1
|
Khurana A, Allawadhi P, Singh V, Khurana I, Yadav P, Sathua KB, Allwadhi S, Banothu AK, Navik U, Bharani KK. Antimicrobial and anti-viral effects of selenium nanoparticles and selenoprotein based strategies: COVID-19 and beyond. J Drug Deliv Sci Technol 2023; 86:104663. [PMID: 37362903 PMCID: PMC10249347 DOI: 10.1016/j.jddst.2023.104663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023]
Abstract
Deficiency of selenium (Se) has been described in a significant number of COVID-19 patients having a higher incidence of mortality, which makes it a pertinent issue to be addressed clinically for effective management of the COVID-19 pandemic. Se nanoparticles (SeNPs) provide a unique option for managing the havoc caused by the COVID-19 pandemic. SeNPs possess promising anti-inflammatory and anti-fibrotic effects by virtue of their nuclear factor kappa-light-chain-stimulator of activated B cells (NFκB), mitogen-activated protein kinase (MAPKs), and transforming growth factor-beta (TGF-β) modulatory activity. In addition, SeNPs possess remarkable immunomodulatory effects, making them a suitable option for supplementation with a much lower risk of toxicity compared to their elemental counterpart. Further, SeNPs have been shown to curtail viral and microbial infections, thus, making it a novel means to halt viral growth. In addition, it can be administered in the form of aerosol spray, direct injection, or infused thin-film transdermal patches to reduce the spread of this highly contagious viral infection. Moreover, a considerable decrease in the expression of selenoprotein along with enhanced expression of IL-6 in COVID-19 suggests a potential association among selenoprotein expression and COVID-19. In this review, we highlight the unique antimicrobial and antiviral properties of SeNPs and the immunomodulatory potential of selenoproteins. We provide the rationale behind their potentially interesting properties and further exploration in the context of microbial and viral infections. Further, the importance of selenoproteins and their role in maintaining a successful immune response along with their association to Se status is summarized.
Collapse
Affiliation(s)
- Amit Khurana
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Rajendranagar, Hyderabad, 500030, PVNRTVU, Telangana, India
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Warangal, 506166, PVNRTVU, Telangana, India
| | - Prince Allawadhi
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Vishakha Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Isha Khurana
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Kshirod Bihari Sathua
- Department of Pharmacology, College of Pharmaceutical Sciences, Konark Marine Drive Road, Puri, 752002, Odisha, India
| | - Sachin Allwadhi
- Department of Computer Science and Engineering, University Institute of Engineering and Technology (UIET), Maharshi Dayanand University (MDU), Rohtak, 124001, Haryana, India
| | - Anil Kumar Banothu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Rajendranagar, Hyderabad, 500030, PVNRTVU, Telangana, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Kala Kumar Bharani
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Warangal, 506166, PVNRTVU, Telangana, India
| |
Collapse
|
2
|
Abstract
Significance: Thioredoxin (Trx) is a powerful antioxidant that reduces protein disulfides to maintain redox stability in cells and is involved in regulating multiple redox-dependent signaling pathways. Recent Advance: The current accumulation of findings suggests that Trx participates in signaling pathways that interact with various proteins to manipulate their dynamic regulation of structure and function. These network pathways are critical for cancer pathogenesis and therapy. Promising clinical advances have been presented by most anticancer agents targeting such signaling pathways. Critical Issues: We herein link the signaling pathways regulated by the Trx system to potential cancer therapeutic opportunities, focusing on the coordination and strengths of the Trx signaling pathways in apoptosis, ferroptosis, immunomodulation, and drug resistance. We also provide a mechanistic network for the exploitation of therapeutic small molecules targeting the Trx signaling pathways. Future Directions: As research data accumulate, future complex networks of Trx-related signaling pathways will gain in detail. In-depth exploration and establishment of these signaling pathways, including Trx upstream and downstream regulatory proteins, will be critical to advancing novel cancer therapeutics. Antioxid. Redox Signal. 38, 403-424.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zhengjia Zhao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | | | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, China
| |
Collapse
|
3
|
Pouremamali F, Pouremamali A, Dadashpour M, Soozangar N, Jeddi F. An update of Nrf2 activators and inhibitors in cancer prevention/promotion. Cell Commun Signal 2022; 20:100. [PMID: 35773670 PMCID: PMC9245222 DOI: 10.1186/s12964-022-00906-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/09/2022] [Indexed: 01/01/2023] Open
Abstract
NF-E2-related factor 2 (Nrf2) protein is a basic-region leucine zipper transcription factor that defends against endogenous or exogenous stressors. By inducing several cytoprotective and detoxifying gene expressions, Nrf2 can increase the sensitivity of the cells to oxidants and electrophiles. Transient Nrf2 activation, by its specific activators, has protective roles against carcinogenesis and cancer development. However, permanent activation of Nrf2 promotes various cancer properties, comprising malignant progression, chemo/radio resistance, and poor patient prognosis. Taken together, these findings suggest that reaching an optimal balance between paradoxical functions of Nrf2 in malignancy may render a selective improvement to identify therapeutic strategies in cancer treatment. In this review, we describe lately discovered Nrf2 inducers and inhibitors, and their chemopreventive and/or anticancer activities. The Nrf2 pathway signifies one of the most significant cell defense procedures against exogenous or endogenous stressors. Certainly, by increasing the expression of several cytoprotective genes, the transcription factor Nrf2 can shelter cells and tissues from multiple sources of damage including electrophilic, xenobiotic, metabolic, and oxidative stress. Notably, the aberrant activation or accumulation of Nrf2, a common event in many tumors, confers a selective advantage to cancer cells and is connected to malignant progression, therapy resistance, and poor prognosis. Therefore, lately, Nrf2 has arisen as a hopeful target in treatment of cancer, and many struggles have been made to detect therapeutic strategies intended at disrupting its pro-oncogenic role. By summarizing the outcomes from past and recent studies, this review provided an overview concerning the Nrf2 pathway and the molecular mechanisms causing Nrf2 hyperactivation in cancer cells. Finally, this paper also described some of the most promising therapeutic approaches that have been successfully employed to counteract Nrf2 activity in tumors, with a particular emphasis on the development of natural compounds and the adoption of drug repurposing strategies. Video abstract
Collapse
Affiliation(s)
- Farhad Pouremamali
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Pouremamali
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.,Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Soozangar
- Digestive Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran. .,Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
4
|
Pourzand C, Albieri-Borges A, Raczek NN. Shedding a New Light on Skin Aging, Iron- and Redox-Homeostasis and Emerging Natural Antioxidants. Antioxidants (Basel) 2022; 11:471. [PMID: 35326121 PMCID: PMC8944509 DOI: 10.3390/antiox11030471] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/10/2022] Open
Abstract
Reactive oxygen species (ROS) are necessary for normal cell signaling and the antimicrobial defense of the skin. However excess production of ROS can disrupt the cellular redox balance and overwhelm the cellular antioxidant (AO) capacity, leading to oxidative stress. In the skin, oxidative stress plays a key role in driving both extrinsic and intrinsic aging. Sunlight exposure has also been a major contributor to extrinsic photoaging of the skin as its oxidising components disrupt both redox- and iron-homeostasis, promoting oxidative damage to skin cells and tissue constituents. Upon oxidative insults, the interplay between excess accumulation of ROS and redox-active labile iron (LI) and its detrimental consequences to the skin are often overlooked. In this review we have revisited the oxidative mechanisms underlying skin damage and aging by focussing on the concerted action of ROS and redox-active LI in the initiation and progression of intrinsic and extrinsic skin aging processes. Based on these, we propose to redefine the selection criteria for skin antiaging and photoprotective ingredients to include natural antioxidants (AOs) exhibiting robust redox-balancing and/or iron-chelating properties. This would promote the concept of natural-based or bio-inspired bifunctional anti-aging and photoprotective ingredients for skincare and sunscreen formulations with both AO and iron-chelating properties.
Collapse
Affiliation(s)
- Charareh Pourzand
- Medicines Design, Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK
- Medicines Development, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Andrea Albieri-Borges
- Research and Development, ASEA LLC., Pleasant Grove, UT 84062, USA; (A.A.-B.); (N.N.R.)
| | - Nico N. Raczek
- Research and Development, ASEA LLC., Pleasant Grove, UT 84062, USA; (A.A.-B.); (N.N.R.)
| |
Collapse
|
5
|
Ren Z, Liang H, Galbo PM, Dharmaratne M, Kulkarni AS, Fard AT, Aoun ML, Martinez-Lopez N, Suyama K, Benard O, Zheng W, Liu Y, Albanese J, Zheng D, Mar JC, Singh R, Prystowsky MB, Norton L, Hazan RB. Redox signaling by glutathione peroxidase 2 links vascular modulation to metabolic plasticity of breast cancer. Proc Natl Acad Sci U S A 2022; 119:e2107266119. [PMID: 35193955 PMCID: PMC8872779 DOI: 10.1073/pnas.2107266119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
In search of redox mechanisms in breast cancer, we uncovered a striking role for glutathione peroxidase 2 (GPx2) in oncogenic signaling and patient survival. GPx2 loss stimulates malignant progression due to reactive oxygen species/hypoxia inducible factor-α (HIF1α)/VEGFA (vascular endothelial growth factor A) signaling, causing poor perfusion and hypoxia, which were reversed by GPx2 reexpression or HIF1α inhibition. Ingenuity Pathway Analysis revealed a link between GPx2 loss, tumor angiogenesis, metabolic modulation, and HIF1α signaling. Single-cell RNA analysis and bioenergetic profiling revealed that GPx2 loss stimulated the Warburg effect in most tumor cell subpopulations, except for one cluster, which was capable of oxidative phosphorylation and glycolysis, as confirmed by coexpression of phosphorylated-AMPK and GLUT1. These findings underscore a unique role for redox signaling by GPx2 dysregulation in breast cancer, underlying tumor heterogeneity, leading to metabolic plasticity and malignant progression.
Collapse
Affiliation(s)
- Zuen Ren
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Huizhi Liang
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Phillip M Galbo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Malindrie Dharmaratne
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, 4072 QLD, Australia
| | - Ameya S Kulkarni
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Atefeh Taherian Fard
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, 4072 QLD, Australia
| | - Marie Louise Aoun
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Nuria Martinez-Lopez
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Kimita Suyama
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Wei Zheng
- Department of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Joseph Albanese
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jessica C Mar
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, 4072 QLD, Australia
| | - Rajat Singh
- Department of Endocrinology, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Larry Norton
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Rachel B Hazan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461;
| |
Collapse
|
6
|
Yan Q. The Yin-Yang Dynamics in Cancer Pharmacogenomics and Personalized Medicine. Methods Mol Biol 2022; 2547:141-163. [PMID: 36068463 DOI: 10.1007/978-1-0716-2573-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The enormous heterogeneity of cancer systems has made it very challenging to overcome drug resistance and adverse reactions to achieve personalized therapies. Recent developments in systems biology, especially the perception of cancer as the complex adaptive system (CAS), may help meet the challenges by deciphering the interactions at various levels from the molecular, cellular, tissue-organ, to the whole organism. The ubiquitous Yin-Yang interactions among the coevolving components, including the genes and proteins, decide their spatiotemporal features at various stages from cancer initiation to metastasis. The Yin-Yang imbalances across different systems levels, from genetic mutations to tumor cells adaptation, have been related to the intra- and inter-tumoral heterogeneity in the micro- and macro-environments. At the molecular and cellular levels, dysfunctional Yin-Yang dynamics in the cytokine networks, mitochondrial activities, redox systems, apoptosis, and metabolism can contribute to tumor cell growth and escape of immune surveillance. Up to the organism and system levels, the Yin-Yang imbalances in the cancer microenvironments can lead to different phenotypes from breast cancer to leukemia. These factors may be considered the systems-based biomarkers and treatment targets. The features of adaptation and nonlinearity in Yin-Yang dynamical interactions should be addressed by individualized drug combinations, dosages, intensities, timing, and frequencies at different cancer stages. The comprehensive "Yin-Yang dynamics" framework would enable powerful approaches for personalized and systems medicine strategies.
