51
|
Poganik JR, Long MJC, Aye Y. Getting the Message? Native Reactive Electrophiles Pass Two Out of Three Thresholds to be Bona Fide Signaling Mediators. Bioessays 2018; 40:e1700240. [PMID: 29603288 PMCID: PMC6488019 DOI: 10.1002/bies.201700240] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/24/2018] [Indexed: 12/11/2022]
Abstract
Precision cell signaling activities of reactive electrophilic species (RES) are arguably among the most poorly-understood means to transmit biological messages. Latest research implicates native RES to be a chemically-distinct subset of endogenous redox signals that influence cell decision making through non-enzyme-assisted modifications of specific proteins. Yet, fundamental questions remain regarding the role of RES as bona fide second messengers. Here, we lay out three sets of criteria we feel need to be met for RES to be considered as true cellular signals that directly mediate information transfer by modifying "first-responding" sensor proteins. We critically assess the available evidence and define the extent to which each criterion has been fulfilled. Finally, we offer some ideas on the future trajectories of the electrophile signaling field taking inspiration from work that has been done to understand canonical signaling mediators. Also see the video abstract here: https://youtu.be/rG7o0clVP0c.
Collapse
Affiliation(s)
- Jesse R. Poganik
- Department of Chemistry and Chemical Biology Cornell University Ithaca, NY 14853, USA
| | - Marcus J. C. Long
- Department of Chemistry and Chemical Biology Cornell University Ithaca, NY 14853, USA
| | - Yimon Aye
- Department of Chemistry and Chemical Biology Cornell University Ithaca, NY 14853, USA
- Department of Biochemistry Weill Cornell Medicine New York, NY 10065, USA
| |
Collapse
|
52
|
Hlusicka J, Loster T, Lischkova L, Vaneckova M, Seidl Z, Diblik P, Kuthan P, Urban P, Navratil T, Kacer P, Zakharov S. Role of activation of lipid peroxidation in the mechanisms of acute methanol poisoning. Clin Toxicol (Phila) 2018; 56:893-903. [DOI: 10.1080/15563650.2018.1455980] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jiri Hlusicka
- Toxicological Information Centre, General University Hospital in Prague, Prague, Czech Republic
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas Loster
- Faculty of Informatics and Statistics, Department of Statistics and Probability, University of Economics, Prague, Czech Republic
| | - Lucie Lischkova
- Toxicological Information Centre, General University Hospital in Prague, Prague, Czech Republic
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Zdenek Seidl
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Diblik
- Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Kuthan
- Department of Ophthalmology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Urban
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Centre for Industrial Hygiene and Occupational Medicine, National Institute of Public Health, Prague, Czech Republic
| | - Tomas Navratil
- J. Heyrovsky Institute of Physical Chemistry, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petr Kacer
- Biocev, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Sergey Zakharov
- Toxicological Information Centre, General University Hospital in Prague, Prague, Czech Republic
- Department of Occupational Medicine, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
53
|
Aidoud N, Delplanque B, Baudry C, Garcia C, Moyon A, Balasse L, Guillet B, Antona C, Darmaun D, Fraser K, Ndiaye S, Leruyet P, Martin JC. A combination of lipidomics, MS imaging, and PET scan imaging reveals differences in cerebral activity in rat pups according to the lipid quality of infant formulas. FASEB J 2018; 32:4776-4790. [DOI: 10.1096/fj.201800034r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Nacima Aidoud
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN)INSERMInstitut National de la Recherche Agricole (INRA)BioMetMarseilleFrance
| | - Bernadette Delplanque
- Laboratoire de Neuroendocrinologie Moléculaire de la Prise Alimentaire (NMPA)Centre de Neurosciences Université Paris-Sud (CNPS)OrsayFrance
| | | | - Cyrielle Garcia
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN)INSERMInstitut National de la Recherche Agricole (INRA)BioMetMarseilleFrance
| | - Anais Moyon
- Centre Européen de Recherche en Imagerie Médicale (CERIMED)Université d'Aix-MarseilleMarseilleFrance
| | - Laure Balasse
- Centre Européen de Recherche en Imagerie Médicale (CERIMED)Université d'Aix-MarseilleMarseilleFrance
| | - Benjamin Guillet
- Centre Européen de Recherche en Imagerie Médicale (CERIMED)Université d'Aix-MarseilleMarseilleFrance
| | - Claudine Antona
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN)INSERMInstitut National de la Recherche Agricole (INRA)BioMetMarseilleFrance
| | - Dominique Darmaun
- INRAPhysiopathologie des Adaptations Nutritionnelles (PHAN)Université de NantesNantesFrance
- Thermo Fisher ScientificCourtaboeufFrance
| | - Karl Fraser
- Food Nutrition and Health TeamFood and Bio-Based Products GroupAgResearch Grasslands Research CentreRiddet InstituteMassey UniversityPalmerston NorthNew Zealand
| | - Sega Ndiaye
- INRAPhysiopathologie des Adaptations Nutritionnelles (PHAN)Université de NantesNantesFrance
- Thermo Fisher ScientificCourtaboeufFrance
| | | | - Jean-Charles Martin
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN)INSERMInstitut National de la Recherche Agricole (INRA)BioMetMarseilleFrance
| |
Collapse
|
54
|
Gallo G, Bruno R, Taranto A, Martino G. Are Polyunsaturated Fatty Acid Metabolites, the Protective Effect of 4-hydroxytyrosol on Human Red Blood Cell Membranes and Oxidative Damage (4-hydroxyalkenals) Compatible in Hypertriglyceridemic Patients? Pharmacogn Mag 2017; 13:S561-S566. [PMID: 29142415 PMCID: PMC5669098 DOI: 10.4103/pm.pm_483_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/08/2016] [Indexed: 12/20/2022] Open
Abstract
Background: Increased levels of malondialdehyde (MDA) and 4-hydroxynonenal (HNE) are demonstrated in plasma of uremic patients. A study showed that the comparison of erythrocytes of healthy and diseased patients (obese, hypertensive, and Type 2 diabetics) with age is associated to a disturbed oxidant/antioxidant balance when obesity is associated with hypertension. 4-hydroxytyrosol is shown to significantly protect red blood cells (RBCs) from oxidative damage (4-HNE). In literature, there are partial discussions on the role of lipids and their oxidation products. The products of degradation of membrane proteins are observed as self-consisting products without interrelations with membrane lipids. Objective: The aim of this study is to evaluate the role of polyunsaturated fatty acid (PUFA) metabolites on oxidative damage (4-hydroxy-alkenals) in RBCs of hypertriglyceridemic patients after membrane treatment with 4-hydroxytyrosol. Materials and Methods: The authors optimize the isolation of RBC ghosts and spectrophotometric method to measure free 4-hydroxyalkenals in human RBC membranes and investigated the effect on oxidative damage in human erythrocyte membranes and in vitro 4-hydroxytyrosol treatment to evaluate the membrane lipids reducible by this phenol. Results: Plasma triglyceride levels in patients are clearly higher than in controls. Moreover, total membrane proteins data are similar to previous described. The normalized alkenals levels are significantly enhanced in hyperlipemic patients in comparison to normoglyceridemic controls. After the 4-hydroxytyrosol action, lipid metabolites substantially decrease. The ratio of oxidized lipids (MDA + HNE) and membrane proteins data are similar to previously described ones. Conclusion: According to experimental data, the accumulation of the alkenals in RBC membrane could be produced either by partial PUFA oxidation contained in glycerides and plasma glycerides and by glycerides into plasma membrane recycled RBC. SUMMARY Hypertriglyceridemia induces oxidative stress in human red blood cell (RBC) membranes Oxidative stress causes increased plasma membrane total protein concentration and hydroxynonenal and malondialdehyde levels The authors optimize the isolation of RBC ghosts and spectrophotometric method to measure free 4-hydroxyalkenals in human RBC membranes After the reduction with 4-hydroxytyrosol, oxidized lipid concentration significantly decrease.
Abbreviations used: RBC: Red blood cell; MDA: Malondialdehyde; HNE\HAE: 4-hydroxyalkenals; LPO: Lipid peroxidation; ROS: Reactive oxygen species; ORAC: Oxygen Radical Absorbance Capacity.
Collapse
Affiliation(s)
- Giuseppe Gallo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende (CS), Italy
| | - Rosalinda Bruno
- Department of Pharmacy and Health Sciences and Nutrition, University of Calabria, Rende (CS), Italy
| | - Adele Taranto
- Regione Calabria ASP Cosenza, Hospital Giovanni Iannelli, Cetraro (CS), Italy
| | - Guglielmo Martino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende (CS), Italy
| |
Collapse
|
55
|
Zuo W, Hu X, Yang Y, Jiang L, Ren L, Huang H. Development of an Improved Method to Determine Saturated Aliphatic Aldehydes in Docosahexaenoic Acid-Rich Oil: A Supplement to p
-Anisidine Value. EUR J LIPID SCI TECH 2017. [DOI: 10.1002/ejlt.201700243] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Wenlu Zuo
- Jiangsu National Synergetic Innovation Center for Advanced Materials, College of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences; Nanjing Tech University; No. 30 South Puzhu Road Nanjing 211816 People's Republic of China
| | - Xuechao Hu
- Jiangsu National Synergetic Innovation Center for Advanced Materials, College of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences; Nanjing Tech University; No. 30 South Puzhu Road Nanjing 211816 People's Republic of China
| | - Yaqiong Yang
- Jiangsu National Synergetic Innovation Center for Advanced Materials, College of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences; Nanjing Tech University; No. 30 South Puzhu Road Nanjing 211816 People's Republic of China
| | - Ling Jiang
- Jiangsu National Synergetic Innovation Center for Advanced Materials, College of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences; Nanjing Tech University; No. 30 South Puzhu Road Nanjing 211816 People's Republic of China
| | - Lujing Ren
- Jiangsu National Synergetic Innovation Center for Advanced Materials, College of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences; Nanjing Tech University; No. 30 South Puzhu Road Nanjing 211816 People's Republic of China
| | - He Huang
- Jiangsu National Synergetic Innovation Center for Advanced Materials, College of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences; Nanjing Tech University; No. 30 South Puzhu Road Nanjing 211816 People's Republic of China
| |
Collapse
|
56
|
Romano A, Serviddio G, Calcagnini S, Villani R, Giudetti AM, Cassano T, Gaetani S. Linking lipid peroxidation and neuropsychiatric disorders: focus on 4-hydroxy-2-nonenal. Free Radic Biol Med 2017; 111:281-293. [PMID: 28063940 DOI: 10.1016/j.freeradbiomed.2016.12.046] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/27/2016] [Accepted: 12/30/2016] [Indexed: 12/25/2022]
Abstract
4-hydroxy-2-nonenal (HNE) is considered to be a strong marker of oxidative stress; the interaction between HNE and cellular proteins leads to the formation of HNE-protein adducts able to alter cellular homeostasis and cause the development of a pathological state. By virtue of its high lipid concentration, oxygen utilization, and the presence of metal ions participating to redox reactions, the brain is highly susceptible to the formation of free radicals and HNE-related compounds. A variety of neuropsychiatric disorders have been associated with elevations of HNE concentration. For example, increased levels of HNE were found in the cortex of bipolar and schizophrenic patients, while HNE plasma concentrations resulted high in patients with major depression. On the same line, high brain concentrations of HNE were found associated with Huntington's inclusions. The incidence of high HNE levels is relevant also in the brain and cerebrospinal fluid of patients suffering from Parkinson's disease. Intriguingly, in this case the increase of HNE was associated with an accumulation of iron in the substantia nigra, a brain region highly affected by the pathology. In the present review we recapitulate the findings supporting the role of HNE in the pathogenesis of different neuropsychiatric disorders to highlight the pathogenic mechanisms ascribed to HNE accumulation. The aim of this review is to offer novel perspectives both for the understanding of etiopathogenetic mechanisms that remain still unclear and for the identification of new useful biological markers. We conclude suggesting that targeting HNE-driven cellular processes may represent a new more efficacious therapeutical intervention.
