1
|
Farooqui AA, Farooqui T. Phospholipids, Sphingolipids, and Cholesterol-Derived Lipid Mediators and Their Role in Neurological Disorders. Int J Mol Sci 2024; 25:10672. [PMID: 39409002 PMCID: PMC11476704 DOI: 10.3390/ijms251910672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Neural membranes are composed of phospholipids, sphingolipids, cholesterol, and proteins. In response to cell stimulation or injury, the metabolism of lipids generates various lipid mediators, which perform many cellular functions. Thus, phospholipids release arachidonic acid or docosahexaenoic acid from the sn-2 position of the glycerol moiety by the action of phospholipases A2. Arachidonic acid is a precursor for prostaglandins, leukotrienes, thromboxane, and lipoxins. Among these mediators, prostaglandins, leukotrienes, and thromboxane produce neuroinflammation. In contrast, lipoxins produce anti-inflammatory and pro-resolving effects. Prostaglandins, leukotrienes, and thromboxane are also involved in cell proliferation, differentiation, blood clotting, and blood vessel permeability. In contrast, DHA-derived lipid mediators are called specialized pro-resolving lipid metabolites (SPMs). They include resolvins, protectins, and maresins. These mediators regulate immune function by producing anti-inflammatory, pro-resolving, and cell protective effects. Sphingolipid-derived metabolites are ceramide, ceramide1-phosphate, sphingosine, and sphingosine 1 phosphate. They regulate many cellular processes, including enzyme activities, cell migration and adhesion, inflammation, and immunity. Cholesterol is metabolized into hydroxycholesterols and 7-ketocholesterol, which not only disrupts membrane fluidity, but also promotes inflammation, oxidative stress, and apoptosis. These processes lead to cellular damage.
Collapse
Affiliation(s)
| | - Tahira Farooqui
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
2
|
Díaz M, Valdés-Baizabal C, de Pablo DP, Marin R. Age-Dependent Changes in Nrf2/Keap1 and Target Antioxidant Protein Expression Correlate to Lipoxidative Adducts, and Are Modulated by Dietary N-3 LCPUFA in the Hippocampus of Mice. Antioxidants (Basel) 2024; 13:206. [PMID: 38397804 PMCID: PMC10886099 DOI: 10.3390/antiox13020206] [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: 10/22/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The brain has a high metabolism rate that may generate reactive oxygen and nitrogen species. Consequently, nerve cells require highly efficient antioxidant defenses in order to prevent a condition of deleterious oxidative stress. This is particularly relevant in the hippocampus, a highly complex cerebral area involved in processing superior cognitive functions. Most current evidence points to hippocampal oxidative damage as a causal effect for neurodegenerative disorders, especially Alzheimer's disease. Nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 (Nrf2/Keap1) is a master key for the transcriptional regulation of antioxidant and detoxifying systems. It is ubiquitously expressed in brain areas, mainly supporting glial cells. In the present study, we have analyzed the relationships between Nrf2 and Keap1 isoforms in hippocampal tissue in response to aging and dietary long-chain polyunsaturated fatty acids (LCPUFA) supplementation. The possible involvement of lipoxidative and nitrosative by-products in the dynamics of the Nrf2/Keap1 complex was examined though determination of protein adducts, namely malondialdehyde (MDA), 4-hydroxynonenal (HNE), and 3-nitro-tyrosine (NTyr) under basal conditions. The results were correlated to the expression of target proteins heme-oxygenase-1 (HO-1) and glutathione peroxidase 4 (GPx4), whose expressions are known to be regulated by Nrf2/Keap1 signaling activation. All variables in this study were obtained simultaneously from the same preparations, allowing multivariate approaches. The results demonstrate a complex modification of the protein expression patterns together with the formation of adducts in response to aging and diet supplementation. Both parameters exhibited a strong interaction. Noticeably, LCPUFA supplementation to aged animals restored the Nrf2/Keap1/target protein patterns to the status observed in young animals, therefore driving a "rejuvenation" of hippocampal antioxidant defense.
Collapse
Affiliation(s)
- Mario Díaz
- Department of Physics, Faculty of Sciences, University of La Laguna, 38200 Tenerife, Spain
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
| | - Catalina Valdés-Baizabal
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Daniel Pereda de Pablo
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Raquel Marin
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
- Associate Research Unit ULL-CSIC “Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases”, 38200 Tenerife, Spain
| |
Collapse
|
3
|
Borgonovi SM, Iametti S, Di Nunzio M. Docosahexaenoic Acid as Master Regulator of Cellular Antioxidant Defenses: A Systematic Review. Antioxidants (Basel) 2023; 12:1283. [PMID: 37372014 DOI: 10.3390/antiox12061283] [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: 05/18/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Docosahexaenoic acid (DHA) is a polyunsaturated fatty acid that benefits the prevention of chronic diseases. Due to its high unsaturation, DHA is vulnerable to free radical oxidation, resulting in several unfavorable effects, including producing hazardous metabolites. However, in vitro and in vivo investigations suggest that the relationship between the chemical structure of DHA and its susceptibility to oxidation may not be as clear-cut as previously thought. Organisms have developed a balanced system of antioxidants to counteract the overproduction of oxidants, and the nuclear factor erythroid 2-related factor 2 (Nrf2) is the key transcription factor identified for transmitting the inducer signal to the antioxidant response element. Thus, DHA might preserve the cellular redox status promoting the transcriptional regulation of cellular antioxidants through Nrf2 activation. Here, we systematically summarize the research on the possible role of DHA in controlling cellular antioxidant enzymes. After the screening process, 43 records were selected and included in this review. Specifically, 29 studies related to the effects of DHA in cell cultures and 15 studies concerned the effects of consumption or treatment with DHA in animal. Despite DHA's promising and encouraging effects at modulating the cellular antioxidant response in vitro/in vivo, some differences observed among the reviewed studies may be accounted for by the different experimental conditions adopted, including the time of supplementation/treatment, DHA concentration, and cell culture/tissue model. Moreover, this review offers potential molecular explanations for how DHA controls cellular antioxidant defenses, including involvement of transcription factors and the redox signaling pathway.
Collapse
Affiliation(s)
- Sara Margherita Borgonovi
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| | - Stefania Iametti
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| | - Mattia Di Nunzio
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Via Celoria 2, 20133 Milan, Italy
| |
Collapse
|
4
|
Davinelli S, Medoro A, Intrieri M, Saso L, Scapagnini G, Kang JX. Targeting NRF2-KEAP1 axis by Omega-3 fatty acids and their derivatives: Emerging opportunities against aging and diseases. Free Radic Biol Med 2022; 193:736-750. [PMID: 36402440 DOI: 10.1016/j.freeradbiomed.2022.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/01/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
The transcription factor NRF2 and its endogenous inhibitor KEAP1 play a crucial role in the maintenance of cellular redox homeostasis by regulating the gene expression of diverse networks of antioxidant, anti-inflammatory, and detoxification enzymes. Therefore, activation of NRF2 provides cytoprotection against numerous pathologies, including age-related diseases. An age-associated loss of NRF2 function may be a key driving force behind the aging phenotype. Recently, numerous NRF2 inducers have been identified and some of them are promising candidates to restore NRF2 transcriptional activity during aging. Emerging evidence indicates that omega-3 (n-3) polyunsaturated fatty acids (PUFAs) and their electrophilic derivatives may trigger a protective response via NRF2 activation, rescuing or maintaining cellular redox homeostasis. In this review, we provide an overview of the NRF2-KEAP1 system and its dysregulation in aging cells. We also summarize current studies on the modulatory role of n-3 PUFAs as potential agents to prevent multiple chronic diseases and restore the age-related impairment of NRF2 function.
Collapse
Affiliation(s)
- Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Alessandro Medoro
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Mariano Intrieri
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy.
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
4-Hydroxynonenal Modulates Blood-Brain Barrier Permeability In Vitro through Changes in Lipid Composition and Oxidative Status of Endothelial Cells and Astrocytes. Int J Mol Sci 2022; 23:ijms232214373. [PMID: 36430852 PMCID: PMC9698020 DOI: 10.3390/ijms232214373] [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: 10/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Blood brain barrier (BBB) is a dynamic interface responsible for proper functioning of brain, but also a major obstacle for effective treatment of neurological diseases. Increased levels of free radicals, in high ferrous and high lipid content surrounding, induce lipid peroxidation, leading to production of 4-hydroxynonenal (HNE). HNE modifies all key proteins responsible for proper brain functioning thus playing a major role in the onset of neurological diseases. To investigate HNE effects on BBB permeability, we developed two in vitro BBB models-'physiological' and 'pathological'. The latter mimicked HNE modified extracellular matrix under oxidative stress conditions in brain pathologies. We showed that exogenous HNE induce activation of antioxidative defense systems by increasing catalase activity and glutathione content as well as reducing lipid peroxide levels in endothelial cells and astrocytes of 'physiological' model. While in 'pathological' model, exogenous HNE further increased lipid peroxidation levels of endothelial cells and astrocytes, followed by increase in Nrf2 and glutathione levels in endothelial cells. At lipid composition level, HNE caused increase in ω3 polyunsaturated fatty acid (PUFA) level in endothelial cells, followed by decrease in ω3 PUFA level and increase in monounsaturated fatty acid level in astrocytes. Using these models, we showed for the first time that HNE in 'pathological' model can reduce BBB permeability.
