodules underlie chromatin regulatory factors driver mutations. Oncotarget 2017; 7:30748-59. [PMID: 27095575 PMCID: PMC5058714 DOI: 10.18632/oncotarget.8752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/31/2016] [Indexed: 11/25/2022] Open
Abstract
Chromatin regulatory factors (CRFs), are known to be involved in tumorigenesis in several cancer types. Nevertheless, the molecular mechanisms through which driver alterations of CRFs cause tumorigenesis remain unknown. Here, we developed a CRFs Oncomodules Discovery approach, which mines several sources of cancer genomics and perturbaomics data. The approach prioritizes sets of genes significantly miss-regulated in primary tumors (oncomodules) bearing mutations of driver CRFs. We applied the approach to eleven TCGA tumor cohorts and uncovered oncomodules potentially associated to mutations of five driver CRFs in three cancer types. Our results revealed, for example, the potential involvement of the mTOR pathway in the development of tumors with loss-of-function mutations of MLL2 in head and neck squamous cell carcinomas. The experimental validation that MLL2 loss-of-function increases the sensitivity of cancer cell lines to mTOR inhibition lends further support to the validity of our approach. The potential oncogenic modules detected by our approach may guide experiments proposing ways to indirectly target driver mutations of CRFs.
Collapse
Affiliation(s)
- Joan Frigola
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Ane Iturbide
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain
| | - Nuria Lopez-Bigas
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Sandra Peiro
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain
| | - Abel Gonzalez-Perez
- Research Program on Biomedical Informatics, IMIM Hospital del Mar Medical Research Institute and Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| |
Collapse
|
84
|
Nakamura T, Abe-Kanoh N, Nakamura Y. Physiological relevance of covalent protein modification by dietary isothiocyanates. J Clin Biochem Nutr 2017; 62:11-19. [PMID: 29371751 PMCID: PMC5773839 DOI: 10.3164/jcbn.17-91] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/01/2017] [Indexed: 12/15/2022] Open
Abstract
Isothiocyanates (ITCs), naturally occurring in abundance in cruciferous vegetables, are the most well-studied organosulfur compounds having an electrophilic reactivity. ITCs have been accepted as major ingredients of these vegetables that afford their health promoting potentials. ITCs are able to modulate protein functions related to drug-metabolizing enzymes, transporters, kinases and phosphatases, etc. One of the most important questions about the molecular basis for the health promoting effects of ITCs is how they modulate cellular target proteins. Although the molecular targets of ITCs remains to be validated, dietary modulation of the target proteins via covalent modification by ITCs should be one of the promising strategies for the protection of cells against oxidative and inflammatory damage. This review discusses the plausible target proteins of dietary ITCs with an emphasis on possible involvement of protein modification in their health promoting effects. The fundamental knowledge of ITCs is also included with consideration of the chemistry, intracellular behavior, and metabolism.
Collapse
Affiliation(s)
- Toshiyuki Nakamura
- Graduate School of Environmental and Life Science, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Naomi Abe-Kanoh
- Graduate School of Environmental and Life Science, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.,Department of Food Science, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yoshimasa Nakamura
- Graduate School of Environmental and Life Science, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
85
|
Menga M, Trotta R, Scrima R, Pacelli C, Silvestri V, Piccoli C, Capitanio N, Liso A. Febrile temperature reprograms by redox-mediated signaling the mitochondrial metabolic phenotype in monocyte-derived dendritic cells. Biochim Biophys Acta Mol Basis Dis 2017; 1864:685-699. [PMID: 29246446 DOI: 10.1016/j.bbadis.2017.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/15/2017] [Accepted: 12/08/2017] [Indexed: 02/06/2023]
Abstract
Fever-like hyperthermia is known to stimulate innate and adaptive immune responses. Hyperthermia-induced immune stimulation is also accompanied with, and likely conditioned by, changes in the cell metabolism and, in particular, mitochondrial metabolism is now recognized to play a pivotal role in this context, both as energy supplier and as signaling platform. In this study we asked if challenging human monocyte-derived dendritic cells with a relatively short-time thermal shock in the fever-range, typically observed in humans, caused alterations in the mitochondrial oxidative metabolism. We found that following hyperthermic stress (3h exposure at 39°C) TNF-α-releasing dendritic cells undergo rewiring of the oxidative metabolism hallmarked by decrease of the mitochondrial respiratory activity and of the oxidative phosphorylation and increase of lactate production. Moreover, enhanced production of reactive oxygen and nitrogen species and accumulation of mitochondrial Ca2+ was consistently observed in hyperthermia-conditioned dendritic cells and exhibited a reciprocal interplay. The hyperthermia-induced impairment of the mitochondrial respiratory activity was (i) irreversible following re-conditioning of cells to normothermia, (ii) mimicked by exposing normothermic cells to the conditioned medium of the hyperthermia-challenged cells, (iii) largely prevented by antioxidant and inhibitors of the nitric oxide synthase and of the mitochondrial calcium porter, which also inhibited release of TNF-α. These observations combined with gene expression analysis support a model based on a thermally induced autocrine signaling, which rewires and sets a metabolism checkpoint linked to immune activation of dendritic cells.
