1
|
Ilaghi M, Kafi F, Shafiei M, Zangiabadian M, Nasiri MJ. Dietary supplementations to mitigate the cardiopulmonary effects of air pollution toxicity: A systematic review of clinical trials. PLoS One 2024; 19:e0304402. [PMID: 38870164 PMCID: PMC11175466 DOI: 10.1371/journal.pone.0304402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/09/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND There is a consistent association between exposure to air pollution and elevated rates of cardiopulmonary illnesses. As public health activities emphasize the paramount need to reduce exposure, it is crucial to examine strategies like the antioxidant diet that could potentially protect individuals who are unavoidably exposed. METHODS A systematic search was performed in PubMed/Medline, EMBASE, CENTRAL, and ClinicalTrials.gov up to March 31, 2023, for clinical trials assessing dietary supplements against cardiovascular (blood pressure, heart rate, heart rate variability, brachial artery diameter, flow-mediated dilation, and lipid profile) or pulmonary outcomes (pulmonary function and airway inflammation) attributed to air pollution exposure. RESULTS After reviewing 4681 records, 18 studies were included. There were contradictory findings on the effects of fish oil and olive oil supplementations on cardiovascular outcomes. Although with limited evidence, fish oil offered protection against pulmonary dysfunction induced by pollutants. Most studies on vitamin C did not find protective cardiovascular effects; however, the combination of vitamin C and E offered protective effects against pulmonary dysfunction but showed conflicting results for cardiovascular outcomes. Other supplements like sulforaphane, L-arginine, n-acetylcysteine, and B vitamins showed potential beneficial effects but need further research due to the limited number of existing trials. CONCLUSIONS Although more research is needed to determine the efficacy and optimal dose of anti-inflammatory and antioxidant dietary supplements against air pollution toxicity, this low-cost preventative strategy has the potential to offer protection against outcomes of air pollution exposure.
Collapse
Affiliation(s)
- Mehran Ilaghi
- Institute of Neuropharmacology, Kerman Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Kafi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohadeseh Shafiei
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Moein Zangiabadian
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Shusterman D. History of pollutant adjuvants in respiratory allergy. FRONTIERS IN ALLERGY 2024; 5:1374771. [PMID: 38533354 PMCID: PMC10964904 DOI: 10.3389/falgy.2024.1374771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Combined exposures to allergens and air pollutants emerged as a topic of concern in scientific circles by the 1980's, when it became clear that parallel increases in respiratory allergies and traffic-related air pollution had been occurring during the 20th century. Although historically there has been a tendency to treat exposure-related symptoms as either allergic or toxicologic in nature, cross-interactions have since been established between the two modalities. For example, exposure to selected air pollutants in concert with a given allergen can increase the likelihood that an individual will become sensitized to that allergen, strongly suggesting that the pollutant acted as an adjuvant. Although not a review of underlying mechanisms, the purpose of this mini-review is to highlight the potential significance of co-exposure to adjuvant chemicals in predicting allergic sensitization in the respiratory tract. The current discussion emphasizes the upper airway as a model for respiratory challenge studies, the results of which may be applicable-not only to allergic rhinitis-but also to conjunctivitis and asthma.
Collapse
Affiliation(s)
- Dennis Shusterman
- Upper Airway Biology Laboratory, Division of Occupational, Environmental and Climate Medicine, Department of Medicine, University of California, San Francisco, CA, United States
| |
Collapse
|
3
|
Peden DB. Respiratory Health Effects of Air Pollutants. Immunol Allergy Clin North Am 2024; 44:15-33. [PMID: 37973257 DOI: 10.1016/j.iac.2023.07.004] [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: 11/19/2023]
Abstract
Air pollution is a risk factor for asthma and respiratory infection. Avoidance of air pollution is the best approach to mitigating the impacts of pollution. Personal preventive strategies are possible, but policy interventions are the most effective ways to prevent pollution and its effect on asthma and respiratory infection.
Collapse
Affiliation(s)
- David B Peden
- Division of Pediatric Allergy & Immunology and, Center for Environmental Medicine, Asthma and Lung Biology, The School of Medicine, The University of North Carolina at Chapel Hill, UNC School of Medicine, 104 Mason Farm Road, CB#7310, Chapel Hill, NC 27599-7310, USA.
| |
Collapse
|
4
|
Li X, Cai Z, Yang F, Wang Y, Pang X, Sun J, Li X, Lu Y. Broccoli Improves Lipid Metabolism and Intestinal Flora in Mice with Type 2 Diabetes Induced by HFD and STZ Diet. Foods 2024; 13:273. [PMID: 38254574 PMCID: PMC10814524 DOI: 10.3390/foods13020273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Globally, type 2 diabetes (T2DM) is on the rise. Maintaining a healthy diet is crucial for both treating and preventing T2DM.As a common vegetable in daily diet, broccoli has antioxidant, anti-inflammatory and anticarcoma physiological activities. We developed a mouse model of type 2 diabetes and carried out a systematic investigation to clarify the function of broccoli in reducing T2DM symptoms and controlling intestinal flora. The findings demonstrated that broccoli could successfully lower fasting blood glucose (FBG), lessen insulin resistance, regulate lipid metabolism, lower the levels of TC, TG, LDL-C, and MDA, stop the expression of IL-1β and IL-6, and decrease the harm that diabetes causes to the pancreas, liver, fat, and other organs and tissues. Furthermore, broccoli altered the intestinal flora's makeup in mice with T2DM. At the genus level, the relative abundance of Allobaculum decreased, and that of Odoribacter and Oscillospira increased; At the family level, the relative abundances of Odoribacteraceae, Rikenellaceae and S24-7 decreased, while the relative abundances of Erysipelotrichaceae and Rikenellaceae increased.
Collapse
Affiliation(s)
- Xin Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; (X.L.); (Z.C.); (Y.W.); (X.P.); (J.S.); (Y.L.)
- Priority Academic Program, Development of Jiangsu Higher Education Institutions (PAPD), Nanjing 210023, China
| | - Zifan Cai
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; (X.L.); (Z.C.); (Y.W.); (X.P.); (J.S.); (Y.L.)
| | - Feiyu Yang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
| | - Yunfan Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; (X.L.); (Z.C.); (Y.W.); (X.P.); (J.S.); (Y.L.)
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; (X.L.); (Z.C.); (Y.W.); (X.P.); (J.S.); (Y.L.)
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; (X.L.); (Z.C.); (Y.W.); (X.P.); (J.S.); (Y.L.)
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; (X.L.); (Z.C.); (Y.W.); (X.P.); (J.S.); (Y.L.)
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; (X.L.); (Z.C.); (Y.W.); (X.P.); (J.S.); (Y.L.)
| |
Collapse
|
5
|
Fang ZF, Fu Y, Peng Y, Song SR, Wang ZN, Yang Y, Nie YC, Han HL, Teng YB, Xiao WM, Chen JP, Zhou BJ, Ou GL, Xie JX, Liu XY, Zhang JJ, Zhong NS. Citrus peel extract protects against diesel exhaust particle-induced chronic obstructive pulmonary disease-like lung lesions and oxidative stress. Food Funct 2023; 14:9841-9856. [PMID: 37850547 DOI: 10.1039/d3fo02010j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide and characterized by emphysema, small airway remodeling and mucus hypersecretion. Citrus peels have been widely used as food spices and in traditional Chinese medicine for chronic lung disease. Given that citrus peels are known for containing antioxidants and anti-inflammatory compounds, we hypothesize that citrus peel intake can suppress oxidative stress and inflammatory response to air pollution exposure, thereby alleviating COPD-like pathologies. This study aimed to investigate the efficacy of citrus peel extract, namely Guang Chenpi (GC), in preventing the development of COPD induced by diesel exhaust particles (DEPs) and its potential mechanism. DEP-induced COPD-like lung pathologies, inflammatory responses and oxidative stress with or without GC treatment were examined in vivo and in vitro. Our in vivo study showed that GC was effective in decreasing inflammatory cell counts and inflammatory mediator (IL-17A and TNF-α) concentrations in bronchoalveolar lavage fluid (BALF). Pretreatment with GC extract also significantly decreased oxidative stress in the serum and lung tissue of DEP-induced COPD rats. Furthermore, GC pretreatment effectively reduced goblet cell hyperplasia (PAS positive cells) and fibrosis of the small airways, decreased macrophage infiltration as well as carbon loading in the peripheral lungs, and facilitated the resolution of emphysema and small airway remodeling in DEP-induced COPD rats. An in vitro free radical scavenging assay revealed robust antioxidant potential of GC in scavenging DPPH free radicals. Moreover, GC demonstrated potent capacities in reducing ROS production and enhancing SOD activity in BEAS-2B cells stimulated by DEPs. GC treatment significantly attenuated the increased level of IL-8 and MUC5AC from DEP-treated BEAS-2B cells. Mechanistically, GC treatment upregulated the protein level of Nrf-2 and could function via MAPK/NF-κB signaling pathways by suppressing the phosphorylation of p38, JNK and p65. Citrus peel extract is effective in decreasing oxidative stress and inflammatory responses of the peripheral lungs to DEP exposure. These protective effects further contributed to the resolution of COPD-like pathologies.
Collapse
Affiliation(s)
- Zhang-Fu Fang
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen 518020, China
- Guangzhou Laboratory, Guangzhou 510320, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yu Fu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Sheng-Ren Song
- Department of Respiratory Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China
| | - Zhao-Ni Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yang Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yi-Chu Nie
- Translational Medicine Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Hai-Long Han
- Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu 215316, China.
| | - Yan-Bo Teng
- Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu 215316, China.
| | - Wei-Min Xiao
- Shenzhen Academy of Metrology & Quality Inspection, Shenzhen 518055, China
| | - Jia-Ping Chen
- Shenzhen Academy of Metrology & Quality Inspection, Shenzhen 518055, China
| | | | - Guo-Liang Ou
- Jiangmen Palace International Food, Inc., Jiangmen 529000, China
| | - Jia-Xing Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Xiao-Yu Liu
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen 518061, China.
| | - Junfeng Jim Zhang
- Nicholas School of the Environment and Global Health Institute, Duke University, Durham, North Carolina 27708, USA
- Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu 215316, China.
| | - Nan-Shan Zhong
- Guangzhou Laboratory, Guangzhou 510320, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
6
|
Treasure K, Harris J, Williamson G. Exploring the anti-inflammatory activity of sulforaphane. Immunol Cell Biol 2023; 101:805-828. [PMID: 37650498 DOI: 10.1111/imcb.12686] [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] [Received: 03/20/2023] [Revised: 07/24/2023] [Accepted: 08/10/2023] [Indexed: 09/01/2023]
Abstract
Dysregulation of innate immune responses can result in chronic inflammatory conditions. Glucocorticoids, the current frontline therapy, are effective immunosuppressive drugs but come with a trade-off of cumulative and serious side effects. Therefore, alternative drug options with improved safety profiles are urgently needed. Sulforaphane, a phytochemical derived from plants of the brassica family, is a potent inducer of phase II detoxification enzymes via nuclear factor-erythroid factor 2-related factor 2 (NRF2) signaling. Moreover, a growing body of evidence suggests additional diverse anti-inflammatory properties of sulforaphane through interactions with mediators of key signaling pathways and inflammatory cytokines. Multiple studies support a role for sulforaphane as a negative regulator of nuclear factor kappa-light chain enhancer of activated B cells (NF-κB) activation and subsequent cytokine release, inflammasome activation and direct regulation of the activity of macrophage migration inhibitory factor. Significantly, studies have also highlighted potential steroid-sparing activity for sulforaphane, suggesting that it may have potential as an adjunctive therapy for some inflammatory conditions. This review discusses published research on sulforaphane, including proposed mechanisms of action, and poses questions for future studies that might help progress our understanding of the potential clinical applications of this intriguing molecule.