Collapse
|
7
|
Cumpstey AF, Clark AD, Santolini J, Jackson AA, Feelisch M. COVID-19: A Redox Disease-What a Stress Pandemic Can Teach Us About Resilience and What We May Learn from the Reactive Species Interactome About Its Treatment. Antioxid Redox Signal 2021; 35:1226-1268. [PMID: 33985343 DOI: 10.1089/ars.2021.0017] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus causing coronavirus disease 2019 (COVID-19), affects every aspect of human life by challenging bodily, socioeconomic, and political systems at unprecedented levels. As vaccines become available, their distribution, safety, and efficacy against emerging variants remain uncertain, and specific treatments are lacking. Recent Advances: Initially affecting the lungs, COVID-19 is a complex multisystems disease that disturbs the whole-body redox balance and can be long-lasting (Long-COVID). Numerous risk factors have been identified, but the reasons for variations in susceptibility to infection, disease severity, and outcome are poorly understood. The reactive species interactome (RSI) was recently introduced as a framework to conceptualize how cells and whole organisms sense, integrate, and accommodate stress. Critical Issues: We here consider COVID-19 as a redox disease, offering a holistic perspective of its effects on the human body, considering the vulnerability of complex interconnected systems with multiorgan/multilevel interdependencies. Host/viral glycan interactions underpin SARS-CoV-2's extraordinary efficiency in gaining cellular access, crossing the epithelial/endothelial barrier to spread along the vascular/lymphatic endothelium, and evading antiviral/antioxidant defences. An inflammation-driven "oxidative storm" alters the redox landscape, eliciting epithelial, endothelial, mitochondrial, metabolic, and immune dysfunction, and coagulopathy. Concomitantly reduced nitric oxide availability renders the sulfur-based redox circuitry vulnerable to oxidation, with eventual catastrophic failure in redox communication/regulation. Host nutrient limitations are crucial determinants of resilience at the individual and population level. Future Directions: While inflicting considerable damage to health and well-being, COVID-19 may provide the ultimate testing ground to improve the diagnosis and treatment of redox-related stress diseases. "Redox phenotyping" of patients to characterize whole-body RSI status as the disease progresses may inform new therapeutic approaches to regain redox balance, reduce mortality in COVID-19 and other redox diseases, and provide opportunities to tackle Long-COVID. Antioxid. Redox Signal. 35, 1226-1268.
Collapse
Affiliation(s)
- Andrew F Cumpstey
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anna D Clark
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jérôme Santolini
- Institute for Integrative Biology of the Cell (I2BC), Biochemistry, Biophysics and Structural Biology, CEA, CNRS, Université Paris-Sud, Universite Paris-Saclay, Gif-sur-Yvette, France
| | - Alan A Jackson
- Human Nutrition, University of Southampton and University Hospital Southampton, Southampton, United Kingdom
| | - Martin Feelisch
- Respiratory and Critical Care Research Group, Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.,Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
8
|
Vaghari-Tabari M, Jafari-Gharabaghlou D, Sadeghsoltani F, Hassanpour P, Qujeq D, Rashtchizadeh N, Ghorbanihaghjo A. Zinc and Selenium in Inflammatory Bowel Disease: Trace Elements with Key Roles? Biol Trace Elem Res 2021; 199:3190-3204. [PMID: 33098076 DOI: 10.1007/s12011-020-02444-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/18/2020] [Indexed: 12/21/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition that may emerge at a young age and often lasts for life. It often goes through phases of recurrence and remission and has a devastating effect on quality of life. The exact etiology of the disease is still unclear, but it appears that an inappropriate immune response to intestinal flora bacteria in people with a genetic predisposition may cause the disease. Managing inflammatory bowel disease is still a serious challenge. Oxidative stress and free radicals appear to be involved in the pathogenesis of this disease, and a number of studies have suggested the use of antioxidants as a therapeutic approach. The antioxidant and anti-inflammatory properties of some trace elements have led some of the research to focus on studying these trace elements in inflammatory bowel disease. Zinc and selenium are among the most important trace elements that have significant anti-inflammatory and antioxidant properties. Some studies have shown the importance of these trace elements in inflammatory bowel disease. In this review, we have attempted to provide a comprehensive overview of the findings of these studies and to gather current knowledge about the association of these trace elements with the inflammatory process and inflammatory bowel disease.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadeghsoltani
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Nadereh Rashtchizadeh
- Connective Tissue Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Amir Ghorbanihaghjo
- Biotechnology Research Center, Tabriz University of Medical Sciences, P.O. Box 14711, Tabriz, 5166614711, Iran.
| |
Collapse
|
9
|
Ferreira RLU, Sena-Evangelista KCM, de Azevedo EP, Pinheiro FI, Cobucci RN, Pedrosa LFC. Selenium in Human Health and Gut Microflora: Bioavailability of Selenocompounds and Relationship With Diseases. Front Nutr 2021; 8:685317. [PMID: 34150830 PMCID: PMC8211732 DOI: 10.3389/fnut.2021.685317] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
This review covers current knowledge of selenium in the dietary intake, its bioavailability, metabolism, functions, biomarkers, supplementation and toxicity, as well as its relationship with diseases and gut microbiota specifically on the symbiotic relationship between gut microflora and selenium status. Selenium is essential for the maintenance of the immune system, conversion of thyroid hormones, protection against the harmful action of heavy metals and xenobiotics as well as for the reduction of the risk of chronic diseases. Selenium is able to balance the microbial flora avoiding health damage associated with dysbiosis. Experimental studies have shown that inorganic and organic selenocompounds are metabolized to selenomethionine and incorporated by bacteria from the gut microflora, therefore highlighting their role in improving the bioavailability of selenocompounds. Dietary selenium can affect the gut microbial colonization, which in turn influences the host's selenium status and expression of selenoproteoma. Selenium deficiency may result in a phenotype of gut microbiota that is more susceptible to cancer, thyroid dysfunctions, inflammatory bowel disease, and cardiovascular disorders. Although the host and gut microbiota benefit each other from their symbiotic relationship, they may become competitors if the supply of micronutrients is limited. Intestinal bacteria can remove selenium from the host resulting in two to three times lower levels of host's selenoproteins under selenium-limiting conditions. There are still gaps in whether these consequences are unfavorable to humans and animals or whether the daily intake of selenium is also adapted to meet the needs of the bacteria.
Collapse
Affiliation(s)
| | - Karine Cavalcanti Maurício Sena-Evangelista
- Postgraduate Program in Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil.,Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Eduardo Pereira de Azevedo
- Graduate Program of Biotechnology, Laureate International Universities - Universidade Potiguar, Natal, Brazil
| | - Francisco Irochima Pinheiro
- Graduate Program of Biotechnology, Laureate International Universities - Universidade Potiguar, Natal, Brazil.,Medical School, Laureate International Universities - Universidade Potiguar, Natal, Brazil
| | - Ricardo Ney Cobucci
- Graduate Program of Biotechnology, Laureate International Universities - Universidade Potiguar, Natal, Brazil.,Medical School, Laureate International Universities - Universidade Potiguar, Natal, Brazil
| | - Lucia Fatima Campos Pedrosa
- Postgraduate Program in Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil.,Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
10
|
McCarty MF, Iloki Assanga SB, Lewis Luján L, O’Keefe JH, DiNicolantonio JJ. Nutraceutical Strategies for Suppressing NLRP3 Inflammasome Activation: Pertinence to the Management of COVID-19 and Beyond. Nutrients 2020; 13:E47. [PMID: 33375692 PMCID: PMC7823562 DOI: 10.3390/nu13010047] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 02/03/2023] Open
Abstract
Inflammasomes are intracellular protein complexes that form in response to a variety of stress signals and that serve to catalyze the proteolytic conversion of pro-interleukin-1β and pro-interleukin-18 to active interleukin-1β and interleukin-18, central mediators of the inflammatory response; inflammasomes can also promote a type of cell death known as pyroptosis. The NLRP3 inflammasome has received the most study and plays an important pathogenic role in a vast range of pathologies associated with inflammation-including atherosclerosis, myocardial infarction, the complications of diabetes, neurological and autoimmune disorders, dry macular degeneration, gout, and the cytokine storm phase of COVID-19. A consideration of the molecular biology underlying inflammasome priming and activation enables the prediction that a range of nutraceuticals may have clinical potential for suppressing inflammasome activity-antioxidants including phycocyanobilin, phase 2 inducers, melatonin, and N-acetylcysteine, the AMPK activator berberine, glucosamine, zinc, and various nutraceuticals that support generation of hydrogen sulfide. Complex nutraceuticals or functional foods featuring a number of these agents may find utility in the prevention and control of a wide range of medical disorders.
Collapse
Affiliation(s)
| | - Simon Bernard Iloki Assanga
- Department of Research and Postgraduate in Food, University of Sonora, Centro 83000, Mexico; (S.B.I.A.); (L.L.L.)
| | - Lidianys Lewis Luján
- Department of Research and Postgraduate in Food, University of Sonora, Centro 83000, Mexico; (S.B.I.A.); (L.L.L.)
| | | | | |
Collapse
|
11
|
McCarty MF, Lerner A. Perspective: Prospects for Nutraceutical Support of Intestinal Barrier Function. Adv Nutr 2020; 12:316-324. [PMID: 33126251 PMCID: PMC8243597 DOI: 10.1093/advances/nmaa139] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/28/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Impairment of intestinal barrier function is linked to certain pathologies and to aging, and can be a cause of bacterial infections, systemic and hepatic inflammation, food allergies, and autoimmune disorders. The formation and maintenance of intestinal tight junctions is supported by glucagon-like peptide-2 (GLP-2), which via insulin-like growth factor I activity boosts phosphoinositide 3-kinase/Akt/mammalian target of rapamycin complex 1 (PI3K/Akt/mTORC1) signaling in enterocytes. 5'-AMP-activated protein kinase (AMPK) activity as well as estrogen receptor-β (ERβ) activity are also protective in this regard. Conversely, activation of mitogen-activated protein kinases (MAPKs) and cellular Src (c-Src) under inflammatory conditions can induce dissociation of tight junctions. Hence, nutraceuticals that promote GLP-2 secretion from L cells-effective pre/probiotics, glycine, and glutamine-as well as diets rich in soluble fiber or resistant starch, can support intestinal barrier function. AMPK activators-notably berberine and the butyric acid produced by health-promoting microflora-are also beneficial in this regard, as are soy isoflavones, which function as selective agonists for ERβ. The adverse impact of MAPK and c-Src overactivation on the intestinal barrier can be combatted with various antioxidant measures, including phycocyanobilin, phase 2-inducer nutraceuticals, and N-acetylcysteine. These considerations suggest that rationally designed functional foods or complex supplementation programs could have clinical potential for supporting and restoring healthful intestinal barrier function.
Collapse
|
12
|
Stolwijk JM, Garje R, Sieren JC, Buettner GR, Zakharia Y. Understanding the Redox Biology of Selenium in the Search of Targeted Cancer Therapies. Antioxidants (Basel) 2020; 9:E420. [PMID: 32414091 PMCID: PMC7278812 DOI: 10.3390/antiox9050420] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/24/2020] [Accepted: 05/10/2020] [Indexed: 12/18/2022] Open
Abstract
Selenium (Se) is an essential trace nutrient required for optimal human health. It has long been suggested that selenium has anti-cancer properties. However, clinical trials have shown inconclusive results on the potential of Se to prevent cancer. The suggested role of Se in the prevention of cancer is centered around its role as an antioxidant. Recently, the potential of selenium as a drug rather than a supplement has been uncovered. Selenium compounds can generate reactive oxygen species that could enhance the treatment of cancer. Transformed cells have high oxidative distress. As normal cells have a greater capacity to meet oxidative challenges than tumor cells, increasing the flux of oxidants with high dose selenium treatment could result in cancer-specific cell killing. If the availability of Se is limited, supplementation of Se can increase the expression and activities of Se-dependent proteins and enzymes. In cell culture, selenium deficiency is often overlooked. We review the importance of achieving normal selenium biology and how Se deficiency can lead to adverse effects. We examine the vital role of selenium in the prevention and treatment of cancer. Finally, we examine the properties of Se-compounds to better understand how each can be used to address different research questions.