Collapse
|
6
|
Wu Y, Zhang Y, Jiao J. The relationship between n-3 polyunsaturated fatty acids and telomere: A review on proposed nutritional treatment against metabolic syndrome and potential signaling pathways. Crit Rev Food Sci Nutr 2022; 64:4457-4476. [PMID: 36330807 DOI: 10.1080/10408398.2022.2142196] [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: 11/06/2022]
Abstract
Metabolic syndrome (MetS), a cluster of metabolic abnormalities composed of central obesity, elevated blood pressure, glucose disturbances, hypercholesterolemia and dyslipidaemia, has increasingly become a public health problem in the 21st century worldwide. The dysfunction of telomeres, the repetitive DNA with highly conserved sequences (5'-TTAGGG-3'), is remarkably correlated with organismal aging, even suggesting a causal relationship with metabolic disorders. The health benefits of n-3 polyunsaturated fatty acids (PUFAs) in multiple disorders are associated with telomere length in evidence, which have recently drawn wide attention. However, functional targets and pathways for the associations of n-3 PUFAs and telomere with MetS remain scare. Few studies have summarized the role of n-3 PUFAs in DNA damage repair pathways, anti-inflammatory pathways, and redox balance, linking with telomere biology, and other potential telomere-related signaling pathways. This review aims to (i) elucidate how n-3 PUFAs ameliorate telomere attrition in the context of anti-oxidation and anti-inflammation; (ii) unravel the role of n-3 PUFAs in modulating telomere-related neuron dysfunction and regulating the neuro-endocrine-immunological network in MetS; (iii) epidemiologically implicate the associations of metabolic disorders and n-3 PUFAs with telomere length; and (iv) suggest promising biochemical approaches and advancing methodologies to overcome the inter-variation problem helpful for future research.
Collapse
Affiliation(s)
- Yuqi Wu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Kalkman HO. Potential Suicide Prophylactic Activity by the Fish Oil Metabolite, 4-Hydroxyhexenal. Int J Mol Sci 2022; 23:ijms23136953. [PMID: 35805959 PMCID: PMC9266565 DOI: 10.3390/ijms23136953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/05/2023] Open
Abstract
Low levels of n-3 poly-unsaturated fatty acids (n-3 PUFAs) and high levels of n-6 PUFAs in the blood circulation are associated with an increased risk for suicide. Clinical studies indicate that docosahexaenoic acid (DHA, a n-3 PUFA found in fish-oil) displays protective effects against suicide. It has recently been proposed that the activation of the transcription factor NRF2 might be the pharmacological activity that is common to current anti-suicidal medications. Oxidation products from fish oil, including those from DHA, are electrophiles that reversibly bind to a protein ‘KEAP1’, which acts as the molecular inhibitor of NRF2 and so indirectly promotes NRF2-transcriptional activity. In the majority of publications, the NRF2-stimulant effect of DHA is ascribed to the metabolite 4-hydroxyhexenal (4HHE). It is suggested to investigate whether 4HHE will display a therapeutically useful anti-suicidal efficacy.
Collapse
|
8
|
Karanikas E. Psychologically Traumatic Oxidative Stress; A Comprehensive Review of Redox Mechanisms and Related Inflammatory Implications. PSYCHOPHARMACOLOGY BULLETIN 2021; 51:65-86. [PMID: 34887600 PMCID: PMC8601764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The organism's energy requirements for homeostatic balance are covered by the redox mechanisms. Yet in case of psychologically traumatic stress, allostatic regulations activate both pro-oxidant and antioxidant molecules as well as respective components of the inflammatory system. Thus a new setpoint of dynamic interactions among redox elements is reached. Similarly, a multifaceted interplay between redox and inflammatory fields is activated with the mediation of major effector systems such as the immune system, Hypothalamic-Pituitary-Adrenal axis, kynurenine, and the glycaemic regulatory one. In case of sustained and/or intense traumatic stress the prophylactic antioxidant components are inadequate to provide the organism with neuroprotection finally culminating in Oxidative Stress and subsequently to cellular apoptosis. In parallel multiple inflammatory systems trigger and/or are triggered by the redox systems in tight fashion so that the causation sequence appears obscure. This exhaustive review aims at the comprehension of the interaction among components of the redox system as well as to the collection of disperse findings relative to the redox-inflammatory interplay in the context of traumatic stress so that new research strategies could be developed.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Karanikas, Department of Psychiatry, General Military Hospital, Thessaloniki, Greece
| |
Collapse
|
9
|
Dynamic Role of Phospholipases A2 in Health and Diseases in the Central Nervous System. Cells 2021; 10:cells10112963. [PMID: 34831185 PMCID: PMC8616333 DOI: 10.3390/cells10112963] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 12/20/2022] Open
Abstract
Phospholipids are major components in the lipid bilayer of cell membranes. These molecules are comprised of two acyl or alkyl groups and different phospho-base groups linked to the glycerol backbone. Over the years, substantial interest has focused on metabolism of phospholipids by phospholipases and the role of their metabolic products in mediating cell functions. The high levels of polyunsaturated fatty acids (PUFA) in the central nervous system (CNS) have led to studies centered on phospholipases A2 (PLA2s), enzymes responsible for cleaving the acyl groups at the sn-2 position of the phospholipids and resulting in production of PUFA and lysophospholipids. Among the many subtypes of PLA2s, studies have centered on three major types of PLA2s, namely, the calcium-dependent cytosolic cPLA2, the calcium-independent iPLA2 and the secretory sPLA2. These PLA2s are different in their molecular structures, cellular localization and, thus, production of lipid mediators with diverse functions. In the past, studies on specific role of PLA2 on cells in the CNS are limited, partly because of the complex cellular make-up of the nervous tissue. However, understanding of the molecular actions of these PLA2s have improved with recent advances in techniques for separation and isolation of specific cell types in the brain tissue as well as development of sensitive molecular tools for analyses of proteins and lipids. A major goal here is to summarize recent studies on the characteristics and dynamic roles of the three major types of PLA2s and their oxidative products towards brain health and neurological disorders.
Collapse
|
10
|
Chirumbolo S, Valdenassi L, Simonetti V, Bertossi D, Ricevuti G, Franzini M, Pandolfi S. Insights on the mechanisms of action of ozone in the medical therapy against COVID-19. Int Immunopharmacol 2021; 96:107777. [PMID: 34020394 PMCID: PMC8112288 DOI: 10.1016/j.intimp.2021.107777] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/28/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
An increasing amount of reports in the literature is showing that medical ozone (O3) is used, with encouraging results, in treating COVID-19 patients, optimizing pain and symptoms relief, respiratory parameters, inflammatory and coagulation markers and the overall health status, so reducing significantly how much time patients underwent hospitalization and intensive care. To date, aside from mechanisms taking into account the ability of O3 to activate a rapid oxidative stress response, by up-regulating antioxidant and scavenging enzymes, no sound hypothesis was addressed to attempt a synopsis of how O3 should act on COVID-19. The knowledge on how O3 works on inflammation and thrombosis mechanisms is of the utmost importance to make physicians endowed with new guns against SARS-CoV2 pandemic. This review tries to address this issue, so to expand the debate in the scientific community.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Luigi Valdenassi
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy
| | - Vincenzo Simonetti
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy
| | - Dario Bertossi
- Department of Surgery, Dentistry, Paediatrics and Gynaecology Unit of Maxillo-Facial Surgery University of Verona, Verona, Italy
| | | | - Marianno Franzini
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy
| | - Sergio Pandolfi
- SIOOT, High School in Oxygen Ozone Therapy, University of Pavia, Italy; SIOOT INTERNATIONAL, Communian Clinic, Gorle Bergamo, Italy; Villa Mafalda Clinics via Monte delle Gioie, Rome, Italy
| |
Collapse
|
11
|
Oxidative Dysregulation in Early Life Stress and Posttraumatic Stress Disorder: A Comprehensive Review. Brain Sci 2021; 11:brainsci11060723. [PMID: 34072322 PMCID: PMC8228973 DOI: 10.3390/brainsci11060723] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic stress may chronically affect master homeostatic systems at the crossroads of peripheral and central susceptibility pathways and lead to the biological embedment of trauma-related allostatic trajectories through neurobiological alterations even decades later. Lately, there has been an exponential knowledge growth concerning the effect of traumatic stress on oxidative components and redox-state homeostasis. This extensive review encompasses a detailed description of the oxidative cascade components along with their physiological and pathophysiological functions and a systematic presentation of both preclinical and clinical, genetic and epigenetic human findings on trauma-related oxidative stress (OXS), followed by a substantial synthesis of the involved oxidative cascades into specific and functional, trauma-related pathways. The bulk of the evidence suggests an imbalance of pro-/anti-oxidative mechanisms under conditions of traumatic stress, respectively leading to a systemic oxidative dysregulation accompanied by toxic oxidation byproducts. Yet, there is substantial heterogeneity in findings probably relative to confounding, trauma-related parameters, as well as to the equivocal directionality of not only the involved oxidative mechanisms but other homeostatic ones. Accordingly, we also discuss the trauma-related OXS findings within the broader spectrum of systemic interactions with other major influencing systems, such as inflammation, the hypothalamic-pituitary-adrenal axis, and the circadian system. We intend to demonstrate the inherent complexity of all the systems involved, but also put forth associated caveats in the implementation and interpretation of OXS findings in trauma-related research and promote their comprehension within a broader context.
