1
|
Curieses Andrés CM, Pérez de la Lastra JM, Andrés Juan C, Plou FJ, Pérez-Lebeña E. From reactive species to disease development: Effect of oxidants and antioxidants on the cellular biomarkers. J Biochem Mol Toxicol 2023; 37:e23455. [PMID: 37437103 DOI: 10.1002/jbt.23455] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023]
Abstract
The influence of modern lifestyle, diet, exposure to chemicals such as phytosanitary substances, together with sedentary lifestyles and lack of exercise play an important role in inducing reactive stress (RS) and disease. The imbalance in the production and scavenging of free radicals and the induction of RS (oxidative, nitrosative, and halogenative) plays an essential role in the etiology of various chronic pathologies, such as cardiovascular diseases, diabetes, neurodegenerative diseases, and cancer. The implication of free radicals and reactive species injury in metabolic disturbances and the onset of many diseases have been accumulating for several decades, and are now accepted as a major cause of many chronic diseases. Exposure to elevated levels of free radicals can cause molecular structural impact on proteins, lipids, and DNA, as well as functional alteration of enzyme homeostasis, leading to aberrations in gene expression. Endogenous depletion of antioxidant enzymes can be mitigated using exogenous antioxidants. The current interest in the use of exogenous antioxidants as adjunctive agents for the treatment of human diseases allows a better understanding of these diseases, facilitating the development of new therapeutic agents with antioxidant activity to improve the treatment of various diseases. Here we examine the role that RS play in the initiation of disease and in the reactivity of free radicals and RS in organic and inorganic cellular components.
Collapse
Affiliation(s)
| | | | - Celia Andrés Juan
- Department of Organic Chemistry, Cinquima Institute, Faculty of Sciences, Valladolid University, Valladolid, Spain
| | - Francisco J Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, Madrid, Spain
| | | |
Collapse
|
2
|
Musetti B, Bahnson EM, Thomson L. Cannabinoids in inflammation and atherosclerosis. MEDICINAL USAGE OF CANNABIS AND CANNABINOIDS 2023:159-169. [DOI: 10.1016/b978-0-323-90036-2.00016-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Impact of Reactive Species on Amino Acids-Biological Relevance in Proteins and Induced Pathologies. Int J Mol Sci 2022; 23:ijms232214049. [PMID: 36430532 PMCID: PMC9692786 DOI: 10.3390/ijms232214049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
This review examines the impact of reactive species RS (of oxygen ROS, nitrogen RNS and halogens RHS) on various amino acids, analyzed from a reactive point of view of how during these reactions, the molecules are hydroxylated, nitrated, or halogenated such that they can lose their capacity to form part of the proteins or peptides, and can lose their function. The reactions of the RS with several amino acids are described, and an attempt was made to review and explain the chemical mechanisms of the formation of the hydroxylated, nitrated, and halogenated derivatives. One aim of this work is to provide a theoretical analysis of the amino acids and derivatives compounds in the possible positions. Tyrosine, methionine, cysteine, and tryptophan can react with the harmful peroxynitrite or •OH and •NO2 radicals and glycine, serine, alanine, valine, arginine, lysine, tyrosine, histidine, cysteine, methionine, cystine, tryptophan, glutamine and asparagine can react with hypochlorous acid HOCl. These theoretical results may help to explain the loss of function of proteins subjected to these three types of reactive stresses. We hope that this work can help to assess the potential damage that reactive species can cause to free amino acids or the corresponding residues when they are part of peptides and proteins.
Collapse
|
4
|
Sisto M, Ribatti D, Lisi S. Molecular Mechanisms Linking Inflammation to Autoimmunity in Sjögren's Syndrome: Identification of New Targets. Int J Mol Sci 2022; 23:13229. [PMID: 36362017 PMCID: PMC9658723 DOI: 10.3390/ijms232113229] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 10/15/2023] Open
Abstract
Sjögren's syndrome (SS) is a systemic autoimmune rheumatic disorder characterized by the lymphocytic infiltration of exocrine glands and the production of autoantibodies to self-antigens. The involvement of the exocrine glands drives the pathognomonic manifestations of dry eyes (keratoconjunctivitis sicca) and dry mouth (xerostomia) that define sicca syndrome. To date, the molecular mechanisms mediating pathological salivary gland dysfunction in SS remain to be elucidated, despite extensive studies investigating the underlying cause of this disease, hampering the development of novel therapeutic strategies. Many researchers have identified a multifactorial pathogenesis of SS, including environmental, genetic, neuroendocrine, and immune factors. In this review, we explore the latest developments in understanding the molecular mechanisms involved in the pathogenesis of SS, which have attracted increasing interest in recent years.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari “Aldo Moro”, Piazza Giulio Cesare 1, I-70124 Bari, Italy
| | | | | |
Collapse
|
5
|
Comparison of Five Oxidative Stress Biomarkers in Vegans and Omnivores from Germany and Finland. Nutrients 2022; 14:nu14142918. [PMID: 35889875 PMCID: PMC9323774 DOI: 10.3390/nu14142918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
When the amount of reactive oxygen species produced by human metabolism cannot be balanced by antioxidants, this phenomenon is commonly referred to as oxidative stress. It is hypothesised that diets with high amounts of plant food products may have a beneficial impact on oxidative stress status. However, few studies have examined whether a vegan diet is associated with lower oxidative stress compared to an omnivorous diet. The present cross-sectional study aimed to compare the levels of five oxidative stress biomarkers in vegans and omnivores. Data of 36 vegans and 36 omnivores from Germany and of 21 vegans and 18 omnivores from Finland were analysed. HPLC coupled with mass spectrometry or fluorescence detection and ELISA methods were used to measure the oxidative stress biomarkers malondialdehyde (MDA), protein carbonyls and 3-nitrotyrosine in plasma and 8-hydroxy-2'-deoxyguanosine (8-OHdG) and 8-iso-prostaglandin F2α (8-iso-PGF2α) in 24 h urine. Analyses of variance and covariance, considering potential confounders, were used. Vegans and omnivores showed no differences in MDA and protein carbonyl concentrations. In Finnish but not in German vegans, the concentrations of 3-nitrotyrosine were lower compared to those in omnivores (p = 0.047). In Germany, vegans showed lower excretion levels of 8-iso-PGF2α than omnivores (p = 0.002) and with a trend also of 8-OHdG (p = 0.05). The sensitivity analysis suggests lower 8-iso-PGF2α excretion levels in women compared to men, independently of the dietary group. The present study contributes to expanding our knowledge of the relationship between diet and oxidative stress and showed that 3-nitrotyrosine, 8-OHdG and 8-iso-PGF2α tended to be lower in vegans. Furthermore, studies are recommended to validate the present findings.
Collapse
|
6
|
Jiang M, Zhao XM, Jiang ZS, Wang GX, Zhang DW. Protein tyrosine nitration in atherosclerotic endothelial dysfunction. Clin Chim Acta 2022; 529:34-41. [PMID: 35149004 DOI: 10.1016/j.cca.2022.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 11/29/2022]
Abstract
Accumulation of reactive oxygen species (ROS) can induce both protein tyrosine nitration and endothelial dysfunction in atherosclerosis. Endothelial dysfunction refers to impaired endothelium-dependent vasorelaxation that can be triggered by an imbalance in nitric oxide (NO) production and consumption. ROS reacts with NO to generate peroxynitrite, decreasing NO bioavailability. Peroxynitrite also promotes protein tyrosine nitration in vivo that can affect protein structure and function and further damage endothelial function. In this review, we discuss the process of protein tyrosine nitration, increased expression of nitrated proteins in cardiovascular disease and their association with endothelial dysfunction, and the interference of tyrosine nitration with antioxidants and the protective role in endothelial dysfunction. These may lead us to the conception that protein tyrosine nitration may be one of the causes of endothelial dysfunction, and help us gain information about the mechanism of endothelial dysfunction underlying atherosclerosis.
Collapse
Affiliation(s)
- Miao Jiang
- Institute of Cardiovascular Disease, Department of Pathophysiology, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic disease, Postdoctoral Research Station of Basic Medicine, University of South China, Hengyang, 421001, China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering Collage of Chongqing University, Chongqing, 400030, China
| | - Xiao-Mei Zhao
- College of Public Health, University of South China, Hengyang, 421001, Hunan, China
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Department of Pathophysiology, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic disease, Postdoctoral Research Station of Basic Medicine, University of South China, Hengyang, 421001, China.
| | - Gui-Xue Wang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering Collage of Chongqing University, Chongqing, 400030, China.
| | - Da-Wei Zhang
- Group on the Molecular and Cell Biology of Lipids, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Pahimi N, Rasool AHG, Sanip Z, Bokti NA, Yusof Z, W. Isa WYH. An Evaluation of the Role of Oxidative Stress in Non-Obstructive Coronary Artery Disease. J Cardiovasc Dev Dis 2022; 9:jcdd9020051. [PMID: 35200704 PMCID: PMC8878238 DOI: 10.3390/jcdd9020051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Approximately half of all women presenting to the emergency department with angina chest pain do not have obstructive coronary artery disease (CAD) on coronary angiography. This condition is termed non-obstructive coronary artery disease (NOCAD), and includes ischemia with no obstructive coronary artery disease (INOCA) and myocardial infarction with non-obstructive coronary arteries (MINOCA). Oxidative stress has been reported to be involved in the development and progression of CAD. However, a scarcity of studies has assessed a correlation between oxidative stress and NOCAD. Thus, a literature review was performed of available reports on the role of oxidative stress in NOCAD. Possible mechanisms involved in oxidative stress that may contribute to NOCAD were identified and evaluated. A key finding of this literature review was that oxidative stress caused vasoconstriction and endothelial damage, and this results in coronary microvascular dysfunction and vasospasm, which, in turn, lead to the pathogenesis of NOCAD.