Collapse
Affiliation(s)
- Jeffrey M. Stolwijk
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA;
| | - Rohan Garje
- Department of Internal Medicine, Division of Medical Oncology and Hematology, The University of Iowa Hospital and Clinics—Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA;
| | - Jessica C. Sieren
- Departments of Radiology and Biomedical Engineering, The University of Iowa, Iowa City, IA 52242, USA;
| | - Garry R. Buettner
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA;
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
| | - Yousef Zakharia
- Department of Internal Medicine, Division of Medical Oncology and Hematology, The University of Iowa Hospital and Clinics—Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA;
| |
Collapse
|
13
|
Stanishevska NV. Selenoproteins and their emerging roles in signaling pathways. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The functional activity of selenoproteins has a wide range of effects on complex pathogenetic processes, including teratogenesis, immuno-inflammatory, neurodegenerative. Being active participants and promoters of many signaling pathways, selenoproteins support the lively interest of a wide scientific community. This review is devoted to the analysis of recent data describing the participation of selenoproteins in various molecular interactions mediating important signaling pathways. Data processing was carried out by the method of complex analysis. For convenience, all selenoproteins were divided into groups depending on their location and function. Among the group of selenoproteins of the ER membrane, selenoprotein N affects the absorption of Ca2+ by the endoplasmic reticulum mediated by oxidoreductin (ERO1), a key player in the CHOP/ERO1 branch, a pathogenic mechanism that causes myopathy. Another selenoprotein of the ER membrane selenoprotein K binding to the DHHC6 protein affects the IP3R receptor that regulates Ca2+ flux. Selenoprotein K is able to affect another protein of the endoplasmic reticulum CHERP, also appearing in Ca2+ transport. Selenoprotein S, associated with the lumen of ER, is able to influence the VCP protein, which ensures the incorporation of selenoprotein K into the ER membrane. Selenoprotein M, as an ER lumen protein, affects the phosphorylation of STAT3 by leptin, which confirms that Sel M is a positive regulator of leptin signaling. Selenoprotein S also related to luminal selenoproteins ER is a modulator of the IRE1α-sXBP1 signaling pathway. Nuclear selenoprotein H will directly affect the suppressor of malignant tumours, p53 protein, the activation of which increases with Sel H deficiency. The same selenoprotein is involved in redox regulation. Among the cytoplasmic selenoproteins, abundant investigations are devoted to SelP, which affects the PI3K/Akt/Erk signaling pathway during ischemia/reperfusion, is transported into the myoblasts through the plasmalemma after binding to the apoER2 receptor, and into the neurons to the megaline receptor and in general, selenoprotein P plays the role of a pool that stores the necessary trace element and releases it, if necessary, for vital selenoproteins. The thioredoxin reductase family plays a key role in the invasion and metastasis of salivary adenoid cystic carcinoma through the influence on the TGF-β-Akt/GSK-3β pathway during epithelial-mesenchymal transition. The deletion of thioredoxin reductase 1 affects the levels of messengers of the Wnt/β-catenin signaling pathway. No less studied is the glutathione peroxidase group, of which GPX3 is able to inhibit signaling in the Wnt/β-catenin pathway and thereby inhibit thyroid metastasis, as well as suppress protein levels in the PI3K/Akt/c-fos pathway. A key observation is that in cases of carcinogenesis, a decrease in GPX3 and its hypermethylation are almost always found. Among deiodinases, deiodinase 3 acts as a promoter of the oncogenes BRAF, MEK or p38, while stimulating a decrease in the expression of cyclin D1. The dependence of the level of deiodinase 3 on the Hedgehog (SHH) signaling pathway is also noted. Methionine sulfoxide reductase A can compete for the uptake of ubiquitin, reduce p38, JNK and ERK promoters of the MAPK signaling pathway; methionine sulfoxide reductase B1 suppresses MAPK signaling messengers, and also increases PARP and caspase 3.
Collapse
|
14
|
Hariharan S, Dharmaraj S. Selenium and selenoproteins: it's role in regulation of inflammation. Inflammopharmacology 2020; 28:667-695. [PMID: 32144521 PMCID: PMC7222958 DOI: 10.1007/s10787-020-00690-x] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 02/06/2020] [Indexed: 12/22/2022]
Abstract
Abstract Selenium is an essential immunonutrient which holds the human’s metabolic activity with its chemical bonds. The organic forms of selenium naturally present in human body are selenocysteine and selenoproteins. These forms have a unique way of synthesis and translational coding. Selenoproteins act as antioxidant warriors for thyroid regulation, male-fertility enhancement, and anti-inflammatory actions. They also participate indirectly in the mechanism of wound healing as oxidative stress reducers. Glutathione peroxidase (GPX) is the major selenoprotein present in the human body, which assists in the control of excessive production of free radical at the site of inflammation. Other than GPX, other selenoproteins include selenoprotein-S that regulates the inflammatory cytokines and selenoprotein-P that serves as an inducer of homeostasis. Previously, reports were mainly focused on the cellular and molecular mechanism of wound healing with reference to various animal models and cell lines. In this review, the role of selenium and its possible routes in translational decoding of selenocysteine, synthesis of selenoproteins, systemic action of selenoproteins and their indirect assimilation in the process of wound healing are explained in detail. Some of the selenium containing compounds which can acts as cancer preventive and therapeutics are also discussed. These compounds directly or indirectly exhibit antioxidant properties which can sustain the intracellular redox status and these activities protect the healthy cells from reactive oxygen species induced oxidative damage. Although the review covers the importance of selenium/selenoproteins in wound healing process, still some unresolved mystery persists which may be resolved in near future. Graphic abstract ![]()
Collapse
Affiliation(s)
- Sneha Hariharan
- Department of Biochemistry, Karpagam Academy of Higher Education, Eachanari Post, Pollachi Main Road, Coimbatore, Tamil Nadu, 641021, India
| | - Selvakumar Dharmaraj
- Department of Biochemistry, Karpagam Academy of Higher Education, Eachanari Post, Pollachi Main Road, Coimbatore, Tamil Nadu, 641021, India.
| |
Collapse
|
15
|
Silvestrini A, Mordente A, Martino G, Bruno C, Vergani E, Meucci E, Mancini A. The Role of Selenium in Oxidative Stress and in Nonthyroidal Illness Syndrome (NTIS): An Overview. Curr Med Chem 2020; 27:423-449. [PMID: 29421998 DOI: 10.2174/0929867325666180201111159] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/12/2018] [Accepted: 01/13/2018] [Indexed: 12/28/2022]
Abstract
Selenium is a trace element, nutritionally classified as an essential micronutrient, involved in maintaining the correct function of several enzymes incorporating the selenocysteine residue, namely the selenoproteins. The human selenoproteome including 25 proteins is extensively described here. The most relevant selenoproteins, including glutathione peroxidases, thioredoxin reductases and iodothyronine deiodinases are required for the proper cellular redox homeostasis as well as for the correct thyroid function, thus preventing oxidative stress and related diseases. This review summarizes the main advances on oxidative stress with a focus on selenium metabolism and transport. Moreover, thyroid-related disorders are discussed, considering that the thyroid gland contains the highest selenium amount per gram of tissue, also for future possible therapeutic implication.
Collapse
Affiliation(s)
- Andrea Silvestrini
- Institute of Biochemistry and Clinical Biochemistry, School of Medicine, Catholic University, Largo F. Vito 1, Rome 00168, Italy
| | - Alvaro Mordente
- Institute of Biochemistry and Clinical Biochemistry, School of Medicine, Catholic University, Largo F. Vito 1, Rome 00168, Italy
| | - Giuseppe Martino
- Operative Unit of Endocrinology, School of Medicine, Catholic University, Largo A. Gemelli 1, Rome, 00168, Italy
| | - Carmine Bruno
- Operative Unit of Endocrinology, School of Medicine, Catholic University, Largo A. Gemelli 1, Rome, 00168, Italy
| | - Edoardo Vergani
- Operative Unit of Endocrinology, School of Medicine, Catholic University, Largo A. Gemelli 1, Rome, 00168, Italy
| | - Elisabetta Meucci
- Institute of Biochemistry and Clinical Biochemistry, School of Medicine, Catholic University, Largo F. Vito 1, Rome 00168, Italy
| | - Antonio Mancini
- Operative Unit of Endocrinology, School of Medicine, Catholic University, Largo A. Gemelli 1, Rome, 00168, Italy
| |
Collapse
|
16
|
Abstract
Ferroptosis is a type of programmed cell death dependent on iron. It is different from other forms of cell death such as apoptosis, classic necrosis and autophagy. Ferroptosis is involved in many neurodegenerative diseases. The role of ferroptosis in glutamate-induced neuronal toxicity is not fully understood. To test its toxicity, glutamate (1.25–20 mM) was applied to HT-22 cells for 12 to 48 hours. The optimal experimental conditions occurred at 12 hours after incubation with 5 mM glutamate. Cells were cultured with 3–12 μM ferrostatin-1, an inhibitor of ferroptosis, for 12 hours before exposure to glutamate. The cell viability was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Autophagy was determined by monodansylcadaverine staining and apoptosis by caspase 3 activity. Damage to cell structures was observed under light and by transmission electron microscopy. The release of lactate dehydrogenase was detected by the commercial kit. Reactive oxygen species were measured by flow cytometry. Glutathione peroxidase activity, superoxide dismutase activity and malondialdehyde level were detected by the appropriate commercial kit. Prostaglandin peroxidase synthase 2 and glutathione peroxidase 4 gene expression was detected by real-time quantitative polymerase chain reaction. Glutathione peroxidase 4 and nuclear factor erythroid-derived-like 2 protein expression was detected by western blot analysis. Results showed that ferrostatin-1 can significantly counter the effects of glutamate on HT-22 cells, improving the survival rate, reducing the release of lactate dehydrogenase and reducing the damage to mitochondrial ultrastructure. However, it did not affect the caspase-3 expression and monodansylcadaverine-positive staining in glutamate-injured HT-22 cells. Ferrostatin-1 reduced the levels of reactive oxygen species and malondialdehyde and enhanced superoxide dismutase activity. It decreased gene expression of prostaglandin peroxidase synthase 2 and increased gene expression of glutathione peroxidase 4 and protein expressions of glutathione peroxidase 4 and nuclear factor (erythroid-derived)-like 2 in glutamate-injured HT-22 cells. Treatment of cultured cells with the apoptosis inhibitor Z-Val-Ala-Asp (OMe)-fluoromethyl ketone (2–8 μM), autophagy inhibitor 3-methyladenine (100–400 μM) or necrosis inhibitor necrostatin-1 (10–40 μM) had no effect on glutamate induced cell damage. However, the iron chelator deferoxamine mesylate salt inhibited glutamate induced cell death. Thus, the results suggested that ferroptosis is caused by glutamate-induced toxicity and that ferrostatin-1 protects HT-22 cells from glutamate-induced oxidative toxicity by inhibiting the oxidative stress.
Collapse
Affiliation(s)
- Jun Chu
- Xin'an Key Laboratory of Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Chen-Xu Liu
- Xin'an Key Laboratory of Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Rui Song
- Xin'an Key Laboratory of Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Qing-Lin Li
- Xin'an Key Laboratory of Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| |
Collapse
|
17
|
The potential effect of methylseleninic acid (MSA) against γ-irradiation induced testicular damage in rats: Impact on JAK/STAT pathway. Arch Biochem Biophys 2019; 679:108205. [PMID: 31758927 DOI: 10.1016/j.abb.2019.108205] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/31/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022]
Abstract
This study suggested that methylseleninic acid (MSA) could respond to the inflammatory signaling associated with ionizing radiation-induced testicular damage. Mature male rats were divided into four groups: negative control, whole body γ-irradiated (IRR) (5 Gy), MSA (0.5 mg/kg, daily for nine consecutive days), and MSA+ IRR groups. MSA increased serum testosterone level and testicular glutathione peroxidase (GPx) as well as decreased the percentage of sperm abnormalities. Radiation prompted inflammatory signaling in the testes through increasing phospho-janus kinase1 (p-JAK1), phospho-signal transducers and activators of transcription 3 (p-STAT3) protein expressions. This induced increment in the inflammatory markers including nuclear factor- kappa B (NF-κB) and interleukin-1beta (IL-1β) levels. Also, radiation induced elevation of nitric oxide (NO) and malondialdhyde (MDA) levels with consequent reduction in testicular reduced glutathione level (GSH) and superoxide dismutase (SOD) activity. MSA significantly counteracted the radiation effect on testicular nuclear factor erythroid-2-related factor-2 (Nrf2) and suppressor of cytokine signaling (Socs3) protein expressions. In summary, this investigation proposed that MSA preserved spermatogenesis through increasing testosterone levels and GPx activity. Additionally, it diminished testicular inflammation by increasing of Nrf2 and Socs3 levels leading to reducing of p-JAK1, p-STAT3 and NF-κB levels. Histopathological examination results of testicular tissues showed a coincidence with the biochemical analysis.
Collapse
|
18
|
Chen M, Li X, Shi Q, Zhang Z, Xu S. Hydrogen sulfide exposure triggers chicken trachea inflammatory injury through oxidative stress-mediated FOS/IL8 signaling. JOURNAL OF HAZARDOUS MATERIALS 2019; 368:243-254. [PMID: 30684762 DOI: 10.1016/j.jhazmat.2019.01.054] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 06/09/2023]
Abstract
Hydrogen sulfide (H2S) is well known to cause irritation and damage to airway following inhalation, but the mechanism by which H2S contributes to airway toxicity is unclear. In order to assess the respiratory toxicity of H2S inhalation in chicken trachea, we investigated the change of oxidative stress parameters, tracheal tissue structure and transcriptome profiles of chicken trachea exposed to H2S for 42 days. The results showed H2S exposure induced oxidative stress and inflammation in trachea. The ultrastructural analysis revealed loss of cilia and accumulation of mucus in tracheal epithelium. Differentially expressed genes (DEGs) analysis indicated 454 genes were significantly changed, including 136 genes upregulated and 318 genes downregulated. Gene ontology and KEGG analysis showed many genes involved in response to oxidative stress, inflammatory and immune response, which might contribute to H2S-induced tracheal inflammatory injury. Among those genes, N-acetyl-L-cysteine (NAC) treatment blocked the H2S-triggered expression of FOS and IL8. Silencing FOS by siRNA inhibited H2S-induced expression of IL8. Taken together, we concluded that H2S induced oxidative stress leads to tracheal inflammation through FOS/IL8 signaling, leading to excessive mucus secretion and absence of cilia. These results provide new insights for unveiling the biological effects of H2S in vivo and in vitro.