Collapse
Affiliation(s)
- Marta Menga
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Rosa Trotta
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Veronica Silvestri
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| | - Arcangelo Liso
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
86
|
Dinkova-Kostova AT, Fahey JW, Kostov RV, Kensler TW. KEAP1 and Done? Targeting the NRF2 Pathway with Sulforaphane. Trends Food Sci Technol 2017; 69:257-269. [PMID: 29242678 PMCID: PMC5725197 DOI: 10.1016/j.tifs.2017.02.002] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/12/2017] [Accepted: 02/14/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Since the re-discovery of sulforaphane in 1992 and the recognition of the bioactivity of this phytochemical, many studies have examined its mode of action in cells, animals and humans. Broccoli, especially as young sprouts, is a rich source of sulforaphane and broccoli-based preparations are now used in clinical studies probing efficacy in health preservation and disease mitigation. Many putative cellular targets are affected by sulforaphane although only one, KEAP1-NRF2 signaling, can be considered a validated target at this time. The transcription factor NRF2 is a master regulator of cell survival responses to endogenous and exogenous stressors. SCOPE AND APPROACH This review summarizes the chemical biology of sulforaphane as an inducer of NRF2 signaling and efficacy as an inhibitor of carcinogenesis. It also provides a summary of the current findings from clinical trials using a suite of broccoli sprout preparations on a series of short-term endpoints reflecting a diversity of molecular actions. KEY FINDINGS AND CONCLUSIONS Sulforaphane, as a pure chemical, protects against chemical-induced skin, oral, stomach, colon, lung and bladder carcinogenesis and in genetic models of colon and prostate carcinogenesis. In many of these settings the antitumorigenic efficacy of sulforaphane is dampened in Nrf2-disrupted animals. Broccoli preparations rich in glucoraphanin or sulforaphane exert demonstrable pharmacodynamic action in over a score of clinical trials. Measures of NRF2 pathway response and function are serving as guideposts for the optimization of dose, schedule and formulation as clinical trials with broccoli-based preparations become more commonplace and more rigorous in design and implementation.
Collapse
Affiliation(s)
- Albena T. Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jed W. Fahey
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Human Nutrition, Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rumen V. Kostov
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W. Kensler
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
87
|
Bogen KT. Linear-No-Threshold Default Assumptions are Unwarranted for Cytotoxic Endpoints Independently Triggered by Ultrasensitive Molecular Switches. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2017; 37:1808-1816. [PMID: 28437864 DOI: 10.1111/risa.12813] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 03/03/2017] [Indexed: 06/07/2023]
Abstract
Crump's response in this issue to my critique of linear-no-threshold (LNT) default assumptions for noncancer and nongenotoxic cancer risks (Risk Analysis 2016; 36(3):589-604) is rebutted herein. Crump maintains that distinguishing between a low-dose linear dose response and a threshold dose response on the basis of dose-response data is impossible even for endpoints involving increased cytotoxicity. My rebuttal relies on descriptions and specific illustrations of two well-characterized ultrasensitive molecular switches that govern two key cytoprotective responses to cellular stress-heat shock response and antioxidant response element activation, respectively-each of which serve to suppress stress-induced apoptotic cell death unless overwhelmed. Because detailed dose-response data for each endpoint is shown to be J- or inverted-J-shaped with high confidence, and because independent pathways can explain background rates of apoptosis, LNT assumptions for this cytotoxic endpoint are unwarranted, at least in some cases and perhaps generally.