Collapse
Affiliation(s)
- Nurnajwa Pahimi
- Pharmacology Vascular Laboratory, Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Malaysia; (N.P.); (A.H.G.R.)
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
| | - Aida Hanum Ghulam Rasool
- Pharmacology Vascular Laboratory, Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Malaysia; (N.P.); (A.H.G.R.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Zulkefli Sanip
- Central Research Laboratory, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia;
| | - Nur Adilah Bokti
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Zurkurnai Yusof
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - W. Yus Haniff W. Isa
- Cardiology Unit, Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia; (N.A.B.); (Z.Y.)
- Department of Internal Medicine, Hospital Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Correspondence: ; Tel.: +60-9767-3987
| |
Collapse
|
8
|
El-Mahdy MA, Ewees MG, Eid MS, Mahgoup EM, Khaleel SA, Zweier JL. Electronic Cigarette Exposure Causes Vascular Endothelial Dysfunction Due to NADPH Oxidase Activation and eNOS Uncoupling. Am J Physiol Heart Circ Physiol 2022; 322:H549-H567. [PMID: 35089811 PMCID: PMC8917923 DOI: 10.1152/ajpheart.00460.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We recently reported a mouse model of chronic electronic cigarette (e-cig) exposure-induced cardiovascular pathology, where long-term exposure to e-cig vape (ECV) induces cardiac abnormalities, impairment of endothelial function, and systemic hypertension. Here, we delineate the underlying mechanisms of ECV-induced vascular endothelial dysfunction (VED), a central trigger of cardiovascular disease. C57/BL6 male mice were exposed to ECV generated from e-cig liquid containing 0, 6, or 24 mg/ml nicotine for 16 and 60 weeks. Time-dependent elevation in blood pressure and systemic vascular resistance were observed, along with an impairment of acetylcholine-induced aortic relaxation in ECV-exposed mice, compared to air-exposed control. Decreased intravascular nitric oxide (NO) levels and increased superoxide generation with elevated 3-nitrotyrosine levels in the aorta of ECV-exposed mice were observed, indicating that ECV-induced superoxide reacts with NO to generate cytotoxic peroxynitrite. Exposure increased NADPH oxidase expression, supporting its role in ECV-induced superoxide generation. Downregulation of endothelial nitric oxide synthase (eNOS) expression and Akt-dependent eNOS phosphorylation occurred in the aorta of ECV-exposed mice, indicating that exposure inhibited de novo NO synthesis. Following ECV exposure, the critical NOS cofactor tetrahydrobiopterin was decreased, with a concomitant loss of its salvage enzyme, dihydrofolate reductase. NADPH oxidase and NOS inhibitors abrogated ECV-induced superoxide generation in the aorta of ECV exposed mice. Together, our data demonstrate that ECV exposure activates NADPH oxidase and uncouples eNOS, causing a vicious cycle of superoxide generation and vascular oxidant stress that triggers VED and hypertension with predisposition to other cardiovascular disease.
Collapse
Affiliation(s)
- Mohamed A El-Mahdy
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Mohamed G Ewees
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Mahmoud S Eid
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Elsayed M Mahgoup
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Sahar A Khaleel
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Jay L Zweier
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
9
|
Yoshida N, Kato Y, Takatsu H, Fukui K. Relationship between Cognitive Dysfunction and Age-Related Variability in Oxidative Markers in Isolated Mitochondria of Alzheimer's Disease Transgenic Mouse Brains. Biomedicines 2022; 10:281. [PMID: 35203488 PMCID: PMC8869326 DOI: 10.3390/biomedicines10020281] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
Many neurodegenerative disorders, including Alzheimer's disease (AD), are strongly associated with the accumulation of oxidative damage. Transgenic animal models are commonly used to elucidate the pathogenic mechanism of AD. Beta amyloid (Aβ) and tau hyperphosphorylation are very famous hallmarks of AD and well-studied, but the relationship between mitochondrial dysfunction and the onset and progression of AD requires further elucidation. In this study we used transgenic mice (the strain name is 5xFAD) at three different ages (3, 6, and 20 months old) as an AD model. Cognitive impairment in AD mice occurred in an age-dependent manner. Aβ1-40 expression significantly increased in an age-dependent manner in all brain regions with or without AD, and Aβ1-42 expression in the hippocampus increased at a young age. In a Western blot analysis using isolated mitochondria from three brain regions (cerebral cortex, cerebellum, and hippocampus), NMNAT-3 expression in the hippocampi of aged AD mice was significantly lower than that of young AD mice. SOD-2 expression in the hippocampi of AD mice was lower than for the age-matched controls. However, 3-NT expression in the hippocampi of AD mice was higher than for the age-matched controls. NQO-1 expression in the cerebral cortex of AD mice was higher than for the age-matched controls at every age that we examined. However, hippocampal NQO-1 expression in 6-month-old AD mice was significantly lower than in 3-month-old AD mice. These results indicate that oxidative stress in the hippocampi of AD mice is high compared to other brain regions and may induce mitochondrial dysfunction via oxidative damage. Protection of mitochondria from oxidative damage may be important to maintain cognitive function.
Collapse
Affiliation(s)
- Naoki Yoshida
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan;
| | - Yugo Kato
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan;
| | - Hirokatsu Takatsu
- Department of Medical Technology, Faculty of Health Sciences, Kyorin University, Shimorenjaku 5-4-1, Mitaka, Tokyo 181-8612, Japan;
| | - Koji Fukui
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan;
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Fukasaku 307, Minuma-ku, Saitama 337-8570, Japan;
| |
Collapse
|
10
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
11
|
Ramazan ZK, Sarı İ, Yıldırım BG, Güntürk İ, Küçük E, Erşan S, Seydel GŞ. The Evaluation of oxidative stress, 3-nitrotyrosine, and HMGB-1 levels in patients with Wet Type Age-Related Macular Degeneration. J Med Biochem 2021; 41:275-281. [PMID: 36042902 PMCID: PMC9375537 DOI: 10.5937/jomb0-32189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/19/2021] [Indexed: 11/04/2022] Open
Abstract
Background This study aims to compare serum HMGB-1, 3-nitrotyrosine (3-NT), TAS, TOS, and OSI levels in Wettype Age-Related Macular Degeneration (wAMD) patients and healthy controls to determine the correlation of these parameters with each other. Methods Thirty patients with Wet-type Age-Related Macular Degeneration (wAMD) and 27 healthy adults, as controls were enrolled in the study. We determined the TAS and TOS levels in serum samples of both groups using commercial kits on a microplate reader. Serum HMGB-1 and 3-NT levels were measured with the enzyme-linked immunosorbent assay method. Results HMGB-1 levels were significantly higher in the patient group (137.51 pg/mL, p=0.001), while there was no difference between the two groups in serum 3-NT levels (p=0.428). A statistically significant difference found in the levels of TOS and OSI (p=0.001 and p=0.045, respectively) between the patients and controls, however, no significant difference was observed between the groups in terms of TAS levels (p=0.228). Conclusions Oxidative stress and HMGB-1 levels were increased in wAMD patients and enhanced oxidative stress may be associated with increased tissue necrosis and inflammation. Thus administration of antioxidant treatment in addition to routine therapy should be considered in wAMD.