Collapse
Affiliation(s)
- Katie Treasure
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- Victorian Heart Hospital, Monash University, Clayton, VIC, Australia
| | - James Harris
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC, Australia
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Gary Williamson
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- Victorian Heart Hospital, Monash University, Clayton, VIC, Australia
| |
Collapse
|
7
|
Royce SG, Licciardi PV, Beh RC, Bourke JE, Donovan C, Hung A, Khurana I, Liang JJ, Maxwell S, Mazarakis N, Pitsillou E, Siow YY, Snibson KJ, Tobin MJ, Ververis K, Vongsvivut J, Ziemann M, Samuel CS, Tang MLK, El-Osta A, Karagiannis TC. Sulforaphane prevents and reverses allergic airways disease in mice via anti-inflammatory, antioxidant, and epigenetic mechanisms. Cell Mol Life Sci 2022; 79:579. [DOI: 10.1007/s00018-022-04609-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022]
|
8
|
Sulforaphane inhibits angiotensin II-induced cardiomyocyte apoptosis by acetylation modification of Nrf2. Aging (Albany NY) 2022; 14:6740-6755. [PMID: 36006435 PMCID: PMC9467410 DOI: 10.18632/aging.204247] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
Oxidative stress is the central cause of angiotensin II (Ang II)-induced myocardial injury, and nuclear factor erythroid 2-related factor (Nrf2) is the core molecule of the anti-oxidant defense system. We have previously demonstrated that sulforaphane (SFN) can prevent Ang II-induced myocardial injury by activating Nrf2; however, the underlying molecular mechanism is still unclear. This study aimed to evaluate whether SFN prevents Ang II-induced cardiomyocyte apoptosis through acetylation modification of <i>Nrf2</i>. Wild-type and <i>Nrf2</i> knockdown embryonic rat cardiomyocytes (H9C2) were exposed to Ang II to induce apoptosis, oxidative stress, and inflammatory responses. SFN treatment significantly reduced Ang II-induced cardiomyocyte apoptosis, inflammation and oxidative stress. Activation of Nrf2 played a critical role in preventing cardiomyocyte apoptosis. After Nrf2 was knockdown, the anti-inflammatory, antioxidant stress of SFN were eliminated. Furthermore, Nrf2 activation by SFN was closely related to the decreased activity of histone deacetylases (HDACs) and increased histone-3 (H3) acetylation levels in <i>Nrf2</i> promoter region. These findings confirm that Nrf2 plays a key role in SFN preventing Ang II-induced cardiomyocyte apoptosis. SFN activates Nrf2 by inhibiting HDACs expression and activation.
Collapse
|
9
|
Bayo Jimenez MT, Frenis K, Hahad O, Steven S, Cohen G, Cuadrado A, Münzel T, Daiber A. Protective actions of nuclear factor erythroid 2-related factor 2 (NRF2) and downstream pathways against environmental stressors. Free Radic Biol Med 2022; 187:72-91. [PMID: 35613665 DOI: 10.1016/j.freeradbiomed.2022.05.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/23/2022] [Accepted: 05/19/2022] [Indexed: 12/14/2022]
Abstract
Environmental risk factors, including noise, air pollution, chemical agents, ultraviolet radiation (UVR) and mental stress have a considerable impact on human health. Oxidative stress and inflammation are key players in molecular pathomechanisms of environmental pollution and risk factors. In this review, we delineate the impact of environmental risk factors and the protective actions of the nuclear factor erythroid 2-related factor 2 (NRF2) in connection to oxidative stress and inflammation. We focus on well-established studies that demonstrate the protective actions of NRF2 and its downstream pathways against different environmental stressors. State-of-the-art mechanistic considerations on NRF2 signaling are discussed in detail, e.g. classical concepts like KEAP1 oxidation/electrophilic modification, NRF2 ubiquitination and degradation. Specific focus is also laid on NRF2-dependent heme oxygenase-1 induction with detailed presentation of the protective down-stream pathways of heme oxygenase-1, including interaction with BACH1 system. The significant impact of all environmental stressors on the circadian rhythm and the interactions of NRF2 with the circadian clock will also be considered here. A broad range of NRF2 activators is discussed in relation to environmental stressor-induced health side effects, thereby suggesting promising new mitigation strategies (e.g. by nutraceuticals) to fight the negative effects of the environment on our health.
Collapse
Affiliation(s)
- Maria Teresa Bayo Jimenez
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Katie Frenis
- Department of Hematology and Oncology, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Leibniz Insitute for Resilience Research (LIR), Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Guy Cohen
- The Skin Research Institute, The Dead Sea and Arava Science Center, Masada, 86910, Israel; Ben Gurion University of the Negev, Eilat Campus, Eilat, 8855630, Israel
| | - Antonio Cuadrado
- Departamento de Bioquímica, Facultad de Medicina, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas 'Alberto Sols' UAM-CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
10
|
Tian JJ, Levy M, Zhang X, Sinnott R, Maddela R. Counteracting Health Risks by Modulating Homeostatic Signaling. Pharmacol Res 2022; 182:106281. [PMID: 35661711 DOI: 10.1016/j.phrs.2022.106281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/14/2022] [Accepted: 05/27/2022] [Indexed: 10/18/2022]
Abstract
Homeostasis was initially conceptualized by Bernard and Cannon around a century ago as a steady state of physiological parameters that vary within a certain range, such as blood pH, body temperature, and heart rate1,2. The underlying mechanisms that maintain homeostasis are explained by negative feedbacks that are executed by the neuronal, endocrine, and immune systems. At the cellular level, homeostasis, such as that of redox and energy steady state, also exists and is regulated by various cell signaling pathways. The induction of homeostatic mechanism is critical for human to adapt to various disruptive insults (stressors); while on the other hand, adaptation occurs at the expense of other physiological processes and thus runs the risk of collateral damages, particularly under conditions of chronic stress. Conceivably, anti-stress protection can be achieved by stressor-mimicking medicinals that elicit adaptive responses prior to an insult and thereby serve as health risk countermeasures; and in situations where maladaptation may occur, downregulating medicinals could be used to suppress the responses and prevent subsequent pathogenesis. Both strategies are preemptive interventions particularly suited for individuals who carry certain lifestyle, environmental, or genetic risk factors. In this article, we will define and characterize a new modality of prophylactic intervention that forestalls diseases via modulating homeostatic signaling. Moreover, we will provide evidence from the literature that support this concept and distinguish it from other homeostasis-related interventions such as adaptogen, hormesis, and xenohormesis.
Collapse
Affiliation(s)
- Junqiang J Tian
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA.
| | - Mark Levy
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| | - Xuekai Zhang
- Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing100029, China; US Center for Chinese Medicine, 14801 Physicians lane, 171 A 2nd Floor, #281, Rockville MD 20850, USA
| | - Robert Sinnott
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| | - Rolando Maddela
- USANA Health Science, Inc., 3838 Parkway Blvd, Salt Lake City, UT 84121, USA
| |
Collapse
|
11
|
Marino M, Martini D, Venturi S, Tucci M, Porrini M, Riso P, Del Bo' C. An Overview of Registered Clinical Trials on Glucosinolates and Human Health: The Current Situation. Front Nutr 2021; 8:730906. [PMID: 34778334 PMCID: PMC8578719 DOI: 10.3389/fnut.2021.730906] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Epidemiological studies suggest a potential role of glucosinolates (GSLs) and isothiocyanates on human health. However, evidence from intervention studies, due to heterogeneity in features of study design, duration, participants, food or food components administered, and outcomes analyzed, is still insufficient. The current review aims to provide an overview of the trials on GSLs and GSL-rich foods registered over the last 20 years with the intention to summarize the main topics and results, but also the existing gaps that still need to be covered. Studies were collected by using ClinicalTrials.gov and the International Standard Randomized Controlled Trial Number (ISRCTN) registry. A total of 87 registered trials were identified with which most of them were performed by using extracts or pure compounds (n = 60) while few were conducted with GSL-rich foods (n = 27). In detail, sulforaphane was the most investigated compound, while broccoli was the most frequent food tested in the trials. The majority of the studies assessed the health effects of GSLs focusing on outcomes related to cancer and cognitive function, even if the current findings are not univocal. Emerging topics also included the study of GSLs and gut microbiota interaction and impact on skin health. Further attention was also drawn to the bioavailability of GSLs and/or derivatives from foods, extracts, and single compounds by also considering the contribution of the different genetic polymorphisms. In conclusion, although considerable efforts have been made to study GSLs and GSL-rich foods, further studies are necessary to provide evidence-based research and to corroborate the findings obtained. The interindividual response due to genetic polymorphisms should be further investigated in order to explore the contribution to the overall beneficial effect.
Collapse
Affiliation(s)
| | | | | | | | | | - Patrizia Riso
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
12
|
McCarty MF, Lerner A, DiNicolantonio JJ, Benzvi C. Nutraceutical Aid for Allergies - Strategies for Down-Regulating Mast Cell Degranulation. J Asthma Allergy 2021; 14:1257-1266. [PMID: 34737578 PMCID: PMC8558634 DOI: 10.2147/jaa.s332307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/24/2021] [Indexed: 11/23/2022] Open
Abstract
Interactions of antigens with the mast cell FcεRI-IgE receptor complex induce degranulation and boost synthesis of pro-inflammatory lipid mediators and cytokines. Activation of spleen tyrosine kinase (Syk) functions as a central hub in this signaling. The tyrosine phosphatase SHP-1 opposes Syk activity; stimulation of NADPH oxidase by FcεRI activation results in the production of oxidants that reversibly inhibit SHP-1, up-regulating the signal from Syk. Activated AMPK can suppress Syk activation by the FcεRI receptor, possibly reflecting its ability to phosphorylate the FcεRI beta subunit. Cyclic GMP, via protein kinase G II, enhances the activity of SHP-1 by phosphorylating its C-terminal region; this may explain its inhibitory impact on mast cell activation. Hydrogen sulfide (H2S) likewise opposes mast cell activation; H2S can boost AMPK activity, up-regulate cGMP production, and trigger Nrf2-mediated induction of Phase 2 enzymes - including heme oxygenase-1, whose generation of bilirubin suppresses NADPH oxidase activity. Phycocyanobilin (PCB), a chemical relative of bilirubin, shares its inhibitory impact on NADPH oxidase, rationalizing reported anti-allergic effects of PCB-rich spirulina ingestion. Phase 2 inducer nutraceuticals can likewise oppose the up-regulatory impact of NADPH oxidase on FcεRI signaling. AMPK can be activated with the nutraceutical berberine. High-dose biotin can boost cGMP levels in mast cells via direct stimulation of soluble guanylate cyclase. Endogenous generation of H2S in mast cells can be promoted by administering N-acetylcysteine and likely by taurine, which increases the expression of H2S-producing enzymes in the vascular system. Mast cell stabilization by benifuuki green tea catechins may reflect the decreased surface expression of FcεRI.
Collapse
Affiliation(s)
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| | - James J DiNicolantonio
- Saint Luke’s Mid America Heart Institute, Kansas City, MO, USA
- Advanced Ingredients for Dietary Products, AIDP, City of Industry, CA, USA
| | - Carina Benzvi
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| |
Collapse
|
13
|
Audousset C, McGovern T, Martin JG. Role of Nrf2 in Disease: Novel Molecular Mechanisms and Therapeutic Approaches - Pulmonary Disease/Asthma. Front Physiol 2021; 12:727806. [PMID: 34658913 PMCID: PMC8511424 DOI: 10.3389/fphys.2021.727806] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a major transcription factor involved in redox homeostasis and in the response induced by oxidative injury. Nrf2 is present in an inactive state in the cytoplasm of cells. Its activation by internal or external stimuli, such as infections or pollution, leads to the transcription of more than 500 elements through its binding to the antioxidant response element. The lungs are particularly susceptible to factors that generate oxidative stress such as infections, allergens and hyperoxia. Nrf2 has a crucial protective role against these ROS. Oxidative stress and subsequent activation of Nrf2 have been demonstrated in many human respiratory diseases affecting the airways, including asthma and chronic obstructive pulmonary disease (COPD), or the pulmonary parenchyma such as acute respiratory distress syndrome (ARDS) and pulmonary fibrosis. Several compounds, both naturally occurring and synthetic, have been identified as Nrf2 inducers and enhance the activation of Nrf2 and expression of Nrf2-dependent genes. These inducers have proven particularly effective at reducing the severity of the oxidative stress-driven lung injury in various animal models. In humans, these compounds offer promise as potential therapeutic strategies for the management of respiratory pathologies associated with oxidative stress but there is thus far little evidence of efficacy through human trials. The purpose of this review is to summarize the involvement of Nrf2 and its inducers in ARDS, COPD, asthma and lung fibrosis in both human and in experimental models.
Collapse
Affiliation(s)
- Camille Audousset
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| | - Toby McGovern
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| | - James G Martin
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| |
Collapse
|
14
|
Aryal A, Harmon AC, Dugas TR. Particulate matter air pollutants and cardiovascular disease: Strategies for intervention. Pharmacol Ther 2021; 223:107890. [PMID: 33992684 PMCID: PMC8216045 DOI: 10.1016/j.pharmthera.2021.107890] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
Air pollution is consistently linked with elevations in cardiovascular disease (CVD) and CVD-related mortality. Particulate matter (PM) is a critical factor in air pollution-associated CVD. PM forms in the air during the combustion of fuels as solid particles and liquid droplets and the sources of airborne PM range from dust and dirt to soot and smoke. The health impacts of PM inhalation are well documented. In the US, where CVD is already the leading cause of death, it is estimated that PM2.5 (PM < 2.5 μm in size) is responsible for nearly 200,000 premature deaths annually. Despite the public health data, definitive mechanisms underlying PM-associated CVD are elusive. However, evidence to-date implicates mechanisms involving oxidative stress, inflammation, metabolic dysfunction and dyslipidemia, contributing to vascular dysfunction and atherosclerosis, along with autonomic dysfunction and hypertension. For the benefit of susceptible individuals and individuals who live in areas where PM levels exceed the National Ambient Air Quality Standard, interventional strategies for mitigating PM-associated CVD are necessary. This review will highlight current state of knowledge with respect to mechanisms for PM-dependent CVD. Based upon these mechanisms, strategies for intervention will be outlined. Citing data from animal models and human subjects, these highlighted strategies include: 1) antioxidants, such as vitamins E and C, carnosine, sulforaphane and resveratrol, to reduce oxidative stress and systemic inflammation; 2) omega-3 fatty acids, to inhibit inflammation and autonomic dysfunction; 3) statins, to decrease cholesterol accumulation and inflammation; 4) melatonin, to regulate the immune-pineal axis and 5) metformin, to address PM-associated metabolic dysfunction. Each of these will be discussed with respect to its potential role in limiting PM-associated CVD.