Collapse
Affiliation(s)
- Menghao Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xiaojing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Qunxiang Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
19
|
Gopalakrishna R, Gundimeda U, Zhou S, Bui H, Holmgren A. Redox regulation of protein kinase C by selenometabolites and selenoprotein thioredoxin reductase limits cancer prevention by selenium. Free Radic Biol Med 2018; 127:55-61. [PMID: 29775743 DOI: 10.1016/j.freeradbiomed.2018.05.062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/10/2018] [Accepted: 05/13/2018] [Indexed: 11/26/2022]
Abstract
The cancer-preventive mechanism of selenium should address the way low concentrations of selenometabolites react with cellular targets without being diffused from the sites of generation, the way selenium selectively kills tumor cells, and the intriguing U-shaped curve that is seen with dietary supplementation of selenium and cancer prevention. Protein kinase C (PKC), a receptor for tumor promoters, is well suited for this mechanism. Due to the catalytic redox cycle, low concentrations of methylselenol, a postulated active metabolite of selenium, react with the tumor-promoting lipid hydroperoxide bound to PKC to form methylseleninic acid (MSA), which selectively reacts with thiol residues present within the vicinity of the PKC catalytic domain to inactivate it. Given that lipid hydroperoxide levels are high in promoting cells, PKC inactivation selectively leads to death in these cells. A biphasic effect of MSA in inducing cell death was observed in certain prostate cancer cell lines; lower concentrations of MSA induced cell death, while higher concentrations failed to do so. Lower concentrations of selenium inactivate more sensitive antiapoptotic isoenzymes of PKC (ε and α), sparing less sensitive proapoptotic isoenzymes (PKCδ and PKCζ). Higher concentrations of selenium also inactivate proapoptotic isoenzymes and consequently make tumor cells resistant to apoptosis. Due to a high-affinity binding of thioredoxin to the PKC catalytic domain, this thiol oxidation is explicitly reversed by thioredoxin reductase (TXNRD), a selenoprotein. Therefore, overexpression of TXNRD in advanced tumor cells could make them resistant to selenium-induced death. Conceivably, this mechanism, at least in part, explains why selenium prevents cancer only in certain cases.
Collapse
Affiliation(s)
- Rayudu Gopalakrishna
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| | - Usha Gundimeda
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Sarah Zhou
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Helena Bui
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Arne Holmgren
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| |
Collapse
|
20
|
Bertz M, Kühn K, Koeberle SC, Müller MF, Hoelzer D, Thies K, Deubel S, Thierbach R, Kipp AP. Selenoprotein H controls cell cycle progression and proliferation of human colorectal cancer cells. Free Radic Biol Med 2018; 127:98-107. [PMID: 29330096 DOI: 10.1016/j.freeradbiomed.2018.01.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/19/2017] [Accepted: 01/07/2018] [Indexed: 12/22/2022]
Abstract
Selenoprotein H (SELENOH) is supposed to be involved in redox regulation as well as in tumorigenesis. However, its role in healthy and transformed cells of the gastrointestinal tract remains elusive. We analyzed SELENOH expression in cells depending on their selenium supply and differentiation status and found that SELENOH expression was increased in tumor tissue, in undifferentiated epithelial cells from mice and in colorectal cancer lines as compared to more differentiated ones. Knockdown studies in human colorectal cancer cells revealed that repression of SELENOH decreased cellular differentiation and increased proliferation and migration. In addition, SELENOH knockdown cells have a higher competence to form colonies or tumor xenografts. In parallel, they show a faster cell cycle transition. The high levels of SELENOH in tumors as well as in undifferentiated, proliferative cells together with its inhibitory effects on proliferation and G1/S phase transition suggest SELENOH as a key regulator for cell cycle progression and for prevention of uncontrolled proliferation. As SELENOH expression is highly dependent on the selenium status, effects of selenium supplementation on cancer initiation and progression appear to involve SELENOH.
Collapse
Affiliation(s)
- Martin Bertz
- Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam-Rehbruecke, D-14558 Nuthetal, Germany
| | - Katrin Kühn
- Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam-Rehbruecke, D-14558 Nuthetal, Germany
| | - Solveigh C Koeberle
- Department of Molecular Nutritional Physiology, Institute of Nutrition, University of Jena, Jena D-07743, Germany
| | - Mike F Müller
- Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam-Rehbruecke, D-14558 Nuthetal, Germany
| | - Doerte Hoelzer
- Department of Human Nutrition, Institute of Nutrition, University of Jena, Jena D-07743, Germany
| | - Karolin Thies
- Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam-Rehbruecke, D-14558 Nuthetal, Germany
| | - Stefanie Deubel
- Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam-Rehbruecke, D-14558 Nuthetal, Germany
| | - René Thierbach
- Department of Human Nutrition, Institute of Nutrition, University of Jena, Jena D-07743, Germany
| | - Anna P Kipp
- Department of Molecular Nutritional Physiology, Institute of Nutrition, University of Jena, Jena D-07743, Germany.
| |
Collapse
|
21
|
Peters KM, Carlson BA, Gladyshev VN, Tsuji PA. Selenoproteins in colon cancer. Free Radic Biol Med 2018; 127:14-25. [PMID: 29793041 PMCID: PMC6168369 DOI: 10.1016/j.freeradbiomed.2018.05.075] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/18/2018] [Accepted: 05/20/2018] [Indexed: 02/07/2023]
Abstract
Selenocysteine-containing proteins (selenoproteins) have been implicated in the regulation of various cell signaling pathways, many of which are linked to colorectal malignancies. In this in-depth excurse into the selenoprotein literature, we review possible roles for human selenoproteins in colorectal cancer, focusing on the typical hallmarks of cancer cells and their tumor-enabling characteristics. Human genome studies of single nucleotide polymorphisms in various genes coding for selenoproteins have revealed potential involvement of glutathione peroxidases, thioredoxin reductases, and other proteins. Cell culture studies with targeted down-regulation of selenoproteins and studies utilizing knockout/transgenic animal models have helped elucidate the potential roles of individual selenoproteins in this malignancy. Those selenoproteins, for which strong links to development or progression of colorectal cancer have been described, may be potential future targets for clinical interventions.
Collapse
Affiliation(s)
- Kristin M Peters
- Dept. of Biological Sciences, Towson University, 8000 York Rd, Towson, MD 21252, United States.
| | - Bradley A Carlson
- National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, United States.
| | - Vadim N Gladyshev
- Dept. of Medicine, Brigham & Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, United States.
| | - Petra A Tsuji
- Dept. of Biological Sciences, Towson University, 8000 York Rd, Towson, MD 21252, United States.
| |
Collapse
|
22
|
Antioxidant effects of Lycium barbarum polysaccharides on photoreceptor degeneration in the light-exposed mouse retina. Biomed Pharmacother 2018; 103:829-837. [PMID: 29684862 DOI: 10.1016/j.biopha.2018.04.104] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/12/2018] [Accepted: 04/13/2018] [Indexed: 01/05/2023] Open
Abstract
We assessed the neuroprotective effects of Lycium barbarum Polysaccharides (LBP) on photoreceptor degeneration and the mechanisms involved in oxidative stress in light-exposed mouse retinas. Mice were given a gavage of LBP (150 mg/kg or 300 mg/kg) or phosphate buffered saline (PBS) for 7 days before exposure to light (5000 lx for 24 h). We found that LBP significantly improved the electroretinography (ERG) amplitudes of the a- and b-waves that had been attenuated by light exposure. In addition, changes caused by light exposure including photoreceptor cell loss, nuclear condensation, an increased number of mitochondria vacuoles, outer membrane disc swelling and cristae fractures were distinctly ameliorated by LBP. LBP treatment also significantly prevented the generation of reactive oxygen species (ROS) compared with PBS treatment. The levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and thioredoxin reductase (TrxR1) mRNA were decreased in PBS-treated mice compared with controls but increased remarkably in LBP-treated mice. The mRNA levels of the DNA repair gene Poly (ADP-ribose) polymerase (PARP14) was increased in PBS-treated mice but decreased significantly in the LBP-treated mice. Our findings indicate that pretreatment with LBP effectively protected photoreceptor cells against light-induced retinal damage probably through the up-regulation of the antioxidative genes Nrf2 and TrxR1, the elimination of oxygen free radicals, and the subsequent reduction in the mitochondrial reaction to oxidative stress and enhancement in antioxidant capacity. In addition, the decreased level of PARP14 mRNA in LBP-treated mice also indicated a protective effect of LBP on delaying photoreceptor in the light-damaged retina.
Collapse
|
23
|
Naiki T, Naiki-Ito A, Iida K, Etani T, Kato H, Suzuki S, Yamashita Y, Kawai N, Yasui T, Takahashi S. GPX2 promotes development of bladder cancer with squamous cell differentiation through the control of apoptosis. Oncotarget 2018; 9:15847-15859. [PMID: 29662611 PMCID: PMC5882302 DOI: 10.18632/oncotarget.24627] [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: 10/27/2017] [Accepted: 02/27/2018] [Indexed: 01/10/2023] Open
Abstract
Herein, we elucidated the molecular mechanisms and therapeutic potential of glutathione peroxidase 2 (GPX2) in bladder cancer. GPX2 expression gradually increased during progression from normal to papillary or nodular hyperplasia (PNHP) and urothelial carcinoma (UC) in a rat N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced bladder carcinogenesis model. GPX2 overexpression was more marked in UC with squamous differentiation (SqD) than in pure UC. Clinical intraepithelial lesions of papillary UC and invasive UC with SqD also had strong GPX2 expression in human radical cystectomy specimens. In addition, prognostic analysis using transurethral specimens revealed that low expression level of GPX2 predicted poor prognosis in patients with pure UC. Further, UC cell lines, BC31 and RT4, cultured in vitro also overexpressed GPX2. Knock-down of GPX2 induced significant inhibition of intracellular reactive oxygen species (ROS) production, in addition to significant growth inhibition and increased apoptosis with activation of caspase 3 or 7 in both BC31 and RT4 cells. Interestingly, tumor growth of BC31 cells subcutaneously transplanted in nude mice was significantly caused the induction of apoptosis, as well as inhibition of angiogenesis and SqD by GPX2 down-regulation. Our findings demonstrated that GPX2 plays an important role in bladder carcinogenesis through the regulation of apoptosis against intracellular ROS, and may be considered as a novel biomarker or therapeutic target in bladder cancer.
Collapse
Affiliation(s)
- Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Keitaro Iida
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshiki Etani
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Shugo Suzuki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoriko Yamashita
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Noriyasu Kawai
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
24
|
|
25
|
Lennicke C, Rahn J, Wickenhauser C, Lichtenfels R, Müller AS, Wessjohann LA, Kipp AP, Seliger B. Loss of epithelium-specific GPx2 results in aberrant cell fate decisions during intestinal differentiation. Oncotarget 2017; 9:539-552. [PMID: 29416634 PMCID: PMC5787487 DOI: 10.18632/oncotarget.22640] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 10/27/2017] [Indexed: 01/07/2023] Open
Abstract
The selenoprotein glutathione peroxidase 2 (GPx2) is expressed in the epithelium of the gastrointestinal tract, where it is thought to be involved in maintaining mucosal homeostasis. To gain novel insights into the role of GPx2, proteomic profiles of colonic tissues either derived from wild type (WT) or GPx2 knockout (KO) mice, maintained under selenium (Se) deficiency or adequate Se supplementation conditions were established and analyzed. Amongst the panel of differentially expressed proteins, the calcium-activated chloride channel regulator 1 (CLCA1) was significantly down-regulated in GPx2 KO versus WT mice regardless of the given Se status. Moreover, transcript levels of the isoforms CLCA2 and CLCA3 showed a similar expression pattern. In the intestine, CLCA1 is usually restricted to mucin-producing goblet cells. However, although -SeKO mice had the highest numbers of goblet cells as confirmed by significantly enhanced mRNA expression levels of the goblet cell marker mucin-2, the observed expression pattern suggests that GPx2 KO goblet cells might be limited in synthesizing CLCA1. Furthermore, transcript levels of differentiation markers such as chromogranin-1 (Chga) for enteroendocrine cells and leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) for stem cells were also downregulated in GPx2 KO mice. Moreover, this was accompanied by a downregulation of the mRNA expression levels of the intestinal hormones glucagon-like peptide 1 (Glp1), ghrelin (Ghrl) and somatostatin (Sst). Thus, it seems that GPx2 might be important for the modulation of cell fate decisions in the murine intestinal epithelium.