Collapse
Affiliation(s)
- Zor Kürşad Ramazan
- Niğde Ömer Halisdemir University School of Medicine Department of Ophthalmology, Bor Yolu, Niğde, Turkey
| | - İsmail Sarı
- Niğde Ömer Halisdemir University School of Medicine Department of Biochemistry, Bor Yolu, Niğde, Turkey
| | - Biçer Gamze Yıldırım
- Niğde Ömer Halisdemir University School of Medicine Department of Ophthalmology, Bor Yolu, Niğde, Turkey
| | - İnayet Güntürk
- Niğde Ömer Halisdemir University, Healthcare Services, Zübeyde Hanım Health Services Vocational High School, Bor Yolu, Niğde, Turkey
| | - Erkut Küçük
- Niğde Ömer Halisdemir University School of Medicine Department of Ophthalmology, Bor Yolu, Niğde, Turkey
| | - Serpil Erşan
- Niğde Ömer Halisdemir University School of Medicine Department of Biochemistry, Bor Yolu, Niğde, Turkey
| | - Gönül Şeyda Seydel
- Niğde Ömer Halisdemir University School of Medicine Department of Medical Biochemistry, Bor Yolu, Niğde, Turkey
| |
Collapse
|
12
|
Igel E, Haller A, Wolfkiel PR, Orr-Asman M, Jaeschke A, Hui DY. Distinct pro-inflammatory properties of myeloid cell-derived apolipoprotein E2 and E4 in atherosclerosis promotion. J Biol Chem 2021; 297:101106. [PMID: 34425108 PMCID: PMC8437825 DOI: 10.1016/j.jbc.2021.101106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 11/25/2022] Open
Abstract
Polymorphisms in the apolipoprotein E (apoE) gene are risk factors for chronic inflammatory diseases including atherosclerosis. The gene product apoE is synthesized in many cell types and has both lipid transport–dependent and lipid transport–independent functions. Previous studies have shown that apoE expression in myeloid cells protects against atherogenesis in hypercholesterolemic ApoE−/− mice. However, the mechanism of this protection is still unclear. Using human APOE gene replacement mice as models, this study showed that apoE2 and apoE4 expressed endogenously in myeloid cells enhanced the inflammatory response via mechanisms independent of plasma lipoprotein transport. The data revealed that apoE2-expressing myeloid cells contained higher intracellular cholesterol levels because of impaired efflux, causing increasing inflammasome activation and myelopoiesis. In contrast, intracellular cholesterol levels were not elevated in apoE4-expressing myeloid cells, and its proinflammatory property was found to be independent of inflammasome signaling and related to enhanced oxidative stress. When ApoE−/− mice were reconstituted with bone marrow from various human APOE gene replacement mice, effective reduction of atherosclerosis was observed with marrow cells obtained from APOE3 but not APOE2 and APOE4 gene replacement mice. Taken together, these results documented that apoE2 and apoE4 expression in myeloid cells promotes inflammation via distinct mechanisms and promotes atherosclerosis in a plasma lipoprotein transport–independent manner.
Collapse
Affiliation(s)
- Emily Igel
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - April Haller
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Patrick R Wolfkiel
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Melissa Orr-Asman
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
13
|
Ferlazzo N, Currò M, Isola G, Maggio S, Bertuccio MP, Trovato-Salinaro A, Matarese G, Alibrandi A, Caccamo D, Ientile R. Changes in the Biomarkers of Oxidative/Nitrosative Stress and Endothelial Dysfunction Are Associated with Cardiovascular Risk in Periodontitis Patients. Curr Issues Mol Biol 2021; 43:704-715. [PMID: 34287264 PMCID: PMC8929118 DOI: 10.3390/cimb43020051] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with cardiovascular disease (CVD) and periodontitis (PT) show shared risk factors as result of the altered molecular mechanisms associated with pathological conditions. The aim of our study was to evaluate if the plasma biomarkers associated with endothelial dysfunction may also be related to alterations in the inflammatory status in peripheral blood mononuclear cells (PBMC). Patients with PT, coronary heart disease (CHD), or both diseases as well as controls were enrolled. Plasma levels of coenzyme Q10 (CoQ10), 3-nitrotyrosine (NT), and asymmetric dimethylarginine (ADMA) were assessed using HPLC. mRNA levels of caspase-1 (CASP1), NLR family pyrin domain containing 3 (NLRP3), and tumor necrosis factor-α (TNF-α) in PBMC from the recruited subjects were quantified using real-time PCR. Patients with PT + CHD showed lower CoQ10 plasma levels and increased concentrations of NT in comparison to healthy subjects. ADMA levels were higher in CHD and PT + CHD patients compared to controls. Transcript levels of CASP1, NLRP3, and TNF-α were up-regulated in PBMC from all patient groups when compared to healthy subjects. Our results suggest a possible causal link between oxidative stress, high levels of NT and ADMA, and inflammasome activation, which may be involved in the endothelial inflammatory dysfunction leading to the pathogenesis and progression of CHD in PT patients.
Collapse
Affiliation(s)
- Nadia Ferlazzo
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
- Correspondence: (N.F.); (M.C.); Tel.: +39-0902213389 (M.C.)
| | - Monica Currò
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
- Correspondence: (N.F.); (M.C.); Tel.: +39-0902213389 (M.C.)
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via S. Sofia 78, 95123 Catania, Italy;
| | - Silvia Maggio
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | - Maria Paola Bertuccio
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | | | - Giovanni Matarese
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | - Angela Alibrandi
- Unit of Statistical and Mathematical Sciences, Department of Economics, University of Messina, 98123 Messina, Italy;
| | - Daniela Caccamo
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| | - Riccardo Ientile
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98123 Messina, Italy; (S.M.); (M.P.B.); (G.M.); (D.C.); (R.I.)
| |
Collapse
|
14
|
Melatonin, Its Metabolites and Their Interference with Reactive Nitrogen Compounds. Molecules 2021; 26:molecules26134105. [PMID: 34279445 PMCID: PMC8271479 DOI: 10.3390/molecules26134105] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Melatonin and several of its metabolites are interfering with reactive nitrogen. With the notion of prevailing melatonin formation in tissues that exceeds by far the quantities in blood, metabolites come into focus that are poorly found in the circulation. Apart from their antioxidant actions, both melatonin and N1-acetyl-5-methoxykynuramine (AMK) downregulate inducible and inhibit neuronal NO synthases, and additionally scavenge NO. However, the NO adduct of melatonin redonates NO, whereas AMK forms with NO a stable product. Many other melatonin metabolites formed in oxidative processes also contain nitrosylatable sites. Moreover, AMK readily scavenges products of the CO2-adduct of peroxynitrite such as carbonate radicals and NO2. Protein AMKylation seems to be involved in protective actions.
Collapse
|
15
|
Kuschman HP, Palczewski MB, Thomas DD. Nitric oxide and hydrogen sulfide: Sibling rivalry in the family of epigenetic regulators. Free Radic Biol Med 2021; 170:34-43. [PMID: 33482335 DOI: 10.1016/j.freeradbiomed.2021.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 01/12/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) were previously only known for their toxic properties. Now they are regarded as potent gaseous messenger molecules (gasotransmitters) that rapidly transverse cell membranes and transduce cellular signals through their chemical reactions and modifications to protein targets. Both are known to regulate numerous physiological functions including angiogenesis, vascular tone, and immune response, to name a few. NO and H2S often work synergistically and in competition to regulate each other's synthesis, target protein activity via posttranslational modifications (PTMs), and chemical interactions. In addition to their canonical modes of action, increasing evidence has demonstrated that NO and H2S share another signaling mechanism: epigenetic regulation. This review will compare and contrast biosynthesis and metabolism of NO and H2S, their individual and shared interactions, and the growing body of evidence for their roles as endogenous epigenetic regulatory molecules.
Collapse
Affiliation(s)
- Hannah Petraitis Kuschman
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Marianne B Palczewski
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Douglas D Thomas
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States.
| |
Collapse
|
16
|
The Effects of Prunus spinosa L. Flower Extracts, Model Polyphenols and Phenolic Metabolites on Oxidative/Nitrative Modifications of Human Plasma Components with Particular Emphasis on Fibrinogen In Vitro. Antioxidants (Basel) 2021; 10:antiox10040581. [PMID: 33918684 PMCID: PMC8069707 DOI: 10.3390/antiox10040581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/28/2022] Open
Abstract
Oxidative post-translational modifications of fibrinogen (a multifunctional blood plasma protein essential for hemostasis) are associated with the pathogenesis of cardiovascular disorders (CVDs). Prunus spinosa flower is a herbal medicine used in an adjuvant treatment of CVDs and rich in polyphenolic antioxidants. In the present study, phytochemically standardized P. spinosa flower extracts, their primary native polyphenols and potential phenolic metabolites were evaluated in vitro for their protective effects on fibrinogen (isolated and in the human plasma matrix) using a panel of complementary methods (SDS-PAGE, western blot, C-ELISA, fluorometry, FRAP, TBARS). The results revealed that the tested analytes at in vivo relevant levels (1–5 µg/mL) considerably reduced the structural changes in the fibrinogen molecule under the oxidative stress conditions induced by peroxynitrite. In particular, they diminished the oxidation and/or nitration of amino acid residues, including tyrosine and tryptophan, as well as the formation of high molecular weight aggregates. The decrease in the levels of 3-nitrotyrosine was about 13.5–33.0% and 58.3–97.1% at 1 µg/mL and 50 µg/mL, respectively. The study indicated that low molecular weight polyphenols were crucial for the protective activity of the extracts toward fibrinogen and other human plasma components. The investigated model compounds effectively protected total plasma proteins and lipids against oxidative damage (by reducing the levels of 3-nitrotyrosine and thiobarbituric acid-reactive substances and normalizing/enhancing the non-enzymatic antioxidant capacity of plasma). The work provides insight into the role of native and metabolized polyphenols as contributory factors to the systemic activity of blackthorn flower extracts within the circulatory system.