Collapse
|
12
|
Sun GY, Appenteng MK, Li R, Woo T, Yang B, Qin C, Pan M, Cieślik M, Cui J, Fritsche KL, Gu Z, Will M, Beversdorf D, Adamczyk A, Han X, Greenlief CM. Docosahexaenoic Acid (DHA) Supplementation Alters Phospholipid Species and Lipid Peroxidation Products in Adult Mouse Brain, Heart, and Plasma. Neuromolecular Med 2021; 23:118-129. [PMID: 32926329 PMCID: PMC9555299 DOI: 10.1007/s12017-020-08616-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/07/2020] [Indexed: 10/23/2022]
Abstract
The abundance of docosahexaenoic acid (DHA) in phospholipids in the brain and retina has generated interest to search for its role in mediating neurological functions. Besides the source of many oxylipins with pro-resolving properties, DHA also undergoes peroxidation, producing 4-hydroxyhexenal (4-HHE), although its function remains elusive. Despite wide dietary consumption, whether supplementation of DHA may alter the peroxidation products and their relationship to phospholipid species in brain and other body organs have not been explored sufficiently. In this study, adult mice were administered a control or DHA-enriched diet for 3 weeks, and phospholipid species and peroxidation products were examined in brain, heart, and plasma. Results demonstrated that this dietary regimen increased (n-3) and decreased (n-6) species to different extent in all major phospholipid classes (PC, dPE, PE-pl, PI and PS) examined. Besides changes in phospholipid species, DHA-enriched diet also showed substantial increases in 4-HHE in brain, heart, and plasma. Among different brain regions, the hippocampus responded to the DHA-enriched diet showing significant increase in 4-HHE. Considering the pro- and anti-inflammatory pathways mediated by the (n-6) and (n-3) polyunsaturated fatty acids, unveiling the ability for DHA-enriched diet to alter phospholipid species and lipid peroxidation products in the brain and in different body organs may be an important step forward towards understanding the mechanism(s) for this (n-3) fatty acid on health and diseases.
Collapse
Affiliation(s)
- Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Michael K Appenteng
- Department of Chemistry, University of Missouri, 125 Chemistry Bldg., Columbia, MO, 65211, USA
| | - Runting Li
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Taeseon Woo
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, 65211, USA
| | - Bo Yang
- Department of Chemistry, University of Missouri, 125 Chemistry Bldg., Columbia, MO, 65211, USA
| | - Chao Qin
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science & Center at San Antonio, San Antonio, TX, 78229, USA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science & Center at San Antonio, San Antonio, TX, 78229, USA
| | - Magdalena Cieślik
- Department of Cellular Signaling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, 65211, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Matthew Will
- Department of Psychological Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - David Beversdorf
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, 65211, USA
- Departments of Radiology, Neurology and Psychological Sciences, and the Thompson Center, University of Missouri, Columbia, MO, 65211, USA
| | - Agata Adamczyk
- Department of Cellular Signaling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science & Center at San Antonio, San Antonio, TX, 78229, USA
| | - C Michael Greenlief
- Department of Chemistry, University of Missouri, 125 Chemistry Bldg., Columbia, MO, 65211, USA.
| |
Collapse
|
13
|
Modulatory role of dietary polyunsaturated fatty acids in Nrf2-mediated redox homeostasis. Prog Lipid Res 2020; 80:101066. [DOI: 10.1016/j.plipres.2020.101066] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
|
14
|
Paciorek P, Żuberek M, Grzelak A. Products of Lipid Peroxidation as a Factor in the Toxic Effect of Silver Nanoparticles. MATERIALS 2020; 13:ma13112460. [PMID: 32481688 PMCID: PMC7321096 DOI: 10.3390/ma13112460] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/11/2020] [Accepted: 05/21/2020] [Indexed: 11/20/2022]
Abstract
In our previous study we have shown that nanoparticles have different effects depending on the energy metabolism of the cell, which is an important factor in the context of oncology and diabetes. Here we assess the influence of AgNPs on cellular lipid components in varying glucose concentrations. To assess the effect of silver nanoparticles on cell lipids, we measured cell viability, the fluidity of the cell membranes, the content of amino groups in proteins, the level of lipid peroxidation products, the concentration of 4-hydroxynonenal (4-HNE), and the concentration of lipid peroxides. The obtained results show differences in the formation of lipid peroxidation products in cells exposed to oxidative stress induced by nanoparticles. In addition, we have shown that the metabolic state of the cell is a factor significantly affecting this process.
Collapse
|
15
|
Lee YY, Galano J, Leung HH, Balas L, Oger C, Durand T, Lee JC. Nonenzymatic oxygenated metabolite of docosahexaenoic acid, 4(RS)‐4‐F4t‐neuroprostane, acts as a bioactive lipid molecule in neuronal cells. FEBS Lett 2020; 594:1797-1808. [DOI: 10.1002/1873-3468.13774] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/30/2020] [Accepted: 03/17/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Yiu Yiu Lee
- School of Biological Sciences The University of Hong Kong Hong Kong
| | - Jean‐Marie Galano
- Institut des Biomolécules Max Mousseron IBMM Université de Montpellier CNRS ENSCM Faculté de Pharmacie Montpellier France
| | - Ho Hang Leung
- School of Biological Sciences The University of Hong Kong Hong Kong
| | - Laurence Balas
- Institut des Biomolécules Max Mousseron IBMM Université de Montpellier CNRS ENSCM Faculté de Pharmacie Montpellier France
| | - Camille Oger
- Institut des Biomolécules Max Mousseron IBMM Université de Montpellier CNRS ENSCM Faculté de Pharmacie Montpellier France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron IBMM Université de Montpellier CNRS ENSCM Faculté de Pharmacie Montpellier France
| | | |
Collapse
|
16
|
Park I, Xun P, Tsinovoi CL, Klemmer P, Liu K, He K. Intakes of long-chain omega-3 polyunsaturated fatty acids and non-fried fish in relation to incidence of chronic kidney disease in young adults: a 25-year follow-up. Eur J Nutr 2020; 59:399-407. [PMID: 31175412 PMCID: PMC6898765 DOI: 10.1007/s00394-019-02022-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 06/04/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE The prevalence of chronic kidney disease (CKD) is increasing rapidly in many countries and has become a major public health concern. Although intakes of long-chain omega-3 polyunsaturated fatty acids (LCω3PUFA) and its food source-fish-may have renal protective effects, little is known about the longitudinal association between these dietary factors and CKD incidence. METHODS A total of 4133 healthy individuals of black and white race aged 18-30 at baseline (1985-1986) from the Coronary Artery Risk Development in Young Adults study were enrolled and followed up over 25 years. LCω3PUFA and fish intake were assessed by an interview-based dietary history questionnaire at baseline, year 7 (1992-1993) and 20 (2005-2006). RESULTS Four hundred and eighty-nine incident cases of CKD were identified. After adjustment for potential confounders, LCω3PUFA intake was inversely associated with CKD incidence [HR = 0.73 (95% CI 0.60-0.89), P = 0.002, with one standard division (0.19 g/day) increment in LCω3PUFA]. This inverse association was persisted among females [0.64 (95% CI 0.48, 0.84; P = 0.002], but not males (Pinteraction = 0.070). A marginal significant inverse association was also found between non-fried fish consumption and CKD incidence (HR = 0.86, 95% CI 0.73, 1.01; P = 0.073). CONCLUSIONS Dietary LCω3PUFA intake was inversely associated with incidence of CKD among American young adults over 25 years of follow-up. The suggestive evidence of the inverse association between non-fried fish consumption with CKD incidence needs further confirmation.
Collapse
Affiliation(s)
- Inwhee Park
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, 1025 E. 7th Street, Bloomington, IN, 47405, USA
- Department of Nephrology, School of Medicine, Ajou University, Suwon, Gyeonggi-do, Republic of Korea
| | - Pengcheng Xun
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, 1025 E. 7th Street, Bloomington, IN, 47405, USA
| | - Cari Lewis Tsinovoi
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, 1025 E. 7th Street, Bloomington, IN, 47405, USA
| | - Philip Klemmer
- Division of Nephrology and Hypertension, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Kiang Liu
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ka He
- Department of Epidemiology and Biostatistics, School of Public Health-Bloomington, Indiana University, 1025 E. 7th Street, Bloomington, IN, 47405, USA.
| |
Collapse
|
17
|
Romana-Souza B, Saguie BO, Pereira de Almeida Nogueira N, Paes M, Dos Santos Valença S, Atella GC, Monte-Alto-Costa A. Oleic acid and hydroxytyrosol present in olive oil promote ROS and inflammatory response in normal cultures of murine dermal fibroblasts through the NF-κB and NRF2 pathways. Food Res Int 2020; 131:108984. [PMID: 32247459 DOI: 10.1016/j.foodres.2020.108984] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/28/2019] [Accepted: 01/05/2020] [Indexed: 12/27/2022]
Abstract
Few studies have evaluated the effects of olive oil on normal tissues like skin and its components. Hence, we investigated whether olive oil could increase the production of ROS and oxidative damage in murine dermal fibroblast cultures in a short-term exposition. In addition, we evaluated the role of oleic acid and hydroxytyrosol, which are the two most important components of olive oil, in the associated mechanisms of action, and the metabolism of long-chain fatty acids from olive oil. To study this, neonatal murine dermal fibroblasts (NMDF) were incubated with olive oil, oleic acid, or hydroxytyrosol for 24 or 72 h. The NMDF incubated with olive oil or oleic acid showed an increase in the production of ROS after 24 h, lipid peroxidation, and protein carbonylation after 72 h, as well as increased expression of nuclear factor-kappa B (NF-κB) p65 and cyclooxygenase-2 (COX-2) after 72 h. However, NMDF treated with olive oil or hydroxytyrosol demonstrated an increase in the expression of nuclear factor-erythroid2-related factor 2 (NRF2) and heme oxygenase-1 (HO-1) after 72 h. In addition, NMDF treated with olive oil also showed an increase in the protein expression of diacylglycerol acyltransferase1 (DGAT1), which promotes triacylglycerol synthesis, and in the levels of triacylglycerols. The microscopic analysis showed Nile red-positive lipid droplets inside olive oil-treated NMDF after 72 h. Moreover, gas chromatography-mass spectrometry demonstrated high levels of oleic acid in the olive oil-treated NMDF after 72 h. In conclusion, oleic acid present in the olive oil promotes the production of ROS and oxidative damage in murine dermal fibroblasts, which leads to NF-κB p65 and COX-2 expression, while hydroxytyrosol promotes NRF2 and HO-1 expression. In addition, NMDF area capable of absorbing long-chain fatty acids derived from olive oil, which promotes the synthesis and the accumulation of triacylglycerols into cytoplasm of NMDF through DGAT1 activation.