Collapse
Affiliation(s)
- Claudia Lennicke
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Jette Rahn
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Rudolf Lichtenfels
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | | | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz-Institute of Plant Biochemistry, 06120 Halle (Saale), Germany
| | - Anna P Kipp
- Institute of Nutrition, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
26
|
Liu Q, Yang J, Cai J, Luan Y, Sattar H, Liu M, Xu S, Zhang Z. Analysis of the Interactions Between Thioredoxin and 20 Selenoproteins in Chicken. Biol Trace Elem Res 2017; 179:304-317. [PMID: 28251482 DOI: 10.1007/s12011-017-0961-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
Abstract
Thioredoxin (Trx) is a small molecular protein with complicated functions in a number of processes, including inflammation, apoptosis, embryogenesis, cardiovascular disease, and redox regulation. Some selenoproteins, such as glutathione peroxidase (Gpx), iodothyronine deiodinase (Dio), and thioredoxin reductase (TR), are involved in redox regulation. However, whether there are interactions between Trx and selenoproteins is still not known. In the present paper, we used a Modeller, Hex 8.0.0, and the KFC2 Server to predict the interactions between Trx and selenoproteins. We used the Modeller to predict the target protein in objective format and assess the accuracy of the results. Molecular interaction studies with Trx and selenoproteins were performed using the molecular docking tools in Hex 8.0.0. Next, we used the KFC2 Server to further test the protein binding sites. In addition to the selenoprotein physiological functions, we also explored potential relationships between Trx and selenoproteins beyond all the results we got. The results demonstrate that Trx has the potential to interact with 19 selenoproteins, including iodothyronine deiodinase 1 (Dio1), iodothyronine deiodinase 3 (Dio3), glutathione peroxidase 1 (Gpx1), glutathione peroxidase 2 (Gpx2), glutathione peroxidase 3 (Gpx3), glutathione peroxidase 4 (Gpx4), selenoprotein H (SelH), selenoprotein I (SelI), selenoprotein M (SelM), selenoprotein N (SelN), selenoprotein T (SelT), selenoprotein U (SelU), selenoprotein W (SelW), selenoprotein 15 (Sep15), methionine sulfoxide reductase B (Sepx1), selenophosphate synthetase 1 (SPS1), TR1, TR2, and TR3, among which TR1, TR2, TR3, SPS1, Sep15, SelN, SelM, SelI, Gpx2, Gpx3, Gpx4, and Dio3 exhibited intense correlations with Trx. However, additional experiments are needed to verify them.
Collapse
Affiliation(s)
- Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Jingzeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yilin Luan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Hamid Sattar
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Man Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
27
|
Abstract
Cancer survival is largely impacted by the dissemination of cancer cells from the original tumor site to secondary tissues or organs through metastasis. Targets for antimetastatic therapies have recently become a focus of research, but progress will require a better understanding of the molecular mechanisms driving metastasis. Selenoproteins play important roles in many of the cellular activities underlying metastasis including cell adhesion, matrix degradation and migration, invasion into the blood and extravasation into secondary tissues, and subsequent proliferation into metastatic tumors along with the angiogenesis required for growth. In this review the roles identified for different selenoproteins in these steps and how they may promote or inhibit metastatic cancers is discussed. These roles include selenoenzyme modulation of redox tone and detoxification of reactive oxygen species, calcium homeostasis and unfolded protein responses regulated by endoplasmic reticulum selenoproteins, and the multiple physiological responses influenced by other selenoproteins.
Collapse
Affiliation(s)
- Michael P Marciel
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Peter R Hoffmann
- John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States.
| |
Collapse
|
28
|
Jeddi F, Soozangar N, Sadeghi MR, Somi MH, Samadi N. Contradictory roles of Nrf2/Keap1 signaling pathway in cancer prevention/promotion and chemoresistance. DNA Repair (Amst) 2017; 54:13-21. [DOI: 10.1016/j.dnarep.2017.03.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 03/25/2017] [Accepted: 03/26/2017] [Indexed: 12/17/2022]
|
29
|
Rusolo F, Capone F, Pasquale R, Angiolillo A, Colonna G, Castello G, Costantini M, Costantini S. Comparison of the seleno-transcriptome expression between human non-cancerous mammary epithelial cells and two human breast cancer cell lines. Oncol Lett 2017; 13:2411-2417. [PMID: 28454412 DOI: 10.3892/ol.2017.5715] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 06/16/2016] [Indexed: 11/06/2022] Open
Abstract
Breast cancer is the second most common cause of mortality in women; therefore, the identification of novel putative markers is required to improve its diagnosis and prognosis. Selenium is known to protect mammary epithelial cells from oxidative DNA damage, and to inhibit the initiation phase of carcinogenesis by stimulating DNA repair and apoptosis regulation. Consequently, the present study has focused attention on the selenoprotein family and their involvement in breast cancer. The present study performed a global analysis of the seleno-transcriptome expression in human breast cancer MCF-7 and MDA-MB231 cell lines compared with healthy breast MCF-10A cells using reverse transcription-quantitative polymerase chain reaction. The present data revealed the presence of differently expressed genes in MCF-7 and MDA-MB231 cells compared with MCF-10A cells: Four downregulated [glutathione peroxidase (GPX)1, GPX4, GPX5 and GPX7] and three upregulated (deiodinase iodothyronine, type II, GPX2 and GPX3) genes. Additionally, interactomic investigation were performed by the present study to evaluate the association between the downregulated and upregulated genes, and to identify putative HUB nodes, which represent the centers of association between the genes that are capable of direct control over the gene networks. Network analysis revealed that all differentially regulated genes, with the exception of selenoprotein T, are implicated in the same network that presents three HUB nodes interconnected to the selenoprotein mRNAs, including TP53, estrogen receptor 1 and catenin-β1 (CTNNB1). Overall, these data demonstrated for the first time, a profile of seleno-mRNAs specific for human breast cells, indicating that these genes alter their expression on the basis of the ER-positivity or negativity of breast cancer cells.
Collapse
Affiliation(s)
- Fabiola Rusolo
- Oncology Research Center of Mercogliano, National Cancer Institute 'G. Pascale Foundation', IRCCS, I-80131 Naples, Italy
| | - Francesca Capone
- Oncology Research Center of Mercogliano, National Cancer Institute 'G. Pascale Foundation', IRCCS, I-80131 Naples, Italy
| | - Raffaella Pasquale
- Oncology Research Center of Mercogliano, National Cancer Institute 'G. Pascale Foundation', IRCCS, I-80131 Naples, Italy
| | - Antonella Angiolillo
- Department of Medicine and Health Sciences, University of Molise, I-86100 Campobasso, Italy
| | - Giovanni Colonna
- Medical Informatics Service, University Hospital, Second University of Naples, I-80131 Naples, Italy
| | - Giuseppe Castello
- Oncology Research Center of Mercogliano, National Cancer Institute 'G. Pascale Foundation', IRCCS, I-80131 Naples, Italy
| | - Maria Costantini
- Department of Biology and Evolution of Marine Organisms, Anton Dohrn Zoological Station, I-80121 Naples, Italy
| | - Susan Costantini
- Oncology Research Center of Mercogliano, National Cancer Institute 'G. Pascale Foundation', IRCCS, I-80131 Naples, Italy
| |
Collapse
|
30
|
Nagakannan P, Iqbal MA, Yeung A, Thliveris JA, Rastegar M, Ghavami S, Eftekharpour E. Perturbation of redox balance after thioredoxin reductase deficiency interrupts autophagy-lysosomal degradation pathway and enhances cell death in nutritionally stressed SH-SY5Y cells. Free Radic Biol Med 2016; 101:53-70. [PMID: 27693380 DOI: 10.1016/j.freeradbiomed.2016.09.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/24/2016] [Accepted: 09/25/2016] [Indexed: 01/25/2023]
Abstract
Oxidative damage and aggregation of cellular proteins is a hallmark of neuronal cell death after neurotrauma and chronic neurodegenerative conditions. Autophagy and ubiquitin protease system are involved in degradation of protein aggregates, and interruption of their function is linked to apoptotic cell death in these diseases. Oxidative modification of cysteine groups in key molecular proteins has been linked to modification of cellular systems and cell death in these conditions. Glutathione and thioredoxin systems provide reducing protons that can effectively reverse protein modifications and promote cell survival. The central role of Thioredoxin in inhibition of apoptosis is well identified. Additionally, its involvement in initiation of autophagy has been suggested recently. We therefore aimed to investigate the involvement of Thioredoxin system in autophagy-apoptosis processes. A model of serum deprivation in SH-SY5Y was used that is associated with autophagy and apoptosis. Using pharmacological and RNA-editing technology we show that Thioredoxin reductase deficiency in this model enhances oxidative stress and interrupts the early protective autophagy and promotes apoptosis. This was associated with decreased protein-degradation in lysosomes due to altered lysosomal acidification and accumulation of autophagosomes as well as impairment in proteasome pathway. We further confirmed that the extent of oxidative stress is a determining factor in autophagy- apoptosis interplay, as upregulation of cellular reducing capacity by N-acetylcysteine prevented impairment in autophagy and proteasome systems thus promoted cell viability. Our study provides evidence that excessive oxidative stress inhibits protein degradation systems and affects the final stages of autophagy by inhibiting autolysosome maturation: a novel mechanistic link between protein aggregation and conversion of autophagy to apoptosis that can be applicable to neurodegenerative diseases.
Collapse
Affiliation(s)
- Pandian Nagakannan
- Spinal Cord Research Center, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Mohamed Ariff Iqbal
- Spinal Cord Research Center, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Albert Yeung
- Spinal Cord Research Center, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - James A Thliveris
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Mojgan Rastegar
- Department of Biochemistry & Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Eftekhar Eftekharpour
- Spinal Cord Research Center, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
31
|
Carlson BA, Tobe R, Yefremova E, Tsuji PA, Hoffmann VJ, Schweizer U, Gladyshev VN, Hatfield DL, Conrad M. Glutathione peroxidase 4 and vitamin E cooperatively prevent hepatocellular degeneration. Redox Biol 2016; 9:22-31. [PMID: 27262435 PMCID: PMC4900515 DOI: 10.1016/j.redox.2016.05.003] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 02/07/2023] Open
Abstract
The selenoenzyme glutathione peroxidase 4 (Gpx4) is an essential mammalian glutathione peroxidase, which protects cells against detrimental lipid peroxidation and governs a novel form of regulated necrotic cell death, called ferroptosis. To study the relevance of Gpx4 and of another vitally important selenoprotein, cytosolic thioredoxin reductase (Txnrd1), for liver function, mice with conditional deletion of Gpx4 in hepatocytes were studied, along with those lacking Txnrd1 and selenocysteine (Sec) tRNA (Trsp) in hepatocytes. Unlike Txnrd1- and Trsp-deficient mice, Gpx4−/− mice died shortly after birth and presented extensive hepatocyte degeneration. Similar to Txnrd1-deficient livers, Gpx4−/− livers manifested upregulation of nuclear factor (erythroid-derived)-like 2 (Nrf2) response genes. Remarkably, Gpx4−/− pups born from mothers fed a vitamin E-enriched diet survived, yet this protection was reversible as subsequent vitamin E deprivation caused death of Gpx4-deficient mice ~4 weeks thereafter. Abrogation of selenoprotein expression in Gpx4−/− mice did not result in viable mice, indicating that the combined deficiency aggravated the loss of Gpx4 in liver. By contrast, combined Trsp/Txnrd1-deficient mice were born, but had significantly shorter lifespans than either single knockout, suggesting that Txnrd1 plays an important role in supporting liver function of mice lacking Trsp. In sum our study demonstrates that the ferroptosis regulator Gpx4 is critical for hepatocyte survival and proper liver function, and that vitamin E can compensate for its loss by protecting cells against deleterious lipid peroxidation. Conditional Gpx4 loss causes hepatocellular degeneration and early death of mice. Dietary vitamin E supplementation rescues death of liver-specific Gpx4 null mice. Nutritional vitamin E content in diet may severely impact experimental outcome.