Collapse
|
88
|
Dayalan Naidu S, Dikovskaya D, Gaurilcikaite E, Knatko EV, Healy ZR, Mohan H, Koh G, Laurell A, Ball G, Olagnier D, de la Vega L, Ganley IG, Talalay P, Dinkova-Kostova AT. Transcription factors NRF2 and HSF1 have opposing functions in autophagy. Sci Rep 2017; 7:11023. [PMID: 28887499 PMCID: PMC5591275 DOI: 10.1038/s41598-017-11262-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/22/2017] [Indexed: 12/19/2022] Open
Abstract
Autophagy plays a critical role in the maintenance of cellular homeostasis by degrading proteins, lipids and organelles. Autophagy is activated in response to stress, but its regulation in the context of other stress response pathways, such as those mediated by heat shock factor 1 (HSF1) and nuclear factor-erythroid 2 p45-related factor 2 (NRF2), is not well understood. We found that the Michael acceptor bis(2-hydoxybenzylidene)acetone (HBB2), a dual activator of NRF2 and HSF1, protects against the development of UV irradiation-mediated cutaneous squamous cell carcinoma in mice. We further show that HBB2 is an inducer of autophagy. In cells, HBB2 increases the levels of the autophagy-cargo protein p62/sequestosome 1, and the lipidated form of microtubule-associated protein light chain 3 isoform B. Activation of autophagy by HBB2 is impaired in NRF2-deficient cells, which have reduced autophagic flux and low basal and induced levels of p62. Conversely, HSF1-deficient cells have increased autophagic flux under both basal as well as HBB2-induced conditions, accompanied by increased p62 levels. Our findings suggest that NRF2 and HSF1 have opposing roles during autophagy, and illustrate the existence of tight mechanistic links between the cellular stress responses.
Collapse
Affiliation(s)
- Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Dina Dikovskaya
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Egle Gaurilcikaite
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Elena V Knatko
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Zachary R Healy
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Pulmonary and Critical Care Medicine and Department of Internal Medicine, Duke University Hospital, Durham, NC, 27705, USA
| | - Hema Mohan
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Glenn Koh
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Axel Laurell
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Graeme Ball
- Dundee Imaging Facility, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - David Olagnier
- Lady Davis Institute-Jewish General Hospital, McGill University, Division of Experimental Medicine, Montreal, QC, Canada
- Department of Biomedicine, Aarhus Research Center for Innate Immunology, Aarhus University, Aarhus, 8000, Denmark
| | - Laureano de la Vega
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Ian G Ganley
- The Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| | - Paul Talalay
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
89
|
Quiles JM, Narasimhan M, Shanmugam G, Milash B, Hoidal JR, Rajasekaran NS. Differential regulation of miRNA and mRNA expression in the myocardium of Nrf2 knockout mice. BMC Genomics 2017; 18:509. [PMID: 28673258 PMCID: PMC5496330 DOI: 10.1186/s12864-017-3875-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022] Open
Affiliation(s)
- Justin M Quiles
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, BMR2 Room 533|901 19th Street South, Birmingham, AL, 35294-2180, USA
| | - Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Gobinath Shanmugam
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, BMR2 Room 533|901 19th Street South, Birmingham, AL, 35294-2180, USA
| | | | | | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Division of Molecular & Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, BMR2 Room 533|901 19th Street South, Birmingham, AL, 35294-2180, USA.
- Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA.