Collapse
Affiliation(s)
- Bruna Romana-Souza
- Tissue Repair Laboratory, Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Bianca Oliveira Saguie
- Tissue Repair Laboratory, Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Marcia Paes
- Laboratory of Trypanosomatids and Vectores Interection, Department of Biochemistry, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Georgia Correa Atella
- Laboratory of Lipid and Lipoprotein Biochemistry, Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andréa Monte-Alto-Costa
- Tissue Repair Laboratory, Department of Histology and Embryology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Effects of Docosahexaenoic Acid and Its Peroxidation Product on Amyloid-β Peptide-Stimulated Microglia. Mol Neurobiol 2019; 57:1085-1098. [PMID: 31677009 DOI: 10.1007/s12035-019-01805-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
Abstract
Growing evidence suggests that docosahexaenoic acid (DHA) exerts neuroprotective effects, although the mechanism(s) underlying these beneficial effects are not fully understood. Here we demonstrate that DHA, but not arachidonic acid (ARA), suppressed oligomeric amyloid-β peptide (oAβ)-induced reactive oxygen species (ROS) production in primary mouse microglia and immortalized mouse microglia (BV2). Similarly, DHA but not ARA suppressed oAβ-induced increases in phosphorylated cytosolic phospholipase A2 (p-cPLA2), inducible nitric oxide synthase (iNOS), and tumor necrosis factor-α (TNF-α) in BV2 cells. LC-MS/MS assay indicated the ability for DHA to cause an increase in 4-hydroxyhexenal (4-HHE) and suppress oAβ-induced increase in 4-hydroxynonenal (4-HNE). Although oAβ did not alter the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, exogenous DHA, ARA as well as low concentrations of 4-HHE and 4-HNE upregulated this pathway and increased production of heme oxygenase-1 (HO-1) in microglial cells. These results suggest that DHA modulates ARA metabolism in oAβ-stimulated microglia through suppressing oxidative and inflammatory pathways and upregulating the antioxidative stress pathway involving Nrf2/HO-1. Understanding the mechanism(s) underlying the beneficial effects of DHA on microglia should shed light into nutraceutical therapy for the prevention and treatment of Alzheimer's disease (AD).
Collapse
|
19
|
Dong J, Feng X, Zhang J, Zhang Y, Xia F, Liu L, Jin Z, Lu C, Xia Y, Papadimos TJ, Xu X. ω-3 fish oil fat emulsion preconditioning mitigates myocardial oxidative damage in rats through aldehydes stress. Biomed Pharmacother 2019; 118:109198. [PMID: 31336342 DOI: 10.1016/j.biopha.2019.109198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/23/2019] [Accepted: 07/02/2019] [Indexed: 11/21/2022] Open
Abstract
ω-3 fish oil fat emulsions contain a considerable quantity of unsaturated carbon-carbon double bonds, which undergo lipid peroxidation to yield low-dose aldehydes. These aldehydes may stimulate the production of antioxidant enzymes, thereby mitigating myocardial oxidative damage. This study aims to (1) verify the cardioprotective effect of ω-3 fish oil fat emulsion in vivo and in vitro, and (2) determine whether aldehyde stress is a protective mechanism. For modeling purposes, we pretreated rats with 2 ml/kg of a 10% ω-3 fish oil fat emulsion for 5 days in order to generate a sufficient aldehyde stress response to trigger the production of antioxidant enzymes, and we obtained similar response with H9C2 cells that were pretreated with a 0.5% ω-3 fish oil fat emulsion for 24 h. ω-3 fish oil fat emulsion pretreatment in vivo reduced the myocardial infarct size, decreased the incidence of arrhythmias, and promoted the recovery of cardiac function after myocardial ischemia/reperfusion injury. Once the expression of nuclear factor E2-related factor 2 (Nrf2) was silenced in H9C2 cells, aldehydes no longer produced enough antioxidant enzymes to reverse the oxidative damage caused by tert-butyl hydroperoxide (TBHP). Our results demonstrated that ω-3 fish oil fat emulsion enhanced the inhibition of oxidation and production of free radicals, and alleviated myocardial oxidative injury via activation of the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Jiaojiao Dong
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaona Feng
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Jingxiong Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yujian Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Fangfang Xia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Le Liu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhousheng Jin
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Caijiao Lu
- Burn Wound Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yun Xia
- Department of Anesthesiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Thomas J Papadimos
- Department of Anesthesiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Xuzhong Xu
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
20
|
Shobako N, Ishikado A, Ogawa Y, Sono Y, Kusakari T, Suwa M, Matsumoto M, Ohinata K. Vasorelaxant and Antihypertensive Effects That Are Dependent on the Endothelial NO System Exhibited by Rice Bran-Derived Tripeptide. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:1437-1442. [PMID: 30609899 DOI: 10.1021/acs.jafc.8b06341] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
We recently identified a novel, potent antihypertensive peptide, Leu-Arg-Ala (LRA; minimum effective dose = 0.25 mg/kg), from rice bran protein. In this study, we found that LRA potently relaxed mesenteric arteries isolated from spontaneously hypertensive rats (SHRs) (EC50 = 0.1 μM). In contrast, the vasorelaxant activity of each amino acid that constitutes the LRA tripeptide was remarkably attenuated. The LRA-induced vasorelaxant activity was inhibited by N(G)-nitro-l-arginine methyl ester (L-NAME; NO synthase [NOS] inhibitor) but not by an antagonist of bradykinin B2 and Mas receptors or by a phosphoinositide 3-kinase inhibitor. The antihypertensive effect induced after the oral administration of LRA was inhibited by L-NAME. LRA also induced the phosphorylation of endothelial NOS in human umbilical vein endothelial cells. Taken together, LRA may exhibit antihypertensive effects via NO-mediated vasorelaxation. LRA is the first example of a NO-dependent vasorelaxant peptide identified from rice bran protein.
Collapse
Affiliation(s)
- Naohisa Shobako
- Health Care R&D , SUNSTAR , Takatsuki , Osaka 569-1195 , Japan
- Division of Food Science and Biotechnology, Graduate School of Agriculture , Kyoto University , Uji , Kyoto 611-0011 , Japan
| | | | - Yutaro Ogawa
- Health Care R&D , SUNSTAR , Takatsuki , Osaka 569-1195 , Japan
| | - Yoko Sono
- Health Care R&D , SUNSTAR , Takatsuki , Osaka 569-1195 , Japan
| | | | - Makoto Suwa
- Health Care R&D , SUNSTAR , Takatsuki , Osaka 569-1195 , Japan
| | | | - Kousaku Ohinata
- Division of Food Science and Biotechnology, Graduate School of Agriculture , Kyoto University , Uji , Kyoto 611-0011 , Japan
| |
Collapse
|
21
|
Narayanankutty A, Gopinath MK, Vakayil M, Ramavarma SK, Babu TD, Raghavamenon AC. Non-enzymatic conversion of primary oxidation products of Docosahexaenoic acid into less toxic acid molecules. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 203:222-228. [PMID: 29870906 DOI: 10.1016/j.saa.2018.05.082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 05/23/2018] [Accepted: 05/26/2018] [Indexed: 06/08/2023]
Abstract
Docosahexaenoic acid (DHA) is long chain omega-3 fatty acid with known health benefits and clinical significance. However, 4-hydroxy hexenal (HHE), an enzymatic oxidation product of DHA has recently been reported to have health-damaging effects. This conflict raises major concern on the long-term clinical use of these fatty acids. Even though the enzymatic and non-enzymatic conversion of HHE to nontoxic acid molecules is possible by the aldehyde detoxification systems, it has not yet studied. To address this, primary oxidation products of DHA in lipoxidase system were subjected to non-enzymatic conversion at physiological temperature over a period of 1 week. The reaction was monitored using HPLC, IR spectroscopy and biochemical assays (based on the loss of conjugated dienes, lipid peroxides aldehydes). Short term and long term cytotoxicity of the compounds generated at various time points were analyzed. IR and HPLC spectra revealed that the level of aldehydes in the primary oxidation products reduced over time, generating acids and acid derivatives within a week period. In short term and long term cytotoxicity analysis, initial decomposition products were found more toxic than the 1-week decomposition products. Further, when primary oxidation products were subjected to aldehyde dehydrogenase mediated oxidation, it generated products that are also less toxic. The study suggests the possible non-enzymatic conversion of primary oxidation products of DHA to less cytotoxic acid molecules. Exploration of the physiological roles of these acid molecules may explain the biological potential of omega-3 fatty acids.