Collapse
Affiliation(s)
- Bradley A Carlson
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ryuta Tobe
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elena Yefremova
- Helmholtz Zentrum München, Institute for Developmental Genetics, Neuherberg, Germany
| | - Petra A Tsuji
- Department of Biological Sciences, Towson University, Towson, MD, USA
| | - Victoria J Hoffmann
- Office of the Director Diagnostic and Research Services Branch, National Institutes of Health, Bethesda, MD, USA
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dolph L Hatfield
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute for Developmental Genetics, Neuherberg, Germany.
| |
Collapse
|
32
|
Gouge J, Satia K, Guthertz N, Widya M, Thompson AJ, Cousin P, Dergai O, Hernandez N, Vannini A. Redox Signaling by the RNA Polymerase III TFIIB-Related Factor Brf2. Cell 2016; 163:1375-87. [PMID: 26638071 PMCID: PMC4671959 DOI: 10.1016/j.cell.2015.11.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/14/2015] [Accepted: 10/23/2015] [Indexed: 01/06/2023]
Abstract
TFIIB-related factor 2 (Brf2) is a member of the family of TFIIB-like core transcription factors. Brf2 recruits RNA polymerase (Pol) III to type III gene-external promoters, including the U6 spliceosomal RNA and selenocysteine tRNA genes. Found only in vertebrates, Brf2 has been linked to tumorigenesis but the underlying mechanisms remain elusive. We have solved crystal structures of a human Brf2-TBP complex bound to natural promoters, obtaining a detailed view of the molecular interactions occurring at Brf2-dependent Pol III promoters and highlighting the general structural and functional conservation of human Pol II and Pol III pre-initiation complexes. Surprisingly, our structural and functional studies unravel a Brf2 redox-sensing module capable of specifically regulating Pol III transcriptional output in living cells. Furthermore, we establish Brf2 as a central redox-sensing transcription factor involved in the oxidative stress pathway and provide a mechanistic model for Brf2 genetic activation in lung and breast cancer. Architectural conservation of TFIIB and TFIIB-related factors Brf2 is a redox-sensing RNA polymerase III core transcription factor Brf2 regulates cellular responses to oxidative stress Brf2 amplification enables cancer cells to evade oxidative stress-induced apoptosis
Collapse
Affiliation(s)
- Jerome Gouge
- Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Karishma Satia
- Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Nicolas Guthertz
- Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Marcella Widya
- Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Andrew James Thompson
- Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Pascal Cousin
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Oleksandr Dergai
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Nouria Hernandez
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Alessandro Vannini
- Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, UK.
| |
Collapse
|
33
|
Abu N, Yeap SK, Pauzi AZM, Akhtar MN, Zamberi NR, Ismail J, Zareen S, Alitheen NB. Dual Regulation of Cell Death and Cell Survival upon Induction of Cellular Stress by Isopimara-7,15-Dien-19-Oic Acid in Cervical Cancer, HeLa Cells In vitro. Front Pharmacol 2016; 7:89. [PMID: 27065873 PMCID: PMC4814465 DOI: 10.3389/fphar.2016.00089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 03/18/2016] [Indexed: 11/14/2022] Open
Abstract
The Fritillaria imperialis is an ornamental flower that can be found in various parts of the world including Iraq, Afghanistan, Pakistan, and the Himalayas. The use of this plant as traditional remedy is widely known. This study aims to unveil the anti-cancer potentials of Isopimara-7,15-Dien-19-Oic Acid, extracted from the bulbs of F. imperialis in cervical cancer cell line, HeLa cells. Flow cytometry analysis of cell death, gene expression analysis via cDNA microarray and protein array were performed. Based on the results, Isopimara-7,15-Dien-19-Oic acid simultaneously induced cell death and promoted cell survival. The execution of apoptosis was apparent based on the flow cytometry results and regulation of both pro and anti-apoptotic genes. Additionally, the regulation of anti-oxidant genes were up-regulated especially thioredoxin, glutathione and superoxide dismutase- related genes. Moreover, the treatment also induced the activation of pro-survival heat shock proteins. Collectively, Isopimara-7,15-Dien-19-Oic Acid managed to induce cellular stress in HeLa cells and activate several anti- and pro survival pathways.
Collapse
Affiliation(s)
- Nadiah Abu
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia Serdang, Malaysia
| | - Swee K Yeap
- Laboratory of Immunotherapeutics and Vaccine (LIVES), Institute of Bioscience, Universiti Putra Malaysia Serdang, Malaysia
| | - Ahmad Z Mat Pauzi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia Serdang, Malaysia
| | - M Nadeem Akhtar
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Kuantan, Malaysia
| | - Nur R Zamberi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia Serdang, Malaysia
| | - Jamil Ismail
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Kuantan, Malaysia
| | - Seema Zareen
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Kuantan, Malaysia
| | - Noorjahan B Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia Serdang, Malaysia
| |
Collapse
|
34
|
Choi EO, Jeong JW, Park C, Hong SH, Kim GY, Hwang HJ, Cho EJ, Choi YH. Baicalein protects C6 glial cells against hydrogen peroxide-induced oxidative stress and apoptosis through regulation of the Nrf2 signaling pathway. Int J Mol Med 2016; 37:798-806. [PMID: 26796879 DOI: 10.3892/ijmm.2016.2460] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/11/2016] [Indexed: 11/06/2022] Open
Abstract
Baicalein, a flavonoid originally obtained from the roots of Scutellaria baicalensis Georgi, has been reported to possess various biological properties. Although several studies have demonstrated the anti-oxidative activity of baicalein, its neuroprotective mechanisms have not been clearly established. The present study aimed to detect the effects of baicalein against hydrogen peroxide (H2O2)-induced neuronal damage in C6 glial cells and to investigate the molecular mechanisms involved in this process. The results demonstrated that baicalein effectively inhibited H2O2-induced growth and reactive oxygen species (ROS) generation. We noted that Baicalein also attenuated the H2O2‑induced formation of comet tail, phosphorylation of p-γH2A.X, loss of mitochondrial membrane potential (MMP or ΔΨm), and changes to apoptosis‑related protein expression, which suggests that it can prevent H2O2‑induced cellular DNA damage and apoptotic cell death. Furthermore, treatment with baicalein effectively induced the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2) as well as heme oxygenase-1 (HO-1) and thioredoxin reductase 1 (TrxR1) in a concentration and time-dependent manner. Moreover, the protective effects of baicalein against H2O2‑induced DNA damage and apoptosis were abolished by zinc protoporphyrin (ZnPP) IX, a HO-1 inhibitor, and auranofin, a TrxR inhibitor. In addition, we noted that the cytoprotective effects of baicalein were attenuated by transient transfection with Nrf2-specific small interfering RNA (siRNA). The findings of our present study suggest that baicalein enhances cellular antioxidant defense capacity through the inhibition of ROS generation and the activation of the Nrf2 signaling pathway, thus protecting C6 cells from H2O2-induced neuronal damage.
Collapse
Affiliation(s)
- Eun-Ok Choi
- Department of Food and Nutrition, College of Human Ecology, Pusan National University, Busan 609-735, Republic of Korea
| | - Jin-Woo Jeong
- Anti-Aging Research Center and Blue-Bio Industry RIC, College of Natural Sciences and Human Ecology, Dongeui University, Busan 614-714, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences and Human Ecology, Dongeui University, Busan 614-714, Republic of Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
| | - Hye-Jin Hwang
- Anti-Aging Research Center and Blue-Bio Industry RIC, College of Natural Sciences and Human Ecology, Dongeui University, Busan 614-714, Republic of Korea
| | - Eun-Ju Cho
- Department of Food and Nutrition, College of Human Ecology, Pusan National University, Busan 609-735, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center and Blue-Bio Industry RIC, College of Natural Sciences and Human Ecology, Dongeui University, Busan 614-714, Republic of Korea
| |
Collapse
|
35
|
Cebula M, Schmidt EE, Arnér ESJ. TrxR1 as a potent regulator of the Nrf2-Keap1 response system. Antioxid Redox Signal 2015; 23:823-53. [PMID: 26058897 PMCID: PMC4589110 DOI: 10.1089/ars.2015.6378] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE All cells must maintain a balance between oxidants and reductants, while allowing for fluctuations in redox states triggered by signaling, altered metabolic flow, or extracellular stimuli. Furthermore, they must be able to rapidly sense and react to various challenges that would disrupt the redox homeostasis. RECENT ADVANCES Many studies have identified Keap1 as a key sensor for oxidative or electrophilic stress, with modification of Keap1 by oxidation or electrophiles triggering Nrf2-mediated transcriptional induction of enzymes supporting reductive and detoxification pathways. However, additional mechanisms for Nrf2 regulation are likely to exist upstream of, or in parallel with, Keap1. CRITICAL ISSUES Here, we propose that the mammalian selenoprotein thioredoxin reductase 1 (TrxR1) is a potent regulator of Nrf2. A high chemical reactivity of TrxR1 and its vital role for the thioredoxin (Trx) system distinguishes TrxR1 as a prime target for electrophilic challenges. Chemical modification of the selenocysteine (Sec) in TrxR1 by electrophiles leads to rapid inhibition of thioredoxin disulfide reductase activity, often combined with induction of NADPH oxidase activity of the derivatized enzyme, thereby affecting many downstream redox pathways. The notion of TrxR1 as a regulator of Nrf2 is supported by many publications on effects in human cells of selenium deficiency, oxidative stress or electrophile exposure, as well as the phenotypes of genetic mouse models. FUTURE DIRECTIONS Investigation of the role of TrxR1 as a regulator of Nrf2 activation will facilitate further studies of redox control in diverse cells and tissues of mammals, and possibly also in animals of other classes.
Collapse
Affiliation(s)
- Marcus Cebula
- 1 Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
| | - Edward E Schmidt
- 2 Microbiology and Immunology, Montana State University , Bozeman, Montana
| | - Elias S J Arnér
- 1 Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
| |
Collapse
|
36
|
Hiller F, Besselt K, Deubel S, Brigelius-Flohé R, Kipp AP. GPx2 Induction Is Mediated Through STAT Transcription Factors During Acute Colitis. Inflamm Bowel Dis 2015; 21:2078-89. [PMID: 26115075 DOI: 10.1097/mib.0000000000000464] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The selenoprotein glutathione peroxidase 2 (GPx2) is highly expressed in the gastrointestinal epithelium. During inflammatory bowel disease and colorectal cancer, GPx2 expression is enhanced. METHODS We analyzed GPx2 expression and transcriptional regulation during the different phases of dextran sulfate sodium (DSS)-induced colitis in mice and in cytokine-treated colorectal cancer cells. RESULTS In the colon of DSS-treated mice, GPx2 was upregulated during the acute and recovery phase. In the latter, it was specifically localized in regenerating ki67-positive crypts next to ulcerations. In cultured cells, endogenous GPx2 expression and GPx2 promoter activity were enhanced by the anti-inflammatory mediators 15-deoxy-Δ(12,14)-prostaglandin J2 (15d-PGJ2) and interleukin-22 (IL-22), while it was unaffected by classical proinflammatory cytokines like IL-1β. Induction of GPx2 expression by 15d-PGJ2 was mediated through Nrf2. In contrast, in DSS-treated Nrf2-KO mice GPx2 expression remained upregulated during recovery, which appeared to be independent of Nrf2. IL-22 activates transcription factors of the signal transducers and activators of transcription (STAT) family. Therefore, we analyzed the GPx2 promoter for putative STAT-responsive elements and identified 4 of them. Point mutation of the binding element next to the transcription start completely abolished promoter activation after IL-22 treatment and after cotransfection of STAT expression plasmids. To show in vivo relevance of the obtained results, we performed immunohistochemistry for phospho-STAT3 and GPx2. Especially during acute colitis, GPx2 and nuclear STAT3 colocalized in inflamed areas. CONCLUSIONS GPx2 is a novel target of STAT transcription factors. The upregulation of GPx2 by IL-22 indicates that GPx2 might be important for the resolution of inflammation.
Collapse
Affiliation(s)
- Franziska Hiller
- Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam, Germany
| | | | | | | | | |
Collapse
|
37
|
Synergy between sulforaphane and selenium in protection against oxidative damage in colonic CCD841 cells. Nutr Res 2015; 35:610-7. [DOI: 10.1016/j.nutres.2015.05.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/18/2015] [Accepted: 05/27/2015] [Indexed: 01/15/2023]
|
38
|
Tsuji PA, Carlson BA, Yoo MH, Naranjo-Suarez S, Xu XM, He Y, Asaki E, Seifried HE, Reinhold WC, Davis CD, Gladyshev VN, Hatfield DL. The 15kDa selenoprotein and thioredoxin reductase 1 promote colon cancer by different pathways. PLoS One 2015; 10:e0124487. [PMID: 25886253 PMCID: PMC4401539 DOI: 10.1371/journal.pone.0124487] [Citation(s) in RCA: 33] [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: 12/30/2014] [Accepted: 03/03/2015] [Indexed: 11/29/2022] Open
Abstract
Selenoproteins mediate much of the cancer-preventive properties of the essential nutrient selenium, but some of these proteins have been shown to also have cancer-promoting effects. We examined the contributions of the 15kDa selenoprotein (Sep15) and thioredoxin reductase 1 (TR1) to cancer development. Targeted down-regulation of either gene inhibited anchorage-dependent and anchorage-independent growth and formation of experimental metastases of mouse colon carcinoma CT26 cells. Surprisingly, combined deficiency of Sep15 and TR1 reversed the anti-cancer effects observed with down-regulation of each single gene. We found that inflammation-related genes regulated by Stat-1, especially interferon-γ-regulated guanylate-binding proteins, were highly elevated in Sep15-deficient, but not in TR1-deficient cells. Interestingly, components of the Wnt/β-catenin signaling pathway were up-regulated in cells lacking both TR1 and Sep15. These results suggest that Sep15 and TR1 participate in interfering regulatory pathways in colon cancer cells. Considering the variable expression levels of Sep15 and TR1 found within the human population, our results provide insights into new roles of selenoproteins in cancer.