Collapse
Affiliation(s)
- Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, Via Luigi Pinto, c/o Ospedali Riuniti, 71122 Foggia, Italy
| | - Silvio Calcagnini
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Rosanna Villani
- Department of Medical and Surgical Sciences, University of Foggia, Via Luigi Pinto, c/o Ospedali Riuniti, 71122 Foggia, Italy
| | - Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Centro Ecotekne, sp Lecce-Monteroni 73100 Lecce, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, Via Luigi Pinto, c/o Ospedali Riuniti, 71122 Foggia, Italy.
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| |
Collapse
|
57
|
Sousa BC, Pitt AR, Spickett CM. Chemistry and analysis of HNE and other prominent carbonyl-containing lipid oxidation compounds. Free Radic Biol Med 2017; 111:294-308. [PMID: 28192230 DOI: 10.1016/j.freeradbiomed.2017.02.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/28/2017] [Accepted: 02/01/2017] [Indexed: 01/02/2023]
Abstract
The process of lipid oxidation generates a diverse array of small aldehydes and carbonyl-containing compounds, which may occur in free form or esterified within phospholipids and cholesterol esters. These aldehydes mostly result from fragmentation of fatty acyl chains following radical oxidation, and the products can be subdivided into alkanals, alkenals (usually α,β-unsaturated), γ-substituted alkenals and bis-aldehydes. Isolevuglandins are non-fragmented di-carbonyl compounds derived from H2-isoprostanes, and oxidation of the ω-3-fatty acid docosahexenoic acid yield analogous 22 carbon neuroketals. Non-radical oxidation by hypochlorous acid can generate α-chlorofatty aldehydes from plasmenyl phospholipids. Most of these compounds are reactive and have generally been considered as toxic products of a deleterious process. The reactivity is especially high for the α,β-unsaturated alkenals, such as acrolein and crotonaldehyde, and for γ-substituted alkenals, of which 4-hydroxy-2-nonenal and 4-oxo-2-nonenal are best known. Nevertheless, in recent years several previously neglected aldehydes have been investigated and also found to have significant reactivity and biological effects; notable examples are 4-hydroxy-2-hexenal and 4-hydroxy-dodecadienal. This has led to substantial interest in the biological effects of all of these lipid oxidation products and their roles in disease, including proposals that HNE is a second messenger or signalling molecule. However, it is becoming clear that many of the effects elicited by these compounds relate to their propensity for forming adducts with nucleophilic groups on proteins, DNA and specific phospholipids. This emphasizes the need for good analytical methods, not just for free lipid oxidation products but also for the resulting adducts with biomolecules. The most informative methods are those utilizing HPLC separations and mass spectrometry, although analysis of the wide variety of possible adducts is very challenging. Nevertheless, evidence for the occurrence of lipid-derived aldehyde adducts in biological and clinical samples is building, and offers an exciting area of future research.
Collapse
Affiliation(s)
- Bebiana C Sousa
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Andrew R Pitt
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Corinne M Spickett
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK.
| |
Collapse
|
58
|
Sasson S. Nutrient overload, lipid peroxidation and pancreatic beta cell function. Free Radic Biol Med 2017; 111:102-109. [PMID: 27600453 DOI: 10.1016/j.freeradbiomed.2016.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 12/16/2022]
Abstract
Since the landmark discovery of α,β-unsaturated 4-hydroxyalkenals by Esterbauer and colleagues most studies have addressed the consequences of the tendency of these lipid peroxidation products to form covalent adducts with macromolecules and modify cellular functions. Many studies describe detrimental and cytotoxic effects of 4-hydroxy-2E-nonenal (4-HNE) in myriad tissues and organs and many pathologies. Other studies similarly assigned unfavorable effects to 4-hydroxy-2E-hexenal (4-HHE) and 4-hydroxy-2E,6Z-dodecadienal (4-HDDE). Nutrient overload (e.g., hyperglycemia, hyperlipidemia) modifies lipid metabolism in cells and promotes lipid peroxidation and the generation of α,β-unsaturated 4-hydroxyalkenals. Advances glycation- and lipoxidation end products (AGEs and ALEs) have been associated with the development of insulin resistance and pancreatic beta cell dysfunction and the etiology of type 2 diabetes and its peripheral complications. Less acknowledged are genuine signaling properties of 4-hydroxyalkenals in hormetic processes that provide defense against the consequences of nutrient overload. This review addresses recent findings on such lipohormetic mechanisms that are associated with lipid peroxidation in pancreatic beta cells. This article is part of a Special Issue entitled SI: LIPID OXIDATION PRODUCTS, edited by Giuseppe Poli.
Collapse
Affiliation(s)
- Shlomo Sasson
- Institute for Drug Research, Section of Pharmacology, Diabetes Research Unit, Hebrew University Faculty of Medicine, Jerusalem 9112001, Israel.
| |
Collapse
|
59
|
Łuczaj W, Gęgotek A, Skrzydlewska E. Antioxidants and HNE in redox homeostasis. Free Radic Biol Med 2017; 111:87-101. [PMID: 27888001 DOI: 10.1016/j.freeradbiomed.2016.11.033] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/11/2022]
Abstract
Under physiological conditions, cells are in a stable state known as redox homeostasis, which is maintained by the balance between continuous ROS/RNS generation and several mechanisms involved in antioxidant activity. ROS overproduction results in alterations in the redox homeostasis that promote oxidative damage to major components of the cell, including the biomembrane phospholipids. Lipid peroxidation subsequently generates a diverse set of products, including α,β-unsaturated aldehydes. Of these products, 4-hydroxy-2-nonenal (HNE) is the most studied aldehyde on the basis of its involvement in cellular physiology and pathology. This review summarizes the current knowledge in the field of HNE generation, metabolism, and detoxification, as well as its interactions with various cellular macromolecules (protein, phospholipid, and nucleic acid). The formation of HNE-protein adducts enables HNE to participate in multi-step regulation of cellular metabolic pathways that include signaling and transcription of antioxidant enzymes, pro-inflammatory factors, and anti-apoptotic proteins. The most widely described roles for HNE in the signaling pathways are associated with its activation of kinases, as well as transcription factors that are responsible for redox homeostasis (Ref-1, Nrf2, p53, NFκB, and Hsf1). Depending on its level, HNE exerts harmful or protective effects associated with the induction of antioxidant defense mechanisms. These effects make HNE a key player in maintaining redox homeostasis, as well as producing imbalances in this system that participate in aging and the development of pathological conditions.
Collapse
Affiliation(s)
- Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2d, 15-222 Bialystok, Poland
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2d, 15-222 Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2d, 15-222 Bialystok, Poland.
| |
Collapse
|
60
|
Cesselli D, Aleksova A, Sponga S, Cervellin C, Di Loreto C, Tell G, Beltrami AP. Cardiac Cell Senescence and Redox Signaling. Front Cardiovasc Med 2017; 4:38. [PMID: 28612009 PMCID: PMC5447053 DOI: 10.3389/fcvm.2017.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
Aging is characterized by a progressive loss of the ability of the organism to cope with stressors and to repair tissue damage. As a result, chronic diseases, including cardiovascular disease, increase their prevalence with aging, underlining the existence of common mechanisms that lead to frailty and age-related diseases. In this frame, the progressive decline of the homeostatic and reparative function of primitive cells has been hypothesized to play a major role in the evolution of cardiac pathology to heart failure. Although initially it was believed that reactive oxygen species (ROS) were produced in an unregulated manner as a byproduct of cellular metabolism, causing macromolecular damage and aging, accumulating evidence indicate the major role played by redox signaling in physiology. Aim of this review is to critically revise evidence linking ROS to cell senescence and aging and to provide evidence of the primary role played by redox signaling, with a particular emphasis on the multifunctional protein APE1/Ref in stem cell biology. Finally, we will discuss evidence supporting the role of redox signaling in cardiovascular cells.
Collapse
Affiliation(s)
| | - Aneta Aleksova
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Trieste, Italy
| | - Sandro Sponga
- Cardiothoracic Surgery, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | | | | | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | | |
Collapse
|
61
|
Nelson MAM, Baba SP, Anderson EJ. Biogenic Aldehydes as Therapeutic Targets for Cardiovascular Disease. Curr Opin Pharmacol 2017; 33:56-63. [PMID: 28528297 DOI: 10.1016/j.coph.2017.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/28/2017] [Accepted: 04/19/2017] [Indexed: 12/21/2022]
Abstract
Aldehydes are continuously formed in biological systems through enzyme-dependent and spontaneous oxidation of lipids, glucose, and primary amines. These highly reactive, biogenic electrophiles can become toxic via covalent modification of proteins, lipids and DNA. Thus, agents that scavenge aldehydes through conjugation have therapeutic value for a number of major cardiovascular diseases. Several commonly-prescribed drugs (e.g., hydralazine) have been shown to have potent aldehyde-conjugating properties which may contribute to their beneficial effects. Herein, we briefly describe the major sources and toxicities of biogenic aldehydes in cardiovascular system, and provide an overview of drugs that are known to have aldehyde-conjugating effects. Some compounds of phytochemical origin, and histidyl-dipeptides with emerging therapeutic value in this area are also discussed.
Collapse
Affiliation(s)
- Margaret-Ann M Nelson
- Department of Pharmacology & Toxicology, East Carolina University, Greenville, NC, USA
| | - Shahid P Baba
- Department of Medicine, Diabetes and Obesity Center, University of Louisville, Louisville, KY, USA
| | - Ethan J Anderson
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
62
|
Zhong CB, Chen X, Zhou XY, Wang XB. The Role of Peroxisome Proliferator-Activated Receptor γ in Mediating Cardioprotection Against Ischemia/Reperfusion Injury. J Cardiovasc Pharmacol Ther 2017; 23:46-56. [PMID: 28466688 DOI: 10.1177/1074248417707049] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Myocardial infarction (MI) is a serious cardiovascular disease resulting in high rates of morbidity and mortality. Although advances have been made in restoring myocardial perfusion in ischemic areas, decreases in cardiomyocyte death and infarct size are still limited, attributing to myocardial ischemia/reperfusion (I/R) injury. It is necessary to develop therapies to restrict myocardial I/R injury and protect cardiomyocytes against further damage after MI. Many studies have suggested that peroxisome proliferator-activated receptor γ (PPARγ), a ligand-inducible nuclear receptor that predominantly regulates glucose and lipid metabolism, is a promising therapeutic target for ameliorating myocardial I/R injury. Thus, this review focuses on the role of PPARγ in cardioprotection during myocardial I/R. The cardioprotective effects of PPARγ, including attenuating oxidative stress, inhibiting inflammatory responses, improving glucose and lipid metabolism, and antagonizing apoptosis, are described. Additionally, the underlying mechanisms of cardioprotective effects of PPARγ, such as regulating the expression of target genes, influencing other transcription factors, and modulating kinase signaling pathways, are further discussed.
Collapse
Affiliation(s)
- Chong-Bin Zhong
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xi Chen
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xu-Yue Zhou
- 1 The Second Clinical Institute of Southern Medical University, Guangzhou, China
| | - Xian-Bao Wang
- 2 Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
63
|
Grasso GI, Bellia F, Arena G, Satriano C, Vecchio G, Rizzarelli E. Multitarget trehalose-carnosine conjugates inhibit Aβ aggregation, tune copper(II) activity and decrease acrolein toxicity. Eur J Med Chem 2017; 135:447-457. [PMID: 28475972 DOI: 10.1016/j.ejmech.2017.04.060] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/06/2017] [Accepted: 04/22/2017] [Indexed: 01/12/2023]
Abstract
Increasing evidence is accumulating, showing that neurodegenerative disorders are somehow associated with the toxicity of amyloid aggregates, metal ion dyshomeostasis as well as with products generated by oxidative stress. Within the biological oxidation products, acrolein does have a prominent role. A promising strategy to deal with the above neurogenerative disorders is to use multi-functions bio-molecules. Herein, we show how a class of bio-conjugates takes advantage of the antiaggregating, antioxidant and antiglycating properties of trehalose and carnosine. Their ability to sequester acrolein and to inhibit both self- and metal-induced aggregation is here reported. The copper(II) coordination properties of a new trehalose-carnosine conjugate and the relative antioxidant effects have also been investigated.