Collapse
Affiliation(s)
- Ankit Aryal
- Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, Baton Rouge, Louisiana 70803, United States of America
| | - Ashlyn C Harmon
- Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, Baton Rouge, Louisiana 70803, United States of America
| | - Tammy R Dugas
- Louisiana State University School of Veterinary Medicine, Department of Comparative Biomedical Sciences, Skip Bertman Drive, Baton Rouge, Louisiana 70803, United States of America.
| |
Collapse
|
15
|
Sulforaphane attenuates oxidative stress and inflammation induced by fine particulate matter in human bronchial epithelial cells. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
16
|
Fahey JW, Kensler TW. The Challenges of Designing and Implementing Clinical Trials With Broccoli Sprouts… and Turning Evidence Into Public Health Action. Front Nutr 2021; 8:648788. [PMID: 33996874 PMCID: PMC8116591 DOI: 10.3389/fnut.2021.648788] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Broccoli sprouts are a convenient and rich source of the glucosinolate glucoraphanin, which can generate the chemopreventive agent sulforaphane through the catalytic actions of plant myrosinase or β-thioglucosidases in the gut microflora. Sulforaphane, in turn, is an inducer of cytoprotective enzymes through activation of Nrf2 signaling, and a potent inhibitor of carcinogenesis in multiple murine models. Sulforaphane is also protective in models of diabetes, neurodegenerative disease, and other inflammatory processes, likely reflecting additional actions of Nrf2 and interactions with other signaling pathways. Translating this efficacy into the design and implementation of clinical chemoprevention trials, especially food-based trials, faces numerous challenges including the selection of the source, placebo, and dose as well as standardization of the formulation of the intervention material. Unlike in animals, purified sulforaphane has had very limited use in clinical studies. We have conducted a series of clinical studies and randomized clinical trials to evaluate the effects of composition (glucoraphanin-rich [± myrosinase] vs. sulforaphane-rich or mixture beverages), formulation (beverage vs. tablet) and dose, on the efficacy of these broccoli sprout-based preparations to evaluate safety, pharmacokinetics, pharmacodynamic action, and clinical benefit. While the challenges for the evaluation of broccoli sprouts in clinical trials are themselves formidable, further hurdles must be overcome to bring this science to public health action.
Collapse
Affiliation(s)
- Jed W. Fahey
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Nutrition and Food Studies, College of Health and Human Services, George Mason University, Fairfax, VA, United States
| | - Thomas W. Kensler
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
17
|
Bousquet J, Le Moing V, Blain H, Czarlewski W, Zuberbier T, de la Torre R, Pizarro Lozano N, Reynes J, Bedbrook A, Cristol JP, Cruz AA, Fiocchi A, Haahtela T, Iaccarino G, Klimek L, Kuna P, Melén E, Mullol J, Samolinski B, Valiulis A, Anto JM. Efficacy of broccoli and glucoraphanin in COVID-19: From hypothesis to proof-of-concept with three experimental clinical cases. World Allergy Organ J 2021; 14:100498. [PMID: 33425204 PMCID: PMC7770975 DOI: 10.1016/j.waojou.2020.100498] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/26/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
COVID-19 is described in a clinical case involving a patient who proposed the hypothesis that Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-interacting nutrients may help to prevent severe COVID-19 symptoms. Capsules of broccoli seeds containing glucoraphanin were being taken before the onset of SARS-CoV-2 infection and were continued daily for over a month after the first COVID-19 symptoms. They were found to reduce many of the symptoms rapidly and for a duration of 6-12 h by repeated dosing. When the patient was stable but still suffering from cough and nasal obstruction when not taking the broccoli capsules, a double-blind induced cough challenge confirmed the speed of onset of the capsules (less than 10 min). A second clinical case with lower broccoli doses carried out during the cytokine storm confirmed the clinical benefits already observed. A third clinical case showed similar effects at the onset of symptoms. In the first clinical trial, we used a dose of under 600 μmol per day of glucoraphanin. However, such a high dose may induce pharmacologic effects that require careful examination before the performance of any study. It is likely that the fast onset of action is mediated through the TRPA1 channel. These experimental clinical cases represent a proof-of-concept confirming the hypothesis that Nrf2-interacting nutrients are effective in COVID-19. However, this cannot be used in practice before the availability of further safety data, and confirmation is necessary through proper trials on efficacy and safety.
Collapse
Key Words
- ACE, Angiotensin converting enzyme
- AT1R, Angiotensin II receptor type 1
- BMI, Body mass index
- Broccoli
- Broccoli, Broccoli seed capsules
- COVID-19
- COVID-19, Coronavirus 19 disease
- Cough challenge
- NAPQI, N-acetyl-p-benzoquinone imine
- Nrf2
- Nrf2, Nuclear factor (erythroid-derived 2)-like 2
- SARS, Severe acute respiratory syndrome
- SARS-Cov-2, Severe acute respiratory syndrome coronavirus 2
- TRP, Transient receptor potential
- TRPA1
- TRPA1, Transient receptor potential ankyrin 1
- TRPV1
- TRPV1, Transient receptor potential vanillin 1
- VAS, Visual analogue scale
Collapse
Affiliation(s)
- Jean Bousquet
- Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Comprehensive Allergy Center, Department of Dermatology and Allergy, Berlin, Germany
- MACVIA France, University Hospital, Montpellier, France
| | | | - Hubert Blain
- Department of Geriatrics, Montpellier University Hospital, Montpellier, France
| | | | - Torsten Zuberbier
- Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Comprehensive Allergy Center, Department of Dermatology and Allergy, Berlin, Germany
| | - Rafael de la Torre
- CIBER Fisiopatologia de La Obesidad y Nutrición (CIBEROBN), Madrid, Spain
- IMIM (Hospital del Mar Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | - Jacques Reynes
- Maladies Infectiouses et Tropicales, CHU Montpellier, France
| | - Anna Bedbrook
- MACVIA France, University Hospital, Montpellier, France
- MASK-air, Montpellier, France
| | - Jean-Paul Cristol
- Laboratoire de Biochimie et Hormonologie, PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de Montpellier, France
| | - Alvaro A. Cruz
- Fundação ProAR, Federal University of Bahia and GARD/WHO Planning Group, Salvador, Brazil
| | - Alessandro Fiocchi
- Division of Allergy, Department of Pediatric Medicine - The Bambino Gesù Children's Research Hospital Holy see, Rome, Italy
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital, And University of Helsinki, Helsinki, Finland
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, Federico II University, Napoli, Italy
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Piotr Kuna
- Division of Internal Medicine, Asthma and Allergy, Barlicki University Hospital, Medical University of Lodz, Poland
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Sachs' Children's Hospital, Stockholm, Sweden
| | - Joaquim Mullol
- Rhinology Unit & Smell Clinic, ENT Department, Hospital Clinic - Clinical & Experimental Respiratory Immunoallergy, IDIBAPS, CIBERES, Universitat de Barcelona, Barcelona, Spain
| | - Boleslaw Samolinski
- Department of Prevention of Environmental Hazards and Allergology, Medical University of Warsaw, Poland
| | - Arunas Valiulis
- Vilnius University Faculty of Medicine, Institute of Clinical Medicine & Institute of Health Sciences, Vilnius, Lithuania
| | - Josep M. Anto
- IMIM (Hospital del Mar Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
- ISGlobal. ISGlobAL, Barcelona, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
| |
Collapse
|
18
|
Hurtado-Lorenzo A, Honig G, Heller C. Precision Nutrition Initiative: Toward Personalized Diet Recommendations for Patients With Inflammatory Bowel Diseases. CROHN'S & COLITIS 360 2020; 2:otaa087. [PMID: 36777761 PMCID: PMC9802167 DOI: 10.1093/crocol/otaa087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
| | - Gerard Honig
- Research Department, Crohn’s & Colitis Foundation, New York, New York, USA
| | - Caren Heller
- Research Department, Crohn’s & Colitis Foundation, New York, New York, USA
| |
Collapse
|
19
|
Yagishita Y, Gatbonton-Schwager TN, McCallum ML, Kensler TW. Current Landscape of NRF2 Biomarkers in Clinical Trials. Antioxidants (Basel) 2020; 9:antiox9080716. [PMID: 32784785 PMCID: PMC7464243 DOI: 10.3390/antiox9080716] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
The transcription factor NF-E2 p45-related factor 2 (NRF2; encoded by NFE2L2) plays a critical role in the maintenance of cellular redox and metabolic homeostasis, as well as the regulation of inflammation and cellular detoxication pathways. The contribution of the NRF2 pathway to organismal homeostasis is seen in many studies using cell lines and animal models, raising intense attention towards targeting its clinical promise. Over the last three decades, an expanding number of clinical studies have examined NRF2 inducers targeting an ever-widening range of diseases. Full understanding of the pharmacokinetic and pharmacodynamic properties of drug candidates rely partly on the identification, validation, and use of biomarkers to optimize clinical applications. This review focuses on results from clinical trials with four agents known to target NRF2 signaling in preclinical studies (dimethyl fumarate, bardoxolone methyl, oltipraz, and sulforaphane), and evaluates the successes and limitations of biomarkers focused on expression of NRF2 target genes and others, inflammation and oxidative stress biomarkers, carcinogen metabolism and adduct biomarkers in unavoidably exposed populations, and targeted and untargeted metabolomics. While no biomarkers excel at defining pharmacodynamic actions in this setting, it is clear that these four lead clinical compounds do touch the NRF2 pathway in humans.
Collapse
|
20
|
Carlsten C, Salvi S, Wong GWK, Chung KF. Personal strategies to minimise effects of air pollution on respiratory health: advice for providers, patients and the public. Eur Respir J 2020; 55:1902056. [PMID: 32241830 PMCID: PMC7270362 DOI: 10.1183/13993003.02056-2019] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/24/2020] [Indexed: 11/11/2022]
Abstract
As global awareness of air pollution rises, so does the imperative to provide evidence-based recommendations for strategies to mitigate its impact. While public policy has a central role in reducing air pollution, exposure can also be reduced by personal choices. Qualified evidence supports limiting physical exertion outdoors on high air pollution days and near air pollution sources, reducing near-roadway exposure while commuting, utilising air quality alert systems to plan activities, and wearing facemasks in prescribed circumstances. Other strategies include avoiding cooking with solid fuels, ventilating and isolating cooking areas, and using portable air cleaners fitted with high-efficiency particulate air filters. We detail recommendations to assist providers and public health officials when advising patients and the public regarding personal-level strategies to mitigate risk imposed by air pollution, while recognising that well-designed prospective studies are urgently needed to better establish and validate interventions that benefit respiratory health in this context.
Collapse
Affiliation(s)
- Christopher Carlsten
- Air Pollution Exposure Laboratory, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Gary W K Wong
- Dept of Pediatrics and School of Public Health, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kian Fan Chung
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| |
Collapse
|
21
|
Barthelemy J, Sanchez K, Miller MR, Khreis H. New Opportunities to Mitigate the Burden of Disease Caused by Traffic Related Air Pollution: Antioxidant-Rich Diets and Supplements. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17020630. [PMID: 31963738 PMCID: PMC7014349 DOI: 10.3390/ijerph17020630] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022]
Abstract
Air pollution is associated with premature mortality and a wide spectrum of diseases. Traffic-related air pollution (TRAP) is one of the most concerning sources of air pollution for human exposure and health. Until TRAP levels can be significantly reduced on a global scale, there is a need for effective shorter-term strategies to prevent the adverse health effects of TRAP. A growing number of studies suggest that increasing antioxidant intake, through diet or supplementation, may reduce this burden of disease. In this paper, we conducted a non-systematic literature review to assess the available evidence on antioxidant-rich diets and antioxidant supplements as a strategy to mitigate adverse health effects of TRAP in human subjects. We identified 11 studies that fit our inclusion criteria; 3 of which investigated antioxidant-rich diets and 8 of which investigated antioxidant supplements. Overall, we found consistent evidence that dietary intake of antioxidants from adherence to the Mediterranean diet and increased fruit and vegetable consumption is effective in mitigating adverse health effects associated with TRAP. In contrast, antioxidant supplements, including fish oil, olive oil, and vitamin C and E supplements, presented conflicting evidence. Further research is needed to determine why antioxidant supplementation has limited efficacy and whether this relates to effective dose, supplement formulation, timing of administration, or population being studied. There is also a need to better ascertain if susceptible populations, such as children, the elderly, asthmatics and occupational workers consistently exposed to TRAP, should be recommended to increase their antioxidant intake to reduce their burden of disease. Policymakers should consider increasing populations' antioxidant intake, through antioxidant-rich diets, as a relatively cheap and easy preventive measure to lower the burden of disease associated with TRAP.