Collapse
|
17
|
Li J, Wei J, Gao Z, Yin G, Li H. The oxidative reactivity of three manganese(III) porphyrin complexes with hydrogen peroxide and nitrite toward catalytic nitration of protein tyrosine. Metallomics 2021; 13:6134099. [PMID: 33576808 DOI: 10.1093/mtomcs/mfab005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/10/2021] [Accepted: 02/04/2021] [Indexed: 11/12/2022]
Abstract
Understanding the toxicological properties of MnIII-porphyrins (MnTPPS, MnTMPyP, or MnTBAP) can provide important biochemical rationales in developing them as the therapeutic drugs against protein tyrosine nitration-induced inflammation diseases. Here, we present a comprehensive understanding of the pH-dependent redox behaviors of these MnIII-porphyrins and their structural effects on catalyzing bovine serum albumin (BSA) nitration in the presence of H2O2 and NO2-. It was found that both MnTPPS and MnTBAP stand out in catalyzing BSA nitration at physiologically close condition (pH 8), yet they are less effective at pH 6 and 10. MnTMPyP was shown to have no ability to catalyze BSA nitration under all tested pHs (pH 6, 8, and 10). The kinetics and active intermediate determination through electrochemistry method revealed that both the pH-dependent redox behavior of the central metal cation and the antioxidant capability of porphin derivative contribute to the catalytic activities of three MnIII-porphyrins in BSA nitration in the presence of H2O2/NO2-. These comprehensive studies on the oxidative reactivity of MnIII-porphyrins toward BSA nitration may provide new clues for searching the manganese-based therapeutic drugs against the inflammation-related diseases.
Collapse
Affiliation(s)
- Jiayu Li
- Hubei Provincial Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Jingjing Wei
- Hubei Provincial Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Zhonghong Gao
- Hubei Provincial Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Guochuan Yin
- Hubei Provincial Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| | - Hailing Li
- Hubei Provincial Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, P. R. China
| |
Collapse
|
18
|
Folkes LK, Bartesaghi S, Trujillo M, Wardman P, Radi R. The effects of nitric oxide or oxygen on the stable products formed from the tyrosine phenoxyl radical. Free Radic Res 2021; 55:141-153. [PMID: 33399021 DOI: 10.1080/10715762.2020.1870684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tyrosine is a critical component of many proteins and can be the subject of oxidative posttranslational modifications. Furthermore, the oxidation of tyrosine residues to phenoxyl radicals, sometimes quite stable, is essential for some enzymatic functions. The lifetime and fate of tyrosine phenoxyl radicals in biological systems are largely driven by the availability and proximity of oxidants and reductants. Tyrosine phenoxyl radicals have extremely low reactivity with molecular oxygen whereas reactions with nitric oxide are diffusion controlled. This is in contrast to equivalent reactions with tryptophanyl and cysteinyl radicals where reactions with oxygen are much faster. Despite, the quite disparate apparent reactivity of tyrosine phenoxyl radicals with oxygen and nitric oxide being known, the products of the reactions are not well established. Changes in the levels from expected basal concentrations of stable products resulting from tyrosine phenoxyl radicals, for example naturally occurring 3,3'-dityrosine, 3-nitrotyrosine, and 3-hydroxytyrosine, can be indicative of oxidative and/or nitrosative stress. Using the radiolytic generation of specific oxidizing radicals to form tyrosine phenoxyl radicals in an aqueous solution at a known rate, we have compared the products in the absence and presence of nitric oxide or oxygen. Possible reactions of the phenoxyl radicals with oxygen remain unclear although we show evidence for a small decrease in the yield of dityrosine and loss of tyrosine in the presence of 20% oxygen. Low concentrations of nitric oxide in anoxic conditions react with tyrosine phenoxyl radicals, by what is most probably through the formation of an unstable intermediate, regenerating tyrosine and forming nitrite.
Collapse
Affiliation(s)
- Lisa K Folkes
- MRC Oxford Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, UK
| | - Silvina Bartesaghi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Peter Wardman
- MRC Oxford Institute for Radiation Oncology and Biology, Department of Oncology, University of Oxford, Oxford, UK
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
19
|
The challenge of detecting modifications on proteins. Essays Biochem 2020; 64:135-153. [PMID: 31957791 DOI: 10.1042/ebc20190055] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
Post-translational modifications (PTMs) are integral to the regulation of protein function, characterising their role in this process is vital to understanding how cells work in both healthy and diseased states. Mass spectrometry (MS) facilitates the mass determination and sequencing of peptides, and thereby also the detection of site-specific PTMs. However, numerous challenges in this field continue to persist. The diverse chemical properties, low abundance, labile nature and instability of many PTMs, in combination with the more practical issues of compatibility with MS and bioinformatics challenges, contribute to the arduous nature of their analysis. In this review, we present an overview of the established MS-based approaches for analysing PTMs and the common complications associated with their investigation, including examples of specific challenges focusing on phosphorylation, lysine acetylation and redox modifications.
Collapse
|
20
|
Zhu Y, Shui X, Liang Z, Huang Z, Qi Y, He Y, Chen C, Luo H, Lei W. Gut microbiota metabolites as integral mediators in cardiovascular diseases (Review). Int J Mol Med 2020; 46:936-948. [PMID: 32705240 PMCID: PMC7388831 DOI: 10.3892/ijmm.2020.4674] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVDs), such as atherosclerosis, hypertension, myocardial infarction and diabetic heart disease, are associated with high morbidity and mortality rates worldwide, and may also induce multiple organ failure in their later stages, greatly reducing the long-term survival of the patients. There are several causes of CVDs, but after nearly a decade of investigation, researchers have found that CVDs are usually accompanied by an imbalance of gut microbiota and a decreased abundance of flora. More importantly, the metabolites produced by intestinal flora, such as trimethylamine and trimethylamine N-oxide, bile acids, short-chain fatty acids and aromatic amino acids, exert different effects on the occurrence and development of CVDs, as observed in the relevant pathways in the cells, which may either promote or protect against CVD occurrence. It is known that changes in the intestinal flora following antibiotic administration, diet supplementation with probiotics, or exercise, can interfere with the composition of the intestinal flora and may represent an effective approach to preventing or treating CVDs. The focus of this review was the analysis of gut microbiota metabolites to elucidate their effects on CVDs and to identify the most cost-effective and beneficial methods for treating CVDs with minimal side effects.
Collapse
Affiliation(s)
- Ying Zhu
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zheng Liang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Zufeng Huang
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Can Chen
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
21
|
Zhao J, Liang H, Shi W. Effect of serum 3-nitrotyrosine on the occurrence and development of carotid atherosclerosis in patients with essential hypertension. Minerva Med 2020; 112:670-671. [PMID: 32338481 DOI: 10.23736/s0026-4806.20.06558-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jing Zhao
- Department of Cardiology, Haijiya Hospital of Heze, Heze, China
| | - Houcai Liang
- Department of ICU, Dongda Hospital of Shanxian, Heze, China
| | - Wenbing Shi
- Department of Cardiology, Dongda Hospital of Shanxian, Heze, China -
| |
Collapse
|
22
|
Hernandez-Baixauli J, Quesada-Vázquez S, Mariné-Casadó R, Gil Cardoso K, Caimari A, Del Bas JM, Escoté X, Baselga-Escudero L. Detection of Early Disease Risk Factors Associated with Metabolic Syndrome: A New Era with the NMR Metabolomics Assessment. Nutrients 2020; 12:E806. [PMID: 32197513 PMCID: PMC7146483 DOI: 10.3390/nu12030806] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
The metabolic syndrome is a multifactorial disease developed due to accumulation and chronification of several risk factors associated with disrupted metabolism. The early detection of the biomarkers by NMR spectroscopy could be helpful to prevent multifactorial diseases. The exposure of each risk factor can be detected by traditional molecular markers but the current biomarkers have not been enough precise to detect the primary stages of disease. Thus, there is a need to obtain novel molecular markers of pre-disease stages. A promising source of new molecular markers are metabolomics standing out the research of biomarkers in NMR approaches. An increasing number of nutritionists integrate metabolomics into their study design, making nutrimetabolomics one of the most promising avenues for improving personalized nutrition. This review highlight the major five risk factors associated with metabolic syndrome and related diseases including carbohydrate dysfunction, dyslipidemia, oxidative stress, inflammation, and gut microbiota dysbiosis. Together, it is proposed a profile of metabolites of each risk factor obtained from NMR approaches to target them using personalized nutrition, which will improve the quality of life for these patients.