Collapse
Affiliation(s)
- Giuseppa Ida Grasso
- Institute of Biostructure and Bioimaging, National Research Council (CNR), via P. Gaifami 18, 95126, Catania, Italy.
| | - Francesco Bellia
- Institute of Biostructure and Bioimaging, National Research Council (CNR), via P. Gaifami 18, 95126, Catania, Italy.
| | - Giuseppe Arena
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Cristina Satriano
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Graziella Vecchio
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Enrico Rizzarelli
- Institute of Biostructure and Bioimaging, National Research Council (CNR), via P. Gaifami 18, 95126, Catania, Italy; Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
64
|
Shotgun lipidomics in substantiating lipid peroxidation in redox biology: Methods and applications. Redox Biol 2017; 12:946-955. [PMID: 28494428 PMCID: PMC5423350 DOI: 10.1016/j.redox.2017.04.030] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 04/10/2017] [Accepted: 04/23/2017] [Indexed: 11/27/2022] Open
Abstract
Multi-dimensional mass spectrometry-based shotgun lipidomics (MDMS-SL) has made profound advances for comprehensive analysis of cellular lipids. It represents one of the most powerful tools in analyzing lipids directly from lipid extracts of biological samples. It enables the analysis of nearly 50 lipid classes and thousands of individual lipid species with high accuracy/precision. The redox imbalance causes oxidative stress, resulting in lipid peroxidation, and alterations in lipid metabolism and homeostasis. Some lipid classes such as oxidized fatty acids, 4-hydroxyalkenal species, and plasmalogen are sensitive to oxidative stress or generated corresponding to redox imbalance. Therefore, accurate assessment of these lipid classes can provide not only the redox states, but also molecular insights into the pathogenesis of diseases. This review focuses on the advances of MDMS-SL in analysis of these lipid classes and molecular species, and summarizes their recent representative applications in biomedical/biological research. We believe that MDMS-SL can make great contributions to redox biology through substantiating the aberrant lipid metabolism, signaling, trafficking, and homeostasis under oxidative stress-related condition.
Collapse
|
65
|
Comparison of protective effect of ascorbic acid on redox and endocannabinoid systems interactions in in vitro cultured human skin fibroblasts exposed to UV radiation and hydrogen peroxide. Arch Dermatol Res 2017; 309:285-303. [PMID: 28285367 PMCID: PMC5387039 DOI: 10.1007/s00403-017-1729-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/27/2017] [Accepted: 02/22/2017] [Indexed: 02/07/2023]
Abstract
The mechanisms of biological activity of commonly used natural compounds are constantly examined. Therefore, the aim of this study was to compare ascorbic acid efficacy in counteracting the consequences of UV and hydrogen peroxide treatment on lipid mediators and their regulative action on antioxidant abilities. Skin fibroblasts exposed to UVA and UVB irradiation, treated with hydrogen peroxide and ascorbic acid. The redox system was estimated through reactive oxygen species (ROS) generation (electron spin resonance spectrometer) and antioxidants level/activity (HPLC/spectrometry) which activity was evaluated by the level of phospholipid metabolites: 4-hydroxynonenal, malondialdehyde, 8-isoprostanes and endocannabinoids (GC/LC-MS) in the human skin fibroblasts. Protein and DNA oxidative modifications were also determined (LC). The expression of nuclear factor erythroid 2-related factor 2 (Nrf2), its activators and inhibitors as well as pro/anti-apoptotic proteins and endocannabinoid receptors was examined (Western blot) and collagen metabolism was evaluated by collagen biosynthesis and prolidase activity (spectrometry). UVA and UVB irradiation and hydrogen peroxide treatment enhanced activity of xanthine and NADPH oxidases resulting in ROS generation as well as diminution of antioxidant phospholipid protection (glutathione peroxidase-glutathione-vitamin E), what led to increased lipid peroxidation and decreased endocannabinoids level. Dysregulation of cannabinoid receptors expression and environment of transcription factor Nrf2 caused apoptosis induction. Ascorbic acid partially prevented ROS generation, antioxidant capacity diminution and endocannabinoid systems disturbances but only slightly protected macromolecules such as phospholipid, protein and DNA against oxidative modifications. However, ascorbic acid significantly prevented decrease in collagen type I biosynthesis. Ascorbic acid in similar degree prevents UV (UVA and UVB) and hydrogen peroxide-dependent redox imbalance. However, this antioxidant cannot efficiently protect cellular macromolecules and avert metabolic dysregulation leading to apoptosis.
Collapse
|
66
|
Bromfield EG, Mihalas BP, Dun MD, Aitken RJ, McLaughlin EA, Walters JL, Nixon B. Inhibition of arachidonate 15-lipoxygenase prevents 4-hydroxynonenal-induced protein damage in male germ cells†. Biol Reprod 2017; 96:598-609. [DOI: 10.1093/biolre/iox005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 01/30/2017] [Indexed: 12/20/2022] Open
|
67
|
Rutin as a Mediator of Lipid Metabolism and Cellular Signaling Pathways Interactions in Fibroblasts Altered by UVA and UVB Radiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4721352. [PMID: 28168010 PMCID: PMC5266866 DOI: 10.1155/2017/4721352] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/05/2016] [Indexed: 02/07/2023]
Abstract
Background. Rutin is a natural nutraceutical that is a promising compound for the prevention of UV-induced metabolic changes in skin cells. The aim of this study was to examine the effects of rutin on redox and endocannabinoid systems, as well as proinflammatory and proapoptotic processes, in UV-irradiated fibroblasts. Methods. Fibroblasts exposed to UVA and UVB radiation were treated with rutin. The activities and levels of oxidants/antioxidants and endocannabinoid system components, as well as lipid, DNA, and protein oxidation products, and the proinflammatory and pro/antiapoptotic proteins expression were measured. Results. Rutin reduced UV-induced proinflammatory response and ROS generation and enhanced the activity/levels of antioxidants (SOD, GSH-Px, vitamin E, GSH, and Trx). Rutin also normalized UV-induced Nrf2 expression. Its biological activity prevented changes in the levels of the lipid mediators: MDA, 4-HNE, and endocannabinoids, as well as the endocannabinoid receptors CB1/2, VR1, and GPR55 expression. Furthermore, rutin prevented the protein modifications (tyrosine derivatives formation in particular) and decreased the levels of the proapoptotic markers—caspase-3 and cytochrome c. Conclusion. Rutin prevents UV-induced inflammation and redox imbalance at protein and transcriptional level which favors lipid, protein, and DNA protection. In consequence rutin regulates endocannabinoid system and apoptotic balance.
Collapse
|
68
|
Salazar P, Cisternas P, Codocedo JF, Inestrosa NC. Induction of hypothyroidism during early postnatal stages triggers a decrease in cognitive performance by decreasing hippocampal synaptic plasticity. Biochim Biophys Acta Mol Basis Dis 2017; 1863:870-883. [PMID: 28088629 DOI: 10.1016/j.bbadis.2017.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/25/2016] [Accepted: 01/04/2017] [Indexed: 01/27/2023]
Abstract
Thyroid hormones are vital in the control of multiple body functions, including the correct performance of the brain. Multiple diseases are associated with thyroid gland functioning, including hypothyroidism. To date, little is known regarding the effects of the establishment of this condition at a young age on brain function. Here, we evaluated the effect of hypothyroidism in an early postnatal stage in cognitive abilities with focus on the hippocampus. In our model, hypothyroidism was induced in young rats at 21days of age using 0.05% 6-propyl-2-thiouracil (PTU) for 4weeks reaching significantly lower levels of fT4 (control: 1.337ng/dL±0.115, PTU: 0.050ng/dL±0.001). Following the induction of hypothyroidism, several cognitive tasks were assessed to investigate the effects of hypothyroidism on cognition performance. We determined that hypothyroidism triggers a significant dysfunction in learning and memory processes observed in the Morris Water Maze were the latency times were higher in PTU rats (controls: 37s; PTU: 57s). The cognitive impairment was correlated with a reduction in hippocampal plasticity with respect to both long-term potentiation (LTP) (control: 1.45, PTU: 1.00) and depression (LTD) (control: 0.71, PTU: 1.01). Furthermore, a decrease in the rate of glucose utilization (control: 223nmol∗mg of protein, PTU:148nmol∗mg of protein) was observed, along with an increase in oxidative stress and a decrease in MAP2 marker in the hippocampus. Our findings suggest that the induction of hypothyroidism in a young rat model alters numerous functions at the level of the hippocampus.
Collapse
Affiliation(s)
- Paulina Salazar
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Universidad de Atacama, Facultad de Ciencias Naturales, Departamento de Química y Biología, Copayapu 485, Copiapó, Chile
| | - Juan Francisco Codocedo
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
69
|
Elvsåshagen T, Zuzarte P, Westlye LT, Bøen E, Josefsen D, Boye B, Hol PK, Malt UF, Young LT, Andreazza AC. Dentate gyrus-cornu ammonis (CA) 4 volume is decreased and associated with depressive episodes and lipid peroxidation in bipolar II disorder: Longitudinal and cross-sectional analyses. Bipolar Disord 2016; 18:657-668. [PMID: 27995733 DOI: 10.1111/bdi.12457] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 11/09/2016] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Reduced dentate gyrus volume and increased oxidative stress have emerged as potential pathophysiological mechanisms in bipolar disorder. However, the relationship between dentate gyrus volume and peripheral oxidative stress markers remains unknown. Here, we examined dentate gyrus-cornu ammonis (CA) 4 volume longitudinally in patients with bipolar II disorder (BD-II) and healthy controls and investigated whether BD-II is associated with elevated peripheral levels of oxidative stress. METHODS We acquired high-resolution structural 3T-magnetic resonance imaging (MRI) images and quantified hippocampal subfield volumes using an automated segmentation algorithm in individuals with BD-II (n=29) and controls (n=33). The participants were scanned twice, at study inclusion and on average 2.4 years later. In addition, we measured peripheral levels of two lipid peroxidation markers (4-hydroxy-2-nonenal [4-HNE] and lipid hydroperoxides [LPH]). RESULTS First, we demonstrated that the automated hippocampal subfield segmentation technique employed in this work reliably measured dentate gyrus-CA4 volume. Second, we found a decreased left dentate gyrus-CA4 volume in patients and that a larger number of depressive episodes between T1 and T2 predicted greater volume decline. Finally, we showed that 4-HNE was elevated in BD-II and that 4-HNE was negatively associated with left and right dentate gyrus-CA4 volumes in patients. CONCLUSIONS These results are consistent with a role for the dentate gyrus in the pathophysiology of bipolar disorder and suggest that depressive episodes and elevated oxidative stress might contribute to hippocampal volume decreases. In addition, these findings provide further support for the hypothesis that peripheral lipid peroxidation markers may reflect brain alterations in bipolar disorders.