Collapse
Affiliation(s)
- Jillian Barthelemy
- Center for Advancing Research in Transportation Emissions, Energy, and Health (CARTEEH), Texas A & M Transportation Institute (TTI), College Station, TX 77843, USA; (J.B.); (K.S.)
| | - Kristen Sanchez
- Center for Advancing Research in Transportation Emissions, Energy, and Health (CARTEEH), Texas A & M Transportation Institute (TTI), College Station, TX 77843, USA; (J.B.); (K.S.)
| | - Mark R. Miller
- Centre for Cardiovascular Science, Queens Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK;
| | - Haneen Khreis
- Center for Advancing Research in Transportation Emissions, Energy, and Health (CARTEEH), Texas A & M Transportation Institute (TTI), College Station, TX 77843, USA; (J.B.); (K.S.)
- Barcelona Institute for Global Health (ISGlobal), Centre for Research in Environmental Epidemiology (CREAL), 08003 Barcelona, Spain
- Correspondence:
| |
Collapse
|
22
|
Combination of Broccoli Sprout Extract and Zinc Provides Better Protection against Intermittent Hypoxia-Induced Cardiomyopathy Than Monotherapy in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2985901. [PMID: 31934264 PMCID: PMC6942874 DOI: 10.1155/2019/2985901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/31/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Abstract
Nuclear factor-E2-related factor 2 (Nrf2) and metallothionein have each been reported to protect against chronic intermittent hypoxia- (IH-) induced cardiomyopathy. Sulforaphane-rich broccoli sprout extract (BSE) and zinc can effectively induce Nrf2 and metallothionein, respectively, to protect against IH-induced cardiomyopathy via antioxidative stress. However, whether the cardiac protective effects of the combination of BSE and zinc can be synergistic or the same has not been evaluated. In this study, we treated 8-week-old C57BL/6J mice with BSE and/or zinc during exposure to IH for 8 weeks. Cardiac dysfunction, as determined by echocardiography, and pathological remodeling and abnormalities, including cardiac fibrosis, inflammation, and oxidative damage, examined by histopathology and western blotting, were clearly observed in IH mice but were not significant in IH mice treated with either BSE, zinc, or zinc/BSE. Furthermore, the effects of the combined treatment with BSE and zinc were always greater than those of single treatments. Nrf2 function and metallothionein expression in the heart increased to a greater extent using the combination of BSE and zinc than using BSE or zinc alone. These findings for the first time indicate that the dual activation of Nrf2 and metallothionein by combined treatment with BSE and zinc may be more effective than monotherapy at preventing the development of IH-induced cardiomyopathy.
Collapse
|
23
|
Pázmándi K, Sütő M, Fekete T, Varga A, Boldizsár E, Boldogh I, Bácsi A. Oxidized base 8-oxoguanine, a product of DNA repair processes, contributes to dendritic cell activation. Free Radic Biol Med 2019; 143:209-220. [PMID: 31408726 PMCID: PMC6848796 DOI: 10.1016/j.freeradbiomed.2019.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 01/14/2023]
Abstract
A growing body of evidence suggests that elevated levels of reactive oxygen species (ROS) in the airways caused by exposure to gas phase pollutants or particulate matter are able to activate dendritic cells (DCs); however, the exact mechanisms are still unclear. When present in excess, ROS can modify macromolecules including DNA. One of the most abundant DNA base lesions is 7,8-dihydro-8-oxoguanine (8-oxoG), which is repaired by the 8-oxoguanine DNA glycosylase 1 (OGG1)-initiated base excision repair (BER) (OGG1-BER) pathway. Studies have also demonstrated that in addition to its role in repairing oxidized purines, OGG1 has guanine nucleotide exchange factor activity when bound to 8-oxoG. In the present study, we tested the hypothesis that exposure to 8-oxoG, the specific product of OGG1-BER, induces functional changes of DCs. Supporting our hypothesis, transcriptome analysis revealed that in mouse lungs, out of 95 genes associated with DCs' function, 22 or 42 were significantly upregulated after a single or multiple intranasal 8-oxoG challenges, respectively. In a murine model of allergic airway inflammation, significantly increased serum levels of ovalbumin (OVA)-specific IgE antibodies were detected in mice sensitized via nasal challenges with OVA+8-oxoG compared to those challenged with OVA alone. Furthermore, exposure of primary human monocyte-derived DCs (moDC) to 8-oxoG base resulted in significantly enhanced expression of cell surface molecules (CD40, CD86, CD83, HLA-DQ) and augmented the secretion of pro-inflammatory mediators IL-6, TNF and IL-8, whereas it did not considerably influence the production of the anti-inflammatory cytokine IL-10. The stimulatory effects of 8-oxoG on human moDCs were abolished upon siRNA-mediated OGG1 depletion. Collectively, these data suggest that OGG1-BER-generated 8-oxoG base-driven cell signaling activates DCs, which may contribute to initiation of both the innate and adaptive immune responses under conditions of oxidative stress.
Collapse
Affiliation(s)
- Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary
| | - Máté Sütő
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary; Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary
| | - Aliz Varga
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary
| | - Eszter Boldizsár
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary; Doctoral School of Molecular Cellular and Immune Biology, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary
| | - István Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, Debrecen, H-4032, Hungary.
| |
Collapse
|
24
|
Houghton CA. Sulforaphane: Its "Coming of Age" as a Clinically Relevant Nutraceutical in the Prevention and Treatment of Chronic Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2716870. [PMID: 31737167 PMCID: PMC6815645 DOI: 10.1155/2019/2716870] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/24/2019] [Accepted: 09/06/2019] [Indexed: 12/17/2022]
Abstract
A growing awareness of the mechanisms by which phytochemicals can influence upstream endogenous cellular defence processes has led to intensified research into their potential relevance in the prevention and treatment of disease. Pharmaceutical medicine has historically looked to plants as sources of the starting materials for drug development; however, the focus of nutraceutical medicine is to retain the plant bioactive in as close to its native state as possible. As a consequence, the potency of a nutraceutical concentrate or an extract may be lower than required for significant gene expression. The molecular structure of bioactive phytochemicals to a large extent determines the molecule's bioavailability. Polyphenols are abundant in dietary phytochemicals, and extensive in vitro research has established many of the signalling mechanisms involved in favourably modulating human biochemical pathways. Such pathways are associated with core processes such as redox modulation and immune modulation for infection control and for downregulating the synthesis of inflammatory cytokines. Although the relationship between oxidative stress and chronic disease continues to be affirmed, direct-acting antioxidants such as vitamins A, C, and E, beta-carotene, and others have not yielded the expected preventive or therapeutic responses, even though several large meta-analyses have sought to evaluate the potential benefit of such supplements. Because polyphenols exhibit poor bioavailability, few of their impressive in vitro findings have been replicated in vivo. SFN, an aliphatic isothiocyanate, emerges as a phytochemical with comparatively high bioavailability. A number of clinical trials have demonstrated its ability to produce favourable outcomes in conditions for which there are few satisfactory pharmaceutical solutions, foreshadowing the potential for SFN as a clinically relevant nutraceutical. Although myrosinase-inert broccoli sprout extracts are widely available, there now exist myrosinase-active broccoli sprout supplements that yield sufficient SFN to match the doses used in clinical trials.
Collapse
|
25
|
Yagishita Y, Fahey JW, Dinkova-Kostova AT, Kensler TW. Broccoli or Sulforaphane: Is It the Source or Dose That Matters? Molecules 2019; 24:E3593. [PMID: 31590459 PMCID: PMC6804255 DOI: 10.3390/molecules24193593] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/02/2019] [Indexed: 12/18/2022] Open
Abstract
There is robust epidemiological evidence for the beneficial effects of broccoli consumption on health, many of them clearly mediated by the isothiocyanate sulforaphane. Present in the plant as its precursor, glucoraphanin, sulforaphane is formed through the actions of myrosinase, a β-thioglucosidase present in either the plant tissue or the mammalian microbiome. Since first isolated from broccoli and demonstrated to have cancer chemoprotective properties in rats in the early 1990s, over 3000 publications have described its efficacy in rodent disease models, underlying mechanisms of action or, to date, over 50 clinical trials examining pharmacokinetics, pharmacodynamics and disease mitigation. This review evaluates the current state of knowledge regarding the relationships between formulation (e.g., plants, sprouts, beverages, supplements), bioavailability and efficacy, and the doses of glucoraphanin and/or sulforaphane that have been used in pre-clinical and clinical studies. We pay special attention to the challenges for better integration of animal model and clinical studies, particularly with regard to selection of dose and route of administration. More effort is required to elucidate underlying mechanisms of action and to develop and validate biomarkers of pharmacodynamic action in humans. A sobering lesson is that changes in approach will be required to implement a public health paradigm for dispensing benefit across all spectrums of the global population.
Collapse
Affiliation(s)
- Yoko Yagishita
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Jed W Fahey
- Department of Medicine, Division of Clinical Pharmacology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
- Cullman Chemoprotection Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| | - Albena T Dinkova-Kostova
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
- Cullman Chemoprotection Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland DD1 9SY, UK.
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
- Cullman Chemoprotection Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Al-Harbi NO, Nadeem A, Ahmad SF, AlThagfan SS, Alqinyah M, Alqahtani F, Ibrahim KE, Al-Harbi MM. Sulforaphane treatment reverses corticosteroid resistance in a mixed granulocytic mouse model of asthma by upregulation of antioxidants and attenuation of Th17 immune responses in the airways. Eur J Pharmacol 2019; 855:276-284. [PMID: 31100413 DOI: 10.1016/j.ejphar.2019.05.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 12/28/2022]
Abstract
Sulforaphane has received considerable attention in recent years due to its antioxidant and anti-inflammatory properties. Its preventive effect in the inhibition of airway inflammation is known; however, whether it affects mixed granulocyte asthma (corticosteroid resistance phenotype) is largely undiscovered. Therefore, we assessed the effect of pharmacological activation of Nrf2, a redox-sensitive transcription factor, using sulforaphane in a mouse model of mixed granulocyte airway inflammation. Mice were sensitized and challenged with cockroach allergen extract (CE), and airway inflammatory parameters and markers of steroid resistance [Nrf2 activity, oxidant-antioxidant balance in airway epithelial cells (AECs)/lung, and IL-17A-related pathway in Th17 cells and dendritic cells (DCs)] were investigated. Our results show that sulforaphane administration reduced neutrophilic airway inflammation, myeloperoxidase (MPO) activity, and Th17 immune responses in a mixed granulocyte mouse model of asthma through Nrf2 activation. On the other hand, corticosteroid treatment decreased Th2/eosinophilic immune responses but had little on Th17/neutrophilic immune responses. However, combined treatment with both almost completely blocked both neutrophilic/eosinophilic and Th17/Th2 immune responses in the lung. Sulforaphane treatment led to induction of antioxidant enzymes (SOD, GPx) in AECs and pulmonary non-enzymatic antioxidants. Further, it led to reduction in inflammatory cytokines (IL-6/IL-23/IL-17A) in Th17 cells/CD11c + DCs during mixed granulocytic inflammation. Collectively, our study presents the evidence that activation of Nrf2 by sulforaphane reduces neutrophilic airway inflammation by upregulation of antioxidants and downregulation of inflammatory cytokines in airways. This is possibly the basis for reversal of corticosteroid resistance in this model. This shows the therapeutic potential of sulforaphane in mixed granulocyte asthma.