Collapse
Affiliation(s)
- Julia Hernandez-Baixauli
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Sergio Quesada-Vázquez
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Roger Mariné-Casadó
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
- Universitat Rovira i Virgili; Department of Biochemistry and Biotechnology, Ctra. De Valls, s/n, 43007 Tarragona, Spain
| | - Katherine Gil Cardoso
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
- Universitat Rovira i Virgili; Department of Biochemistry and Biotechnology, Ctra. De Valls, s/n, 43007 Tarragona, Spain
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Xavier Escoté
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| | - Laura Baselga-Escudero
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain; (J.H.-B.); (S.Q.-V.); (R.M.-C.); (K.G.C.); (A.C.); (J.M.D.B.)
| |
Collapse
|
23
|
Sabadashka M, Nagalievska M, Sybirna N. Tyrosine nitration as a key event of signal transduction that regulates functional state of the cell. Cell Biol Int 2020; 45:481-497. [PMID: 31908104 DOI: 10.1002/cbin.11301] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/04/2020] [Indexed: 12/21/2022]
Abstract
This review is dedicated to the role of nitration of proteins by tyrosine residues in physiological and pathological conditions. First of all, we analyze the biochemical evidence of peroxynitrite formation and reactions that lead to its formation, types of posttranslational modifications (PTMs) induced by reactive nitrogen species, as well as three biological pathways of tyrosine nitration. Then, we describe two possible mechanisms of protein nitration that are involved in intracellular signal transduction, as well as its interconnection with phosphorylation/dephosphorylation of tyrosine. Next part of the review is dedicated to the role of proteins nitration in different pathological conditions. In this section, special attention is devoted to the role of nitration in changes of functional properties of actin-protein that undergoes PTMs both in normal and pathological conditions. Overall, this review is devoted to the main features of protein nitration by tyrosine residue and the role of this process in intracellular signal transduction in basal and pathological conditions.
Collapse
Affiliation(s)
- Mariya Sabadashka
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4, Hrushevskyi St., Lviv, 79005, Ukraine
| | - Mariia Nagalievska
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4, Hrushevskyi St., Lviv, 79005, Ukraine
| | - Nataliia Sybirna
- Department of Biochemistry, Faculty of Biology, Ivan Franko National University of Lviv, 4, Hrushevskyi St., Lviv, 79005, Ukraine
| |
Collapse
|
24
|
Mastrogiovanni M, Trostchansky A, Rubbo H. Fatty acid nitration in human low-density lipoprotein. Arch Biochem Biophys 2020; 679:108190. [PMID: 31738891 DOI: 10.1016/j.abb.2019.108190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 10/25/2022]
Abstract
Lipid nitration occurs during physiological and pathophysiological conditions, generating a variety of biomolecules capable to modulate inflammatory cell responses. Low-density lipoprotein (LDL) oxidation has been extensively related to atherosclerotic lesion development while oxidative modifications confer the particle pro-atherogenic features. Herein, we reviewed the oxidation versus nitration of human LDL protein and lipid fractions. We propose that unsaturated fatty acids present in LDL can be nitrated under mild nitration conditions, suggesting an anti-atherogenic role for LDL carrying nitro-fatty acids (NFA).
Collapse
Affiliation(s)
- Mauricio Mastrogiovanni
- Departamento de Bioquímica, Facultad de Medicina and Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Andrés Trostchansky
- Departamento de Bioquímica, Facultad de Medicina and Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay
| | - Homero Rubbo
- Departamento de Bioquímica, Facultad de Medicina and Centro de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
25
|
Aguilar EC, Navia-Pelaez JM, Fernandes-Braga W, Soares FLP, Dos Santos LC, Leonel AJ, Capettini LDSA, de Oliveira RP, de Faria AMC, Lemos VS, Alvarez-Leite JI. Gluten exacerbates atherosclerotic plaque formation in ApoE -/- mice with diet-induced obesity. Nutrition 2019; 75-76:110658. [PMID: 32305657 DOI: 10.1016/j.nut.2019.110658] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/25/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Atherosclerosis is an underlying cause of cardiovascular disease, and obesity is one of the risk factors for atherogenesis. Although a gluten-free diet (GFD) has gained popularity as a strategy for weight loss, little is known about the effects of gluten on obesity. We have previously shown a negative effect of gluten on obesity in mice. However, its effects on atherogenesis are still unknown. Therefore, the aim of this study was to determine the effects of gluten on atherosclerosis progression during obesity. METHODS Atherosclerosis-susceptible ApoE knockout mice were subjected to an obesogenic GFD or a diet with 4.5% gluten (GD) for 10 wk. RESULTS Results from the study found that food intake and lipid profile were similar between the groups. However, GD promoted an increase in weight gain, adiposity, and plasma glucose. Pro-inflammatory factors such as tumor necrosis factor, interleukin-6, chemokine ligand-2, and matrix metalloproteinase-2 and -9 also were increased in the adipose tissue of gluten-fed mice. This inflammatory profile was associated with reduced phosphorylation of Akt, and consequently with the intensification of insulin resistance. The GD-enhanced vascular inflammation contributed to the worsening of atherosclerosis in the aorta and aortic root. Inflammatory cells, such as monocyte/macrophage and natural killer cells, and oxidative stress markers, such as superoxide and nitrotyrosine, were increased in atherosclerotic lesions of the GD group. Furthermore, the lesions presented higher necrotic core and lower collagen content, characterizing the less stable plaques. CONCLUSION The gluten-containing high-fat diet was associated with a more severe proatherogenic profile than the gluten-free high-fat diet owing to increased inflammatory and oxidative status at atherosclerotic lesions in obese mice.
Collapse
Affiliation(s)
- Edenil Costa Aguilar
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil.
| | | | - Weslley Fernandes-Braga
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | | | - Alda Jusceline Leonel
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | | | | | - Virginia Soares Lemos
- Departamento de Fisiologia, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | |
Collapse
|
26
|
Bartoloni E, Alunno A, Cafaro G, Valentini V, Bistoni O, Bonifacio AF, Gerli R. Subclinical Atherosclerosis in Primary Sjögren's Syndrome: Does Inflammation Matter? Front Immunol 2019; 10:817. [PMID: 31110500 PMCID: PMC6499202 DOI: 10.3389/fimmu.2019.00817] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022] Open
Abstract
Sjögren's syndrome (SS) is a systemic autoimmune disease mainly characterized by inflammatory involvement of exocrine gland. Atherosclerosis is a complex process leading to plaque formation in arterial wall with subsequent cardiovascular (CV) events. Recently, numerous studies demonstrated that SS patients bear an increased CV risk. Since activation of immune system is a key element in atherosclerosis, it is interesting to analyze whether and how the autoimmune and inflammatory events characterizing SS pathogenesis directly or indirectly contribute to atherosclerosis risk in these patients. An increase in circulating endothelial microparticles and integrins, which may be a consequence of endothelial damage and impaired repair mechanisms, has been demonstrated in SS. Increased endothelial expression of adhesion molecules with subsequent infiltration of inflammatory cells into arterial wall is also a critical event in atherosclerosis. The early inflammatory events taking place in the atherosclerotic plaque cause an increase in alarmins, such as S100A8/A9, which seems to be associated with SS disease activity and, in turn, induce up-regulation of interleukin (IL)-1β and other pro-atherogenic cytokines. Interestingly, increased IL-1β levels were also detected in tertiary lymphoid structures developing in vessel adventitia adjacent to the atherosclerotic plaque, suggesting a direct role of IL-1β in this process. Similar to these structures, germinal center-like structures arising in SS exocrine glands are also tertiary lymphoid systems where T-helper (Th) cell subsets govern the adaptive immune response. Th1 cells are the most prevalent subtype and have been shown to be strongly involved in both SS pathogenesis and atherosclerosis. Th17 cells are attracting great interest and few studies showed its importance in SS development. Albeit in low amounts, a Th17 signature was also detected in atherosclerotic plaques and some animal models demonstrated a significant pro-atherogenic role and positive effects of IL-17A blockade. Despite the fact that T cells have a pivotal role in the inflammatory process that ultimately leads to atherosclerosis, B cells have also been detected in atherosclerotic plaques, although their exact role is still mostly unknown with studies showing contrasting results. In this scenario, the role of inflammation in atherosclerosis pathogenesis in patients with SS needs to be further explored.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | - Alessia Alunno
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | - Valentina Valentini
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | - Onelia Bistoni
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Roberto Gerli
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
27
|
Wang Z, Wu Y, Zhang S, Zhao Y, Yin X, Wang W, Ma X, Liu H. The role of NO-cGMP pathway inhibition in vascular endothelial-dependent smooth muscle relaxation disorder of AT1-AA positive rats: protective effects of adiponectin. Nitric Oxide 2019; 87:10-22. [PMID: 30831264 DOI: 10.1016/j.niox.2019.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/16/2019] [Accepted: 02/19/2019] [Indexed: 12/24/2022]
Abstract
Angiotensin II type 1 receptor autoantibodies (AT1-AA) cause endothelial-dependent smooth muscle relaxation disorder. It is well understood that impairment of the NO-cGMP signaling pathway is one of the mechanisms of endothelial-dependent smooth muscle relaxation disorder. However, it is still unclear whether AT1-AA induces endothelial-dependent smooth muscle relaxation disorder via the impairment of the NO-cGMP signaling pathway. In addition, adiponectin exerts vascular endothelial protection through the NO-cGMP signaling pathway. Therefore, the purpose of this investigation was to assess the mechanism of vascular endothelial-dependent smooth muscle relaxation disorder induced by AT1-AA and the role of adiponectin in attenuating this dysregulation. Serum endothelin-1 (ET-1), adiponectin and AT1-AA were detected by enzyme-linked immunosorbent assay. In preeclamptic patients, there was an increased level of AT1-AA, which was positively correlated with ET-1 and negatively correlated with adiponectin, as elevated levels of ET-1 suggested endothelial injury. AT1-AA-positive animal models were actively immunized with the second extracellular loop of the angiotensin II type 1 receptor (AT1R-ECII) for eight weeks. In thoracic aortas of AT1-AA positive rats, ET-1 was elevated, endothelium-dependent vasodilation was decreased. Paradoxically, as the upstream element of the NO-cGMP signaling pathway, NO production was not decreased but increased, and the ratio of p-VASP/VASP, an established biochemical endpoint of NO-cGMP signaling pathway, was reduced. Moreover, the levels of nitrotyrosine and gp91phox which indicate the presence of peroxynitrite (ONOO-) and superoxide anion (O2·-) were increased. Pretreatment with the ONOO- scavenger FeTMPyP or O2·-scavenger Tempol normalized vasorelaxation. Key enzymes responsible for NO synthesis were also assessed. iNOS protein expression was increased, but p-eNOS(Ser1177)/eNOS was reduced. Preincubation with the iNOS inhibitor 1400 W or eNOS agonist nebivolol restored vasorelaxation. Further experiments showed levels of p-AMPKα (Thr172)/AMPKα, which controls iNOS expression and eNOS activity, to have been reduced. Furthermore, levels of the upstream component of AMPK, adiponectin, was reduced in sera of AT1-AA positive rats and supplementation of adiponectin significantly decreased ET-1 contents, improved endothelial-dependent vasodilation, reduced NO production, elevated p-VASP/VASP, inhibited protein expression of nitrotyrosine and gp91phox, reduced iNOS overexpression, and increased eNOS phosphorylation at Ser1177 in the thoracic aorta of AT1-AA positive rats. These results established that impairment NO-cGMP pathway may aggravate the endothelial-dependent smooth muscle relaxation disorder in AT1-AA positive rats and adiponectin improved endothelial-dependent smooth muscle relaxation disorder by enhancing NO-cGMP pathway. This discovery may shed a novel light on clinical treatment of vascular diseases associated with AT1-AA.