Collapse
Affiliation(s)
- Torbjørn Elvsåshagen
- Department of Neurology, Oslo University Hospital, Oslo, Norway.,Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pedro Zuzarte
- Department of Psychiatry, Santa Maria's University Hospital, University of Lisbon, Lisbon, Portugal.,Department of Pharmacology and Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Department of Psychology, University of Oslo, Oslo, Norway
| | - Erlend Bøen
- Department of Psychiatry, Diakonhjemmet Hospital, Oslo, Norway
| | - Dag Josefsen
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Birgitte Boye
- Section of Psychosocial Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway.,Department of Behavioural Sciences in Medicine, University of Oslo, Oslo, Norway
| | - Per K Hol
- The Intervention Centre, Oslo University Hospital, Oslo, Norway
| | - Ulrik F Malt
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Research and Education, Oslo University Hospital, Oslo, Norway
| | - L Trevor Young
- Department of Pharmacology and Psychiatry, University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Ana C Andreazza
- Department of Pharmacology and Psychiatry, University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
70
|
Murdolo G, Piroddi M, Tortoioli C, Bartolini D, Schmelz M, Luchetti F, Canonico B, Papa S, Zerbinati C, Iuliano L, Galli F. Free Radical-derived Oxysterols: Novel Adipokines Modulating Adipogenic Differentiation of Adipose Precursor Cells. J Clin Endocrinol Metab 2016; 101:4974-4983. [PMID: 27710239 DOI: 10.1210/jc.2016-2918] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
CONTEXT Increased oxidative stress in adipose tissue emerges as an inducer of obesity-linked insulin resistance. Here we tested whether free-radical derived oxysterols are formed by, and accumulate in, human adipocytes. Moreover, we asked whether increased accumulation of oxysterols characterizes the adipose cells of obese patients with type 2 diabetes (T2D) (OBT2D) compared with lean, nondiabetic controls (CTRLs). Finally, we studied the effects of the free radical-derived oxysterols on adipogenic differentiation of adipose-derived stem cells (ASCs). MAIN OUTCOME MEASURES Adipocytes and ASCs were isolated from sc abdominal adipose tissue biopsy in four OBT2D and four CTRL subjects. Oxysterols in adipocytes were detected by gas chromatography/mass spectrometry. The cellular and molecular effects of oxysterols were then evaluated on primary cultures of ASCs focusing on cell viability, adipogenic differentiation, and "canonical" WNT and MAPK signaling pathways. RESULTS 7-ketocholesterol (7κ-C) and 7β-hydroxycholesterol were unambiguously detected in adipocytes, which showed higher oxysterol accumulation (P < .01) in OBT2D, as compared with CTRL individuals. Notably, the accumulation of oxysterols in adipocytes was predicted by the adipose cell size of the donor (R2 = 0.582; P < .01). Challenging ASCs with free radical-derived type I (7κ-C) and type II (5,6-Secosterol) oxysterols led to a time- and concentration-dependent decrease of cell viability. Meaningfully, at a non-toxic concentration (1μM), these bioactive lipids hampered adipogenic differentiation of ASCs by sequential activation of WNT/β-catenin, p38-MAPK, ERK1/2, and JNK signaling pathways. CONCLUSION Free radical-derived oxysterols accumulate in the "diabetic" fat and may act as novel adipokines modulating the adipogenic potential of undifferentiated adipose precursor cells.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Marta Piroddi
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Cristina Tortoioli
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Desirée Bartolini
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Martin Schmelz
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Francesca Luchetti
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Barbara Canonico
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Stefano Papa
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Chiara Zerbinati
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Luigi Iuliano
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| | - Francesco Galli
- Department of Internal Medicine (G.M.), Assisi Hospital, 06081 Assisi (Perugia), Italy; Department of Internal Medicine, Section of Internal Medicine, Endocrine and Metabolic Sciences (G.M., C.T.), University of Perugia, 06126 Perugia Italy; Department of Pharmaceutical Sciences (M.P., D.B., F.G.), University of Perugia, 06126 Perugia, Italy; Department of Anesthesiology and Intensive Care Medicine Mannheim (M.S.), Heidelberg University, 69117 Heidelberg, Germany; Department of Earth, Life and Environmental Sciences (F.L., B.C., S.P.), University Carlo Bo, 61029 Urbino, Italy; and Department of Medico-Surgical Sciences and Biotechnologies, Unit of Vascular Medicine (C.Z., L.I.), Sapienza University of Rome, 00185 Latina, Italy
| |
Collapse
|
71
|
Maulucci G, Cohen O, Daniel B, Sansone A, Petropoulou PI, Filou S, Spyridonidis A, Pani G, De Spirito M, Chatgilialoglu C, Ferreri C, Kypreos KE, Sasson S. Fatty acid-related modulations of membrane fluidity in cells: detection and implications. Free Radic Res 2016; 50:S40-S50. [PMID: 27593084 DOI: 10.1080/10715762.2016.1231403] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Metabolic homeostasis of fatty acids is complex and well-regulated in all organisms. The biosynthesis of saturated fatty acids (SFA) in mammals provides substrates for β-oxidation and ATP production. Monounsaturated fatty acids (MUFA) are products of desaturases that introduce a methylene group in cis geometry in SFA. Polyunsaturated fatty acids (n-6 and n-3 PUFA) are products of elongation and desaturation of the essential linoleic acid and α-linolenic acid, respectively. The liver processes dietary fatty acids and exports them in lipoproteins for distribution and storage in peripheral tissues. The three types of fatty acids are integrated in membrane phospholipids and determine their biophysical properties and functions. This study was aimed at investigating effects of fatty acids on membrane biophysical properties under varying nutritional and pathological conditions, by integrating lipidomic analysis of membrane phospholipids with functional two-photon microscopy (fTPM) of cellular membranes. This approach was applied to two case studies: first, pancreatic beta-cells, to investigate hormetic and detrimental effects of lipids. Second, red blood cells extracted from a genetic mouse model defective in lipoproteins, to understand the role of lipids in hepatic diseases and metabolic syndrome and their effect on circulating cells.
Collapse
Affiliation(s)
- G Maulucci
- a Institute of Physics, Università Cattolica del Sacro Cuore , Roma , Italy
| | - O Cohen
- b Institute for Drug Research, Section of Pharmacology, Diabetes Research Unit, Faculty of Medicine , The Hebrew University , Jerusalem , Israel
| | - B Daniel
- b Institute for Drug Research, Section of Pharmacology, Diabetes Research Unit, Faculty of Medicine , The Hebrew University , Jerusalem , Israel
| | - A Sansone
- c ISOF, BioFreeRadicals Group, Consiglio Nazionale delle Ricerche , Bologna , Italy
| | - P I Petropoulou
- d Department of Pharmacology , University of Patras Medical School , Rio , Greece
| | - S Filou
- d Department of Pharmacology , University of Patras Medical School , Rio , Greece
| | - A Spyridonidis
- e Hematology Department , University of Patras Medical School , Rio , Greece
| | - G Pani
- f Institute of General Pathology, Università Cattolica del Sacro Cuore , Roma , Italy
| | - M De Spirito
- a Institute of Physics, Università Cattolica del Sacro Cuore , Roma , Italy
| | - C Chatgilialoglu
- c ISOF, BioFreeRadicals Group, Consiglio Nazionale delle Ricerche , Bologna , Italy
| | - C Ferreri
- c ISOF, BioFreeRadicals Group, Consiglio Nazionale delle Ricerche , Bologna , Italy
| | - K E Kypreos
- d Department of Pharmacology , University of Patras Medical School , Rio , Greece
| | - S Sasson
- b Institute for Drug Research, Section of Pharmacology, Diabetes Research Unit, Faculty of Medicine , The Hebrew University , Jerusalem , Israel
| |
Collapse
|
72
|
Ferreri C, Golding BT, Jahn U, Ravanat JL. COST Action CM1201 "Biomimetic Radical Chemistry": free radical chemistry successfully meets many disciplines. Free Radic Res 2016; 50:S112-S128. [PMID: 27750460 DOI: 10.1080/10715762.2016.1248961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The COST Action CM1201 "Biomimetic Radical Chemistry" has been active since December 2012 for 4 years, developing research topics organized into four working groups: WG1 - Radical Enzymes, WG2 - Models of DNA damage and consequences, WG3 - Membrane stress, signalling and defenses, and WG4 - Bio-inspired synthetic strategies. International collaborations have been established among the participating 80 research groups with brilliant interdisciplinary achievements. Free radical research with a biomimetic approach has been realized in the COST Action and are summarized in this overview by the four WG leaders.
Collapse
Affiliation(s)
- Carla Ferreri
- a ISOF, Consiglio Nazionale delle Ricerche, BioFreeRadicals Group , Bologna , Italy
| | - Bernard T Golding
- b School of Chemistry, Bedson Building, Newcastle University , Newcastle-upon-Tyne , UK
| | - Ullrich Jahn
- c Institute of Organic Chemistry and Biochemistry , Czech Academy of Sciences , Prague , Czech Republic
| | - Jean-Luc Ravanat
- d INAC-SCIB & CEA, INAC-SyMMES Laboratoire des Lésions des Acides Nucléiques , Université Grenoble Alpes , Grenoble , France
| |
Collapse
|
73
|
4-Hydroxyalkenal-activated PPARδ mediates hormetic interactions in diabetes. Biochimie 2016; 136:85-89. [PMID: 27768859 DOI: 10.1016/j.biochi.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022]
Abstract
Activated peroxisome proliferator-activated receptor-δ (PPARδ) induces the expression of genes encoding enzymes that metabolize fatty acids and carbohydrate. Attempts to identify cellular activators of PPARδ produced large lists of various fatty acids and their metabolic derivatives; however, there is no consensus on specific and selective binding interactions of natural ligands with PPARδ. Most models on binding interactions within the ligand binding domain (LBD) of PPARδ have been derived from analyses of PPARδ-LBD crystals formed with synthetic low molecular weight ligands. Nonetheless, crystals of the whole receptor with natural ligands or of its heterodimer with its cognate retinoid X receptor (RXR) are not yet available for analysis. We have found that 4-hydroxyalkenals, non-enzymatic peroxidation products of polyunsaturated fatty acids (PUFA), namely, 4-hydroxy-2E,6Z-dodecadienal (4-HDDE) and 4-hydroxy-2E-nonenal (4-HNE), activate PPARδ in vascular endothelial cells and insulin-secreting beta cells, respectively. In both cases activated PPARδ induced adaptive responses that allowed the cells to adjust to ambient stressful metabolic conditions. This review article addresses the interactions of 4-hydroxyalkenals with PPARδ and the resulting hormetic interactions in cells exposed to nutrient overload conditions.
Collapse
|
74
|
Gęgotek A, Nikliński J, Žarković N, Žarković K, Waeg G, Łuczaj W, Charkiewicz R, Skrzydlewska E. Lipid mediators involved in the oxidative stress and antioxidant defence of human lung cancer cells. Redox Biol 2016; 9:210-219. [PMID: 27567474 PMCID: PMC5007445 DOI: 10.1016/j.redox.2016.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/22/2016] [Accepted: 08/18/2016] [Indexed: 01/24/2023] Open
Abstract
Background The oxidative modifications of bioactive macromolecules have important roles in carcinogenesis. Of particular interest are lipid peroxidation products, which are involved in the activation of Nrf2 and endocannabinoids that affect cancer progression. Methods In lung cancer tissues (squamous cell lung carcinoma - SCC and adenocarcinoma - AC), the glutathione peroxidase and catalase activity and glutathione level, together with the expression of Nrf2 and its activators/inhibitors were estimated. The oxidative modifications of DNA (8-hydroxy-2′-deoxyguanosine and N7-methylguanine), endocannabinoids (anandamide and 2- arachidonylglyceriol), their receptors (CB1/2, TRV1, GPR55), phospholipid fatty acids (arachidonic, linoleic and docosahexaenoic), and reactive aldehydes (4-hydroxynonenal, 4-oxononenal and malondialdehyde) were determined. Results Tumour tissues showed lower antioxidant capacity than healthy tissues, which was accompanied by lower levels of fatty acids and higher levels of reactive aldehydes. Disturbances in antioxidant capacity and enhanced DNA oxidative modifications were observed in 88% of AC patients and 81% of SCC patients. The 4-hydroxynonenal-Histidine adducts were detected in the necrotic and stromal cells in all tumours. These findings were associated with the enhanced Nrf2 activity, especially in AC. The strong difference between the cancer subtypes was evident in the levels of endocannabinoids, with an increase in 89% of SCC and a decrease in 85% of AC patients being observed. Additionally, the increase in the expression of CB1/2 receptors was observed only in 82% of AC, while the expression of VR1 and GPR55 was enhanced in 79% of SCC and 82% of AC patients. Conclusions This study shows significant differences in the redox status, Nrf2 pathway and endocannabinoid system between SCC and AC tissues. Understanding the relation between the various lipid mediators and antioxidants in different lung cancer subtypes may be beginning for further research on the effective anticancer therapy. AC tissues show lower antioxidant capacity than SCC. Nrf2/ARE pathway is activated stronger in AC than in SCC. AC exhibits more lipid oxidative modifications than SCC. HNE-protein adducts are detected in the necrotic and stromal cells in SCC and AC. SCC exhibits increase in endocannabinoids level while in AC reverse effect is observed.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Departments of Analytical Chemistry, Medical University of Białystok, Białystok, Poland
| | - Jacek Nikliński
- Clinical Molecular Biology, Medical University of Białystok, Białystok, Poland
| | | | - Kamelija Žarković
- University of Zagreb School of Medicine, Clinical Hospital Centre Division of Pathology, Zagreb, Croatia
| | - Georg Waeg
- Institute of Molecular Biosciences, Karl Franzens University in Graz, Austria
| | - Wojciech Łuczaj
- Departments of Analytical Chemistry, Medical University of Białystok, Białystok, Poland
| | | | - Elżbieta Skrzydlewska
- Departments of Analytical Chemistry, Medical University of Białystok, Białystok, Poland.
| |
Collapse
|
75
|
Sieprath T, Corne TDJ, Willems PHGM, Koopman WJH, De Vos WH. Integrated High-Content Quantification of Intracellular ROS Levels and Mitochondrial Morphofunction. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2016; 219:149-77. [PMID: 27207366 DOI: 10.1007/978-3-319-28549-8_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxidative stress arises from an imbalance between the production of reactive oxygen species (ROS) and their removal by cellular antioxidant systems. Especially under pathological conditions, mitochondria constitute a relevant source of cellular ROS. These organelles harbor the electron transport chain, bringing electrons in close vicinity to molecular oxygen. Although a full understanding is still lacking, intracellular ROS generation and mitochondrial function are also linked to changes in mitochondrial morphology. To study the intricate relationships between the different factors that govern cellular redox balance in living cells, we have developed a high-content microscopy-based strategy for simultaneous quantification of intracellular ROS levels and mitochondrial morphofunction. Here, we summarize the principles of intracellular ROS generation and removal, and we explain the major considerations for performing quantitative microscopy analyses of ROS and mitochondrial morphofunction in living cells. Next, we describe our workflow, and finally, we illustrate that a multiparametric readout enables the unambiguous classification of chemically perturbed cells as well as laminopathy patient cells.