- Center for Free Radical Biology, University of Alabama at Birmingham, BMR2 Room 533|901 19th Street South, Birmingham, AL, 35294-2180, USA.
| |
Collapse
|
90
|
Clausen BH, Lundberg L, Yli-Karjanmaa M, Martin NA, Svensson M, Alfsen MZ, Flæng SB, Lyngsø K, Boza-Serrano A, Nielsen HH, Hansen PB, Finsen B, Deierborg T, Illes Z, Lambertsen KL. Fumarate decreases edema volume and improves functional outcome after experimental stroke. Exp Neurol 2017; 295:144-154. [PMID: 28602832 DOI: 10.1016/j.expneurol.2017.06.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/07/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Oxidative stress and inflammation exacerbate tissue damage in the brain after ischemic stroke. Dimethyl-fumarate (DMF) and its metabolite monomethyl-fumarate (MMF) are known to stimulate anti-oxidant pathways and modulate inflammatory responses. Considering these dual effects of fumarates, we examined the effect of MMF treatment after ischemic stroke in mice. METHODS Permanent middle cerebral artery occlusion (pMCAO) was performed using adult, male C57BL/6 mice. Thirty minutes after pMCAO, 20mg/kg MMF was administered intravenously. Outcomes were evaluated 6, 24 and 48h after pMCAO. First, we examined whether a bolus of MMF was capable of changing expression of kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1) and nuclear factor erythroid 2-related factor (Nrf)2 in the infarcted brain. Next, we studied the effect of MMF on functional recovery. To explore mechanisms potentially influencing functional changes, we examined infarct volumes, edema formation, the expression of heat shock protein (Hsp)72, hydroxycarboxylic acid receptor 2 (Hcar2), and inducible nitric oxide synthase (iNOS) in the infarcted brain using real-time PCR and Western blotting. Concentrations of a panel of pro- and anti-inflammatory cytokines (IFNγ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12p70, TNF) were examined in both the infarcted brain tissue and plasma samples 6, 24 and 48h after pMCAO using multiplex electrochemoluminiscence analysis. RESULTS Administration of MMF increased the protein level of Nrf2 6h after pMCAO, and improved functional outcome at 24 and 48h after pMCAO. MMF treatment did not influence infarct size, however reduced edema volume at both 24 and 48h after pMCAO. MMF treatment resulted in increased Hsp72 expression in the brain 6h after pMCAO. Hcar2 mRNA levels increased significantly 24h after pMCAO, but were not different between saline- and MMF-treated mice. MMF treatment also increased the level of the anti-inflammatory cytokine IL-10 in the brain and plasma 6h after pMCAO, and additionally reduced the level of the pro-inflammatory cytokine IL-12p70 in the brain at 24 and 48h after pMCAO. CONCLUSIONS A single intravenous bolus of MMF improved sensory-motor function after ischemic stroke, reduced edema formation, and increased the levels of the neuroprotective protein Hsp72 in the brain. The early increase in IL-10 and reduction in IL-12p70 in the brain combined with changes in systemic cytokine levels may also contribute to the functional recovery after pMCAO.
Collapse
Affiliation(s)
- Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark.
| | - Louise Lundberg
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark
| | - Minna Yli-Karjanmaa
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark.
| | - Nellie Anne Martin
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark; Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark.
| | - Martina Svensson
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Sölveg 19, Lund University, 22100 Lund, Sweden.
| | - Maria Zeiler Alfsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark.
| | - Simon Bertram Flæng
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark.
| | - Kristina Lyngsø
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21 3rd, DK-5000 Odense C, Denmark.
| | - Antonio Boza-Serrano
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Sölveg 19, Lund University, 22100 Lund, Sweden.
| | - Helle H Nielsen
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark.
| | - Pernille B Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21 3rd, DK-5000 Odense C, Denmark.
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark; BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsloewsvej 19, DK-5000 Odense C, Denmark.
| | - Tomas Deierborg
- Department of Experimental Medical Sciences, Experimental Neuroinflammation Laboratory, Sölveg 19, Lund University, 22100 Lund, Sweden.
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark; BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsloewsvej 19, DK-5000 Odense C, Denmark.