Collapse
Affiliation(s)
- Arunaksharan Narayanankutty
- Amala Cancer Research Centre (Recognized Centre of University of Calicut), Amala Nagar, Thrissur, 680 555, Kerala, India
| | - Midhun K Gopinath
- Amala Cancer Research Centre (Recognized Centre of University of Calicut), Amala Nagar, Thrissur, 680 555, Kerala, India
| | - Muneera Vakayil
- Amala Cancer Research Centre (Recognized Centre of University of Calicut), Amala Nagar, Thrissur, 680 555, Kerala, India
| | - Smitha K Ramavarma
- Amala Cancer Research Centre (Recognized Centre of University of Calicut), Amala Nagar, Thrissur, 680 555, Kerala, India
| | - Thekkekara Devassy Babu
- Amala Cancer Research Centre (Recognized Centre of University of Calicut), Amala Nagar, Thrissur, 680 555, Kerala, India
| | - Achuthan C Raghavamenon
- Amala Cancer Research Centre (Recognized Centre of University of Calicut), Amala Nagar, Thrissur, 680 555, Kerala, India.
| |
Collapse
|
22
|
Sun GY, Simonyi A, Fritsche KL, Chuang DY, Hannink M, Gu Z, Greenlief CM, Yao JK, Lee JC, Beversdorf DQ. Docosahexaenoic acid (DHA): An essential nutrient and a nutraceutical for brain health and diseases. Prostaglandins Leukot Essent Fatty Acids 2018; 136:3-13. [PMID: 28314621 PMCID: PMC9087135 DOI: 10.1016/j.plefa.2017.03.006] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 01/01/2023]
Abstract
Docosahexaenoic acid (DHA), a polyunsaturated fatty acid (PUFA) enriched in phospholipids in the brain and retina, is known to play multi-functional roles in brain health and diseases. While arachidonic acid (AA) is released from membrane phospholipids by cytosolic phospholipase A2 (cPLA2), DHA is linked to action of the Ca2+-independent iPLA2. DHA undergoes enzymatic conversion by 15-lipoxygenase (Alox 15) to form oxylipins including resolvins and neuroprotectins, which are powerful lipid mediators. DHA can also undergo non-enzymatic conversion by reacting with oxygen free radicals (ROS), which cause the production of 4-hydoxyhexenal (4-HHE), an aldehyde derivative which can form adducts with DNA, proteins and lipids. In studies with both animal models and humans, there is evidence that inadequate intake of maternal n-3 PUFA may lead to aberrant development and function of the central nervous system (CNS). What is less certain is whether consumption of n-3 PUFA is important in maintaining brain health throughout one's life span. Evidence mostly from non-human studies suggests that DHA intake above normal nutritional requirements might modify the risk/course of a number of diseases of the brain. This concept has fueled much of the present interest in DHA research, in particular, in attempts to delineate mechanisms whereby DHA may serve as a nutraceutical and confer neuroprotective effects. Current studies have revealed ability for the oxylipins to regulation of cell redox homeostasis through the Nuclear factor (erythroid-derived 2)-like 2/Antioxidant response element (Nrf2/ARE) anti-oxidant pathway, and impact signaling pathways associated with neurotransmitters, and modulation of neuronal functions involving brain-derived neurotropic factor (BDNF). This review is aimed at describing recent studies elaborating these mechanisms with special regard to aging and Alzheimer's disease, autism spectrum disorder, schizophrenia, traumatic brain injury, and stroke.
Collapse
Affiliation(s)
- Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - Agnes Simonyi
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Dennis Y Chuang
- Department of Neurology, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH, United States
| | - Mark Hannink
- Biochemistry Department, University of Missouri, Columbia, MO, United States
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, United States
| | | | - Jeffrey K Yao
- Medical Research Service, VA Pittsburgh Healthcare System, and Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - David Q Beversdorf
- Department of Radiology, Neurology, and Psychological Sciences, and the Thompson Center, William and Nancy Thompson Endowed Chair in Radiology, University of Missouri School of Medicine, Columbia, MO, United States
| |
Collapse
|
23
|
Leung KS, Galano JM, Durand T, Lee JCY. Profiling of Omega-Polyunsaturated Fatty Acids and Their Oxidized Products in Salmon after Different Cooking Methods. Antioxidants (Basel) 2018; 7:antiox7080096. [PMID: 30042286 PMCID: PMC6116150 DOI: 10.3390/antiox7080096] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 01/15/2023] Open
Abstract
Consumption of food containing n-3 PUFAs, namely EPA and DHA, are known to benefit health and protect against chronic diseases. Both are richly found in marine-based food such as fatty fish and seafood that are commonly cooked prior to consumption. However, the elevated temperature during cooking potentially degrades the EPA and DHA through oxidation. To understand the changes during different cooking methods, lipid profiles of raw, boiled, pan-fried and baked salmon were determined by LC-MS/MS. Our results showed that pan-frying and baking elevated the concentration of peroxides in salmon, whereas only pan-frying increased the MDA concentration, indicating it to be the most severe procedure to cause oxidation among the cooking methods. Pan-frying augmented oxidized products of n-3 and n-6 PUFAs, while only those of n-3 PUFA were elevated in baked salmon. Notably, pan-frying and baking increased bioactive oxidized n-3 PUFA products, in particular F-4t-neuroprostanes derived from DHA. The results of this study provided a new insight into the application of heat and its effect on PUFAs and the release of its oxidized products in salmon.
Collapse
Affiliation(s)
- Kin Sum Leung
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS Université de Montpellier, ENSCM, F-34093 Montpellier, France.
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS Université de Montpellier, ENSCM, F-34093 Montpellier, France.
| | - Jetty Chung-Yung Lee
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| |
Collapse
|
24
|
Yang B, Li R, Michael Greenlief C, Fritsche KL, Gu Z, Cui J, Lee JC, Beversdorf DQ, Sun GY. Unveiling anti-oxidative and anti-inflammatory effects of docosahexaenoic acid and its lipid peroxidation product on lipopolysaccharide-stimulated BV-2 microglial cells. J Neuroinflammation 2018; 15:202. [PMID: 29986724 PMCID: PMC6038194 DOI: 10.1186/s12974-018-1232-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/20/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Phospholipids in the central nervous system are enriched in n-3 and n-6 polyunsaturated fatty acids (PUFA), especially docosahexaenoic acid (DHA) and arachidonic acid (ARA). These PUFA can undergo enzymatic reactions to produce lipid mediators, as well as reaction with oxygen free radicals to produce 4-hydroxyhexenal (4-HHE) from DHA and 4-hydroxynonenal (4-HNE) from ARA. Recent studies demonstrated pleiotropic properties of these peroxidation products through interaction with oxidative and anti-oxidant response pathways. In this study, BV-2 microglial cells were used to investigate ability for DHA, 4-HHE, and 4-HNE to stimulate the anti-oxidant stress responses involving the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway and synthesis of heme oxygenase (HO-1), as well as to mitigate lipopolysaccharide (LPS)-induced nitric oxide (NO), reactive oxygen species (ROS), and cytosolic phospholipase A2 (cPLA2). In addition, LC-MS/MS analysis was carried out to examine effects of exogenous DHA and LPS stimulation on endogenous 4-HHE and 4-HNE levels in BV-2 microglial cells. METHODS Effects of DHA, 4-HHE, and 4-HNE on LPS-induced NO production was determined using the Griess reagent. LPS-induced ROS production was measured using CM-H2DCFDA. Western blots were used to analyze expression of p-cPLA2, Nrf2, and HO-1. Cell viability and cytotoxicity were measured using the WST-1 assay, and cell protein concentrations were measured using the BCA protein assay kit. An ultra-high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis was used to determine levels of free 4-HHE and 4-HNE in cells. RESULTS DHA (12.5-100 μM), 4-HHE (1.25-10 μM), and 4-HNE (1.25-10 μM) dose dependently suppressed LPS-induced production of NO, ROS, and as p-cPLA2 in BV-2 microglial cells. With the same concentrations, these compounds could enhance Nrf2 and HO-1 expression in these cells. Based on the estimated IC50 values, 4-HHE and 4-HNE were five- to tenfold more potent than DHA in inhibiting LPS-induced NO, ROS, and p-cPLA2. LC-MS/MS analysis indicated ability for DHA (10-50 μM) to increase levels of 4-HHE and attenuate levels of 4-HNE in BV-2 microglial cells. Stimulation of cells with LPS caused an increase in 4-HNE which could be abrogated by cPLA2 inhibitor. In contrast, bromoenol lactone (BEL), a specific inhibitor for the Ca2+-independent phospholipase A2 (iPLA2), could only partially suppress levels of 4-HHE induced by DHA or DHA + LPS. CONCLUSIONS This study demonstrated the ability of DHA and its lipid peroxidation products, namely, 4-HHE and 4-HNE at 1.25-10 μM, to enhance Nrf2/HO-1 and mitigate LPS-induced NO, ROS, and p-cPLA2 in BV-2 microglial cells. In addition, LC-MS/MS analysis of the levels of 4-HHE and 4-HNE in microglial cells demonstrates that increases in production of 4-HHE from DHA and 4-HNE from LPS are mediated by different mechanisms.
Collapse
Affiliation(s)
- Bo Yang
- Chemistry Department, University of Missouri, Columbia, MO, USA
| | - Runting Li
- Biochemistry Department, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | | | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - David Q Beversdorf
- Departments of Radiology, Neurology and Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA. .,Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
25
|
Sottero B, Leonarduzzi G, Testa G, Gargiulo S, Poli G, Biasi F. Lipid Oxidation Derived Aldehydes and Oxysterols Between Health and Disease. EUR J LIPID SCI TECH 2018. [DOI: 10.1002/ejlt.201700047] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Barbara Sottero
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Simona Gargiulo
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, San Luigi Hospital, University of Torino; Regione Gonzole 10 10043 Orbassano (Torino) Italy
| |
Collapse
|
26
|
Romani A, Cervellati C, Muresan XM, Belmonte G, Pecorelli A, Cervellati F, Benedusi M, Evelson P, Valacchi G. Keratinocytes oxidative damage mechanisms related to airbone particle matter exposure. Mech Ageing Dev 2017; 172:86-95. [PMID: 29103985 DOI: 10.1016/j.mad.2017.11.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/30/2017] [Accepted: 11/01/2017] [Indexed: 12/17/2022]
Abstract
Epidemiological evidences have correlated airbone particulate matter (PM) to adverse health effects, mainly linking to pulmonary and cardiovascular disease. Nevertheless, only recently, some studies reported detrimental effects of PM on other organs such as skin. In a recent work, we have reported increased oxidative and inflammatory responses in Reconstituted Human Epidermis (RHE) exposed to ambient particles (CAPs) and we also demonstrated the ability of CAPs to penetrate the skin tissue. The present study was aimed to better understand the cellular mechanisms beyond the oxidative changes induced by CAPs (5-10-25μg/mL) in human immortalized keratinocytes (HaCaT). After 24h of treatment, CAPs were able to enter the cells leading to a decrease in viability, increased levels of 4-hydroxinonenal products (4-HNE) and IL-1α release. Overall these data, suggest lipid and protein oxidative damage, as well as an increase of inflammatory response after being challenged with CAPs. In addition, 3h after CAPs exposure we found a significant increase in NF-kB and Nrf2 translocation into the nucleus. In contrast, no differences in gene expression and enzymatic activity of Nrf2 target genes were detected. This last finding could be explained by the ability of CAPs to possibly alter the binding of Nrf2 to the ARE DNA sequence.