Collapse
Affiliation(s)
- Tom Sieprath
- Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University, Ghent, Belgium.,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Tobias D J Corne
- Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University, Ghent, Belgium.,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter H G M Willems
- Department of Biochemistry (286), Radboud University Medical Centre (RUMC), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Biochemistry (286), Radboud University Medical Centre (RUMC), Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Winnok H De Vos
- Cell Systems and Imaging Research Group (CSI), Department of Molecular Biotechnology, Ghent University, Ghent, Belgium. .,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
76
|
Shapiro H, Singer P, Ariel A. Beyond the classic eicosanoids: Peripherally-acting oxygenated metabolites of polyunsaturated fatty acids mediate pain associated with tissue injury and inflammation. Prostaglandins Leukot Essent Fatty Acids 2016; 111:45-61. [PMID: 27067460 DOI: 10.1016/j.plefa.2016.03.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
Pain is a complex sensation that may be protective or cause undue suffering and loss of function, depending on the circumstances. Peripheral nociceptor neurons (PNs) innervate most tissues, and express ion channels, nocisensors, which depolarize the cell in response to intense stimuli and numerous substances. Inflamed tissues manifest inflammatory hyperalgesia in which the threshold for pain and the response to painful stimuli are decreased and increased, respectively. Constituents of the inflammatory milieu sensitize PNs, thereby contributing to hyperalgesia. Polyunsaturated fatty acids undergo enzymatic and free radical-mediated oxygenation into an array of bioactive metabolites, oxygenated polyunsaturated fatty acids (oxy-PUFAs), including the classic eicosanoids. Oxy-PUFA production is enhanced during inflammation. Pioneering studies by Vane and colleagues from the early 1970s first implicated classic eicosanoids in the pain associated with inflammation. Here, we review the production and action of oxy-PUFAs that are not classic eicosanoids, but nevertheless are produced in injured/ inflamed tissues and activate or sensitize PNs. In general, oxy-PUFAs that sensitize PNs may do so directly, by activation of nocisensors, ion channels or GPCRs expressed on the surface of PNs, or indirectly, by increasing the production of inflammatory mediators that activate or sensitize PNs. We focus on oxy-PUFAs that act directly on PNs. Specifically, we discuss the role of arachidonic acid-derived 12S-HpETE, HNE, ONE, PGA2, iso-PGA2 and 15d-PGJ2, 5,6-and 8,9-EET, PGE2-G and 8R,15S-diHETE, as well as the linoleic acid-derived 9-and 13-HODE in inducing acute nocifensive behavior and/or inflammatory hyperalgesia in rodents. The nocisensors TRPV1, TRPV4 and TRPA1, and putative Gαs-type GPCRs are the PN targets of these oxy-PUFAs.
Collapse
Affiliation(s)
- Haim Shapiro
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Abba Khoushy Ave, Mount Carmel, Haifa 3498838, Israel.
| | - Pierre Singer
- Department of General Intensive Care, Institute for Nutrition Research, Rabin Medical Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva 49100, Israel
| | - Amiram Ariel
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Abba Khoushy Ave, Mount Carmel, Haifa 3498838, Israel
| |
Collapse
|
77
|
Tie C, Hu T, Jia ZX, Zhang JL. Derivatization Strategy for the Comprehensive Characterization of Endogenous Fatty Aldehydes Using HPLC-Multiple Reaction Monitoring. Anal Chem 2016; 88:7762-8. [DOI: 10.1021/acs.analchem.6b01756] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Cai Tie
- State Key Laboratory
of Bioactive Substance
and Function of Natural Medicines, Institute of Materia Medica, Peking
Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, PR China
| | - Ting Hu
- State Key Laboratory
of Bioactive Substance
and Function of Natural Medicines, Institute of Materia Medica, Peking
Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, PR China
| | - Zhi-Xin Jia
- State Key Laboratory
of Bioactive Substance
and Function of Natural Medicines, Institute of Materia Medica, Peking
Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, PR China
| | - Jin-Lan Zhang
- State Key Laboratory
of Bioactive Substance
and Function of Natural Medicines, Institute of Materia Medica, Peking
Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, PR China
| |
Collapse
|
78
|
The Role of Omega-3 Polyunsaturated Fatty Acids in Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6906712. [PMID: 27433289 PMCID: PMC4940554 DOI: 10.1155/2016/6906712] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/16/2016] [Accepted: 05/26/2016] [Indexed: 12/15/2022]
Abstract
Stroke is the third commonest cause of death following cardiovascular diseases and cancer. In particular, in recent years, the morbidity and mortality of stroke keep remarkable growing. However, stroke still captures people attention far less than cardiovascular diseases and cancer. Past studies have shown that oxidative stress and inflammation play crucial roles in the progress of cerebral injury induced by stroke. Evidence is accumulating that the dietary supplementation of fish oil exhibits beneficial effects on several diseases, such as cardiovascular diseases, metabolic diseases, and cancer. Omega-3 polyunsaturated fatty acids (n-3 PUFAs), the major component of fish oil, have been found against oxidative stress and inflammation in cardiovascular diseases. And the potential of n-3 PUFAs in stroke treatment is attracting more and more attention. In this review, we will review the effects of n-3 PUFAs on stroke and mainly focus on the antioxidant and anti-inflammatory effects of n-3 PUFAs.
Collapse
|
79
|
Myhrstad MCW, Ottestad I, Günther CC, Ryeng E, Holden M, Nilsson A, Brønner KW, Kohler A, Borge GIA, Holven KB, Ulven SM. The PBMC transcriptome profile after intake of oxidized versus high-quality fish oil: an explorative study in healthy subjects. GENES AND NUTRITION 2016; 11:16. [PMID: 27551317 PMCID: PMC4968435 DOI: 10.1186/s12263-016-0530-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 04/06/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Marine long-chain polyunsaturated fatty acids are susceptible to oxidation, generating a range of different oxidation products with suggested negative health effects. The aim of the present study was to utilize sensitive high-throughput transcriptome analyses to investigate potential unfavorable effects of oxidized fish oil (PV: 18 meq/kg; AV: 9) compared to high-quality fish oil (PV: 4 meq/kg; AV: 3). METHODS In a double-blinded randomized controlled study for seven weeks, 35 healthy subjects were assigned to 8 g of either oxidized fish oil or high quality fish oil. The daily dose of EPA+DHA was 1.6 g. Peripheral blood mononuclear cells were isolated at baseline and after 7 weeks and transcriptome analyses were performed with the illuminaHT-12 v4 Expression BeadChip. RESULTS No gene transcripts, biological processes, pathway or network were significantly changed in the oxidized fish oil group compared to the fish oil group. Furthermore, gene sets related to oxidative stress and cardiovascular disease were not differently regulated between the groups. Within group analyses revealed a more prominent effect after intake of high quality fish oil as 11 gene transcripts were significantly (FDR < 0.1) changed from baseline versus three within the oxidized fish oil group. CONCLUSION The suggested concern linking lipid oxidation products to short-term unfavorable health effects may therefore not be evident at a molecular level in this explorative study. TRIAL REGISTRATION ClinicalTrials.gov, NCT01034423.
Collapse
Affiliation(s)
- Mari C W Myhrstad
- Department of Health, Nutrition and Management, Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, P.O. Box 4, St. Olavs plass, 0130 Oslo, Norway
| | - Inger Ottestad
- Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317 Oslo, Norway ; Department of Health, Nutrition and Management, Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, P.O. Box 4, St. Olavs plass, 0130 Oslo, Norway
| | | | - Einar Ryeng
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | | | - Astrid Nilsson
- Nofima AS, Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, Aas, N-1431 Norway
| | - Kirsti W Brønner
- TINE SA, Centre for Research and Development, P.O. Box 7, Kalbakken, 0902 Oslo, Norway
| | - Achim Kohler
- Nofima AS, Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, Aas, N-1431 Norway ; Department of Mathematical Sciences and Technology (IMT), Norwegian University of Life Sciences, 1432 Ås, Norway
| | - Grethe I A Borge
- Nofima AS, Norwegian Institute of Food, Fisheries and Aquaculture Research, PB 210, Aas, N-1431 Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317 Oslo, Norway ; Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Rikshospitalet, P.O Box 4950, Nydalen, Oslo, Norway
| | - Stine M Ulven
- Department of Health, Nutrition and Management, Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, P.O. Box 4, St. Olavs plass, 0130 Oslo, Norway ; Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, P.O. Box 1046, Blindern, 0317 Oslo, Norway
| |
Collapse
|
80
|
Hormetic and regulatory effects of lipid peroxidation mediators in pancreatic beta cells. Mol Aspects Med 2016; 49:49-77. [PMID: 27012748 DOI: 10.1016/j.mam.2016.03.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/23/2016] [Accepted: 03/09/2016] [Indexed: 12/12/2022]
Abstract
Nutrient sensing mechanisms of carbohydrates, amino acids and lipids operate distinct pathways that are essential for the adaptation to varying metabolic conditions. The role of nutrient-induced biosynthesis of hormones is paramount for attaining metabolic homeostasis in the organism. Nutrient overload attenuate key metabolic cellular functions and interfere with hormonal-regulated inter- and intra-organ communication, which may ultimately lead to metabolic derangements. Hyperglycemia and high levels of saturated free fatty acids induce excessive production of oxygen free radicals in tissues and cells. This phenomenon, which is accentuated in both type-1 and type-2 diabetic patients, has been associated with the development of impaired glucose tolerance and the etiology of peripheral complications. However, low levels of the same free radicals also induce hormetic responses that protect cells against deleterious effects of the same radicals. Of interest is the role of hydroxyl radicals in initiating peroxidation of polyunsaturated fatty acids (PUFA) and generation of α,β-unsaturated reactive 4-hydroxyalkenals that avidly form covalent adducts with nucleophilic moieties in proteins, phospholipids and nucleic acids. Numerous studies have linked the lipid peroxidation product 4-hydroxy-2E-nonenal (4-HNE) to different pathological and cytotoxic processes. Similarly, two other members of the family, 4-hydroxyl-2E-hexenal (4-HHE) and 4-hydroxy-2E,6Z-dodecadienal (4-HDDE), have also been identified as potential cytotoxic agents. It has been suggested that 4-HNE-induced modifications in macromolecules in cells may alter their cellular functions and modify signaling properties. Yet, it has also been acknowledged that these bioactive aldehydes also function as signaling molecules that directly modify cell functions in a hormetic fashion to enable cells adapt to various stressful stimuli. Recent studies have shown that 4-HNE and 4-HDDE, which activate peroxisome proliferator-activated receptor δ (PPARδ) in vascular endothelial cells and insulin secreting beta cells, promote such adaptive responses to ameliorate detrimental effects of high glucose and diabetes-like conditions. In addition, due to the electrophilic nature of these reactive aldehydes they form covalent adducts with electronegative moieties in proteins, phosphatidylethanolamine and nucleotides. Normally these non-enzymatic modifications are maintained below the cytotoxic range due to efficient cellular neutralization processes of 4-hydroxyalkenals. The major neutralizing enzymes include fatty aldehyde dehydrogenase (FALDH), aldose reductase (AR) and alcohol dehydrogenase (ADH), which transform the aldehyde to the corresponding carboxylic acid or alcohols, respectively, or by biding to the thiol group in glutathione (GSH) by the action of glutathione-S-transferase (GST). This review describes the hormetic and cytotoxic roles of oxygen free radicals and 4-hydroxyalkenals in beta cells exposed to nutritional challenges and the cellular mechanisms they employ to maintain their level at functional range below the cytotoxic threshold.