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21-25, DK-5000 Odense C, Denmark; Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, DK-5000 Odense C, Denmark; BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, J.B. Winsloewsvej 19, DK-5000 Odense C, Denmark.
| |
Collapse
|
91
|
Dayalan Naidu S, Dinkova-Kostova AT. Regulation of the mammalian heat shock factor 1. FEBS J 2017; 284:1606-1627. [PMID: 28052564 DOI: 10.1111/febs.13999] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/17/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022]
Abstract
Living organisms are endowed with the capability to tackle various forms of cellular stress due to the presence of molecular chaperone machinery complexes that are ubiquitous throughout the cell. During conditions of proteotoxic stress, the transcription factor heat shock factor 1 (HSF1) mediates the elevation of heat shock proteins, which are crucial components of the chaperone complex machinery and function to ameliorate protein misfolding and aggregation and restore protein homeostasis. In addition, HSF1 orchestrates a versatile transcriptional programme that includes genes involved in repair and clearance of damaged macromolecules and maintenance of cell structure and metabolism, and provides protection against a broad range of cellular stress mediators, beyond heat shock. Here, we discuss the structure and function of the mammalian HSF1 and its regulation by post-translational modifications (phosphorylation, sumoylation and acetylation), proteasomal degradation, and small-molecule activators and inhibitors.
Collapse
Affiliation(s)
- Sharadha Dayalan Naidu
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, UK
| | - Albena T Dinkova-Kostova
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, UK
- Department of Pharmacology and Molecular Sciences, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
92
|
Sihvola V, Levonen AL. Keap1 as the redox sensor of the antioxidant response. Arch Biochem Biophys 2017; 617:94-100. [DOI: 10.1016/j.abb.2016.10.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 12/30/2022]
|
93
|
Yi M, Yang J, Li W, Li X, Xiong W, McCarthy JB, Li G, Xiang B. The NOR1/OSCP1 proteins in cancer: from epigenetic silencing to functional characterization of a novel tumor suppressor. J Cancer 2017; 8:626-635. [PMID: 28367242 PMCID: PMC5370506 DOI: 10.7150/jca.17579] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/27/2016] [Indexed: 12/16/2022] Open
Abstract
NOR1 (Oxidored-nitro domain-containing protein 1), also known as OSCP1, was first identified in nasopharyngeal carcinoma (NPC) cells in 2003. NOR1 is evolutionarily conserved among species with its expression is restricted to brain, testis and respiratory epithelial cells. NOR1 was downregulated in NPC and the downregulation associates with poor prognosis. Previous study demonstrated that hypermethylation of NOR1 promoter was observed in NPC and hematological malignancies, which has been believed to be the main epigenetic cause for NOR1 silencing in these cancers. Recently, the NOR1 tumor suppressor status has been fully established. NOR1 inhibited cancer cell growth by disturbing tumor cell energe metabolism. NOR1 also promote tumor cells apoptosis in oxidative stress and hypoxia by inhibition of stress induced autophagy. Moreover, NOR1 suppressed cancer cell epithelial-mesenchymal transition, invasion and metastasis via activation of FOXA1/HDAC2-slug regulatory network. Deciphering the molecular mechanisms underlying NOR1 mediated tumor suppressive role would be helpful to a deeper understanding of carcinogenesis and, furthermore, to the development of new therapeutic approaches. Here we summarize the current knowledge on NOR1 focusing on its expression pattern, epigenetic and genetic association with human cancers and its biological functions. This review will also elucidate the potential application of NOR1/OSCP1 for some human malignancies.