Collapse
Affiliation(s)
- Arianna Romani
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Carlo Cervellati
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara Italy
| | - Ximena M Muresan
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giuseppe Belmonte
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Alessandra Pecorelli
- Department of Animal Science, North Carolina State University, Plants for Human Health Institute, NC Research Center, 28081, Kannapolis NC, USA
| | - Franco Cervellati
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Mascia Benedusi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Pablo Evelson
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - G Valacchi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Department of Animal Science, North Carolina State University, Plants for Human Health Institute, NC Research Center, 28081, Kannapolis NC, USA.
| |
Collapse
|
27
|
Nègre-Salvayre A, Garoby-Salom S, Swiader A, Rouahi M, Pucelle M, Salvayre R. Proatherogenic effects of 4-hydroxynonenal. Free Radic Biol Med 2017; 111:127-139. [PMID: 28040472 DOI: 10.1016/j.freeradbiomed.2016.12.038] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/22/2016] [Accepted: 12/24/2016] [Indexed: 01/08/2023]
Abstract
4-hydroxy-2-nonenal (HNE) is a α,β-unsaturated hydroxyalkenal generated by peroxidation of n-6 polyunsaturated fatty acid. This reactive carbonyl compound exhibits a huge number of biological properties that result mainly from the formation of HNE-adducts on free amino groups and thiol groups in proteins. In the vascular system, HNE adduct accumulation progressively leads to cellular dysfunction and tissue damages that are involved in the progression of atherosclerosis and related diseases. HNE contributes to the atherogenicity of oxidized LDL, by forming HNE-apoB adducts that deviate the LDL metabolism to the scavenger receptor pathway of macrophagic cells, and lead to the formation of foam cells. HNE activates transcription factors (Nrf2, NF-kappaB) that (dys)regulate various cellular responses ranging from hormetic and survival signaling at very low concentrations, to inflammatory and apoptotic effects at higher concentrations. Among a variety of cellular targets, HNE can modify signaling proteins involved in atherosclerotic plaque remodeling, particularly growth factor receptors (PDGFR, EGFR), cell cycle proteins, mitochondrial and endoplasmic reticulum components or extracellular matrix proteins, which progressively alters smooth muscle cell proliferation, angiogenesis and induces apoptosis. HNE adducts accumulate in the lipidic necrotic core of advanced atherosclerotic lesions, and may locally contribute to macrophage and smooth muscle cell apoptosis, which may induce plaque destabilization and rupture, thereby increasing the risk of athero-thrombotic events.
Collapse
Affiliation(s)
| | | | | | | | | | - Robert Salvayre
- Inserm UMR-1048, France; University of Toulouse, Faculty of Medicine, Biochemistry Dept, Toulouse, France; CHU Toulouse, Rangueil, Toulouse, France
| |
Collapse
|
28
|
Nagahora N, Yamada H, Kikuchi S, Hakozaki M, Yano A. Nrf2 Activation by 5-lipoxygenase Metabolites in Human Umbilical Vascular Endothelial Cells. Nutrients 2017; 9:nu9091001. [PMID: 28892009 PMCID: PMC5622761 DOI: 10.3390/nu9091001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022] Open
Abstract
5-hydroxyeicosatetraenoic acid (5-HETE) and 5-hydroxyeicosapentaenoic acid (5-HEPE) are major metabolites produced by 5-lipoxygenase (5-LOX) from arachidonic acid (AA) and eicosapentaenoic acid (EPA). Effects of hydroxides on endothelial cells are unclear, although 5-LOX is known to increase at arteriosclerotic lesions. To investigate the effects of hydroxides on human umbilical vein endothelial cells (HUVECs), the cells were treated with 50 μM each of AA, EPA, 5-HETE, and 5-HEPE. Treatment of HUVECs with 5-HETE and 5-HEPE, rather than with AA and EPA, increased the nuclear translocation of NF-E2 related factor 2 (Nrf2) and upregulated the expression of heme oxygenase-1 and cystine/glutamate transporter regulated by Nrf2. Reactive oxygen species (ROS) generation was markedly elevated in HUVECs after treatment with 5-HETE and 5-HEPE, and the pretreatment with α-tocopherol abrogated ROS levels similar to those in the vehicle control. However, ROS generation was independent of Nrf2 activation induced by 5-HETE and 5-HEPE. 5-HETE was converted to 5-oxo-eicosatetraenoic acid (5-oxo-ETE) in HUVECs, and 5-oxo-ETE increased Nrf2 activation. These results suggest that 5-HETE works as an Nrf2 activator through the metabolite 5-oxo-ETE in HUVECs. Similarly, 5-HEPE works in the same way, because 5-HEPE is metabolized to 5-oxo-eicosapentaenoic acid through the same pathway as that for 5-HETE.
Collapse
Affiliation(s)
- Nozomi Nagahora
- Iwate Biotechnology Research Center, 22-174-4 Narita, Kitakami, Iwate 024-0003, Japan.
| | - Hidetoshi Yamada
- Iwate Biotechnology Research Center, 22-174-4 Narita, Kitakami, Iwate 024-0003, Japan.
| | - Sayaka Kikuchi
- Iwate Biotechnology Research Center, 22-174-4 Narita, Kitakami, Iwate 024-0003, Japan.
| | - Mayuka Hakozaki
- Iwate Biotechnology Research Center, 22-174-4 Narita, Kitakami, Iwate 024-0003, Japan.
| | - Akira Yano
- Iwate Biotechnology Research Center, 22-174-4 Narita, Kitakami, Iwate 024-0003, Japan.
| |
Collapse
|
29
|
Silver nanoparticles can attenuate nitrative stress. Redox Biol 2017; 11:646-652. [PMID: 28157664 PMCID: PMC5279695 DOI: 10.1016/j.redox.2017.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/06/2017] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
We have reported previously that glucose availability can modify toxicity of silver nanoparticles (AgNPs) via elevation of antioxidant defence triggered by increased mitochondrial generation of reactive oxygen species. In this study, we examined the effect of glucose availability on the production of reactive nitrogen species in HepG2 cells and modification of nitrative stress by AgNPs. We found that lowering the glucose concentration increased expression of genes coding for inducible nitric oxide syntheas, NOS2 and NOS2A resulting in enhanced production of nitric oxide. Surprisingly, AgNPs decreased the level of nitric oxide accelerated denitration of proteins nitrated by exogenous peroxynitrite in cells grown in the presence of lowered glucose concentration, apparently due to further induction of protective proteins.
Collapse
|
30
|
Narayanankutty A, Kottekkat A, Mathew SE, Illam SP, Suseela IM, Raghavamenon AC. Vitamin E supplementation modulates the biological effects of omega-3 fatty acids in naturally aged rats. Toxicol Mech Methods 2017; 27:207-214. [DOI: 10.1080/15376516.2016.1273431] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Arunaksharan Narayanankutty
- Department of Biochemistry, Amala Cancer Research Centre (recognized by University of Calicut), Thrissur, Kerala, India
| | - Anagha Kottekkat
- Department of Biochemistry, Amala Cancer Research Centre (recognized by University of Calicut), Thrissur, Kerala, India
| | - Shaji E. Mathew
- Department of Biochemistry, Amala Cancer Research Centre (recognized by University of Calicut), Thrissur, Kerala, India
| | - Soorya P. Illam
- Department of Biochemistry, Amala Cancer Research Centre (recognized by University of Calicut), Thrissur, Kerala, India
| | - Indu M. Suseela
- Department of Biochemistry, Amala Cancer Research Centre (recognized by University of Calicut), Thrissur, Kerala, India
| | - Achuthan C. Raghavamenon
- Department of Biochemistry, Amala Cancer Research Centre (recognized by University of Calicut), Thrissur, Kerala, India
| |
Collapse
|
31
|
Polus A, Zapala B, Razny U, Gielicz A, Kiec-Wilk B, Malczewska-Malec M, Sanak M, Childs CE, Calder PC, Dembinska-Kiec A. Omega-3 fatty acid supplementation influences the whole blood transcriptome in women with obesity, associated with pro-resolving lipid mediator production. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1746-1755. [PMID: 27531277 DOI: 10.1016/j.bbalip.2016.08.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 08/07/2016] [Accepted: 08/10/2016] [Indexed: 12/14/2022]
Abstract
The n-3 polyunsaturated fatty acids (PUFAs) eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) may reduce low-grade inflammation associated with obesity. The relationship between therapeutic response to n-3 PUFAs and modification of the transcriptome in obesity or metabolic syndrome remains to be explored. Blood samples were obtained from women with obesity before and after three-months supplementation with a moderate dose of n-3 PUFAs (1.8g EPA+DHA per day) or from controls. n-3 PUFAs (GC) and plasma concentrations of lipoxins, resolvins, protectin X (GC-MS/MS) and inflammatory markers (ELISA) were measured. Whole blood transcriptome was assayed using microarray. Women supplemented with n-3 PUFAs for 3months had significantly higher levels of EPA and DHA in plasma phosphatidylcholine. n-3 PUFA supplementation, in contrast to placebo, significantly decreased the concentrations of several inflammatory markers (SELE, MCP-1, sVCAM-1, sPECAM-1, and hsCRP), fasting triglycerides and insulin and increased the concentrations of pro-resolving DHA derivatives in plasma. The microarray data demonstrated effects of n-3 PUFAs on PPAR-α, NRF2 and NF-κB target genes. N-3 PUFAs increased DHA-derived pro-resolving mediators in women with obesity. Elevated resolvins and up-regulation of the resolvin receptor occurred in parallel with activation of PPAR-α target genes related to lipid metabolism and of NRF2 up-regulated antioxidant enzymes.