Collapse
|
81
|
Wang M, Wang C, Han RH, Han X. Novel advances in shotgun lipidomics for biology and medicine. Prog Lipid Res 2016; 61:83-108. [PMID: 26703190 PMCID: PMC4733395 DOI: 10.1016/j.plipres.2015.12.002] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022]
Abstract
The field of lipidomics, as coined in 2003, has made profound advances and been rapidly expanded. The mass spectrometry-based strategies of this analytical methodology-oriented research discipline for lipid analysis are largely fallen into three categories: direct infusion-based shotgun lipidomics, liquid chromatography-mass spectrometry-based platforms, and matrix-assisted laser desorption/ionization mass spectrometry-based approaches (particularly in imagining lipid distribution in tissues or cells). This review focuses on shotgun lipidomics. After briefly introducing its fundamentals, the major materials of this article cover its recent advances. These include the novel methods of lipid extraction, novel shotgun lipidomics strategies for identification and quantification of previously hardly accessible lipid classes and molecular species including isomers, and novel tools for processing and interpretation of lipidomics data. Representative applications of advanced shotgun lipidomics for biological and biomedical research are also presented in this review. We believe that with these novel advances in shotgun lipidomics, this approach for lipid analysis should become more comprehensive and high throughput, thereby greatly accelerating the lipidomics field to substantiate the aberrant lipid metabolism, signaling, trafficking, and homeostasis under pathological conditions and their underpinning biochemical mechanisms.
Collapse
Affiliation(s)
- Miao Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Chunyan Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Rowland H Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA; College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
82
|
Abstract
One of the prerequisites for cell growth and proliferation is the synthesis of macromolecules, including proteins, nucleic acids and lipids. Cells have to alter their metabolism to allow the production of metabolic intermediates that are the precursors for biomass production. It is now evident that oncogenic signalling pathways target metabolic processes on several levels and metabolic reprogramming has emerged as a hallmark of cancer. The increased metabolic demand of cancer cells also produces selective dependencies that could be targeted for therapeutic intervention. Understanding the role of glucose and lipid metabolism in supporting cancer cell growth and survival is crucial to identify essential processes that could provide therapeutic windows for cancer therapy.
Collapse
|
83
|
HLAVÁČOVÁ M, GUMULEC J, STRAČINA T, FOJTŮ M, RAUDENSKÁ M, MASAŘÍK M, NOVÁKOVÁ M, PAULOVÁ H. Different Doxorubicin Formulations Affect Plasma 4-Hydroxy-2-Nonenal and Gene Expression of Aldehyde Dehydrogenase 3A1 and Thioredoxin Reductase 2 in Rat. Physiol Res 2015; 64:S653-60. [DOI: 10.33549/physiolres.933223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Increased oxidative stress is indisputably an important mechanism of doxorubicin side effects, especially its cardiotoxicity. To prevent impairment of non-tumorous tissue and to improve the specificity in targeting the tumor tissue, new drug nanotransporters are developed. In many cases preclinical therapeutic advantage has been shown when compared with the administration of conventional drug solution. Three forms of doxorubicin – conventional (DOX), encapsulated in liposomes (lipoDOX) and in apoferritin (apoDOX) were applied to Wistar rats. After 24 h exposition, the plasma level of 4-hydroxy-2-nonenal (4-HNE) as a marker of lipoperoxidation and tissue gene expression of thioredoxin reductase 2 (TXNRD2) and aldehyde dehydrogenase 3A1 (ALDH3A1) as an important part of antioxidative system were determined. Only conventional DOX significantly increases the level of 4-HNE; encapsulated forms on the other hand show significant decrease in plasma levels of 4 HNE in comparison with DOX. They also cause significant decrease in gene expression of ALDH3A1 and TXNRD2 in liver as a main detoxification organ, and a mild influence on the expression of these enzymes in left heart ventricle as a potential target of toxicity. Thus, 4-HNE seems to be a good potential biomarker of oxidative stress induced by various forms of doxorubicin.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - H. PAULOVÁ
- Department of Biochemistry, Faculty of Medicine, Masaryk University in Brno, Brno, Czech Republic
| |
Collapse
|
84
|
Viau M, Genot C, Ribourg L, Meynier A. Amounts of the reactive aldehydes, malonaldehyde, 4-hydroxy-2-hexenal, and 4-hydroxy-2-nonenal in fresh and oxidized edible oils do not necessary reflect their peroxide and anisidine values. EUR J LIPID SCI TECH 2015. [DOI: 10.1002/ejlt.201500103] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Michèle Viau
- INRA, UR1268 Biopolymères Interactions Assemblages; Nantes France
| | - Claude Genot
- INRA, UR1268 Biopolymères Interactions Assemblages; Nantes France
| | - Lucie Ribourg
- INRA, UR1268 Biopolymères Interactions Assemblages; Nantes France
| | - Anne Meynier
- INRA, UR1268 Biopolymères Interactions Assemblages; Nantes France
| |
Collapse
|
85
|
Lynch TL, Sivaguru M, Velayutham M, Cardounel AJ, Michels M, Barefield D, Govindan S, dos Remedios C, van der Velden J, Sadayappan S. Oxidative Stress in Dilated Cardiomyopathy Caused by MYBPC3 Mutation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:424751. [PMID: 26508994 PMCID: PMC4609873 DOI: 10.1155/2015/424751] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 07/01/2015] [Accepted: 08/09/2015] [Indexed: 01/23/2023]
Abstract
Cardiomyopathies can result from mutations in genes encoding sarcomere proteins including MYBPC3, which encodes cardiac myosin binding protein-C (cMyBP-C). However, whether oxidative stress is augmented due to contractile dysfunction and cardiomyocyte damage in MYBPC3-mutated cardiomyopathies has not been elucidated. To determine whether oxidative stress markers were elevated in MYBPC3-mutated cardiomyopathies, a previously characterized 3-month-old mouse model of dilated cardiomyopathy (DCM) expressing a homozygous MYBPC3 mutation (cMyBP-C((t/t))) was used, compared to wild-type (WT) mice. Echocardiography confirmed decreased percentage of fractional shortening in DCM versus WT hearts. Histopathological analysis indicated a significant increase in myocardial disarray and fibrosis while the second harmonic generation imaging revealed disorganized sarcomeric structure and myocyte damage in DCM hearts when compared to WT hearts. Intriguingly, DCM mouse heart homogenates had decreased glutathione (GSH/GSSG) ratio and increased protein carbonyl and lipid malondialdehyde content compared to WT heart homogenates, consistent with elevated oxidative stress. Importantly, a similar result was observed in human cardiomyopathy heart homogenate samples. These results were further supported by reduced signals for mitochondrial semiquinone radicals and Fe-S clusters in DCM mouse hearts measured using electron paramagnetic resonance spectroscopy. In conclusion, we demonstrate elevated oxidative stress in MYPBC3-mutated DCM mice, which may exacerbate the development of heart failure.
Collapse
Affiliation(s)
- Thomas L. Lynch
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL 60153, USA
| | - Mayandi Sivaguru
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Murugesan Velayutham
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Arturo J. Cardounel
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Michelle Michels
- Department of Cardiology, Thoraxcenter, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, Netherlands
| | - David Barefield
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL 60153, USA
| | - Suresh Govindan
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL 60153, USA
| | - Cristobal dos Remedios
- Bosch Institute, Discipline of Anatomy and Histology, University of Sydney, Sydney, NSW 2006, Australia
| | - Jolanda van der Velden
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, Netherlands
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
86
|
Schaur RJ, Siems W, Bresgen N, Eckl PM. 4-Hydroxy-nonenal-A Bioactive Lipid Peroxidation Product. Biomolecules 2015; 5:2247-337. [PMID: 26437435 PMCID: PMC4693237 DOI: 10.3390/biom5042247] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/24/2015] [Accepted: 07/29/2015] [Indexed: 12/23/2022] Open
Abstract
This review on recent research advances of the lipid peroxidation product 4-hydroxy-nonenal (HNE) has four major topics: I. the formation of HNE in various organs and tissues, II. the diverse biochemical reactions with Michael adduct formation as the most prominent one, III. the endogenous targets of HNE, primarily peptides and proteins (here the mechanisms of covalent adduct formation are described and the (patho-) physiological consequences discussed), and IV. the metabolism of HNE leading to a great number of degradation products, some of which are excreted in urine and may serve as non-invasive biomarkers of oxidative stress.
Collapse
Affiliation(s)
- Rudolf J Schaur
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 33a, 8010 Graz, Austria.
| | - Werner Siems
- Institute for Medical Education, KortexMed GmbH, Hindenburgring 12a, 38667 Bad Harzburg, Germany.
| | - Nikolaus Bresgen
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| | - Peter M Eckl
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| |
Collapse
|
87
|
Wang CZ, Ayadi AE, Goswamy J, Finnerty CC, Mifflin R, Sousse L, Enkhbaatar P, Papaconstantinou J, Herndon DN, Ansari NH. Topically applied metal chelator reduces thermal injury progression in a rat model of brass comb burn. Burns 2015; 41:1775-1787. [PMID: 26392023 DOI: 10.1016/j.burns.2015.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/26/2015] [Accepted: 08/07/2015] [Indexed: 01/03/2023]
Abstract
UNLABELLED Oxidative stress may be involved in the cellular damage and tissue destruction as burn wounds continues to progress after abatement of the initial insult. Since iron and calcium ions play key roles in oxidative stress, this study tested whether topical application of Livionex formulation (LF) lotion, that contains disodium EDTA as a metal chelator and methyl sulfonyl methane (MSM) as a permeability enhancer, would prevent or reduce burns. METHODS We used an established brass comb burn model with some modifications. Topical application of LF lotion was started 5 min post-burn, and repeated every 8 h for 3 consecutive days. Rats were euthanized and skin harvested for histochemistry and immunohistochemistry. Formation of protein adducts of 4-hydroxynonenal (HNE), malonadialdehyde (MDA) and acrolein (ACR) and expression of aldehyde dehydrogenase (ALDH) isozymes, ALDH1 and ALDH2 were assessed. RESULTS LF lotion-treated burn sites and interspaces showed mild morphological improvement compared to untreated burn sites. Furthermore, the lotion significantly decreased the immunostaining of lipid aldehyde-protein adducts including protein -HNE, -MDA and -ACR adducts, and restored the expression of aldehyde dehydrogenase isozymes in the unburned interspaces. CONCLUSION This data, for the first time, demonstrates that a topically applied EDTA-containing lotion protects burns progression with a concomitant decrease in the accumulation of reactive lipid aldehydes and protection of aldehyde dehydrogenase isozymes. Present studies are suggestive of therapeutic intervention of burns by this novel lotion.