Collapse
Affiliation(s)
- Mei Yi
- Department of Dermatology, Xiangya Hospital, The Central South University, Changsha, 410008, Hunan, China;; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan410078, China;; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan410078, China
| | - Jianbo Yang
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Wenjuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan410078, China;; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan410078, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan410078, China;; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan410078, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan410078, China;; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan410078, China
| | - James B McCarthy
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan410078, China;; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan410078, China
| | - Bo Xiang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan410078, China;; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan410078, China
| |
Collapse
|
94
|
Wang X, Liu D, He D, Suo S, Xia X, He X, Han JDJ, Zheng P. Transcriptome analyses of rhesus monkey preimplantation embryos reveal a reduced capacity for DNA double-strand break repair in primate oocytes and early embryos. Genome Res 2017; 27:567-579. [PMID: 28223401 PMCID: PMC5378175 DOI: 10.1101/gr.198044.115] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 02/10/2017] [Indexed: 12/31/2022]
Abstract
Preimplantation embryogenesis encompasses several critical events including genome reprogramming, zygotic genome activation (ZGA), and cell-fate commitment. The molecular basis of these processes remains obscure in primates in which there is a high rate of embryo wastage. Thus, understanding the factors involved in genome reprogramming and ZGA might help reproductive success during this susceptible period of early development and generate induced pluripotent stem cells with greater efficiency. Moreover, explaining the molecular basis responsible for embryo wastage in primates will greatly expand our knowledge of species evolution. By using RNA-seq in single and pooled oocytes and embryos, we defined the transcriptome throughout preimplantation development in rhesus monkey. In comparison to archival human and mouse data, we found that the transcriptome dynamics of monkey oocytes and embryos were very similar to those of human but very different from those of mouse. We identified several classes of maternal and zygotic genes, whose expression peaks were highly correlated with the time frames of genome reprogramming, ZGA, and cell-fate commitment, respectively. Importantly, comparison of the ZGA-related network modules among the three species revealed less robust surveillance of genomic instability in primate oocytes and embryos than in rodents, particularly in the pathways of DNA damage signaling and homology-directed DNA double-strand break repair. This study highlights the utility of monkey models to better understand the molecular basis for genome reprogramming, ZGA, and genomic stability surveillance in human early embryogenesis and may provide insights for improved homologous recombination-mediated gene editing in monkey.
Collapse
Affiliation(s)
- Xinyi Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China.,Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Denghui Liu
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dajian He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China.,Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Shengbao Suo
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xian Xia
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiechao He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ping Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| |
Collapse
|
95
|
Mills MG, Gallagher EP. A targeted gene expression platform allows for rapid analysis of chemical-induced antioxidant mRNA expression in zebrafish larvae. PLoS One 2017; 12:e0171025. [PMID: 28212397 PMCID: PMC5315391 DOI: 10.1371/journal.pone.0171025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 01/13/2017] [Indexed: 12/11/2022] Open
Abstract
Chemical-induced oxidative stress and the biochemical pathways that protect against oxidative damage are of particular interest in the field of toxicology. To rapidly identify oxidative stress-responsive gene expression changes in zebrafish, we developed a targeted panel of antioxidant genes using the Affymetrix QuantiGene Plex (QGP) platform. The genes contained in our panel include eight putative Nrf2 (Nfe2l2a)-dependent antioxidant genes (hmox1a, gstp1, gclc, nqo1, prdx1, gpx1a, sod1, sod2), a stress response gene (hsp70), an inducible DNA damage repair gene (gadd45bb), and three reference genes (actb1, gapdh, hprt1). We tested this platform on larval zebrafish exposed to tert-butyl hydroperoxide (tBHP) and cadmium (Cd), two model oxidative stressors with different modes of action, and compared our results with those obtained using the more common quantitative PCR (qPCR) method. Both methods showed that exposure to tBHP and Cd induced expression of prdx1, gstp1, and hmox1a (2- to 12-fold increase via QGP), indicative of an activated Nrf2 response in larval zebrafish. Both compounds also elicited a general stress response as reflected by elevation of hsp70 and gadd45bb, with Cd being the more potent inducer. Transient changes were observed in sod2 and gpx1a expression, whereas nqo1, an Nrf2-responsive gene in mammalian cells, was minimally affected by either tBHP or Cd chemical exposures. Developmental expression analysis of the target genes by QGP revealed marked upregulation of sod2 between 0-96hpf, and to a lesser extent, of sod1 and gstp1. Once optimized, QGP analysis of these experiments was accomplished more rapidly, using far less tissue, and at lower total costs than qPCR analysis. In summary, the QGP platform as applied to higher-throughput zebrafish studies provides a reasonable cost-effective alternative to qPCR or more comprehensive transcriptomics approaches to rapidly assess the potential for chemicals to elicit oxidative stress as a mechanism of chemical toxicity.