Collapse
Affiliation(s)
- Naif O Al-Harbi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Sheikh F Ahmad
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sultan S AlThagfan
- Clinical and Hospital Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed M Al-Harbi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
27
|
Lim CC, Hayes RB, Ahn J, Shao Y, Silverman DT, Jones RR, Thurston GD. Mediterranean Diet and the Association Between Air Pollution and Cardiovascular Disease Mortality Risk. Circulation 2019; 139:1766-1775. [PMID: 30700142 PMCID: PMC6453737 DOI: 10.1161/circulationaha.118.035742] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 12/13/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND Recent experimental evidence suggests that nutritional supplementation can blunt adverse cardiopulmonary effects induced by acute air pollution exposure. However, whether usual individual dietary patterns can modify the association between long-term air pollution exposure and health outcomes has not been previously investigated. We assessed, in a large cohort with detailed diet information at the individual level, whether a Mediterranean diet modifies the association between long-term exposure to ambient air pollution and cardiovascular disease mortality risk. METHODS The National Institutes of Health-American Association for Retired Persons Diet and Health Study, a prospective cohort (N=548 845) across 6 states and 2 cities in the United States and with a follow-up period of 17 years (1995-2011), was linked to estimates of annual average exposures to fine particulate matter and nitrogen dioxide at the residential census-tract level. The alternative Mediterranean Diet Index, which uses a 9-point scale to assess conformity with a Mediterranean-style diet, was constructed for each participant from information in cohort baseline dietary questionnaires. We evaluated mortality risks for cardiovascular disease, ischemic heart disease, cerebrovascular disease, or cardiac arrest associated with long-term air pollution exposure. Effect modification of the associations between exposure and the mortality outcomes by alternative Mediterranean Diet Index was examined via interaction terms. RESULTS For fine particulate matter, we observed elevated and significant associations with cardiovascular disease (hazard ratio [HR] per 10 μg/m3, 1.13; 95% CI, 1.08-1.18), ischemic heart disease (HR, 1.16; 95% CI, 1.10-1.23), and cerebrovascular disease (HR, 1.15; 95% CI, 1.03-1.28). For nitrogen dioxide, we found significant associations with cardiovascular disease (HR per 10 ppb, 1.06; 95% CI, 1.04-1.08) and ischemic heart disease (HR, 1.08; 95% CI, 1.05-1.11). Analyses indicated that Mediterranean diet modified these relationships, as those with a higher alternative Mediterranean Diet Index score had significantly lower rates of cardiovascular disease mortality associated with long-term air pollution exposure ( P-interaction<0.05). CONCLUSIONS A Mediterranean diet reduced cardiovascular disease mortality risk related to long-term exposure to air pollutants in a large prospective US cohort. Increased consumption of foods rich in antioxidant compounds may aid in reducing the considerable disease burden associated with ambient air pollution.
Collapse
Affiliation(s)
- Chris C. Lim
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
| | - Richard B. Hayes
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
- Department of Population Health, New York University School of Medicine, New York, NY
| | - Jiyoung Ahn
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
- Department of Population Health, New York University School of Medicine, New York, NY
| | - Yongzhao Shao
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
- Department of Population Health, New York University School of Medicine, New York, NY
| | - Debra T. Silverman
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health,
Rockville, MD
| | - Rena R. Jones
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health,
Rockville, MD
| | - George D. Thurston
- Department of Environmental Medicine, New York University School of Medicine, New York, NY
- Department of Population Health, New York University School of Medicine, New York, NY
| |
Collapse
|
28
|
Mazarakis N, Snibson K, Licciardi PV, Karagiannis TC. The potential use of l-sulforaphane for the treatment of chronic inflammatory diseases: A review of the clinical evidence. Clin Nutr 2019; 39:664-675. [PMID: 30954362 DOI: 10.1016/j.clnu.2019.03.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/07/2019] [Accepted: 03/15/2019] [Indexed: 12/15/2022]
Abstract
According to the World Health Organisation, 70% of all deaths globally can be attributed to chronic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, respiratory conditions, cardiovascular diseases, diabetes and cancer. Chronic inflammation has a significant impact on the quality of life of affected individuals with an increased risk of developing other chronic inflammatory diseases. Given the limitations of current pharmaceuticals, there is an intense research interest in identifying novel dietary interventions that can regulate and alleviate inflammation. A diet rich in cruciferous vegetables has been extensively studied for its immediate and long-term health benefits, particularly in the context of cardiovascular disease and cancer. Cruciferous vegetables contain the precursor glucoraphanin, which is hydrolysed upon consumption to form l-sulforaphane (LSF), the primary active compound that mediates potential cardio-protective and anti-carcinogenic effects. LSF has been shown to have beneficial effects in vitro and in animal studies through its classical antioxidant and anti-inflammatory properties, and more recently its chromatin modifying effects. This review discusses the clinical evidence to date in relation to the use of LSF in the context of chronic inflammatory diseases as well as provide key mechanistic insights for these effects.
Collapse
Affiliation(s)
- Nadia Mazarakis
- Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, Australia; Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Kenneth Snibson
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Tom C Karagiannis
- Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, Melbourne, VIC, Australia.
| |
Collapse
|
29
|
Abellán Á, Domínguez-Perles R, Moreno DA, García-Viguera C. Sorting out the Value of Cruciferous Sprouts as Sources of Bioactive Compounds for Nutrition and Health. Nutrients 2019; 11:E429. [PMID: 30791362 PMCID: PMC6412956 DOI: 10.3390/nu11020429] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/19/2022] Open
Abstract
Edible sprouts with germinating seeds of a few days of age are naturally rich in nutrients and other bioactive compounds. Among them, the cruciferous (Brassicaceae) sprouts stand out due to their high contents of glucosinolates (GLSs) and phenolic compounds. In order to obtain sprouts enriched in these phytochemicals, elicitation is being increasing used as a sustainable practice. Besides, the evidence regarding the bioavailability and the biological activity of these compounds after their dietary intake has also attracted growing interest in recent years, supporting the intake of the natural food instead of enriched ingredients or extracts. Also, there is a growing interest regarding their uses, consumption, and applications for health and wellbeing, in different industrial sectors. In this context, the present review aims to compile and update the available knowledge on the fundamental aspects of production, enrichment in composition, and the benefits upon consumption of diverse edible cruciferous sprouts, which are sources of phenolic compounds and glucosinolates, as well as the evidence on their biological actions in diverse pathophysiological situations and the molecular pathways involved.
Collapse
Affiliation(s)
- Ángel Abellán
- Phytochemistry and Healthy Foods Lab, Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS (CSIC), Campus Universitario de Espinardo 25, 30100 Murcia, Spain.
| | - Raúl Domínguez-Perles
- Phytochemistry and Healthy Foods Lab, Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS (CSIC), Campus Universitario de Espinardo 25, 30100 Murcia, Spain.
| | - Diego A Moreno
- Phytochemistry and Healthy Foods Lab, Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS (CSIC), Campus Universitario de Espinardo 25, 30100 Murcia, Spain.
| | - Cristina García-Viguera
- Phytochemistry and Healthy Foods Lab, Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS (CSIC), Campus Universitario de Espinardo 25, 30100 Murcia, Spain.
| |
Collapse
|
30
|
Cho HY, Miller-DeGraff L, Blankenship-Paris T, Wang X, Bell DA, Lih F, Deterding L, Panduri V, Morgan DL, Yamamoto M, Reddy AJ, Talalay P, Kleeberger SR. Sulforaphane enriched transcriptome of lung mitochondrial energy metabolism and provided pulmonary injury protection via Nrf2 in mice. Toxicol Appl Pharmacol 2018; 364:29-44. [PMID: 30529165 DOI: 10.1016/j.taap.2018.12.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022]
Abstract
Nrf2 is essential to antioxidant response element (ARE)-mediated host defense. Sulforaphane (SFN) is a phytochemical antioxidant known to affect multiple cellular targets including Nrf2-ARE pathway in chemoprevention. However, the role of SFN in non-malignant airway disorders remain unclear. To test if pre-activation of Nrf2-ARE signaling protects lungs from oxidant-induced acute injury, wild-type (Nrf2+/+) and Nrf2-deficient (Nrf2-/-) mice were given SFN orally or as standardized broccoli sprout extract diet (SBE) before hyperoxia or air exposure. Hyperoxia-induced pulmonary injury and oxidation indices were significantly reduced by SFN or SBE in Nrf2+/+ mice but not in Nrf2-/- mice. SFN upregulated a large cluster of basal lung genes that are involved in mitochondrial oxidative phosphorylation, energy metabolism, and cardiovascular protection only in Nrf2+/+ mice. Bioinformatic analysis elucidated ARE-like motifs on these genes. Transcript abundance of the mitochondrial machinery genes remained significantly higher after hyperoxia exposure in SFN-treated Nrf2+/+ mice than in SFN-treated Nrf2-/- mice. Nuclear factor-κB was suggested to be a central molecule in transcriptome networks affected by SFN. Minor improvement of hyperoxia-caused lung histopathology and neutrophilia by SFN in Nrf2-/- mice implies Nrf2-independent or alternate effector mechanisms. In conclusion, SFN is suggested to be as a preventive intervention in a preclinical model of acute lung injury by linking mitochondria and Nrf2. Administration of SFN alleviated acute lung injury-like pathogenesis in a Nrf2-dependent manner. Potential AREs in the SFN-inducible transcriptome for mitochondria bioenergetics provided a new insight into the downstream mechanisms of Nrf2-mediated pulmonary protection.
Collapse
Affiliation(s)
- Hye-Youn Cho
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| | - Laura Miller-DeGraff
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Terry Blankenship-Paris
- Comparative Medicine Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xuting Wang
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Douglas A Bell
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Fred Lih
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Leesa Deterding
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Vijayalakshmi Panduri
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Daniel L Morgan
- National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | | - Anita J Reddy
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Paul Talalay
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, MD 21205, USA
| | - Steven R Kleeberger
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
31
|
Palliyaguru DL, Yuan JM, Kensler TW, Fahey JW. Isothiocyanates: Translating the Power of Plants to People. Mol Nutr Food Res 2018; 62:e1700965. [PMID: 29468815 PMCID: PMC6226005 DOI: 10.1002/mnfr.201700965] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/13/2018] [Indexed: 12/19/2022]
Abstract
Isothiocyanates from cruciferous vegetables have been studied extensively in cells and in animals for their disease preventive and therapeutic effects. However, translating their utility to human populations has been both limited and challenging. Herein, clinical trials employing two isothiocyanates, sulforaphane (SFN; 1-isothiocyanato-4-(methylsulfinyl) butane) and phenethyl isothiocyanate (PEITC; 2-isothiocyanatoethylbenzene) that are isolated principally from broccoli and watercress, respectively, are summarized and discussed. Both of these compounds have been used in small human clinical trials, either within food matrices or as single agents, against a variety of diseases ranging from cancer to autism. Results suggest an opportunity to incorporate them, or more likely preparations derived from their source plants, into larger human disease mitigation efforts. The context for the applications of these compounds and plants in evidence-based food and nutritional policy is also evaluated.
Collapse
Affiliation(s)
- Dushani L Palliyaguru
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Thomas W Kensler
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jed W Fahey
- Departments of Medicine, Pharmacology and Molecular Sciences, International Health and Cullman Chemoprotection Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
32
|
Aqueous Extract of Glucoraphanin-Rich Broccoli Sprouts Inhibits Formation of Advanced Glycation End Products and Attenuates Inflammatory Reactions in Endothelial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:9823141. [PMID: 30174716 PMCID: PMC6106845 DOI: 10.1155/2018/9823141] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/27/2018] [Accepted: 08/01/2018] [Indexed: 11/21/2022]
Abstract
We have previously shown that sulforaphane not only inhibits formation of advanced glycation end products (AGEs) but also exerts anti-inflammatory effects on AGE-exposed human umbilical vein endothelial cells (HUVECs) and AGE-injected rat aortae. Here we examined the effects of aqueous extract of glucoraphanin-rich broccoli sprouts on formation of AGEs and then investigated whether the extract could attenuate inflammatory or oxidative stress reactions in tumor necrosis factor-alpha (TNF-α)- or AGE-exposed HUVECs. Fresh broccoli sprouts were homogenized in phosphate-buffered saline and filtered through a gauze. After centrifugation, clear extract was obtained. AGE formation was measured by enzyme-linked immunosorbent assay. Gene expression was evaluated by real-time reverse transcription-polymerase chain reaction. Reactive oxygen species (ROS) generation were measured using a fluorescent dye. Five percent broccoli sprout extract inhibited the formation of AGEs, reduced basal gene expressions of monocyte chemoattractant protein-1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1,) and receptor for AGEs (RAGE), and upregulated endothelial nitric oxide synthase (eNOS) mRNA levels in HUVECs. TNF-α upregulated MCP-1, ICAM-1, and RAGE mRNA levels in HUVECs, all of which were attenuated by the treatment with 1% broccoli sprout extract. Pretreatment of 1% broccoli sprout extract prevented the ROS generation in HUVECs evoked by AGEs. The present study demonstrates that sulforaphane-rich broccoli sprout extract could inhibit the AGE-RAGE axis and exhibit anti-inflammatory actions in HUVECs. Supplementation of sulforaphane-rich broccoli sprout extract may play a protective role against vascular injury.