Collapse
Affiliation(s)
- Petra A. Tsuji
- Department of Biological Sciences, Towson University, Towson, Maryland, United States of America
| | - Bradley A. Carlson
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Min-Hyuk Yoo
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Salvador Naranjo-Suarez
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xue-Ming Xu
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yiwen He
- Center for Information Technology, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Esther Asaki
- Center for Information Technology, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Harold E. Seifried
- Nutritional Science Research Group, National Institutes of Health, Rockville, Maryland, United States of America
| | - William C. Reinhold
- Genomics & Informatics Group, Laboratory of Molecular Pharmacology, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cindy D. Davis
- Office of Dietary Supplements, National Institutes of Health, Rockville, Maryland, United States of America
| | - Vadim N. Gladyshev
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dolph L. Hatfield
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
39
|
Reszka E, Wieczorek E, Jablonska E, Janasik B, Fendler W, Wasowicz W. Association between plasma selenium level and NRF2 target genes expression in humans. J Trace Elem Med Biol 2015; 30:102-6. [PMID: 25524402 DOI: 10.1016/j.jtemb.2014.11.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/07/2014] [Accepted: 11/17/2014] [Indexed: 11/21/2022]
Abstract
Animal studies in rodent and in vitro studies indicate compensatory role of nuclear factor (erythroid-derived 2)-like (Nrf2) and Nrf2-regulated antioxidant and phase II biotransformation enzymes for the dietary selenium (Se) deficiency or for the loss of selenoproteins. To explore associations between plasma Se level and NRF2-regulated cytoprotective genes expression, an observational study was conducted in a population of 96 healthy non-smoking men living in Central Poland aged 18-83 years with relatively low plasma Se level. NRF2, KEAP2, CAT, EPHX1, GCLC, GCLM, GPX2, GSR, GSTA1, GSTM1, GSTP1, GSTT1, HMOX1, NQO1, PRDX1, SOD1, SOD2, TXNRD1 transcript levels in peripheral blood leukocytes and polymorphism of NRF2-617C/A (rs6721961) in blood genomic DNA were determined by means of quantitative real-time PCR. Mean plasma Se level was found to be 51.10±15.25μg/L (range 23.86-96.18μg/L). NRF2 mRNA level was positively correlated with expression of investigated NRF2-target genes. The multivariate linear regression adjusting for selenium status showed that plasma Se level was significantly inversely associated only with expression of GSTP1 (β-coef.=-0.270, p=0.009), PRDXR1 (β-coef.=-0.245, p=0.017) and SOD2 with an inverse trend toward significance (β-coef.=-0.186, p=0.074), but without an effect of NRF2 gene variants. NRF2 expression was inversely associated with age (r=-0.23, p=0.03) and body mass index (r=-0.29, p<0.001). The findings may suggest a possible link between plasma Se level and cytoprotective response at gene level in humans.
Collapse
Affiliation(s)
- Edyta Reszka
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, 8 Teresy Street, 91-348 Lodz, Poland.
| | - Edyta Wieczorek
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, 8 Teresy Street, 91-348 Lodz, Poland
| | - Ewa Jablonska
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, 8 Teresy Street, 91-348 Lodz, Poland
| | - Beata Janasik
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, 8 Teresy Street, 91-348 Lodz, Poland
| | - Wojciech Fendler
- Department of Pediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, 36/50 Sporna Street, 91-738 Lodz, Poland
| | - Wojciech Wasowicz
- Department of Toxicology and Carcinogenesis, Nofer Institute of Occupational Medicine, 8 Teresy Street, 91-348 Lodz, Poland
| |
Collapse
|
40
|
Bao Y, Wang W, Zhou Z, Sun C. Benefits and risks of the hormetic effects of dietary isothiocyanates on cancer prevention. PLoS One 2014; 9:e114764. [PMID: 25532034 PMCID: PMC4273949 DOI: 10.1371/journal.pone.0114764] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 11/13/2014] [Indexed: 01/16/2023] Open
Abstract
The isothiocyanate (ITC) sulforaphane (SFN) was shown at low levels (1–5 µM) to promote cell proliferation to 120–143% of the controls in a number of human cell lines, whilst at high levels (10–40 µM) it inhibited such cell proliferation. Similar dose responses were observed for cell migration, i.e. SFN at 2.5 µM increased cell migration in bladder cancer T24 cells to 128% whilst high levels inhibited cell migration. This hormetic action was also found in an angiogenesis assay where SFN at 2.5 µM promoted endothelial tube formation (118% of the control), whereas at 10–20 µM it caused significant inhibition. The precise mechanism by which SFN influences promotion of cell growth and migration is not known, but probably involves activation of autophagy since an autophagy inhibitor, 3-methyladenine, abolished the effect of SFN on cell migration. Moreover, low doses of SFN offered a protective effect against free-radical mediated cell death, an effect that was enhanced by co-treatment with selenium. These results suggest that SFN may either prevent or promote tumour cell growth depending on the dose and the nature of the target cells. In normal cells, the promotion of cell growth may be of benefit, but in transformed or cancer cells it may be an undesirable risk factor. In summary, ITCs have a biphasic effect on cell growth and migration. The benefits and risks of ITCs are not only determined by the doses, but are affected by interactions with Se and the measured endpoint.
Collapse
Affiliation(s)
- Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom
- * E-mail:
| | - Wei Wang
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom
| | - Zhigang Zhou
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, United Kingdom
- Department of Cardiovascular Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, P. R. China
| | - Changhao Sun
- School of Public Health, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| |
Collapse
|
41
|
Stefanson AL, Bakovic M. Dietary regulation of Keap1/Nrf2/ARE pathway: focus on plant-derived compounds and trace minerals. Nutrients 2014; 6:3777-801. [PMID: 25244368 PMCID: PMC4179188 DOI: 10.3390/nu6093777] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 08/13/2014] [Accepted: 08/14/2014] [Indexed: 01/10/2023] Open
Abstract
It has become increasingly evident that chronic inflammation underpins the development of many chronic diseases including cancer, cardiovascular disease and type 2 diabetes. Oxidative stress is inherently a biochemical dysregulation of the redox status of the intracellular environment, which under homeostatic conditions is a reducing environment, whereas inflammation is the biological response to oxidative stress in that the cell initiates the production of proteins, enzymes, and other compounds to restore homeostasis. At the center of the day-to-day biological response to oxidative stress is the Keap1/Nrf2/ARE pathway, which regulates the transcription of many antioxidant genes that preserve cellular homeostasis and detoxification genes that process and eliminate carcinogens and toxins before they can cause damage. The Keap1/Nrf2/ARE pathway plays a major role in health resilience and can be made more robust and responsive by certain dietary factors. Transient activation of Nrf2 by dietary electrophilic phytochemicals can upregulate antioxidant and chemopreventive enzymes in the absence of actual oxidative stress inducers. Priming the Keap1/Nrf2/ARE pathway by upregulating these enzymes prior to oxidative stress or xenobiotic encounter increases cellular fitness to respond more robustly to oxidative assaults without activating more intense inflammatory NFκB-mediated responses.
Collapse
Affiliation(s)
- Amanda L Stefanson
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road E, Guelph, Ontario, Canada N1G 2W1.
| | - Marica Bakovic
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road E, Guelph, Ontario, Canada N1G 2W1.
| |
Collapse
|
42
|
Labunskyy VM, Hatfield DL, Gladyshev VN. Selenoproteins: molecular pathways and physiological roles. Physiol Rev 2014; 94:739-77. [PMID: 24987004 DOI: 10.1152/physrev.00039.2013] [Citation(s) in RCA: 850] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Selenium is an essential micronutrient with important functions in human health and relevance to several pathophysiological conditions. The biological effects of selenium are largely mediated by selenium-containing proteins (selenoproteins) that are present in all three domains of life. Although selenoproteins represent diverse molecular pathways and biological functions, all these proteins contain at least one selenocysteine (Sec), a selenium-containing amino acid, and most serve oxidoreductase functions. Sec is cotranslationally inserted into nascent polypeptide chains in response to the UGA codon, whose normal function is to terminate translation. To decode UGA as Sec, organisms evolved the Sec insertion machinery that allows incorporation of this amino acid at specific UGA codons in a process requiring a cis-acting Sec insertion sequence (SECIS) element. Although the basic mechanisms of Sec synthesis and insertion into proteins in both prokaryotes and eukaryotes have been studied in great detail, the identity and functions of many selenoproteins remain largely unknown. In the last decade, there has been significant progress in characterizing selenoproteins and selenoproteomes and understanding their physiological functions. We discuss current knowledge about how these unique proteins perform their functions at the molecular level and highlight new insights into the roles that selenoproteins play in human health.
Collapse
Affiliation(s)
- Vyacheslav M Labunskyy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; and Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dolph L Hatfield
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; and Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; and Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
43
|
Mine N, Yamamoto S, Kufe DW, Von Hoff DD, Kawabe T. Activation of Nrf2 pathways correlates with resistance of NSCLC cell lines to CBP501 in vitro. Mol Cancer Ther 2014; 13:2215-25. [PMID: 25053821 DOI: 10.1158/1535-7163.mct-13-0808] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CBP501 is an anticancer drug candidate that was investigated in two randomized phase II clinical trials for patients with nonsquamous non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). CBP501 has been shown to have two mechanisms of action, namely calmodulin modulation and G2 checkpoint abrogation. Here, we searched for a biomarker to predict sensitivity to CBP501. Twenty-eight NSCLC cell lines were classified into two subgroups, CBP501-sensitive and -insensitive, by quantitatively analyzing the cis-diamminedichloro-platinum (II) (CDDP)-enhancing activity of CBP501 through treatments with short-term (1 hour) coexposure to CDDP and CBP501 or to either alone. Microarray analysis was performed on these cell lines to identify gene expression patterns that correlated with CBP501 sensitivity. We found that multiple nuclear factor erythroid-2-related factor 2 (Nrf2) target genes showed high expression in CBP501-insensitive cell lines. Western blot and immunocytochemical analysis for Nrf2 in NSCLC cell lines also indicated higher protein level in CBP501-insensitive cell lines. Moreover, CBP501 sensitivity is modulated by silencing or sulforaphane-induced overexpression of Nrf2. These results indicate that Nrf2 transcription factor is a potential candidate as a biomarker for resistance to CBP501. This study might help to identify those subpopulations of patients who would respond well to the CBP501 and CDDP combination treatment of NSCLC.
Collapse
Affiliation(s)
| | | | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
44
|
Zhu J, Wang H, Fan Y, Lin Y, Zhang L, Ji X, Zhou M. Targeting the NF-E2-related factor 2 pathway: a novel strategy for glioblastoma (review). Oncol Rep 2014; 32:443-50. [PMID: 24926991 DOI: 10.3892/or.2014.3259] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 05/26/2014] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma is the most common and malignant subtype among all brain tumors. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an essential component of cellular defense against a variety of endogenous and exogenous stresses. A marked increase in research over the past few decades focusing on Nrf2 and its role in regulating glioblastoma has revealed the potential value of Nrf2 in the treatment of glioblastoma. In the present review, we discuss a novel framework of Nrf2 in the regulation of glioblastoma and the mechanisms regarding the downregulation of Nrf2 in treating glioblastoma. The candidate mechanisms include direct and indirect means. Direct mechanisms target tumor molecular pathways in order to overcome resistance to chemotherapy and radiotherapy, to inhibit proliferation, to block invasion and migration, to induce apoptosis, to promote differentiation, to enhance autophagy and to target glioblastoma stem cells. Indirect mechanisms target the reaction between glioblastoma cells and the surrounding microenvironment. Overall, the value of the Nrf2 pathway in glioblastoma provides a promising opportunity for new approaches by which to treat glioblastoma.