Collapse
Affiliation(s)
- Anna Polus
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, Krakow, Poland.
| | - Barbara Zapala
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Urszula Razny
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Anna Gielicz
- Department of Molecular Biology and Clinical Genetics Laboratory, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Beata Kiec-Wilk
- Department of Metabolic Disorders, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | | | - Marek Sanak
- Department of Molecular Biology and Clinical Genetics Laboratory, Collegium Medicum, Jagiellonian University, Krakow, Poland
| | - Caroline E Childs
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Philip C Calder
- Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, University of Southampton, Southampton, United Kingdom
| | - Aldona Dembinska-Kiec
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, Krakow, Poland
| |
Collapse
|
32
|
Cebula M, Schmidt EE, Arnér ESJ. TrxR1 as a potent regulator of the Nrf2-Keap1 response system. Antioxid Redox Signal 2015; 23:823-53. [PMID: 26058897 PMCID: PMC4589110 DOI: 10.1089/ars.2015.6378] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE All cells must maintain a balance between oxidants and reductants, while allowing for fluctuations in redox states triggered by signaling, altered metabolic flow, or extracellular stimuli. Furthermore, they must be able to rapidly sense and react to various challenges that would disrupt the redox homeostasis. RECENT ADVANCES Many studies have identified Keap1 as a key sensor for oxidative or electrophilic stress, with modification of Keap1 by oxidation or electrophiles triggering Nrf2-mediated transcriptional induction of enzymes supporting reductive and detoxification pathways. However, additional mechanisms for Nrf2 regulation are likely to exist upstream of, or in parallel with, Keap1. CRITICAL ISSUES Here, we propose that the mammalian selenoprotein thioredoxin reductase 1 (TrxR1) is a potent regulator of Nrf2. A high chemical reactivity of TrxR1 and its vital role for the thioredoxin (Trx) system distinguishes TrxR1 as a prime target for electrophilic challenges. Chemical modification of the selenocysteine (Sec) in TrxR1 by electrophiles leads to rapid inhibition of thioredoxin disulfide reductase activity, often combined with induction of NADPH oxidase activity of the derivatized enzyme, thereby affecting many downstream redox pathways. The notion of TrxR1 as a regulator of Nrf2 is supported by many publications on effects in human cells of selenium deficiency, oxidative stress or electrophile exposure, as well as the phenotypes of genetic mouse models. FUTURE DIRECTIONS Investigation of the role of TrxR1 as a regulator of Nrf2 activation will facilitate further studies of redox control in diverse cells and tissues of mammals, and possibly also in animals of other classes.
Collapse
Affiliation(s)
- Marcus Cebula
- 1 Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
| | - Edward E Schmidt
- 2 Microbiology and Immunology, Montana State University , Bozeman, Montana
| | - Elias S J Arnér
- 1 Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet , Stockholm, Sweden
| |
Collapse
|
33
|
Nagayama K, Morino K, Sekine O, Nakagawa F, Ishikado A, Iwasaki H, Okada T, Tawa M, Sato D, Imamura T, Nishio Y, Ugi S, Kashiwagi A, Okamura T, Maegawa H. Duality of n-3 Polyunsaturated Fatty Acids on Mcp-1 Expression in Vascular Smooth Muscle: A Potential Role of 4-Hydroxy Hexenal. Nutrients 2015; 7:8112-26. [PMID: 26402697 PMCID: PMC4586576 DOI: 10.3390/nu7095381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/24/2015] [Accepted: 08/31/2015] [Indexed: 01/12/2023] Open
Abstract
N-3 polyunsaturated fatty acids such as docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) have protective effects against atherosclerosis. Monocyte chemotactic protein (MCP)-1 is a major inflammatory mediator in the progression of atherosclerosis. However, little is known about the regulation of MCP-1 by DHA and EPA in vessels and vascular smooth muscle cells (VSMCs). In this study, we compared the effect of DHA and EPA on the expression of Mcp-1 in rat arterial strips and rat VSMCs. DHA, but not EPA, suppressed Mcp-1 expression in arterial strips. Furthermore, DHA generated 4-hydroxy hexenal (4-HHE), an end product of n-3 polyunsaturated fatty acids (PUFAs), in arterial strips as measured by liquid chromatography-tandem mass spectrometry. In addition, 4-HHE treatment suppressed Mcp-1 expression in arterial strips, suggesting 4-HHE derived from DHA may be involved in the mechanism of this phenomenon. In contrast, Mcp-1 expression was stimulated by DHA, EPA and 4-HHE through p38 kinase and the Keap1-Nuclear factor erythroid-derived 2-like 2 (Nrf2) pathway in VSMCs. In conclusion, there is a dual effect of n-3 PUFAs on the regulation of Mcp-1 expression. Further study is necessary to elucidate the pathological role of this phenomenon.
Collapse
MESH Headings
- Aldehydes/metabolism
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chromatography, Liquid
- Docosahexaenoic Acids/pharmacology
- Dose-Response Relationship, Drug
- Down-Regulation
- Eicosapentaenoic Acid/pharmacology
- In Vitro Techniques
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NF-E2-Related Factor 2/metabolism
- RNA Interference
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Tandem Mass Spectrometry
- Time Factors
- Transfection
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Kohji Nagayama
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Katsutaro Morino
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Osamu Sekine
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Fumiyuki Nakagawa
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
- Osaka Laboratory, JCL Bioassay Corporation, 5-16-26, Minamisuita, Suita-shi, Osaka 564-0043, Japan.
| | - Atsushi Ishikado
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
- Sunstar Inc., 3-1 Asahi-machi, Takatsuki, Osaka 569-1195, Japan.
- Joslin Diabetes Centre, Harvard Medical School, MA 02115, USA.
| | - Hirotaka Iwasaki
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Takashi Okada
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Masashi Tawa
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Daisuke Sato
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Takeshi Imamura
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Yoshihiko Nishio
- Department of Diabetes and Endocrine Medicine, Kagoshima University, Kagoshima 890-8580, Japan.
| | - Satoshi Ugi
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Atsunori Kashiwagi
- Kusatsu General Hospital, 1660, Yabase-cho, Kusatsu, Shiga 525-8585, Japan.
| | - Tomio Okamura
- Department of Pharmacology, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Shiga 520-2192, Japan.
| |
Collapse
|
34
|
ω-3 Fatty Acids and Cardiovascular Diseases: Effects, Mechanisms and Dietary Relevance. Int J Mol Sci 2015; 16:22636-61. [PMID: 26393581 PMCID: PMC4613328 DOI: 10.3390/ijms160922636] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/01/2015] [Accepted: 09/09/2015] [Indexed: 02/06/2023] Open
Abstract
ω-3 fatty acids (n-3 FA) have, since the 1970s, been associated with beneficial health effects. They are, however, prone to lipid peroxidation due to their many double bonds. Lipid peroxidation is a process that may lead to increased oxidative stress, a condition associated with adverse health effects. Recently, conflicting evidence regarding the health benefits of intake of n-3 from seafood or n-3 supplements has emerged. The aim of this review was thus to examine recent literature regarding health aspects of n-3 FA intake from fish or n-3 supplements, and to discuss possible reasons for the conflicting findings. There is a broad consensus that fish and seafood are the optimal sources of n-3 FA and consumption of approximately 2-3 servings per week is recommended. The scientific evidence of benefits from n-3 supplementation has diminished over time, probably due to a general increase in seafood consumption and better pharmacological intervention and acute treatment of patients with cardiovascular diseases (CVD).
Collapse
|
35
|
Gęgotek A, Skrzydlewska E. The role of transcription factor Nrf2 in skin cells metabolism. Arch Dermatol Res 2015; 307:385-96. [PMID: 25708189 PMCID: PMC4469773 DOI: 10.1007/s00403-015-1554-2] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/06/2015] [Accepted: 02/12/2015] [Indexed: 12/22/2022]
Abstract
Skin, which is a protective layer of the body, is in constant contact with physical and chemical environmental factors. Exposure of the skin to highly adverse conditions often leads to oxidative stress. Moreover, it has been observed that skin cells are also exposed to reactive oxygen species generated during cell metabolism particularly in relation to the synthesis of melanin or the metabolism in immune system cells. However, skin cells have special features that protect them against oxidative modifications including transcription factor Nrf2, which is responsible for the transcription of the antioxidant protein genes such as antioxidant enzymes, small molecular antioxidant proteins or interleukins, and multidrug response protein. In the present study, the mechanisms of Nrf2 activation have been compared in the cells forming the various layers of the skin: keratinocytes, melanocytes, and fibroblasts. The primary mechanism of control of Nrf2 activity is its binding by cytoplasmic inhibitor Keap1, while cells have also other controlling mechanisms, such as phosphorylation of Nrf2 and modifications of its activators (e.g., Maf, IKKβ) or inhibitors (e.g., Bach1, caveolae, TGF-β). Moreover, there are a number of drugs (e.g., ketoconazole) used in the pharmacotherapy of skin diseases based on the activation of Nrf2, but they may also induce oxidative stress. Therefore, it is important to look for compounds that cause a selective activation of Nrf2 particularly natural substances such as curcumin, sulforaphane, or extracts from the broccoli leaves without side effects. These findings could be helpful in the searching for new drugs for people with vitiligo or even melanoma.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Departments of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland,
| | | |
Collapse
|
36
|
Milkovic L, Cipak Gasparovic A, Zarkovic N. Overview on major lipid peroxidation bioactive factor 4-hydroxynonenal as pluripotent growth-regulating factor. Free Radic Res 2015; 49:850-60. [PMID: 25532703 DOI: 10.3109/10715762.2014.999056] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The reactive aldehyde 4-hydroxynonenal (HNE) is major bioactive marker of lipid peroxidation generated under oxidative stress from polyunsaturated fatty acids. Biomedical significance of HNE was first revealed in pathogenesis of various degenerative and malignant diseases. Thus, HNE was considered for decades only as cytotoxic molecule, "second toxic messenger of free radicals" responsible for numerous undesirable consequences of oxidative stress. However, the increase of knowledge on physiology of redox signaling revealed also desirable, physiological roles of HNE, especially in the field of cellular signaling pathways regulating proliferation, differentiation, and apoptosis. These pluripotent effects of HNE can be explained by its concentration-dependent interactions with the cytokine networks and complex cellular antioxidant systems also showing cell and tissue specificities. Therefore, this paper gives a comprehensive, yet short overview on HNE as pluripotent growth-regulating factor.