Collapse
|
33
|
Patel B, Mann GE, Chapple SJ. Concerted redox modulation by sulforaphane alleviates diabetes and cardiometabolic syndrome. Free Radic Biol Med 2018; 122:150-160. [PMID: 29427794 DOI: 10.1016/j.freeradbiomed.2018.02.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023]
Abstract
Diabetes and cardiometabolic disorders such as hypertension and obesity are major risk factors for the development of cardiovascular disease, with a wealth of evidence suggesting that oxidative stress is linked to the initiation and pathogenesis of these disease processes. With yearly increases in the global incidence of cardiovascular diseases (CVD) and diabetes, numerous studies have focused on characterizing whether upregulating antioxidant defenses through exogenous antioxidants (e.g. vitamin E, vitamin C) or activation of endogenous defenses (e.g. the Nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant defense pathway) may be of benefit. The dietary isothiocyanate sulforaphane (SFN) is currently the subject of several clinical trials for a variety of disease states, including the evaluation of its therapeutic potential to ameliorate diabetic and cardiometabolic complications. SFN is a well characterized and potent Nrf2 inducer, however recent studies suggest its protective actions may be in part mediated by its modulation of various pro-inflammatory (e.g. Nuclear factor-kappa B (NFκB)) and metabolic (e.g. Peroxisome Proliferator-Activator Receptor Gamma (PPARγ)) signaling pathways. The focus of this review is to provide a detailed analysis of the known mechanisms by which SFN modulates Nrf2, NFκB and PPARγ signaling and crosstalk and to provide a critical evaluation of the evidence linking these transcriptional pathways with diabetic and cardiometabolic complications and SFN mediated cytoprotection. To allow comparison between rodent and human studies, we discuss the published bioavailability of SFN metabolites achieved in rodents and man in the context of Nrf2, NFκB and PPARγ signaling. Furthermore, we provide an update on the functional outcomes and implicated signaling pathways reported in recent clinical trials with SFN in Type 2 diabetic patients.
Collapse
Affiliation(s)
- Bijal Patel
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Giovanni E Mann
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Sarah J Chapple
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom.
| |
Collapse
|
34
|
Zhang Z, Bergan R, Shannon J, Slatore CG, Bobe G, Takata Y. The Role of Cruciferous Vegetables and Isothiocyanates for Lung Cancer Prevention: Current Status, Challenges, and Future Research Directions. Mol Nutr Food Res 2018; 62:e1700936. [PMID: 29663679 DOI: 10.1002/mnfr.201700936] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 04/06/2018] [Indexed: 01/07/2023]
Abstract
Lung cancer remains a leading cause of cancer-related deaths in the United States. Although smoking and air pollution exposure are primary risk factors of lung cancer, diet has also been reported to contribute to lung cancer risk. Cruciferous vegetables contain many bioactive compounds that alter the detoxification process of air-borne carcinogenic compounds and, thereby, may decrease lung cancer risk. In the meta-analysis of 31 observational studies, cruciferous vegetable intake is inversely associated with lung cancer risk (summary odds ratio/relative risk = 0.81 and 95% confidence interval = 0.74-0.89 for comparing the highest with lowest intake categories). More observational studies need to measure urinary isothiocyanate (ITC) concentrations and investigate their association with lung cancer risk in populations with relatively high intake of cruciferous vegetables. Current evidence is limited to two phase 2 clinical trials with incomplete reporting. Hence, more short-term clinical phase 2 trials need to examine effects of various amounts and types of cruciferous vegetables on biomarkers of risk and efficacy before a large phase 3 trial can be conducted to assess effects upon lung cancer risk. This would help further elucidate whether the inverse association observed with self-reported cruciferous vegetable intake is indeed due to ITC content or other bioactive compounds.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- OHSU-PSU School of Public Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Raymond Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jackilen Shannon
- OHSU-PSU School of Public Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Christopher G Slatore
- Health Services Research & Development and Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, OR, 97239, USA.,Department of Medicine and Department of Radiation Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Gerd Bobe
- Linus Pauling Institute, Department of Animal Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Yumie Takata
- College of Public Health and Human Sciences, School of Biological and Population Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
35
|
Fahey JW, Olson ME, Stephenson KK, Wade KL, Chodur GM, Odee D, Nouman W, Massiah M, Alt J, Egner PA, Hubbard WC. The Diversity of Chemoprotective Glucosinolates in Moringaceae (Moringa spp.). Sci Rep 2018; 8:7994. [PMID: 29789618 PMCID: PMC5964242 DOI: 10.1038/s41598-018-26058-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/03/2018] [Indexed: 01/04/2023] Open
Abstract
Glucosinolates (GS) are metabolized to isothiocyanates that may enhance human healthspan by protecting against a variety of chronic diseases. Moringa oleifera, the drumstick tree, produces unique GS but little is known about GS variation within M. oleifera, and even less in the 12 other Moringa species, some of which are very rare. We assess leaf, seed, stem, and leaf gland exudate GS content of 12 of the 13 known Moringa species. We describe 2 previously unidentified GS as major components of 6 species, reporting on the presence of simple alkyl GS in 4 species, which are dominant in M. longituba. We document potent chemoprotective potential in 11 of 12 species, and measure the cytoprotective activity of 6 purified GS in several cell lines. Some of the unique GS rank with the most powerful known inducers of the phase 2 cytoprotective response. Although extracts of most species induced a robust phase 2 cytoprotective response in cultured cells, one was very low (M. longituba), and by far the highest was M. arborea, a very rare and poorly known species. Our results underscore the importance of Moringa as a chemoprotective resource and the need to survey and conserve its interspecific diversity.
Collapse
Affiliation(s)
- Jed W Fahey
- Cullman Chemoprotection Center, Johns Hopkins University, Baltimore, Maryland, USA. .,Johns Hopkins University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Baltimore, Maryland, USA. .,Johns Hopkins University School of Medicine, Department of Pharmacology and Molecular Sciences, Baltimore, Maryland, USA. .,Johns Hopkins University Bloomberg School of Public Health, Department of International Health, Center for Human Nutrition, Baltimore, Maryland, USA.
| | - Mark E Olson
- Instituto de Biología, Universidad Nacional Autónoma de México, Tercer Circuito de Ciudad Universitaria, Ciudad de México, 04510, Mexico.,The International Moringa Germplasm Collection, Ejido de la Reforma Agraria, Jalisco, Mexico
| | - Katherine K Stephenson
- Cullman Chemoprotection Center, Johns Hopkins University, Baltimore, Maryland, USA.,Johns Hopkins University School of Medicine, Department of Pharmacology and Molecular Sciences, Baltimore, Maryland, USA
| | - Kristina L Wade
- Cullman Chemoprotection Center, Johns Hopkins University, Baltimore, Maryland, USA.,Johns Hopkins University School of Medicine, Department of Pharmacology and Molecular Sciences, Baltimore, Maryland, USA
| | - Gwen M Chodur
- Cullman Chemoprotection Center, Johns Hopkins University, Baltimore, Maryland, USA.,Johns Hopkins University Bloomberg School of Public Health, Department of International Health, Center for Human Nutrition, Baltimore, Maryland, USA.,Graduate Group in Nutritional Biology, UC Davis, Davis, California, USA
| | - David Odee
- Biotechnology Laboratory, Kenya Forestry Research Institute, Nairobi, Kenya
| | - Wasif Nouman
- Department of Forestry, Range, and Wildlife Management, Bahauddin Zakariya University, Multan, Pakistan
| | - Michael Massiah
- George Washington University, Department of Chemistry, Columbian College of Arts and Sciences, Washington DC, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Baltimore, Maryland, USA
| | - Patricia A Egner
- Johns Hopkins University Bloomberg School of Public Health, Department of Environmental Health and Engineering, Baltimore, Maryland, USA
| | - Walter C Hubbard
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Clinical Pharmacology, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Dinkova-Kostova AT, Fahey JW, Kostov RV, Kensler TW. KEAP1 and Done? Targeting the NRF2 Pathway with Sulforaphane. Trends Food Sci Technol 2017; 69:257-269. [PMID: 29242678 PMCID: PMC5725197 DOI: 10.1016/j.tifs.2017.02.002] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/12/2017] [Accepted: 02/14/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Since the re-discovery of sulforaphane in 1992 and the recognition of the bioactivity of this phytochemical, many studies have examined its mode of action in cells, animals and humans. Broccoli, especially as young sprouts, is a rich source of sulforaphane and broccoli-based preparations are now used in clinical studies probing efficacy in health preservation and disease mitigation. Many putative cellular targets are affected by sulforaphane although only one, KEAP1-NRF2 signaling, can be considered a validated target at this time. The transcription factor NRF2 is a master regulator of cell survival responses to endogenous and exogenous stressors. SCOPE AND APPROACH This review summarizes the chemical biology of sulforaphane as an inducer of NRF2 signaling and efficacy as an inhibitor of carcinogenesis. It also provides a summary of the current findings from clinical trials using a suite of broccoli sprout preparations on a series of short-term endpoints reflecting a diversity of molecular actions. KEY FINDINGS AND CONCLUSIONS Sulforaphane, as a pure chemical, protects against chemical-induced skin, oral, stomach, colon, lung and bladder carcinogenesis and in genetic models of colon and prostate carcinogenesis. In many of these settings the antitumorigenic efficacy of sulforaphane is dampened in Nrf2-disrupted animals. Broccoli preparations rich in glucoraphanin or sulforaphane exert demonstrable pharmacodynamic action in over a score of clinical trials. Measures of NRF2 pathway response and function are serving as guideposts for the optimization of dose, schedule and formulation as clinical trials with broccoli-based preparations become more commonplace and more rigorous in design and implementation.
Collapse
Affiliation(s)
- Albena T. Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jed W. Fahey
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Human Nutrition, Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rumen V. Kostov
- Jacqui Wood Cancer Centre, Division of Cancer Research, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W. Kensler
- Lewis B. and Dorothy Cullman Chemoprotection Center, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
37
|
Fahey JW, Wade KL, Wehage SL, Holtzclaw WD, Liu H, Talalay P, Fuchs E, Stephenson KK. Stabilized sulforaphane for clinical use: Phytochemical delivery efficiency. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600766] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/21/2016] [Accepted: 11/24/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Jed W. Fahey
- The Cullman Chemoprotection Center; Johns Hopkins University; Baltimore MD USA
- Division of Clinical Pharmacology; Department of Medicine; Johns Hopkins University School of Medicine; Baltimore MD USA
- Department of Pharmacology and Molecular Sciences; Johns Hopkins University School of Medicine; Baltimore MD USA
- Department of International Health; Center for Human Nutrition; Bloomberg School of Public Health; Johns Hopkins University; Baltimore MD USA
| | - Kristina L. Wade
- The Cullman Chemoprotection Center; Johns Hopkins University; Baltimore MD USA
- Department of Pharmacology and Molecular Sciences; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Scott L. Wehage
- The Cullman Chemoprotection Center; Johns Hopkins University; Baltimore MD USA
- Department of Pharmacology and Molecular Sciences; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Walter David Holtzclaw
- The Cullman Chemoprotection Center; Johns Hopkins University; Baltimore MD USA
- Department of Pharmacology and Molecular Sciences; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Hua Liu
- The Cullman Chemoprotection Center; Johns Hopkins University; Baltimore MD USA
- Department of Pharmacology and Molecular Sciences; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Paul Talalay
- The Cullman Chemoprotection Center; Johns Hopkins University; Baltimore MD USA
- Department of Pharmacology and Molecular Sciences; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Edward Fuchs
- Division of Clinical Pharmacology; Department of Medicine; Johns Hopkins University School of Medicine; Baltimore MD USA
| | - Katherine K. Stephenson
- The Cullman Chemoprotection Center; Johns Hopkins University; Baltimore MD USA
- Department of Pharmacology and Molecular Sciences; Johns Hopkins University School of Medicine; Baltimore MD USA
| |
Collapse
|
38
|
Wise RA, Holbrook JT, Criner G, Sethi S, Rayapudi S, Sudini KR, Sugar EA, Burke A, Thimmulappa R, Singh A, Talalay P, Fahey JW, Berenson CS, Jacobs MR, Biswal S. Lack of Effect of Oral Sulforaphane Administration on Nrf2 Expression in COPD: A Randomized, Double-Blind, Placebo Controlled Trial. PLoS One 2016; 11:e0163716. [PMID: 27832073 PMCID: PMC5104323 DOI: 10.1371/journal.pone.0163716] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/12/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND COPD patients have high pulmonary and systemic oxidative stress that correlates with severity of disease. Sulforaphane has been shown to induce expression of antioxidant genes via activation of a transcription factor, nuclear factor erythroid-2 related factor 2 (Nrf2). METHODS This parallel, placebo-controlled, phase 2, randomized trial was conducted at three US academic medical centers. Patients who met GOLD criteria for COPD and were able to tolerate bronchoscopies were randomly assigned (1:1:1) to receive placebo, 25 μmoles, or 150 μmoles sulforaphane daily by mouth for four weeks. The primary outcomes were changes in Nrf2 target gene expression (NQ01, HO1, AKR1C1 and AKR1C3) in alveolar macrophages and bronchial epithelial cells. Secondary outcomes included measures of oxidative stress and airway inflammation, and pulmonary function tests. RESULTS Between July 2011 and May 2013, 89 patients were enrolled and randomized. Sulforaphane was absorbed in the patients as evident from their plasma metabolite levels. Changes in Nrf2 target gene expression relative to baseline ranged from 0.79 to 1.45 and there was no consistent pattern among the three groups; the changes were not statistically significantly different from baseline. Changes in measures of inflammation and pulmonary function tests were not different among the groups. Sulforaphane was well tolerated at both dose levels. CONCLUSION Sulforaphane administered for four weeks at doses of 25 μmoles and 150 μmoles to patients with COPD did not stimulate the expression of Nrf2 target genes or have an effect on levels of other anti-oxidants or markers of inflammation. TRIAL REGISTRATION Clinicaltrials.gov: NCT01335971.