Collapse
Affiliation(s)
- Margaret G. Mills
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, United States of America
| | - Evan P. Gallagher
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
96
|
Glory A, Averill-Bates DA. The antioxidant transcription factor Nrf2 contributes to the protective effect of mild thermotolerance (40°C) against heat shock-induced apoptosis. Free Radic Biol Med 2016; 99:485-497. [PMID: 27591796 DOI: 10.1016/j.freeradbiomed.2016.08.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 08/07/2016] [Accepted: 08/31/2016] [Indexed: 10/21/2022]
Abstract
The exposure of cells to low doses of stress induces adaptive survival responses that protect cells against subsequent exposure to toxic stress. The ability of cells to resist subsequent toxic stress following exposure to low dose heat stress at 40°C is known as mild thermotolerance. Mild thermotolerance involves increased expression of heat shock proteins and antioxidants, but the initiating factors in this response are not understood. This study aims to understand the role of the Nrf2 antioxidant pathway in acquisition of mild thermotolerance at 40°C, and secondly, whether the Nrf2 pathway could be involved in the protective effect of thermotolerance against heat-shock (42°C)-induced apoptosis. During cell preconditioning at 40°C, protein expression of the Nrf2 transcription factor increased after 15-60min. In addition, levels of the Nrf2 targets MnSOD, catalase, heme oxygenase-1, glutamate cysteine ligase and Hsp70 increased at 40°C. Levels of these Nrf2 targets were enhanced by Nrf2 activator oltipraz and decreased by shRNA targeting Nrf2. Levels of pro-oxidants increased after 30-60min at 40°C. Pro-oxidant levels were decreased by oltipraz and increased by knockdown of Nrf2. Increased Nrf2 expression and catalase activity at 40°C were inhibited by the antioxidant PEG-catalase and by p53 inhibitor pifithrin-α. These results suggest that mild thermotolerance (40°C) increases cellular pro-oxidant levels, which in turn activate Nrf2 and its target genes. Moreover, Nrf2 contributes to the protective effect of thermotolerance against heat-shock (42°C)-induced apoptosis, because Nrf2 activation by oltipraz enhanced thermotolerance, whereas Nrf2 knockdown partly reversed thermotolerance. Improved knowledge about the different protective mechanisms that mild thermotolerance can activate is crucial for the potential use of this adaptive survival response to treat stress-related diseases.
Collapse
Affiliation(s)
- Audrey Glory
- Département des Sciences Biologiques (TOXEN), Université du Québec à Montréal, CP 8888, Succursale Center-Ville Montréal, Montréal, Québec, Canada H3C 3P8
| | - Diana A Averill-Bates
- Département des Sciences Biologiques (TOXEN), Université du Québec à Montréal, CP 8888, Succursale Center-Ville Montréal, Montréal, Québec, Canada H3C 3P8.
| |
Collapse
|
97
|
Heat Shock Factor 1 Is a Substrate for p38 Mitogen-Activated Protein Kinases. Mol Cell Biol 2016; 36:2403-17. [PMID: 27354066 PMCID: PMC5007788 DOI: 10.1128/mcb.00292-16] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/22/2016] [Indexed: 12/04/2022] Open
Abstract
Heat shock factor 1 (HSF1) monitors the structural integrity of the proteome. Phosphorylation at S326 is a hallmark for HSF1 activation, but the identity of the kinase(s) phosphorylating this site has remained elusive. We show here that the dietary agent phenethyl isothiocyanate (PEITC) inhibits heat shock protein 90 (Hsp90), the main negative regulator of HSF1; activates p38 mitogen-activated protein kinase (MAPK); and increases S326 phosphorylation, trimerization, and nuclear translocation of HSF1, and the transcription of a luciferase reporter, as well as the endogenous prototypic HSF1 target Hsp70. In vitro, all members of the p38 MAPK family rapidly and stoichiometrically catalyze the S326 phosphorylation. The use of stable knockdown cell lines and inhibitors indicated that among the p38 MAPKs, p38γ is the principal isoform responsible for the phosphorylation of HSF1 at S326 in cells. A protease-mass spectrometry approach confirmed S326 phosphorylation and unexpectedly revealed that p38 MAPK also catalyzes the phosphorylation of HSF1 at S303/307, previously known repressive posttranslational modifications. Thus, we have identified p38 MAPKs as highly efficient catalysts for the phosphorylation of HSF1. Furthermore, our findings suggest that the magnitude and persistence of activation of p38 MAPK are important determinants of the extent and duration of the heat shock response.