Collapse
Affiliation(s)
- L Milkovic
- Laboratory for Oxidative Stress, Rudjer Boskovic Institute , Zagreb , Croatia
| | | | | |
Collapse
|
37
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
38
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
39
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438\] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
40
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438;] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
41
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438"] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
42
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [PMID: 24999379 DOI: 10.1155/2014/360438-- or] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
43
|
Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:360438. [PMID: 24999379 PMCID: PMC4066722 DOI: 10.1155/2014/360438] [Citation(s) in RCA: 3147] [Impact Index Per Article: 314.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/24/2014] [Indexed: 02/07/2023]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970-1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010-2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews of in vivo mammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
44
|
Tang Y, Jacobi A, Vater C, Zou X, Stiehler M. Salvianolic acid B protects human endothelial progenitor cells against oxidative stress-mediated dysfunction by modulating Akt/mTOR/4EBP1, p38 MAPK/ATF2, and ERK1/2 signaling pathways. Biochem Pharmacol 2014; 90:34-49. [PMID: 24780446 DOI: 10.1016/j.bcp.2014.04.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/14/2014] [Accepted: 04/14/2014] [Indexed: 12/01/2022]
Abstract
The vascular endothelium is specifically sensitive to oxidative stress, and this is one of the mechanisms that causes widespread endothelial dysfunction in most cardiovascular diseases and disorders. Protection against reactive oxygen species (ROS)-mediated oxidative damage via antioxidant mechanisms is essential for tissue maintenance and shows therapeutic potential for patients suffering from cardiovascular and metabolic disorders. Salvianolic acid B (SalB), a natural bioactive component known from Traditional Chinese Medicine, has been reported to exert cellular protection in various types of cells. However, the underlying mechanisms involved are not fully understood. Here, we showed that SalB significantly promoted the migratory and tube formation abilities of human bone marrow derived-endothelial progenitor cells (BM-EPCs) in vitro, and substantially abrogated hydrogen peroxide (H2O2)-induced cell damage. SalB down-regulated Nox4 and eNOS, as well as nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase expression upon H2O2 induction that in turn prevents oxidative-induced endothelial dysfunction. Moreover, SalB suppressed the Bax/Bcl-xL ratio and caspase-3 activation after H2O2 induction. Furthermore, our results provide mechanistic evidence that activation of the mTOR/p70S6K/4EBP1 pathways is required for both SalB-mediated angiogenic and protective effects against oxidative stress-induced cell injury in BM-EPCs. Suppression of MKK3/6-p38 MAPK-ATF2 and ERK1/2 signaling pathways by SalB significantly protected BM-EPCs against cell injury caused by oxidative stress via reduction of intracellular ROS levels and apoptosis. Taken together, by providing a mechanistic insight into the modulation of redox states in BM-EPCs by SalB, we suggest that SalB has a strong potential of being a new proangiogenic and cytoprotective therapeutic agent with applications in the field of endothelial injury-mediated vascular diseases.
Collapse
Affiliation(s)
- Yubo Tang
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany; Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, China.
| | - Angela Jacobi
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany.
| | - Corina Vater
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany.
| | - Xuenong Zou
- Department of Spinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, China.
| | - Maik Stiehler
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
45
|
Calay D, Mason JC. The multifunctional role and therapeutic potential of HO-1 in the vascular endothelium. Antioxid Redox Signal 2014; 20:1789-809. [PMID: 24131232 DOI: 10.1089/ars.2013.5659] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. In the vasculature, particular interest has focused on antioxidant and anti-inflammatory properties of the inducible HO-1 isoform in the vascular endothelium. This review will present evidence that illustrates the potential therapeutic significance of HO-1 and its products, with special emphasis placed on their beneficial effects on the endothelium in vascular diseases. RECENT ADVANCES The understanding of the molecular basis for the regulation and functions of HO-1 has led to the identification of a variety of drugs that increase HO-1 activity in the vascular endothelium. Moreover, therapeutic delivery of HO-1 products CO, biliverdin, and bilirubin has been shown to have favorable effects, notably on endothelial cells and in animal models of vascular disease. CRITICAL ISSUES To date, mechanistic data identifying the downstream target genes utilized by HO-1 and its products to exert their actions remain relatively sparse. Likewise, studies in man to investigate the efficacy of therapeutics known to induce HO-1 or the consequences of the tissue-specific delivery of CO or biliverdin/bilirubin are rarely performed. FUTURE DIRECTIONS Based on the promising in vivo data from animal models, clinical trials to explore the safety and efficacy of the therapeutic induction of HO-1 and the delivery of its products should now be pursued further, targeting, for example, patients with severe atherosclerotic disease, ischemic limbs, restenosis injury, or at high risk of organ rejection.
Collapse
Affiliation(s)
- Damien Calay
- Vascular Sciences Unit, National Heart and Lung Institute , Imperial Centre for Translational & Experimental Medicine, Imperial College London Hammersmith Hospital, London, United Kingdom
| | | |
Collapse
|
46
|
Aldini G, Carini M, Yeum KJ, Vistoli G. Novel molecular approaches for improving enzymatic and nonenzymatic detoxification of 4-hydroxynonenal: toward the discovery of a novel class of bioactive compounds. Free Radic Biol Med 2014; 69:145-56. [PMID: 24456906 DOI: 10.1016/j.freeradbiomed.2014.01.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/10/2014] [Accepted: 01/10/2014] [Indexed: 11/18/2022]
Abstract
4-Hydroxy-trans-2-nonenal (HNE), an α,β-unsaturated aldehyde generated endogenously by the radical-mediated peroxidation of ω-6 polyunsaturated fatty acids, is a bioactive molecule acting in several physiopathological mechanisms and most of its activity is due to the covalent modification of biomolecules. Although at low and physiological levels HNE acts as an endogenous signaling molecule, a growing bulk of evidence indicates that at high and toxic concentrations, HNE is involved in the onset and propagation of several human diseases. To get more conclusive evidence of HNE as a pathogenetic factor, a pharmacological tool able to inhibit the HNE-induced cellular response is required. Such compound is currently not available, although several molecular strategies have so far been reported with the aim of inhibiting HNE formation or catalyzing its removal. Although most of these are not selective, such strategies have been found to induce several biological responses and would merit further investigation. In this review the various strategies are reported and discussed together with their limits and potentials.
Collapse
Affiliation(s)
- Giancarlo Aldini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy.
| | - Marina Carini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Kyung-Jin Yeum
- Division of Food Bioscience, College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Republic of Korea
| | - Giulio Vistoli
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
47
|
Lipid Peroxidation: Production, Metabolism, and Signaling Mechanisms of Malondialdehyde and 4-Hydroxy-2-Nonenal. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014. [DOI: 10.1155/2014/360438 and (select 9530 from(select count(*),concat(0x716b6b7171,(select (elt(9530=9530,1))),0x7178627171,floor(rand(0)*2))x from information_schema.plugins group by x)a)] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970–1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010–2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews ofin vivomammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
48
|
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970–1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010–2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews ofin vivomammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
49
|
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970–1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010–2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews ofin vivomammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|
50
|
Abstract
Lipid peroxidation can be described generally as a process under which oxidants such as free radicals attack lipids containing carbon-carbon double bond(s), especially polyunsaturated fatty acids (PUFAs). Over the last four decades, an extensive body of literature regarding lipid peroxidation has shown its important role in cell biology and human health. Since the early 1970s, the total published research articles on the topic of lipid peroxidation was 98 (1970–1974) and has been increasing at almost 135-fold, by up to 13165 in last 4 years (2010–2013). New discoveries about the involvement in cellular physiology and pathology, as well as the control of lipid peroxidation, continue to emerge every day. Given the enormity of this field, this review focuses on biochemical concepts of lipid peroxidation, production, metabolism, and signaling mechanisms of two main omega-6 fatty acids lipid peroxidation products: malondialdehyde (MDA) and, in particular, 4-hydroxy-2-nonenal (4-HNE), summarizing not only its physiological and protective function as signaling molecule stimulating gene expression and cell survival, but also its cytotoxic role inhibiting gene expression and promoting cell death. Finally, overviews ofin vivomammalian model systems used to study the lipid peroxidation process, and common pathological processes linked to MDA and 4-HNE are shown.
Collapse
|