Collapse
Affiliation(s)
- Robert A. Wise
- Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Janet T. Holbrook
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Gerard Criner
- Temple University, Philadelphia, Pennsylvania, United States of America
| | - Sanjay Sethi
- University at Buffalo, SUNY and VA WNY Healthcare system, Buffalo, New York, United States of America
| | - Sobharani Rayapudi
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Kuladeep R. Sudini
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Elizabeth A. Sugar
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Alyce Burke
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Rajesh Thimmulappa
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Anju Singh
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Paul Talalay
- Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Jed W. Fahey
- Johns Hopkins University, School of Medicine, Baltimore, Maryland, United States of America
| | - Charles S. Berenson
- University at Buffalo, SUNY and VA WNY Healthcare system, Buffalo, New York, United States of America
| | - Michael R. Jacobs
- Temple University, Philadelphia, Pennsylvania, United States of America
| | - Shyam Biswal
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | | |
Collapse
|
39
|
Cho HY, Wang X, Li J, Bell DA, Kleeberger SR. Potential therapeutic targets in Nrf2-dependent protection against neonatal respiratory distress disease predicted by cDNA microarray analysis and bioinformatics tools. CURRENT OPINION IN TOXICOLOGY 2016; 1:125-133. [PMID: 28920101 DOI: 10.1016/j.cotox.2016.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hyperoxia exposure of newborn rodents has served as a model for bronchopulmonary dysplasia (BPD) phenotypes found in a sub-population of human premature infants. We previously demonstrated that Nrf2 modulates molecular events during saccular-to-alveolar lung maturation and also has a protective role in the pathogenesis of hyperoxia-induced acute lung injury, mortality, arrest of saccular-to-alveolar transition, and lung injury, using Nrf2-deficient and wild-type neonate mice. In this review, we describe how whole-genome transcriptome analyses can identify the means through which Nrf2 transcriptionally modulates organ injury and morphology, cellular growth/proliferation, vasculature development, and immune response during BPD-like pathogenesis. We illustrate how recently developed bioinformatics tools can be used to identify sets of Nrf2-dependently modulated genes in the BPD model, and elucidate direct Nrf2 downstream targets and chemicals/drugs that may act on them. These approaches will provide significant insights into promising therapeutic agents for Nrf2-dependent treatments of complications of preterm birth like BPD.
Collapse
Affiliation(s)
- Hye-Youn Cho
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Xuting Wang
- Genomic Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Jianying Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709.,Integrative Bioinformatics Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Douglas A Bell
- Genomic Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Steven R Kleeberger
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| |
Collapse
|
40
|
Jude J, Koziol-White C, Scala J, Yoo E, Jester W, Maute C, Dalton P, Panettieri R. Formaldehyde Induces Rho-Associated Kinase Activity to Evoke Airway Hyperresponsiveness. Am J Respir Cell Mol Biol 2016; 55:542-553. [PMID: 27149505 DOI: 10.1165/rcmb.2015-0254oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Formaldehyde, a common indoor air pollutant, exacerbates asthma and synergizes with allergen to induce airway hyperresponsiveness (AHR) in animal models. The mechanisms mediating formaldehyde-induced AHR remain poorly understood. We posit that formaldehyde modulates agonist-induced contractile response of human airway smooth muscle (HASM) cells to elicit AHR. HASM cells were exposed to formaldehyde or vehicle and agonist-induced intracellular Ca2+ ([Ca2+]i) and myosin light-chain phosphatase (MYPT1) phosphorylation were determined. Air-liquid interface-differentiated human bronchial epithelial (HBE) cells were exposed to formaldehyde or vehicle and cocultured with HASM cells. Agonist-induced [Ca2+]i and MYPT1 phosphorylation were determined in the cocultured HASM cells. Precision-cut human lung slices were exposed to PBS or varying concentrations of formaldehyde, and then carbachol-induced airway narrowing was determined 24 hours after exposure. HASM cells were transfected with nontargeting or nuclear factor erythroid-derived 2, like 2 (Nrf-2)-targeting small interfering RNA and exposed to formaldehyde or vehicle, followed by determination of antioxidant response (quinone oxido-reductase 1 and thioredoxin 1) and basal and agonist-induced MYPT1 phosphorylation. Formaldehyde enhanced the basal Rho-kinase activity and MYPT1 phosphorylation with little effect on agonist-induced [Ca2+]i in HASM cells. Formaldehyde induced Nrf-2-dependent antioxidant response in HASM cells, although the MYPT1 phosphorylation was independent of Nrf-2 induction. Although HBE cells exposed to formaldehyde had little effect on agonist-induced [Ca2+]i or MYPT1 phosphorylation in cocultured HASM cells, formaldehyde enhanced carbachol-induced airway responsiveness in precision-cut human lung slices. In conclusion, formaldehyde induces phosphorylation of the regulatory subunit of MYPT1, independent of formaldehyde-induced Nrf-2 activation in HASM cells. The findings suggest that the Rho kinase-dependent Ca2+ sensitization pathway plays a role in formaldehyde-induced AHR.
Collapse
Affiliation(s)
- Joseph Jude
- 1 Rutgers Institute for Translational Medicine & Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and
| | - Cynthia Koziol-White
- 1 Rutgers Institute for Translational Medicine & Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and
| | - Jacqueline Scala
- 1 Rutgers Institute for Translational Medicine & Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and
| | - Edwin Yoo
- 1 Rutgers Institute for Translational Medicine & Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and
| | - William Jester
- 1 Rutgers Institute for Translational Medicine & Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and
| | | | - Pamela Dalton
- 2 Monell Chemical Senses Center, Philadelphia, Pennsylvania
| | - Reynold Panettieri
- 1 Rutgers Institute for Translational Medicine & Science, Child Health Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; and
| |
Collapse
|
41
|
Xu Z, Wang S, Ji H, Zhang Z, Chen J, Tan Y, Wintergerst K, Zheng Y, Sun J, Cai L. Broccoli sprout extract prevents diabetic cardiomyopathy via Nrf2 activation in db/db T2DM mice. Sci Rep 2016; 6:30252. [PMID: 27457280 PMCID: PMC4960533 DOI: 10.1038/srep30252] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/01/2016] [Indexed: 12/17/2022] Open
Abstract
To develop a clinic-relevant protocol for systemic up-regulation of NFE2-related factor 2 (Nrf2) to prevent diabetic cardiomyopathy (DCM), male db/db and age-matched wild-type (WT) mice were given sulforaphane (SFN, an Nrf2 activator) and its natural source, broccoli sprout extract (BSE) by gavage every other day for 3 months, with four groups: vehicle (0.1 ml/10 g), BSE-low dose (estimated SFN availability at 0.5 mg/kg), BSE-high dose (estimated SFN availability at 1.0 mg/kg), and SFN (0.5 mg/kg). Cardiac function and pathological changes (hypertrophy, fibrosis, inflammation and oxidative damage) were assessed by echocardiography and histopathological examination along with Western blot and real-time PCR, respectively. Both BSE and SFN significantly prevented diabetes-induced cardiac dysfunction, hypertrophy and fibrosis. Mechanistically, BSE, like SFN, significantly up-regulated Nrf2 transcriptional activity, evidenced by the increased Nrf2 nuclear accumulation and its downstream gene expression. This resulted in a significant prevention of cardiac oxidative damage and inflammation. For all these preventive effects, BSE at high dose provided a similar effect as did SFN. These results indicated that BSE at high dose prevents DCM in a manner congruent with SFN treatment. Therefore, it suggests that BSE could potentially be used as a natural and safe treatment against DCM via Nrf2 activation.
Collapse
Affiliation(s)
- Zheng Xu
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, China.,Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, USA
| | - Shudong Wang
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, China.,Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, USA
| | - Honglei Ji
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, China
| | - Zhiguo Zhang
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, China
| | - Jing Chen
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, USA
| | - Yi Tan
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, USA.,Wendy Novak Diabetes Care Center, University of Louisville, Louisville, KY, USA
| | - Kupper Wintergerst
- Wendy Novak Diabetes Care Center, University of Louisville, Louisville, KY, USA.,Division of Endocrinology, Department of Pediatrics, the University of Louisville, Louisville, KY, USA
| | - Yang Zheng
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, China
| | - Jian Sun
- Cardiovascular Center, the First Hospital of Jilin University, Changchun, China
| | - Lu Cai
- Kosair Children's Hospital Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY, USA.,Wendy Novak Diabetes Care Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
42
|
Duran CG, Burbank AJ, Mills KH, Duckworth HR, Aleman MM, Kesic MJ, Peden DB, Pan Y, Zhou H, Hernandez ML. A proof-of-concept clinical study examining the NRF2 activator sulforaphane against neutrophilic airway inflammation. Respir Res 2016; 17:89. [PMID: 27450419 PMCID: PMC4957339 DOI: 10.1186/s12931-016-0406-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/14/2016] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Sulforaphane (SFN), a naturally occurring isothiocyanate found in cruciferous vegetables, is implicated as a possible therapy for airway inflammation via induction of the transcription factor NF-E2-related factor 2 (NRF2). In this proof-of-concept clinical study, we show that supplementation of SFN with broccoli sprout homogenate in healthy human subjects did not induce expression of antioxidant genes or protect against neutrophilic airway inflammation in an ozone-exposure model. Therefore, dietary sulforaphane supplementation is not a promising candidate for larger scale clinical trials targeting airway inflammation. TRIAL REGISTRATION NCT01625130 . Registered 19 June, 2012.
Collapse
Affiliation(s)
- Charity G Duran
- UNC Center for Environmental Medicine, Asthma, & Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Allison J Burbank
- UNC Center for Environmental Medicine, Asthma, & Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine H Mills
- UNC Center for Environmental Medicine, Asthma, & Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather R Duckworth
- UNC Center for Environmental Medicine, Asthma, & Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maria M Aleman
- UNC Center for Environmental Medicine, Asthma, & Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew J Kesic
- Department of Physical Therapy, Methodist University, Fayetteville, NC, USA
| | - David B Peden
- UNC Center for Environmental Medicine, Asthma, & Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yinghao Pan
- Department of Biostatistics, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Haibo Zhou
- Department of Biostatistics, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michelle L Hernandez
- UNC Center for Environmental Medicine, Asthma, & Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Division of Allergy, Immunology & Rheumatology, UNC School of Medicine, 104 Mason Farm Road, CB #7310, Chapel Hill, NC, 27599-7310, USA.
| |
Collapse
|
43
|
Li S, Williams G, Guo Y. Health benefits from improved outdoor air quality and intervention in China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2016; 214:17-25. [PMID: 27061471 DOI: 10.1016/j.envpol.2016.03.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/25/2016] [Indexed: 06/05/2023]
Abstract
China is at its most critical stage of outdoor air quality management. In order to prevent further deterioration of air quality and to protect human health, the Chinese government has made a series of attempts to reduce ambient air pollution. Unlike previous literature reviews on the widespread hazards of air pollution on health, this review article firstly summarized the existing evidence of human health benefits from intermittently improved outdoor air quality and intervention in China. Contents of this paper provide concrete and direct clue that improvement in outdoor air quality generates various health benefits in China, and confirm from a new perspective that it is worthwhile for China to shift its development strategy from economic growth to environmental economic sustainability. Greater emphasis on sustainable environment design, consistently strict regulatory enforcement, and specific monitoring actions should be regarded in China to decrease the health risks and to avoid long-term environmental threats.