Collapse
|
98
|
Ding Y, Adachi H, Katsuno M, Sahashi K, Kondo N, Iida M, Tohnai G, Nakatsuji H, Sobue G. BIIB021, a synthetic Hsp90 inhibitor, induces mutant ataxin-1 degradation through the activation of heat shock factor 1. Neuroscience 2016; 327:20-31. [PMID: 27058144 DOI: 10.1016/j.neuroscience.2016.03.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/14/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease caused by the expansion of a polyglutamine (polyQ) tract in ataxin-1 (ATXN1). The pathological hallmarks of SCA1 are the loss of cerebellar Purkinje cells and neurons in the brainstem and the presence of nuclear aggregates containing the polyQ-expanded ATXN1 protein. Heat shock protein 90 (Hsp90) inhibitors have been shown to reduce polyQ-induced toxicity. This study was designed to examine the therapeutic effects of BIIB021, a purine-scaffold Hsp90 inhibitor, on the protein homeostasis of polyQ-expanded mutant ATXN1 in a cell culture model of SCA1. Our results demonstrated that BIIB021 activated heat shock factor 1 (HSF1) and suppressed the abnormal accumulation of ATXN1 and its toxicity. The pharmacological degradation of mutant ATXN1 via activated HSF1 was dependent on both the proteasome and autophagy systems. These findings indicate that HSF1 is a key molecule in the regulation of the protein homeostasis of the polyQ-expanded mutant ATXN1 and that Hsp90 has potential as a novel therapeutic target in patients with SCA1.
Collapse
Affiliation(s)
- Ying Ding
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiroaki Adachi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Naohide Kondo
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Madoka Iida
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hideaki Nakatsuji
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
99
|
Hahn ME, Timme-Laragy AR, Karchner SI, Stegeman JJ. Nrf2 and Nrf2-related proteins in development and developmental toxicity: Insights from studies in zebrafish (Danio rerio). Free Radic Biol Med 2015; 88:275-289. [PMID: 26130508 PMCID: PMC4698826 DOI: 10.1016/j.freeradbiomed.2015.06.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 12/14/2022]
Abstract
Oxidative stress is an important mechanism of chemical toxicity, contributing to developmental toxicity and teratogenesis as well as to cardiovascular and neurodegenerative diseases and diabetic embryopathy. Developing animals are especially sensitive to effects of chemicals that disrupt the balance of processes generating reactive species and oxidative stress, and those anti-oxidant defenses that protect against oxidative stress. The expression and inducibility of anti-oxidant defenses through activation of NFE2-related factor 2 (Nrf2) and related proteins is an essential process affecting the susceptibility to oxidants, but the complex interactions of Nrf2 in determining embryonic response to oxidants and oxidative stress are only beginning to be understood. The zebrafish (Danio rerio) is an established model in developmental biology and now also in developmental toxicology and redox signaling. Here we review the regulation of genes involved in protection against oxidative stress in developing vertebrates, with a focus on Nrf2 and related cap'n'collar (CNC)-basic-leucine zipper (bZIP) transcription factors. Vertebrate animals including zebrafish share Nfe2, Nrf1, Nrf2, and Nrf3 as well as a core set of genes that respond to oxidative stress, contributing to the value of zebrafish as a model system with which to investigate the mechanisms involved in regulation of redox signaling and the response to oxidative stress during embryolarval development. Moreover, studies in zebrafish have revealed nrf and keap1 gene duplications that provide an opportunity to dissect multiple functions of vertebrate NRF genes, including multiple sensing mechanisms involved in chemical-specific effects.
Collapse
Affiliation(s)
- Mark E Hahn
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America.
| | - Alicia R Timme-Laragy
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America; Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Sibel I Karchner
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America
| | - John J Stegeman
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts, United States of America
| |
Collapse
|
100
|
Marcelino TB, de Lemos Rodrigues PI, Miguel PM, Netto CA, Pereira Silva LO, Matté C. Effect of maternal exercise on biochemical parameters in rats submitted to neonatal hypoxia-ischemia. Brain Res 2015; 1622:91-101. [DOI: 10.1016/j.brainres.2015.06.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 01/25/2023]
|