Collapse
Affiliation(s)
- Zhiyuan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Yuhui Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Xiaochen Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China
| | - Xinliang Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China; Department of Emergency Medicine, Thomas Jefferson University, Philadephia, Pennsylvania, USA.
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
28
|
Cataldo N, Musetti B, Celano L, Carabio C, Cassina A, Cerecetto H, González M, Thomson L. Inhibition of LDL oxidation and inflammasome assembly by nitroaliphatic derivatives. Potential use as anti-inflammatory and anti-atherogenic agents. Eur J Med Chem 2018; 159:178-186. [PMID: 30292895 DOI: 10.1016/j.ejmech.2018.09.062] [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] [Received: 07/04/2018] [Revised: 09/04/2018] [Accepted: 09/25/2018] [Indexed: 10/28/2022]
Abstract
We have previously shown the antioxidant and anti-inflammatory properties of several para-substituted arylnitroalkenes. Since oxidative stress and inflammation are key processes that drive the initiation and progression of atherosclerosis, in the present work the antioxidant, anti-inflammatory and anti-atherogenic properties of an extended library of aryl-nitroaliphatic derivatives, including several newly designed nitroalkanes, was explored. The antioxidant capacity of the nitroaliphatic compounds, measured using the oxygen radical absorbance capacity assay (ORAC) showed that the p-methylthiophenyl-derivatives were about three times more effective than Trolox to prevent fluorescein oxidation, independently of the presence or the absence of the double bond next to the nitro group. The peroxyl radical scavenger capacity of the p-dimethylaminophenyl-derivatives was even higher, being the reduced form of these compounds even more active. In fact, while the antioxidant capacity of 1-dimethylamino-4-(2-nitro-1Z-ethenyl)benzene and 1-dimethylamino-4-(2-nitro-1Z-propenyl)benzene was 4.2 ± 0.1 and 5.4 ± 0.1 Trolox Eq/mol, respectively; ORAC values obtained with the ethyl and the propyl derivatives were 10 ± 1 and 13 ± 2 Trolox Eq/mol, respectively. The p-dimethylamino-derivatives, especially the nitroalkanes, were also able to prevent LDL oxidation mediated by peroxyl radicals. Oxygen consumption due to the oxidation of fatty acids was delayed in the presence of the dimethylamino substituted compounds, only the alkanes interrupted the chain of lipid oxidations decreasing the rate of oxygen consumption. Although the formation of foam cells in the presence of oxidized-LDL (oxLDL) remained unaffected, the molecules containing the dimethylamino moiety were able to decrease the expression of IL-1β in LPS/INF-γ challenged macrophages.
Collapse
Affiliation(s)
- Nicolás Cataldo
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay; Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Bruno Musetti
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay; Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Laura Celano
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Claudio Carabio
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Adriana Cassina
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, General Flores 2125, Montevideo, 11800, Uruguay; Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, General Flores 2125, Montevideo, 11800, Uruguay
| | - Hugo Cerecetto
- Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Mercedes González
- Grupo de Química Medicinal, Laboratorio de Química Orgánica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay
| | - Leonor Thomson
- Laboratorio de Enzimología, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Iguá 4225, Montevideo, 11400, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, General Flores 2125, Montevideo, 11800, Uruguay.
| |
Collapse
|
29
|
The histone deacetylase inhibitor SAHA induces HSP60 nitration and its extracellular release by exosomal vesicles in human lung-derived carcinoma cells. Oncotarget 2018; 7:28849-67. [PMID: 26700624 PMCID: PMC5045361 DOI: 10.18632/oncotarget.6680] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/22/2015] [Indexed: 12/14/2022] Open
Abstract
HSP60 undergoes changes in quantity and distribution in some types of tumors suggesting a participation of the chaperonin in the mechanism of transformation and cancer progression. Suberoylanilide hydroxamic acid (SAHA), a member of a family of histone deacetylase inhibitors (HDACi), has anti-cancer potential but its interaction, if any, with HSP60 has not been elucidated. We investigated the effects of SAHA in a human lung-derived carcinoma cell line (H292). We analysed cell viability and cycle; oxidative stress markers; mitochondrial integrity; HSP60 protein and mRNA levels; and HSP60 post-translational modifications, and its secretion. We found that SAHA is cytotoxic for H292 cells, interrupting the cycle at the G2/M phase, which is followed by death; cytotoxicity is associated with oxidative stress, mitochondrial damage, and diminution of intracellular levels of HSP60; HSP60 undergoes a post-translational modification and becomes nitrated; and nitrated HSP60 is exported via exosomes. We propose that SAHA causes ROS overproduction and mitochondrial dysfunction, which leads to HSP60 nitration and release into the intercellular space and circulation to interact with the immune system. These successive steps might constitute the mechanism of the anti-tumor action of SAHA and provide a basis to design supplementary therapeutic strategies targeting HSP60, which would be more efficacious than the compound alone.
Collapse
|
30
|
Matczak M, Marchelak A, Michel P, Owczarek A, Piszczan A, Kolodziejczyk-Czepas J, Nowak P, Olszewska MA. Sorbus domestica L. leaf extracts as functional products: phytochemical profiling, cellular safety, pro-inflammatory enzymes inhibition and protective effects against oxidative stress in vitro. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.10.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
31
|
Marchelak A, Owczarek A, Matczak M, Pawlak A, Kolodziejczyk-Czepas J, Nowak P, Olszewska MA. Bioactivity Potential of Prunus spinosa L. Flower Extracts: Phytochemical Profiling, Cellular Safety, Pro-inflammatory Enzymes Inhibition and Protective Effects Against Oxidative Stress In Vitro. Front Pharmacol 2017; 8:680. [PMID: 29085295 PMCID: PMC5649189 DOI: 10.3389/fphar.2017.00680] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/11/2017] [Indexed: 01/06/2023] Open
Abstract
Flower extracts of Prunus spinosa L. (blackthorn)-a traditional medicinal plant of Central and Eastern Europe indicated for the treatment of urinary tract disorders, inflammation, and adjunctive therapy of cardiovascular diseases-were evaluated in terms of chemical composition, antioxidant activity, potential anti-inflammatory effects, and cellular safety in function of fractionated extraction. The UHPLC-PDA-ESI-MS3 fingerprinting led to full or partial identification of 57 marker constituents (36 new for the flowers), mostly flavonoids, A-type proanthocyanidins, and phenolic acids, and provided the basis for authentication and standardization of the flower extracts. With the contents up to 584.07 mg/g dry weight (dw), 490.63, 109.43, and 66.77 mg/g dw of total phenolics (TPC), flavonoids, proanthocyanidins, and phenolic acids, respectively, the extracts were proven to be rich sources of polyphenols. In chemical in vitro tests of antioxidant (DPPH, FRAP, TBARS) and enzyme (lipoxygenase and hyaluronidase) inhibitory activity, the extracts effects were profound, dose-, phenolic-, and extraction solvent-dependent. Moreover, at in vivo-relevant levels (1-5 μg/mL) the extracts effectively protected the human plasma components against peroxynitrite-induced damage (reduced the levels of oxidative stress biomarkers: 3-nitrotyrosine, lipid hydroperoxides, and thiobarbituric acid-reactive substances) and enhanced the total antioxidant status of plasma. The effects observed in biological models were in general dose- and TPC-dependent; only for protein nitration the relationships were not significant. Furthermore, in cytotoxicity tests, the extracts did not affect the viability of human peripheral blood mononuclear cells (PBMC), and might be regarded as safe. Among extracts, the defatted methanol-water (7:3, v/v) extract and its diethyl ether and ethyl acetate fractions appear to be the most advantageous for biological applications. As compared to the positive controls, activity of the extracts was favorable, which might be attributed to some synergic effects of their constituents. In conclusion, this research proves that the antioxidant and enzyme inhibitory capacity of phenolic fractions should be counted as one of the mechanisms behind the activity of the flowers reported by traditional medicine and demonstrates the potential of the extracts as alternative ingredients for functional products supporting the treatment of oxidative stress-related pathologies cross-linked with inflammatory changes, especially in cardiovascular protection.