Collapse
Affiliation(s)
- Cheng Z Wang
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-0647, United States
| | - Amina El Ayadi
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555-0647, United States; Shriners Hospital for Children, Galveston, TX, United States
| | - Juhi Goswamy
- University of Miami Miller School of Medicine, Miami, FL 33124, United States
| | - Celeste C Finnerty
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555-0647, United States; Institute for Translational Sciences, Galveston, TX, United States; Shriners Hospital for Children, Galveston, TX, United States
| | - Randy Mifflin
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555-0647, United States; Shriners Hospital for Children, Galveston, TX, United States
| | - Linda Sousse
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555-0647, United States; Shriners Hospital for Children, Galveston, TX, United States
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555-0647, United States
| | - John Papaconstantinou
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-0647, United States
| | - David N Herndon
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555-0647, United States; Shriners Hospital for Children, Galveston, TX, United States
| | - Naseem H Ansari
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-0647, United States.
| |
Collapse
|
88
|
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: 7.2] [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
|
89
|
Cisternas P, Salazar P, Serrano FG, Montecinos-Oliva C, Arredondo SB, Varela-Nallar L, Barja S, Vio CP, Gomez-Pinilla F, Inestrosa NC. Fructose consumption reduces hippocampal synaptic plasticity underlying cognitive performance. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2379-90. [PMID: 26300486 DOI: 10.1016/j.bbadis.2015.08.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/05/2015] [Accepted: 08/19/2015] [Indexed: 01/15/2023]
Abstract
Metabolic syndrome (MetS) is a global epidemic, which involves a spectrum of metabolic disorders comprising diabetes and obesity. The impact of MetS on the brain is becoming to be a concern, however, the poor understanding of mechanisms involved has limited the development of therapeutic strategies. We induced a MetS-like condition by exposing mice to fructose feeding for 7weeks. There was a dramatic deterioration in the capacity of the hippocampus to sustain synaptic plasticity in the forms of long-term potentiation (LTP) and long-term depression (LTD). Mice exposed to fructose showed a reduction in the number of contact zones and the size of postsynaptic densities (PSDs) in the hippocampus, as well as a decrease in hippocampal neurogenesis. There was an increase in lipid peroxidation likely associated with a deficiency in plasma membrane excitability. Consistent with an overall hippocampal dysfunction, there was a subsequent decrease in hippocampal dependent learning and memory performance, i.e., spatial learning and episodic memory. Most of the pathological sequel of MetS in the brain was reversed three month after discontinue fructose feeding. These results are novel to show that MetS triggers a cascade of molecular events, which disrupt hippocampal functional plasticity, and specific aspects of learning and memory function. The overall information raises concerns about the risk imposed by excessive fructose consumption on the pathology of neurological disorders.
Collapse
Affiliation(s)
- Pedro Cisternas
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Salazar
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe G Serrano
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carla Montecinos-Oliva
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sebastián B Arredondo
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Lorena Varela-Nallar
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Salesa Barja
- Departamento de Pediatria, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos P Vio
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile; Centro UC Síndrome de Down, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
90
|
Breath carbonyl compounds as biomarkers of lung cancer. Lung Cancer 2015; 90:92-7. [PMID: 26233567 DOI: 10.1016/j.lungcan.2015.07.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/15/2015] [Accepted: 07/12/2015] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Lung cancer dysregulations impart oxidative stress which results in important metabolic products in the form of volatile organic compounds (VOCs) in exhaled breath. The objective of this work is to use statistical classification models to determine specific carbonyl VOCs in exhaled breath as biomarkers for detection of lung cancer. MATERIALS AND METHODS Exhaled breath samples from 85 patients with untreated lung cancer, 34 patients with benign pulmonary nodules and 85 healthy controls were collected. Carbonyl compounds in exhaled breath were captured by silicon microreactors and analyzed by Fourier transform ion cyclotron resonance mass spectrometry (FT-ICR-MS). The concentrations of carbonyl compounds were analyzed using a variety of statistical classification models to determine which compounds best differentiated between the patient sub-populations. Predictive accuracy of each of the models was assessed on a separate test data set. RESULTS Six carbonyl compounds (C(4)H(8)O, C(5)H(10)O, C(2)H(4)O(2), C(4)H(8)O(2), C(6)H(10)O(2), C(9)H(16)O(2)) had significantly elevated concentrations in lung cancer patients vs. CONTROLS A model based on counting the number of elevated compounds out of these six achieved an overall classification accuracy on the test data of 97% (95% CI 92%-100%), 95% (95% CI 88%-100%), and 89% (95% CI 79%-99%) for classifying lung cancer patients vs. non-smokers, current smokers, and patients with benign nodules, respectively. These results were comparable to benchmarking based on established statistical and machine-learning methods. The sensitivity in each case was 96% or higher, with specificity ranging from 64% for benign nodule patients to 86% for smokers and 100% for non-smokers. CONCLUSION A model based on elevated levels of the six carbonyl VOCs effectively discriminates lung cancer patients from healthy controls as well as patients with benign pulmonary nodules.
Collapse
|
91
|
Manea SA, Constantin A, Manda G, Sasson S, Manea A. Regulation of Nox enzymes expression in vascular pathophysiology: Focusing on transcription factors and epigenetic mechanisms. Redox Biol 2015; 5:358-366. [PMID: 26133261 PMCID: PMC4501559 DOI: 10.1016/j.redox.2015.06.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 06/19/2015] [Accepted: 06/22/2015] [Indexed: 02/06/2023] Open
Abstract
NADPH oxidases (Nox) represent a family of hetero-oligomeric enzymes whose exclusive biological function is the generation of reactive oxygen species (ROS). Nox-derived ROS are essential modulators of signal transduction pathways that control key physiological activities such as cell growth, proliferation, migration, differentiation, and apoptosis, immune responses, and biochemical pathways. Enhanced formation of Nox-derived ROS, which is generally associated with the up-regulation of different Nox subtypes, has been established in various pathologies, namely cardiovascular diseases, diabetes, obesity, cancer, and neurodegeneration. The detrimental effects of Nox-derived ROS are related to alterations in cell signalling and/or direct irreversible oxidative damage of nucleic acids, proteins, carbohydrates, and lipids. Thus, understanding of transcriptional regulation mechanisms of Nox enzymes have been extensively investigated in an attempt to find ways to counteract the excessive formation of Nox-derived ROS in various pathological states. Despite the numerous existing data, the molecular pathways responsible for Nox up-regulation are not completely understood. This review article summarizes some of the recent advances and concepts related to the regulation of Nox expression in the vascular pathophysiology. It highlights the role of transcription factors and epigenetic mechanisms in this process. Identification of the signalling molecules involved in Nox up-regulation, which is associated with the onset and development of cardiovascular dysfunction may contribute to the development of novel strategies for the treatment of cardiovascular diseases. Nox is a unique class of enzymes whose sole function is the generation of ROS. Nox-derived ROS play a major role in cell physiology. Enhanced expression and activation of Nox has been reported in numerous pathologies. Nox expression is regulated via complex transcription factor-epigenetic mechanisms. Understanding of Nox regulation is essential to counteract ROS-induced cell damage.
Collapse
Affiliation(s)
- Simona-Adriana Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania
| | - Alina Constantin
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania
| | - Gina Manda
- "Victor Babes" National Institute of Pathology, Bucharest, Romania
| | - Shlomo Sasson
- The Institute for Drug Research, Department of Pharmacology, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania.
| |
Collapse
|
92
|
Cohen G, Shamni O, Avrahami Y, Cohen O, Broner EC, Filippov-Levy N, Chatgilialoglu C, Ferreri C, Kaiser N, Sasson S. Beta cell response to nutrient overload involves phospholipid remodelling and lipid peroxidation. Diabetologia 2015; 58:1333-43. [PMID: 25810039 DOI: 10.1007/s00125-015-3566-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 03/11/2015] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Membrane phospholipids are the major intracellular source for fatty acid-derived mediators, which regulate myriad cell functions. We showed previously that high glucose levels triggered the hydrolysis of polyunsaturated fatty acids from beta cell phospholipids. These fatty acids were subjected to free radical-catalysed peroxidation to generate the bioactive aldehyde 4-hydroxy-2E-nonenal (4-HNE). The latter activated the nuclear peroxisome proliferator-activated receptor-δ (PPARδ), which in turn augmented glucose-stimulated insulin secretion. The present study aimed at investigating the combined effects of glucose and fatty acid overload on phospholipid turnover and the subsequent generation of lipid mediators, which affect insulin secretion and beta cell viability. METHODS INS-1E cells were incubated with increasing glucose concentrations (5-25 mmol/l) without or with palmitic acid (PA; 50-500 μmol/l) and taken for fatty acid-based lipidomic analysis and functional assays. Rat isolated islets of Langerhans were used similarly. RESULTS PA was incorporated into membrane phospholipids in a concentration- and time-dependent manner; incorporation was highest at 25 mmol/l glucose. This was coupled to a rapid exchange with saturated, mono-unsaturated and polyunsaturated fatty acids. Importantly, released arachidonic acid and linoleic acid were subjected to peroxidation, resulting in the generation of 4-HNE, which further augmented insulin secretion by activating PPARδ in beta cells. However, this adaptive increase in insulin secretion was abolished at high glucose and PA levels, which induced endoplasmic reticulum stress, apoptosis and cell death. CONCLUSIONS/INTERPRETATION These findings highlight a key role for phospholipid remodelling and fatty acid peroxidation in mediating adaptive and cytotoxic interactions induced by nutrient overload in beta cells.
Collapse
Affiliation(s)
- Guy Cohen
- Department of Pharmacology, Institute for Drug Research, Faculty of Medicine, The Hebrew University, Jerusalem, 9112102, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Wojewoda M, Walczak J, Duszyński J, Szczepanowska J. Selenite activates the ATM kinase-dependent DNA repair pathway in human osteosarcoma cells with mitochondrial dysfunction. Biochem Pharmacol 2015; 95:170-6. [DOI: 10.1016/j.bcp.2015.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/30/2015] [Indexed: 01/22/2023]
|
94
|
Lin HY, Haegele JA, Disare MT, Lin Q, Aye Y. A generalizable platform for interrogating target- and signal-specific consequences of electrophilic modifications in redox-dependent cell signaling. J Am Chem Soc 2015; 137:6232-44. [PMID: 25909755 PMCID: PMC4528680 DOI: 10.1021/ja5132648] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite the known propensity of small-molecule electrophiles to react with numerous cysteine-active proteins, biological actions of individual signal inducers have emerged to be chemotype-specific. To pinpoint and quantify the impacts of modifying one target out of the whole proteome, we develop a target-protein-personalized "electrophile toolbox" with which specific intracellular targets can be selectively modified at a precise time by specific reactive signals. This general methodology, T-REX (targetable reactive electrophiles and oxidants), is established by (1) constructing a platform that can deliver a range of electronic and sterically different bioactive lipid-derived signaling electrophiles to specific proteins in cells; (2) probing the kinetics of targeted delivery concept, which revealed that targeting efficiency in cells is largely driven by initial on-rate of alkylation; and (3) evaluating the consequences of protein-target- and small-molecule-signal-specific modifications on the strength of downstream signaling. These data show that T-REX allows quantitative interrogations into the extent to which the Nrf2 transcription factor-dependent antioxidant response element (ARE) signaling is activated by selective electrophilic modifications on Keap1 protein, one of several redox-sensitive regulators of the Nrf2-ARE axis. The results document Keap1 as a promiscuous electrophile-responsive sensor able to respond with similar efficiencies to discrete electrophilic signals, promoting comparable strength of Nrf2-ARE induction. T-REX is also able to elicit cell activation in cases in which whole-cell electrophile flooding fails to stimulate ARE induction prior to causing cytotoxicity. The platform presents a previously unavailable opportunity to elucidate the functional consequences of small-molecule-signal- and protein-target-specific electrophilic modifications in an otherwise unaffected cellular background.