Collapse
Affiliation(s)
- Shanshan Li
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia.
| | - Gail Williams
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| | - Yuming Guo
- School of Public Health, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
44
|
Chen J, Wang S, Luo M, Zhang Z, Dai X, Kong M, Cai L, Wang Y, Shi B, Tan Y. From the Cover: Zinc Deficiency Worsens and Supplementation Prevents High-Fat Diet Induced Vascular Inflammation, Oxidative Stress, and Pathological Remodeling. Toxicol Sci 2016; 153:124-36. [DOI: 10.1093/toxsci/kfw110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
45
|
Dietary and pharmacological intervention to mitigate the cardiopulmonary effects of air pollution toxicity. Biochim Biophys Acta Gen Subj 2016; 1860:2891-8. [PMID: 27189803 DOI: 10.1016/j.bbagen.2016.05.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/06/2016] [Accepted: 05/12/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Exposure to air pollution contributes importantly to excess morbidity and mortality. And while regulatory actions under the "Clean Air Act" have saved millions of lives by improving air quality, there are still millions of people in the U.S. who live in areas where particulate air pollution (PM) levels exceed the U.S. Environmental Protection Agency's National Ambient Air Quality Standards. Therefore, apart from such localities working to attain such standards the protection of the health of public and in particular those at high risk might benefit from interventional strategies that would ameliorate air pollution's adverse health effects. Because inflammation and oxidative stress appear to mediate the health effects of air pollution, one interventional approach to consider is the use of dietary supplementation or medication with anti-inflammatory or antioxidant properties to block the biological responses that initiate the pathophysiological process that culminates in adverse health effects. SCOPE OF REVIEW This article reviews the capability of dietary supplementation, such as antioxidant vitamins, polyunsaturated fatty acids, and medications as a strategy to mitigate air pollution-induced subclinical cardiopulmonary effects. MAJOR CONCLUSIONS Antioxidant vitamins C and E protect the lungs against short-term ozone and PM exposure. Polyunsaturated fatty acids, such as fish oil and olive oil appear to offer protection against short-term air pollution-induced adverse cardiovascular responses. GENERAL SIGNIFICANCE Taking dietary supplements or medications with antioxidant or anti-inflammatory properties has the potential to provide at least partial protection against air pollution-induced adverse health effects in those individuals who are known to be most susceptible, namely those with pre-existing respiratory and cardiovascular diseases. This article is part of a Special Issue entitled Air Pollution, edited by Wenjun Ding, Andrew J. Ghio and Weidong Wu.
Collapse
|
46
|
Sudini K, Diette GB, Breysse PN, McCormack MC, Bull D, Biswal S, Zhai S, Brereton N, Peng RD, Matsui EC. A Randomized Controlled Trial of the Effect of Broccoli Sprouts on Antioxidant Gene Expression and Airway Inflammation in Asthmatics. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2016; 4:932-40. [PMID: 27130714 DOI: 10.1016/j.jaip.2016.03.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/13/2016] [Accepted: 03/29/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Broccoli sprouts (BS) are the richest source of sulforaphane (SFN), which is a potent inducer of phase II enzymes, which play a critical role in preventing oxidative stress (OS) and inflammation. OBJECTIVES The objective of this study was to determine if ingestion of whole BS improves airway inflammatory and physiologic outcomes, and OS in adults with asthma and allergic sensitization to an indoor allergen. METHODS The study is a double-blind, placebo-controlled, randomized trial to compare the effects of BS with placebo (alfalfa sprouts [AS]) on airway inflammation and markers of OS. Forty adults (aged 18-50 years) were randomized to eat either (a) 100 g of BS daily or (b) 100 g of AS daily for 3 days. Fractional exhaled nitric oxide (FENO), forced expiratory volume 1, nasal epithelial and PBMC gene expression, inflammatory and OS biomarkers, and symptoms were assessed both before and after ingestion of the sprouts. The primary outcome variable was the change in FENO. Secondary outcome measures included rhinitis and asthma symptoms, lung function, and OS and inflammatory biomarkers. RESULTS BS ingestion for 3 consecutive days did not reduce FENO, despite resulting in a marked increase in serum SFN concentrations (21 vs 22 parts per billion, P = .76). Furthermore, BS consumption did not induce cytoprotective antioxidant genes in either PBMCs or nasal epithelial cells, reduce OS and inflammatory markers, or improve lung function. CONCLUSIONS Ingestion of whole BS for 3 days does not appear to improve eosinophilic pulmonary inflammation, inflammatory and OS biomarkers, or clinical features of asthma among atopic adults with asthma despite resulting in a marked increase in serum SFN levels.
Collapse
Affiliation(s)
- Kuladeep Sudini
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Md
| | - Gregory B Diette
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Patrick N Breysse
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD
| | - Meredith C McCormack
- Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Deborah Bull
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Shyam Biswal
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Md; Division of Pulmonary and Critical Care, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Shuyan Zhai
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Nga Brereton
- Institute for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Roger D Peng
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md
| | - Elizabeth C Matsui
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
47
|
Abbas A, Hall JA, Patterson WL, Ho E, Hsu A, Al-Mulla F, Georgel PT. Sulforaphane modulates telomerase activity via epigenetic regulation in prostate cancer cell lines. Biochem Cell Biol 2016; 94:71-81. [DOI: 10.1139/bcb-2015-0038] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Epidemiologic studies have revealed that diets rich in sulforaphane (SFN), an isothiocyanate present in cruciferous vegetables, are associated with a marked decrease in prostate cancer incidence. The chemo-preventive role of SFN is associated with its histone de-acetylase inhibitor activity. However, the effect of SFN on chromatin composition and dynamic folding, especially in relation to HDAC inhibitor activity, remains poorly understood. In this study, we found that SFN can inhibit the expression and activity of human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, in 2 prostate cancer cell lines. This decrease in gene expression is correlated with SFN-induced changes in chromatin structure and composition. The SFN-mediated changes in levels of histone post-translational modifications, more specifically acetylation of histone H3 lysine 18 and di-methylation of histone H3 lysine 4, 2 modifications linked with high risk of prostate cancer recurrence, were associated with regulatory elements within the hTERT promoter region. Chromatin condensation may also play a role in SFN-mediated hTERT repression, since expression and recruitment of MeCP2, a known chromatin compactor, were altered in SFN treated prostate cancer cells. Chromatin immuno-precipitation (ChIP) of MeCP2 showed enrichment over regions of the hTERT promoter with increased nucleosome density. These combined results strongly support a role for SFN in the mediation of epigenetic events leading to the repression of hTERT in prostate cancer cells. This ability of SFN to modify chromatin composition and structure associated with target gene expression provides a new model by which dietary phytochemicals may exert their chemoprevention activity.
Collapse
Affiliation(s)
- Ata Abbas
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
| | - J. Adam Hall
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| | - William L. Patterson
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| | - Emily Ho
- Oregon State University, School of Biological and Population Health Sciences, Linus Pauling Institute, Corvallis, OR, USA
| | - Anna Hsu
- Oregon State University, School of Biological and Population Health Sciences, Linus Pauling Institute, Corvallis, OR, USA
| | - Fahd Al-Mulla
- Kuwait University, Health Sciences Center, Faculty of Medicine, Molecular Pathology Unit, Kuwait City, Kuwait
| | - Philippe T. Georgel
- Department of Biological Sciences, Marshall University, 1 John Marshall Drive, Huntington, WV 25755, USA
- Cell Differentiation and Development Center, Marshall University, Huntington, WV, USA
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, Huntington, WV, USA
| |
Collapse
|
48
|
Müller L, Meyer M, Bauer RN, Zhou H, Zhang H, Jones S, Robinette C, Noah TL, Jaspers I. Effect of Broccoli Sprouts and Live Attenuated Influenza Virus on Peripheral Blood Natural Killer Cells: A Randomized, Double-Blind Study. PLoS One 2016; 11:e0147742. [PMID: 26820305 PMCID: PMC4731143 DOI: 10.1371/journal.pone.0147742] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/05/2016] [Indexed: 12/25/2022] Open
Abstract
Enhancing antiviral host defense responses through nutritional supplementation would be an attractive strategy in the fight against influenza. Using inoculation with live attenuated influenza virus (LAIV) as an infection model, we have recently shown that ingestion of sulforaphane-containing broccoli sprout homogenates (BSH) reduces markers of viral load in the nose. To investigate the systemic effects of short-term BSH supplementation in the context of LAIV-inoculation, we examined peripheral blood immune cell populations in non-smoking subjects from this study, with a particular focus on NK cells. We carried out a randomized, double-blinded, placebo-controlled study measuring the effects of BSH (N = 13) or placebo (alfalfa sprout homogenate, ASH; N = 16) on peripheral blood mononuclear cell responses to a standard nasal vaccine dose of LAIV in healthy volunteers. Blood was drawn prior to (day-1) and post (day2, day21) LAIV inoculation and analyzed for neutrophils, monocytes, macrophages, T cells, NKT cells, and NK cells. In addition, NK cells were enriched, stimulated, and assessed for surface markers, intracellular markers, and cytotoxic potential by flow cytometry. Overall, LAIV significantly reduced NKT (day2 and day21) and T cell (day2) populations. LAIV decreased NK cell CD56 and CD158b expression, while significantly increasing CD16 expression and cytotoxic potential (on day2). BSH supplementation further increased LAIV-induced granzyme B production (day2) in NK cells compared to ASH and in the BSH group granzyme B levels appeared to be negatively associated with influenza RNA levels in nasal lavage fluid cells. We conclude that nasal influenza infection may induce complex changes in peripheral blood NK cell activation, and that BSH increases virus-induced peripheral blood NK cell granzyme B production, an effect that may be important for enhanced antiviral defense responses. Trial Registration: ClinicalTrials.gov NCT01269723
Collapse
Affiliation(s)
- Loretta Müller
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- University Children’s Hospital Basel, Basel, Switzerland
| | - Megan Meyer
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Rebecca N. Bauer
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatric Allergy and Immunology, Stanford University, Stanford, California, United States of America
| | - Haibo Zhou
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Hongtao Zhang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Shannon Jones
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Carole Robinette
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Terry L. Noah
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Ilona Jaspers
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
49
|
Nutritional Solutions to Reduce Risks of Negative Health Impacts of Air Pollution. Nutrients 2015; 7:10398-416. [PMID: 26690474 PMCID: PMC4690091 DOI: 10.3390/nu7125539] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/22/2015] [Accepted: 12/02/2015] [Indexed: 12/20/2022] Open
Abstract
Air pollution worldwide has been associated with cardiovascular and respiratory morbidity and mortality, particularly in urban settings with elevated concentrations of primary pollutants. Air pollution is a very complex mixture of primary and secondary gases and particles, and its potential to cause harm can depend on multiple factors—including physical and chemical characteristics of pollutants, which varies with fine-scale location (e.g., by proximity to local emission sources)—as well as local meteorology, topography, and population susceptibility. It has been hypothesized that the intake of anti-oxidant and anti-inflammatory nutrients may ameliorate various respiratory and cardiovascular effects of air pollution through reductions in oxidative stress and inflammation. To date, several studies have suggested that some harmful effects of air pollution may be modified by intake of essential micronutrients (such as B vitamins, and vitamins C, D, and E) and long-chain polyunsaturated fatty acids. Here, we review the existing literature related to the potential for nutrition to modify the health impacts of air pollution, and offer a framework for examining these interactions.
Collapse
|
50
|
Jackson SJT, Singletary KW, Murphy LL, Venema RC, Young AJ. Phytonutrients Differentially Stimulate NAD(P)H:Quinone Oxidoreductase, Inhibit Proliferation, and Trigger Mitotic Catastrophe in Hepa1c1c7 Cells. J Med Food 2015; 19:47-53. [PMID: 26623679 DOI: 10.1089/jmf.2015.0079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Phytonutrients have rapidly emerged as natural food chemicals possessing multifaceted biological actions that may support beneficial health outcomes. Among the vast array of phytonutrients currently being studied, sulforaphane, curcumin, quercetin, and resveratrol have been frequently reported to stimulate the expression of endogenous detoxification enzymes and may thereby facilitate the neutralization of otherwise harmful environmental agents. Some of these same phytonutrients, however, have also been implicated in disrupting normal cell proliferation and hence may possess toxic properties in and of themselves. In this study, we characterize the respective minimum threshold concentrations of the aforementioned phytonutrients in Hepa1c1c7 cells that stimulate NAD(P)H quinone oxidoreductase (NQO1), a key enzyme in the hepatic neutralization of menadione, other biological oxidants, and some environmental carcinogens. Moreover, our findings demonstrate that relatively low concentrations of either sulforaphane or curcumin significantly (P < .05) increase NQO1 protein expression and activity without triggering G2/M cell cycle arrest or mitotic catastrophe. The minimal quercetin concentration inducing NQO1, however, was 100-fold higher than that which disrupted mitosis. Also, while resveratrol modestly stimulated NQO1, the minimally effective resveratrol concentration concomitantly induced evidence of cellular apoptosis. Taken together, these findings indicate that only particular phytonutrients are likely efficacious in upregulating NQO1 activity without also leading to hepatic cytotoxicity.
Collapse
Affiliation(s)
- Steven J T Jackson
- 1 U.S. Army Aeromedical Research Laboratory, Aircrew Health and Performance Division , Fort Rucker, Alabama, USA
| | - Keith W Singletary
- 2 Department of Food Science and Human Nutrition, University of Illinois , Urbana, Illinois, USA
| | - Laura L Murphy
- 3 Department of Physiology, Southern Illinois University , Carbondale, Illinois, USA
| | - Richard C Venema
- 4 Medical College of Georgia, Vascular Biology Center, Georgia Regents University , Augusta, Georgia , USA
| | - Andrew J Young
- 5 Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine , Natick, Massachusetts, USA
| |
Collapse
|