Collapse
Affiliation(s)
- Anna Marchelak
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| | - Aleksandra Owczarek
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| | - Magdalena Matczak
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| | - Adam Pawlak
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| | - Joanna Kolodziejczyk-Czepas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Pawel Nowak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Monika A Olszewska
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
32
|
Nox2 contributes to hyperinsulinemia-induced redox imbalance and impaired vascular function. Redox Biol 2017; 13:288-300. [PMID: 28600985 PMCID: PMC5466665 DOI: 10.1016/j.redox.2017.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/17/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance promotes vascular endothelial dysfunction and subsequent development of cardiovascular disease. Previously we found that skeletal muscle arteriolar flow-induced dilation (FID) was reduced following a hyperinsulinemic clamp in healthy adults. Therefore, we hypothesized that hyperinsulinemia, a hallmark of insulin resistance, contributes to microvascular endothelial cell dysfunction via inducing oxidative stress that is mediated by NADPH oxidase (Nox) system. We examined the effect of insulin, at levels that are comparable with human hyperinsulinemia on 1) FID of isolated arterioles from human skeletal muscle tissue in the presence and absence of Nox inhibitors and 2) human adipose microvascular endothelial cell (HAMECs) expression of nitric oxide (NO), endothelial NO synthase (eNOS), and Nox-mediated oxidative stress. In six lean healthy participants (mean age 25.5±1.6 y, BMI 21.8±0.9), reactive oxygen species (ROS) were increased while NO and arteriolar FID were reduced following 60 min of ex vivo insulin incubation. These changes were reversed after co-incubation with the Nox isoform 2 (Nox2) inhibitor, VAS2870. In HAMECs, insulin-induced time-dependent increases in Nox2 expression and P47phox phosphorylation were echoed by elevations of superoxide production. In contrast, phosphorylation of eNOS and expression of superoxide dismutase (SOD2 and SOD3) isoforms showed a biphasic response with an increased expression at earlier time points followed by a steep reduction phase. Insulin induced eNOS uncoupling that was synchronized with a drop of NO and a surge of ROS production. These effects were reversed by Tempol (SOD mimetic), Tetrahydrobiopterin (BH4; eNOS cofactor), and VAS2870. Finally, insulin induced nitrotyrosine formation which was reversed by inhibiting NO or superoxide generation. In conclusions, hyperinsulinemia may reduce FID via inducing Nox2-mediated superoxide production in microvascular endothelial cells which reduce the availability of NO and enhances peroxynitrite formation. Therefore, the Nox2 pathway should be considered as a target for the prevention of oxidative stress-associated endothelial dysfunction during hyperinsulinemia. Hyperinsulinemia impairs FID and induces ROS production in human muscle arterioles. Insulin-induced ROS production in endotelial cells is mediated by NADPH oxidase. Long exposure to high insulin levels reduces eNOS phosphorylation and NO production.
Collapse
|
33
|
Bartesaghi S, Herrera D, Martinez DM, Petruk A, Demicheli V, Trujillo M, Martí MA, Estrín DA, Radi R. Tyrosine oxidation and nitration in transmembrane peptides is connected to lipid peroxidation. Arch Biochem Biophys 2017; 622:9-25. [PMID: 28412156 DOI: 10.1016/j.abb.2017.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 12/30/2022]
Abstract
Tyrosine nitration is an oxidative post-translational modification that can occur in proteins associated to hydrophobic bio-structures such as membranes and lipoproteins. In this work, we have studied tyrosine nitration in membranes using a model system consisting of phosphatidylcholine liposomes with pre-incorporated tyrosine-containing 23 amino acid transmembrane peptides. Tyrosine residues were located at positions 4, 8 or 12 of the amino terminal, resulting in different depths in the bilayer. Tyrosine nitration was accomplished by exposure to peroxynitrite and a peroxyl radical donor or hemin in the presence of nitrite. In egg yolk phosphatidylcholine liposomes, nitration was highest for the peptide with tyrosine at position 8 and dramatically increased as a function of oxygen levels. Molecular dynamics studies support that the proximity of the tyrosine phenolic ring to the linoleic acid peroxyl radicals contributes to the efficiency of tyrosine oxidation. In turn, α-tocopherol inhibited both lipid peroxidation and tyrosine nitration. The mechanism of tyrosine nitration involves a "connecting reaction" by which lipid peroxyl radicals oxidize tyrosine to tyrosyl radical and was fully recapitulated by computer-assisted kinetic simulations. Altogether, this work underscores unique characteristics of the tyrosine oxidation and nitration process in lipid-rich milieu that is fueled via the lipid peroxidation process.
Collapse
Affiliation(s)
- Silvina Bartesaghi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Departamento de Educación Médica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay.
| | - Daniel Herrera
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Débora M Martinez
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Ariel Petruk
- Departamento de Química Inorgánica, Analítica y Química-Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cuidad Universitaria, Pab 2, C1428EHA, Buenos Aires, Argentina
| | - Verónica Demicheli
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay
| | - Marcelo A Martí
- Departamento de Química Biológica and IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cuidad Universitaria, Pab 2, C1428EHA, Buenos Aires, Argentina
| | - Darío A Estrín
- Departamento de Química Inorgánica, Analítica y Química-Física and INQUIMAE-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cuidad Universitaria, Pab 2, C1428EHA, Buenos Aires, Argentina
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay; Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Avda. Gral. Flores 2125, Montevideo 11800, Uruguay.
| |
Collapse
|
34
|
Merz T, Stenzel T, Nußbaum B, Wepler M, Szabo C, Wang R, Radermacher P, McCook O. Cardiovascular disease and resuscitated septic shock lead to the downregulation of the H 2S-producing enzyme cystathionine-γ-lyase in the porcine coronary artery. Intensive Care Med Exp 2017; 5:17. [PMID: 28321823 PMCID: PMC5359268 DOI: 10.1186/s40635-017-0131-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/13/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Downregulation of the hydrogen sulfide (H2S)-producing enzymes cystathionine-γ-lyase (CSE), cystathionine-β-synthase (CBS), and/or 3-mercaptopyruvate sulfurtransferase (3-MST) is associated with chronic cardiovascular pathologies. Nevertheless, equivocal data are available on both the expression and function of these enzymes in coronary arteries (CA). We recently reported that atherosclerotic pigs subjected to sepsis developed impaired cardiac function, which coincided with decreased myocardial CSE expression and increased nitrotyrosine formation. To define the endogenous source(s) of H2S in the CA, we studied the expression of CBS, CSE, or 3-MST in the CA of pigs subjected to septic shock with/without pre-existing cardiovascular co-morbidity. METHODS Anesthetized and instrumented FBM "familial hypercholesterolemia Bretoncelles Meishan" pigs with high-fat diet-induced hypercholesterolemia and atherosclerosis were subjected to polymicrobial septic shock, or sham procedure, and subsequent intensive care therapy for 24 h. Young German domestic pigs were used as naïve controls. CSE, CBS, 3-MST, HO-1, eNOS, and nitrotyrosine expression was quantified by immunohistochemistry of formalin-fixed paraffin sections. RESULTS FBM pigs, in the absence of septic shock, showed decreased CSE expression in the media. This decrease became more pronounced after sepsis. The expression pattern of HO-1 resembled the pattern of CSE expression. CBS protein was not detected in the media of any of the CA examined but was localized to the adventitia and only in the atheromatous plaques containing foam cells of the CA, in regions that also displayed abundant nitrotyrosine formation. The CBS expression in the adventitia was not associated with nitrotyrosine formation. 3-MST expression was not found in any of the CA samples. CONCLUSIONS We hypothesize that (i) the reduced CSE expression in FBM pigs may contribute to their cardiovascular disease phenotype and moreover (ii) the further decrease in CA CSE expression in sepsis may contribute to the sepsis-associated cardiac dysfunction.