Collapse
Affiliation(s)
- Hong-Yu Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA
| | - Joseph A. Haegele
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA
| | - Michael T. Disare
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA
| | - Qishan Lin
- Proteomics/Mass Spectrometry Facility, Center for Functional Genomics, University of Albany, Rensselaer, New York, 12144, USA
| | - Yimon Aye
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, 14853, USA
- Department of Biochemistry, Weill Cornell Medical College, New York, New York, 10065, USA
| |
Collapse
|
95
|
Zambelli VO, Gross ER, Chen CH, Gutierrez VP, Cury Y, Mochly-Rosen D. Aldehyde dehydrogenase-2 regulates nociception in rodent models of acute inflammatory pain. Sci Transl Med 2015; 6:251ra118. [PMID: 25163478 DOI: 10.1126/scitranslmed.3009539] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Exogenous aldehydes can cause pain in animal models, suggesting that aldehyde dehydrogenase-2 (ALDH2), which metabolizes many aldehydes, may regulate nociception. To test this hypothesis, we generated a knock-in mouse with an inactivating point mutation in ALDH2 (ALDH2*2), which is also present in human ALDH2 of ~540 million East Asians. The ALDH2*1/*2 heterozygotic mice exhibited a larger response to painful stimuli than their wild-type littermates, and this heightened nociception was inhibited by an ALDH2-selective activator (Alda-1). No effect on inflammation per se was observed. Using a rat model, we then showed that nociception tightly correlated with ALDH activity (R(2) = 0.90) and that reduced nociception was associated with less early growth response protein 1 (EGR1) in the spinal cord and less reactive aldehyde accumulation at the insult site (including acetaldehyde and 4-hydroxynonenal). Further, acetaldehyde- and formalin-induced nociceptive behavior was greater in the ALDH2*1/*2 mice than in the wild-type mice. Finally, Alda-1 treatment was even beneficial when given after the inflammatory agent was administered. Our data in rodent models suggest that the mitochondrial enzyme ALDH2 regulates nociception and could serve as a molecular target for pain control, with ALDH2 activators, such as Alda-1, as potential non-narcotic, cardiac-safe analgesics. Furthermore, our results suggest a possible genetic basis for East Asians' apparent lower pain tolerance.
Collapse
Affiliation(s)
- Vanessa O Zambelli
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. Laboratory of Pain and Signaling, Butantan Institute, São Paulo 05503-900, Brazil
| | - Eric R Gross
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vanessa P Gutierrez
- Laboratory of Pain and Signaling, Butantan Institute, São Paulo 05503-900, Brazil
| | - Yara Cury
- Laboratory of Pain and Signaling, Butantan Institute, São Paulo 05503-900, Brazil
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
96
|
Gabbanini S, Matera R, Valvassori A, Valgimigli L. Rapid liquid chromatography–tandem mass spectrometry analysis of 4-hydroxynonenal for the assessment of oxidative degradation and safety of vegetable oils. Anal Chim Acta 2015; 869:50-8. [DOI: 10.1016/j.aca.2015.02.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 02/13/2015] [Accepted: 02/16/2015] [Indexed: 10/24/2022]
|
97
|
Riahi Y, Kaiser N, Cohen G, Abd-Elrahman I, Blum G, Shapira OM, Koler T, Simionescu M, Sima AV, Zarkovic N, Zarkovic K, Orioli M, Aldini G, Cerasi E, Leibowitz G, Sasson S. Foam cell-derived 4-hydroxynonenal induces endothelial cell senescence in a TXNIP-dependent manner. J Cell Mol Med 2015; 19:1887-99. [PMID: 25754218 PMCID: PMC4549039 DOI: 10.1111/jcmm.12561] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 01/15/2015] [Indexed: 01/01/2023] Open
Abstract
Vascular endothelial cell (VEC) senescence is considered an early event in the development of atherosclerotic lesions. Stressful stimuli, in particular oxidative stress, have been linked to premature senescence in the vasculature. Foam cells are a major source of reactive oxygen species and may play a role in the induction of VEC senescence; hence, we investigated their involvement in the induction of VEC senescence in a co-culture transwell system. Primary bovine aortic endothelial cells, exposed to the secretome of THP-1 monocyte-derived foam cells, were analysed for the induction of senescence. Senescence associated β-galactosidase activity and the expression of p16 and p21 were increased, whereas phosphorylated retinoblastoma protein was reduced. This senescent phenotype was mediated by 4-hydroxnonenal (4-HNE), a lipid peroxidation product secreted from foam cells; scavenging of 4-HNE in the co-culture medium blunted this effect. Furthermore, both foam cells and 4-HNE increased the expression of the pro-oxidant thioredoxin-interacting protein (TXNIP). Molecular manipulation of TXNIP expression confirmed its involvement in foam cell-induced senescence. Previous studies showed that peroxisome proliferator-activated receptor (PPAR)δ was activated by 4-hydroalkenals, such as 4-HNE. Pharmacological interventions supported the involvement of the 4-HNE-PPARδ axis in the induction of TXNIP and VEC senescence. The association of TXNIP with VEC senescence was further supported by immunofluorescent staining of human carotid plaques in which the expression of both TXNIP and p21 was augmented in endothelial cells. Collectively, these findings suggest that foam cell-released 4-HNE activates PPARδ in VEC, leading to increased TXNIP expression and consequently to senescence.
Collapse
Affiliation(s)
- Yael Riahi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel.,Endocrinology and Metabolism Service, Department of Medicine, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Nurit Kaiser
- Endocrinology and Metabolism Service, Department of Medicine, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Guy Cohen
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Ihab Abd-Elrahman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Galia Blum
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Oz M Shapira
- Department of Cardiothoracic Surgery, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Tomer Koler
- Department of Cardiothoracic Surgery, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology N. Simionescu of The Romanian Academy, Bucharest, Romania
| | - Anca V Sima
- Institute of Cellular Biology and Pathology N. Simionescu of The Romanian Academy, Bucharest, Romania
| | - Neven Zarkovic
- Laboratory for Oxidative Stress, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Kamelija Zarkovic
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Marica Orioli
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Erol Cerasi
- Endocrinology and Metabolism Service, Department of Medicine, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Gil Leibowitz
- Endocrinology and Metabolism Service, Department of Medicine, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Shlomo Sasson
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
98
|
Manea A, Manea SA, Todirita A, Albulescu IC, Raicu M, Sasson S, Simionescu M. High-glucose-increased expression and activation of NADPH oxidase in human vascular smooth muscle cells is mediated by 4-hydroxynonenal-activated PPARα and PPARβ/δ. Cell Tissue Res 2015; 361:593-604. [PMID: 25722086 DOI: 10.1007/s00441-015-2120-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 12/29/2014] [Indexed: 01/04/2023]
Abstract
High glucose induces vascular smooth muscle cell (SMC) dysfunction by generating oxidative stress attributable, in part, to the up-regulated NADPH oxidases (Nox). We have attempted to elucidate the high-glucose-generated molecular signals that mediate this effect and hypothesize that products of high-glucose-induced lipid peroxidation regulate Nox by activating peroxisome proliferator-activated receptors (PPARs). Human aortic SMCs were exposed to glucose (5.5-25 mM) or 4-hydroxynonenal (1-25 μM, 4-HNE). Lucigenin assay, real-time polymerase chain reaction, western blot, and promoter analyses were employed to investigate Nox. We found that high glucose generated an increase in Nox activity and expression. It also promoted oxidative stress that consequently induced lipid peroxidation, which resulted in the production of 4-HNE. Pharmacological inhibition of Nox activity significantly reduced the formation of high-glucose-induced 4-HNE. Exposure of SMCs to non-cytotoxic concentrations (1-10 μM) of 4-HNE alone mimicked the effect of high glucose incubation, whereas scavenging of 4-HNE by N-acetyl L-cysteine completely abolished both the effects of high glucose and 4-HNE. The latter exerted its effect by activating PPARα and PPARβ/δ, but not PPARγ, as assessed pharmacologically by the inhibitory effect of selective antagonists and following the silencing of the expression of these receptors. These new data indicate that 4-HNE, generated following Nox activation, functions as an endogenous activator of PPARα and PPARβ/δ. The newly discovered "lipid peroxidation products-PPARs-Nox axis" represents a novel mechanism of Nox regulation and an additional therapeutic target for oxidative stress in diabetes.
Collapse
Affiliation(s)
- Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8 B.P. Hasdeu Street, 050568, Bucharest, Romania,
| | | | | | | | | | | | | |
Collapse
|
99
|
Huang SK, Zhang Q, Qiu Z, Chung KF. Mechanistic impact of outdoor air pollution on asthma and allergic diseases. J Thorac Dis 2015; 7:23-33. [PMID: 25694815 DOI: 10.3978/j.issn.2072-1439.2014.12.13] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 12/03/2014] [Indexed: 12/28/2022]
Abstract
Over the past decades, asthma and allergic diseases, such as allergic rhinitis and eczema, have become increasingly common, but the reason for this increased prevalence is still unclear. It has become apparent that genetic variation alone is not sufficient to account for the observed changes; rather, the changing environment, together with alterations in lifestyle and eating habits, are likely to have driven the increase in prevalence, and in some cases, severity of disease. This is particularly highlighted by recent awareness of, and concern about, the exposure to ubiquitous environmental pollutants, including chemicals with oxidant-generating capacities, and their impact on the human respiratory and immune systems. Indeed, several epidemiological studies have identified a variety of risk factors, including ambient pollutant gases and airborne particles, for the prevalence and the exacerbation of allergic diseases. However, the responsible pollutants remain unclear and the causal relationship has not been established. Recent studies of cellular and animal models have suggested several plausible mechanisms, with the most consistent observation being the direct effects of particle components on the generation of reactive oxygen species (ROS) and the resultant oxidative stress and inflammatory responses. This review attempts to highlight the experimental findings, with particular emphasis on several major mechanistic events initiated by exposure to particulate matters (PMs) in the exposure-disease relationship.
Collapse
Affiliation(s)
- Shau-Ku Huang
- 1 Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 115 Zhunan, Taiwan ; 2 Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA ; 3 State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China ; 4 National Heart & Lung Institute, Imperial College London & Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK
| | - Qingling Zhang
- 1 Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 115 Zhunan, Taiwan ; 2 Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA ; 3 State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China ; 4 National Heart & Lung Institute, Imperial College London & Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK
| | - Zhiming Qiu
- 1 Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 115 Zhunan, Taiwan ; 2 Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA ; 3 State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China ; 4 National Heart & Lung Institute, Imperial College London & Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK
| | - Kian Fan Chung
- 1 Division of Environmental Health and Occupational Medicine, National Health Research Institutes, 115 Zhunan, Taiwan ; 2 Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA ; 3 State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510120, China ; 4 National Heart & Lung Institute, Imperial College London & Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK
| |
Collapse
|
100
|
Hermann PM, Watson SN, Wildering WC. Phospholipase A2 - nexus of aging, oxidative stress, neuronal excitability, and functional decline of the aging nervous system? Insights from a snail model system of neuronal aging and age-associated memory impairment. Front Genet 2014; 5:419. [PMID: 25538730 PMCID: PMC4255604 DOI: 10.3389/fgene.2014.00419] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 11/13/2014] [Indexed: 02/02/2023] Open
Abstract
The aging brain undergoes a range of changes varying from subtle structural and physiological changes causing only minor functional decline under healthy normal aging conditions, to severe cognitive or neurological impairment associated with extensive loss of neurons and circuits due to age-associated neurodegenerative disease conditions. Understanding how biological aging processes affect the brain and how they contribute to the onset and progress of age-associated neurodegenerative diseases is a core research goal in contemporary neuroscience. This review focuses on the idea that changes in intrinsic neuronal electrical excitability associated with (per)oxidation of membrane lipids and activation of phospholipase A2 (PLA2) enzymes are an important mechanism of learning and memory failure under normal aging conditions. Specifically, in the context of this special issue on the biology of cognitive aging we portray the opportunities offered by the identifiable neurons and behaviorally characterized neural circuits of the freshwater snail Lymnaea stagnalis in neuronal aging research and recapitulate recent insights indicating a key role of lipid peroxidation-induced PLA2 as instruments of aging, oxidative stress and inflammation in age-associated neuronal and memory impairment in this model system. The findings are discussed in view of accumulating evidence suggesting involvement of analogous mechanisms in the etiology of age-associated dysfunction and disease of the human and mammalian brain.
Collapse
Affiliation(s)
- Petra M Hermann
- Department of Biological Sciences, University of Calgary Calgary, AB, Canada ; Department of Physiology and Pharmacology, University of Calgary Calgary, AB, Canada
| | - Shawn N Watson
- Department of Biological Sciences, University of Calgary Calgary, AB, Canada
| | - Willem C Wildering
- Department of Biological Sciences, University of Calgary Calgary, AB, Canada ; Department of Physiology and Pharmacology, University of Calgary Calgary, AB, Canada ; Hotchkiss Brain Institute, University of Calgary Calgary, AB, Canada
| |
Collapse
|