Collapse
Affiliation(s)
- Jianhong Zhu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Youwu Fan
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yixing Lin
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Li Zhang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xiangjun Ji
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Mengliang Zhou
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
45
|
Barroso M, Florindo C, Kalwa H, Silva Z, Turanov AA, Carlson BA, de Almeida IT, Blom HJ, Gladyshev VN, Hatfield DL, Michel T, Castro R, Loscalzo J, Handy DE. Inhibition of cellular methyltransferases promotes endothelial cell activation by suppressing glutathione peroxidase 1 protein expression. J Biol Chem 2014; 289:15350-62. [PMID: 24719327 PMCID: PMC4140892 DOI: 10.1074/jbc.m114.549782] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
S-adenosylhomocysteine (SAH) is a negative regulator of most methyltransferases and the precursor for the cardiovascular risk factor homocysteine. We have previously identified a link between the homocysteine-induced suppression of the selenoprotein glutathione peroxidase 1 (GPx-1) and endothelial dysfunction. Here we demonstrate a specific mechanism by which hypomethylation, promoted by the accumulation of the homocysteine precursor SAH, suppresses GPx-1 expression and leads to inflammatory activation of endothelial cells. The expression of GPx-1 and a subset of other selenoproteins is dependent on the methylation of the tRNA(Sec) to the Um34 form. The formation of methylated tRNA(Sec) facilitates translational incorporation of selenocysteine at a UGA codon. Our findings demonstrate that SAH accumulation in endothelial cells suppresses the expression of GPx-1 to promote oxidative stress. Hypomethylation stress, caused by SAH accumulation, inhibits the formation of the methylated isoform of the tRNA(Sec) and reduces GPx-1 expression. In contrast, under these conditions, the expression and activity of thioredoxin reductase 1, another selenoprotein, is increased. Furthermore, SAH-induced oxidative stress creates a proinflammatory activation of endothelial cells characterized by up-regulation of adhesion molecules and an augmented capacity to bind leukocytes. Taken together, these data suggest that SAH accumulation in endothelial cells can induce tRNA(Sec) hypomethylation, which alters the expression of selenoproteins such as GPx-1 to contribute to a proatherogenic endothelial phenotype.
Collapse
Affiliation(s)
- Madalena Barroso
- From the Cardiovascular and ,the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and
| | - Cristina Florindo
- the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and
| | | | - Zélia Silva
- the Chronic Diseases Research Center, Departamento de Imunologia, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1099-085 Lisbon, Portugal
| | - Anton A. Turanov
- Genetics Divisions, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Bradley A. Carlson
- the Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Isabel Tavares de Almeida
- the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and ,Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-004 Lisbon, Portugal
| | - Henk J. Blom
- the Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital, 79106 Freiburg, Germany
| | - Vadim N. Gladyshev
- Genetics Divisions, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Dolph L. Hatfield
- the Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, and
| | | | - Rita Castro
- the Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL) and ,Department of Biochemistry and Human Biology, Faculty of Pharmacy, University of Lisbon, 1649-004 Lisbon, Portugal
| | | | - Diane E. Handy
- From the Cardiovascular and , To whom correspondence should be addressed: Cardiovascular Div., Dept. of Medicine, Brigham and Women's Hospital and Harvard Medical School, 77 Ave. Louis Pasteur, Boston, MA, 02115. Tel.: 617-525-4845; Fax: 617-525-4830; E-mail:
| |
Collapse
|
46
|
Touat-Hamici Z, Legrain Y, Bulteau AL, Chavatte L. Selective up-regulation of human selenoproteins in response to oxidative stress. J Biol Chem 2014; 289:14750-61. [PMID: 24706762 DOI: 10.1074/jbc.m114.551994] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Selenocysteine is inserted into selenoproteins via the translational recoding of a UGA codon, normally used as a stop signal. This process depends on the nature of the selenocysteine insertion sequence element located in the 3' UTR of selenoprotein mRNAs, selenium bioavailability, and, possibly, exogenous stimuli. To further understand the function and regulation of selenoproteins in antioxidant defense and redox homeostasis, we investigated how oxidative stress influences selenoprotein expression as a function of different selenium concentrations. We found that selenium supplementation of the culture media, which resulted in a hierarchical up-regulation of selenoproteins, protected HEK293 cells from reactive oxygen species formation. Furthermore, in response to oxidative stress, we identified a selective up-regulation of several selenoproteins involved in antioxidant defense (Gpx1, Gpx4, TR1, SelS, SelK, and Sps2). Interestingly, the response was more efficient when selenium was limiting. Although a modest change in mRNA levels was noted, we identified a novel translational control mechanism stimulated by oxidative stress that is characterized by up-regulation of UGA-selenocysteine recoding efficiency and relocalization of SBP2, selenocysteine-specific elongation factor, and L30 recoding factors from the cytoplasm to the nucleus.
Collapse
Affiliation(s)
- Zahia Touat-Hamici
- From the Centre de Génétique Moléculaire, CNRS, UPR3404, 91198 Gif-sur-Yvette, France
| | - Yona Legrain
- From the Centre de Génétique Moléculaire, CNRS, UPR3404, 91198 Gif-sur-Yvette, France
| | - Anne-Laure Bulteau
- the Centre de Recherche Institut Cochin, INSERM U567, CNRS UMR 8104, 75005 Paris, France, and the Laboratoire de Chimie Analytique Bio-Inorganique et Environnement, CNRS/UPPA, UMR5254, 64000 Pau, France
| | - Laurent Chavatte
- From the Centre de Génétique Moléculaire, CNRS, UPR3404, 91198 Gif-sur-Yvette, France, the Laboratoire de Chimie Analytique Bio-Inorganique et Environnement, CNRS/UPPA, UMR5254, 64000 Pau, France
| |
Collapse
|
47
|
Hatfield DL, Tsuji PA, Carlson BA, Gladyshev VN. Selenium and selenocysteine: roles in cancer, health, and development. Trends Biochem Sci 2014; 39:112-20. [PMID: 24485058 PMCID: PMC3943681 DOI: 10.1016/j.tibs.2013.12.007] [Citation(s) in RCA: 430] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 12/24/2013] [Accepted: 12/27/2013] [Indexed: 01/08/2023]
Abstract
The many biological and biomedical effects of selenium are relatively unknown outside the selenium field. This fascinating element, initially described as a toxin, was subsequently shown to be essential for health and development. By the mid-1990s selenium emerged as one of the most promising cancer chemopreventive agents, but subsequent human clinical trials yielded contradictory results. However, basic research on selenium continued to move at a rapid pace, elucidating its many roles in health, development, and in cancer prevention and promotion. Dietary selenium acts principally through selenoproteins, most of which are oxidoreductases involved in diverse cellular functions.
Collapse
Affiliation(s)
- Dolph L Hatfield
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Petra A Tsuji
- Department of Biological Sciences, Towson University, Towson, MD 21252, USA
| | - Bradley A Carlson
- Molecular Biology of Selenium Section, Mouse Cancer Genetics Program, Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Lippmann D, Lehmann C, Florian S, Barknowitz G, Haack M, Mewis I, Wiesner M, Schreiner M, Glatt H, Brigelius-Flohé R, Kipp AP. Glucosinolates from pak choi and broccoli induce enzymes and inhibit inflammation and colon cancer differently. Food Funct 2014; 5:1073-81. [DOI: 10.1039/c3fo60676g] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Feeding a glucosinolate-enriched pak choi diet reduced colitis and tumor numbers. No effects were observed by a glucosinolate-enriched broccoli diet.
Collapse
Affiliation(s)
- Doris Lippmann
- German Institute of Human Nutrition Potsdam-Rehbruecke
- D-14558 Nuthetal, Germany
| | - Carsten Lehmann
- German Institute of Human Nutrition Potsdam-Rehbruecke
- D-14558 Nuthetal, Germany
| | - Simone Florian
- German Institute of Human Nutrition Potsdam-Rehbruecke
- D-14558 Nuthetal, Germany
| | - Gitte Barknowitz
- German Institute of Human Nutrition Potsdam-Rehbruecke
- D-14558 Nuthetal, Germany
| | - Michael Haack
- German Institute of Human Nutrition Potsdam-Rehbruecke
- D-14558 Nuthetal, Germany
| | - Inga Mewis
- Leibniz-Institute of Vegetable and Ornamental Crops
- Grossbeeren and Erfurt e.V
- Germany
| | - Melanie Wiesner
- Leibniz-Institute of Vegetable and Ornamental Crops
- Grossbeeren and Erfurt e.V
- Germany
| | - Monika Schreiner
- Leibniz-Institute of Vegetable and Ornamental Crops
- Grossbeeren and Erfurt e.V
- Germany
| | - Hansruedi Glatt
- German Institute of Human Nutrition Potsdam-Rehbruecke
- D-14558 Nuthetal, Germany
| | | | - Anna P. Kipp
- German Institute of Human Nutrition Potsdam-Rehbruecke
- D-14558 Nuthetal, Germany
| |
Collapse
|
49
|
Humann-Ziehank E, Renko K, Bruegmann ML, Devi VR, Hewicker-Trautwein M, Andreae A, Ganter M. Long-term study of ovine pulmonary adenocarcinogenesis in sheep with marginal vs. sufficient nutritional selenium supply: results from computed tomography, pathology, immunohistochemistry, JSRV-PCR and lung biochemistry. J Trace Elem Med Biol 2013; 27:391-9. [PMID: 23623247 DOI: 10.1016/j.jtemb.2013.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 01/09/2013] [Accepted: 03/04/2013] [Indexed: 02/04/2023]
Abstract
The impact of selenium (Se) in carcinogenesis is still debatable due to inconsistent results of observational studies, recent suspicion of diabetic side effects and e.g. dual roles of glutathione peroxidases (GPx). Previously, our group introduced long-term studies on lung carcinogenesis using the jaagtsiekte sheep retrovirus (JSRV) induced ovine pulmonary adenocarcinoma (OPA) as an innovative animal model. The present report describes the results of sufficient (0.2 mg Se/kg dry weight (dw)) vs. marginal (<0.05 mg Se/kg dw) nutritional Se supply on cancer progression over a two-year period in 16 animals. Computed tomography (CT) evaluation of lung cancer progression, final pathological examination, evidence of pro-viral JSRV-DNA in lung, lymph nodes and broncho-alveolar lavage cells as well as biochemical analysis of Se, GPx1 and thioredoxin reductase (TrxR) activity in lung tissue were recorded. Additionally, immunohistochemical determination of GPx1 expression in unaffected and neoplastic lung cells was implemented. The feeding regime caused significant differences in Se concentration and GPx1 activity in lung tissue between groups, whereas TrxR activity remained unaffected. JSRV was evident in broncho-alveolar lavage cells, lung tissue and lung lymph nodes. Quarterly executed CT could not demonstrate differences in lung cancer proliferation intensity. Necropsy and histopathology substantiated CT findings. Immunohistochemical analysis of GPx1 in lung tissue suggested a coherency of GPx1 immunolabelling intensity in dependence of tumour size. It was concluded that the model proved to be suitable for long-term studies of lung cancer proliferation including the impact of modifiable nutritional factors. Proliferation of OPA was unaffected by marginal vs. sufficient nutritional Se supply.
Collapse
Affiliation(s)
- Esther Humann-Ziehank
- Klinik für kleine Klauentiere und Forensische Medizin und Ambulatorische Klinik, Stiftung Tierärztliche Hochschule Hannover, Bischofsholer Damm 15, D-30173 Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
50
|
Bindoli A, Rigobello MP. Principles in redox signaling: from chemistry to functional significance. Antioxid Redox Signal 2013; 18:1557-93. [PMID: 23244515 DOI: 10.1089/ars.2012.4655] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reactive oxygen and nitrogen species are currently considered not only harmful byproducts of aerobic respiration but also critical mediators of redox signaling. The molecules and the chemical principles sustaining the network of cellular redox regulated processes are described. Special emphasis is placed on hydrogen peroxide (H(2)O(2)), now considered as acting as a second messenger, and on sulfhydryl groups, which are the direct targets of the oxidant signal. Cysteine residues of some proteins, therefore, act as sensors of redox conditions and are oxidized in a reversible reaction. In particular, the formation of sulfenic acid and disulfide, the initial steps of thiol oxidation, are described in detail. The many cell pathways involved in reactive oxygen species formation are reported. Central to redox signaling processes are the glutathione and thioredoxin systems controlling H(2)O(2) levels and, hence, the thiol/disulfide balance. Lastly, some of the most important redox-regulated processes involving specific enzymes and organelles are described. The redox signaling area of research is rapidly expanding, and future work will examine new pathways and clarify their importance in cellular pathophysiology.
Collapse
Affiliation(s)
- Alberto Bindoli
- Institute of Neuroscience (CNR), Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | | |
Collapse
|