Collapse
Affiliation(s)
- Tamara Merz
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany
| | - Tatjana Stenzel
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany
| | - Benedikt Nußbaum
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany.,Universitätsklinik Ulm, Klinik für Anästhesiologie, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Martin Wepler
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany.,Universitätsklinik Ulm, Klinik für Anästhesiologie, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch at Galveston, 601 Harborside Drive, Galveston, TX, 77555, USA
| | - Rui Wang
- Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Peter Radermacher
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany
| | - Oscar McCook
- Universitätsklinik Ulm, Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Helmholtzstrasse 8, 89081, Ulm, Germany. .,Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.
| |
Collapse
|
35
|
Cruz DF, Fardilha M. Relevance of peroxynitrite formation and 3-nitrotyrosine on spermatozoa physiology. Porto Biomed J 2016; 1:129-135. [PMID: 32258563 PMCID: PMC6806996 DOI: 10.1016/j.pbj.2016.07.004] [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: 05/09/2016] [Accepted: 07/18/2016] [Indexed: 01/08/2023] Open
Abstract
HIGHLIGHTS Male fertility decline has been attributed, in part, to increased oxidative stress.Here we will focus on spermatozoa ROS, namely O2•-, NO and ONOO- and their contribution to protein tyrosine nitration, namely by 3-NT formation.An in depth review will be made on the methods used to detect protein oxidation.Detecting 3-NT in sperm proteins will have a crucial clinical impact, namely on the follow up of anti-oxidant therapies. ABSTRACT Infertility is a clinical condition that affects around 15% of reproductive-aged couples worldwide. Around half of these cases are due to male factors, the most owing to idiopathic causes. The increase of reactive oxygen species (ROS), which leads to oxidative stress (OS), has been discussed in the last years as a possible cause of male idiopathic infertility. Superoxide anion (O2 •-) and nitric oxide (NO) can react with each other contributing to the formation of peroxynitrite (ONOO-). This molecule can then act on spermatozoa proteins, leading to nitration of protein tyrosines - addition of a nitro (NO2) group - that is then manifested by the formation of 3-nitrotyrosine (3-NT). In turn, 3-NT may be responsible for the alteration or inactivation of the protein function.This review will focus on the description of spermatozoa ROS, namely O2 •-, NO and ONOO- and in their contribution to protein tyrosine nitration, namely by 3-NT formation. Previous results about the effect of ONOO- and 3-NT in spermatozoa will be presented, as well as, the methods that can be performed to detect the protein oxidation by these species. The impact of measuring, at the clinical level, 3-NT, considered a marker of OS, in spermatozoa will be discussed.
Collapse
Affiliation(s)
- Daniel Filipe Cruz
- Signal Transduction Laboratory, Institute for Research in Biomedicine - iBiMED, Health Sciences Program, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Margarida Fardilha
- Signal Transduction Laboratory, Institute for Research in Biomedicine - iBiMED, Health Sciences Program, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
36
|
Nußbaum BL, McCook O, Hartmann C, Matallo J, Wepler M, Antonucci E, Kalbitz M, Huber-Lang M, Georgieff M, Calzia E, Radermacher P, Hafner S. Left ventricular function during porcine-resuscitated septic shock with pre-existing atherosclerosis. Intensive Care Med Exp 2016; 4:14. [PMID: 27271248 PMCID: PMC4894859 DOI: 10.1186/s40635-016-0089-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/31/2016] [Indexed: 02/07/2023] Open
Abstract
Background Reversible, depressed cardiac function is frequently encountered during septic shock and commonly called septic cardiomyopathy. Previous studies demonstrated reduced ejection fraction and left ventricular dilatation in both humans and animal models. However, the majority of the studies in humans excluded pre-existing cardiac disease and animal studies were performed on healthy specimen and/or without vasopressor support during sepsis. In order to more closely mimic the actual patients’ conditions on intensive care units and to assess the influence of both cardiac comorbidity and vasopressor support on septic cardiomyopathy, we evaluated the left ventricular function in a porcine model of resuscitated septic shock with pre-existing atherosclerosis. Methods Hypercholesterolaemic, atherosclerotic pigs due to homozygous low-density lipoprotein receptor mutation and high-fat diet were anaesthetised and surgically instrumented. Faecal peritonitis was induced by inoculation of autologous faeces into the peritoneal cavity in n = 8 animals; n = 5 pigs underwent sham procedure. Sepsis resuscitation included administration of fluids and noradrenaline. Left ventricular function was analysed via pressure-conductance catheters before, 12 and 24 h after the induction of sepsis. Results The main findings were impaired ventricular dilatation (no significant change in the left ventricular end-diastolic volume) and unchanged ejection fraction in septic pigs with pre-existing atherosclerosis. The relaxation time constant τ decreased while dp/dtmax increased. Cardiac nitrotyrosine formation increased while expression of the endogenous hydrogen sulphide (H2S)-producing enzyme cystathionine γ-lyase (CSE) decreased. Conclusions The data of the present study are in conflict with previously published data from healthy animal models, most likely as a result of ongoing resuscitation including noradrenaline treatment or intrinsic pathophysiologic processes of the pre-existing atherosclerosis. Moreover, increased nitrotyrosine formation and decreased expression of CSE suggest the implication of augmented oxidative/nitrosative stress and/or reduced bioavailability of nitric oxide as well as diminished endogenous H2S release in the pathophysiology of septic cardiomyopathy. Electronic supplementary material The online version of this article (doi:10.1186/s40635-016-0089-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Benedikt L Nußbaum
- Klinik für Anästhesiologie, Universitätsklinik Ulm, Ulm, Germany. .,Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany.
| | - Oscar McCook
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Clair Hartmann
- Klinik für Anästhesiologie, Universitätsklinik Ulm, Ulm, Germany.,Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - José Matallo
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Martin Wepler
- Klinik für Anästhesiologie, Universitätsklinik Ulm, Ulm, Germany.,Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Elena Antonucci
- Department of Surgical Sciences and Integrated Diagnostics, IRCCS San Martino IST, University of Genova, Genova, Italy
| | - Miriam Kalbitz
- Klinik für Unfall-, Hand-, Plastische- und Wiederherstellungschirurgie, Universitätsklinik Ulm, Ulm, Germany
| | - Markus Huber-Lang
- Klinik für Unfall-, Hand-, Plastische- und Wiederherstellungschirurgie, Universitätsklinik Ulm, Ulm, Germany
| | | | - Enrico Calzia
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Sebastian Hafner
- Klinik für Anästhesiologie, Universitätsklinik Ulm, Ulm, Germany.,Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinik Ulm, Helmholtzstraße 8/1, 89081, Ulm, Germany
| |
Collapse
|
37
|
Gong D, Cheng HP, Xie W, Zhang M, Liu D, Lan G, Huang C, Zhao ZW, Chen LY, Yao F, Tan YL, Li L, Xia XD, Zheng XL, Wang ZB, Tang CK. Cystathionine γ-lyase(CSE)/hydrogen sulfide system is regulated by miR-216a and influences cholesterol efflux in macrophages via the PI3K/AKT/ABCA1 pathway. Biochem Biophys Res Commun 2016; 470:107-116. [PMID: 26772887 DOI: 10.1016/j.bbrc.2016.01.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 01/01/2016] [Indexed: 12/22/2022]
Abstract
This study was designed to evaluate whether CSE/H2S system, which is regulated by miR-216a, regulated ABCA1-mediated cholesterol efflux and cholesterol contents in THP-1 macrophages-derived foam cells. Our qPCR and western blotting results showed that CSE/H2S significantly up-regulated the expression of ATP-binding cassette transporter A1 (ABCA1) mRNA and protein via PI3K/AKT pathway in foam cells derived from human THP-1 macrophages. The miR-216a directly targeted 3' untranslated region of CSE. It significantly reduced CSE and ABCA1 expression, and also decreased the phosphorylation of PI3K and AKT. Additionally, cholesterol efflux decreased, and cholesterol levels increased in THP-1 macrophage-derived foam cells in response to treatment with miR-216a. Our study demonstrates that CSE/H2S system is regulated by miR-216a, and regulates ABCA1-mediated cholesterol efflux and cholesterol levels through the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Duo Gong
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Hai-Peng Cheng
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Wei Xie
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Min Zhang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Dan Liu
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Gang Lan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Chong Huang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Ling-Yan Chen
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Feng Yao
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Yu-Lin Tan
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Liang Li
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Xiao-Dan Xia
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary, Alberta, T2N 4N1, Canada
| | - Zong-Bao Wang
- School of Pharmacy and Life Science College, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, 421001, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Medical Research Center, Hunan Province Cooperative Innovation Center for Molecular Target NewDrug Study, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|