1
|
Medina Y, Fernandez N, Sierra MN, Castro Parodi M, Damiano AE. Nitrative stress-induced dysregulation of placental AQUAPORIN-9: A potential key player in preeclampsia pathogenesis. Placenta 2025:S0143-4004(25)00034-7. [PMID: 39955250 DOI: 10.1016/j.placenta.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/16/2025] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
Preeclampsia is associated with increased oxidative and nitrative stress, resulting in elevated protein nitration and potential functional impairment. Previously, we found an increased expression of AQP9 protein with a loss of function in preeclamptic placentas. However, the link between nitrative stress and AQP9 has not yet been explored. Here, we aimed to evaluate the effect of nitrative stress on placental AQP9 and its role in the pathogenesis of preeclampsia. In silico analysis was conducted on the amino acid sequences of AQP9 to identify potential nitration sites. Levels of 3NyT-AQP9 were assessed by immunoprecipitation in normal and preeclamptic placentas. AQP9 expression and function were evaluated by culturing normal placental explants with 0, 25, 50, 100, and 200 μM ONOO- to induce nitrative stress. Viability and integrity of the explants and stress markers were determined. Water uptake and utilization of lactate mediated by AQP9 were studied along with the molecular expression of AQP9 and 3-NyT-AQP9. The in silico analysis showed that AQP9 is more susceptible to nitration than other AQPs. The abundance of nitrated AQP9 significantly increased in preeclamptic placentas compared to normal ones (n = 4; p < 0.05). Peroxynitrite treatment also increased AQP9 protein expression without altering its gene expression and impaired the transport of water and lactate mediated by this protein. Our findings provide evidence that nitrative stress induces the nitration of AQP9 protein, leading to the accumulation of a non-functional protein in the syncytiotrophoblasts. Therefore, this altered protein may play a pivotal role in the pathogenesis of preeclampsia by disrupting cellular homeostasis.
Collapse
Affiliation(s)
- Yollyseth Medina
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Laboratorio de Biología de la Reproducción, Buenos Aires, Argentina
| | - Nazarena Fernandez
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Laboratorio de Biología de la Reproducción, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina
| | - Matías N Sierra
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Laboratorio de Biología de la Reproducción, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina
| | - Mauricio Castro Parodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina
| | - Alicia E Damiano
- Universidad de Buenos Aires, CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay), Laboratorio de Biología de la Reproducción, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Kumari R, Kapoor P, Mir BA, Singh M, Parrey ZA, Rakhra G, Parihar P, Khan MN, Rakhra G. Unlocking the versatility of nitric oxide in plants and insights into its molecular interplays under biotic and abiotic stress. Nitric Oxide 2024; 150:1-17. [PMID: 38972538 DOI: 10.1016/j.niox.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
In plants, nitric oxide (NO) has become a versatile signaling molecule essential for mediating a wide range of physiological processes under various biotic and abiotic stress conditions. The fundamental function of NO under various stress scenarios has led to a paradigm shift in which NO is now seen as both a free radical liberated from the toxic product of oxidative metabolism and an agent that aids in plant sustenance. Numerous studies on NO biology have shown that NO is an important signal for germination, leaf senescence, photosynthesis, plant growth, pollen growth, and other processes. It is implicated in defense responses against pathogensas well as adaptation of plants in response to environmental cues like salinity, drought, and temperature extremes which demonstrates its multifaceted role. NO can carry out its biological action in a variety of ways, including interaction with protein kinases, modifying gene expression, and releasing secondary messengers. In addition to these signaling events, NO may also be in charge of the chromatin modifications, nitration, and S-nitrosylation-induced posttranslational modifications (PTM) of target proteins. Deciphering the molecular mechanism behind its essential function is essential to unravel the regulatory networks controlling the responses of plants to various environmental stimuli. Taking into consideration the versatile role of NO, an effort has been made to interpret its mode of action based on the post-translational modifications and to cover shreds of evidence for increased growth parameters along with an altered gene expression.
Collapse
Affiliation(s)
- Ritu Kumari
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Preedhi Kapoor
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Bilal Ahmad Mir
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Maninder Singh
- Department of Biotechnology and Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Zubair Ahmad Parrey
- Plant Physiology and Biochemistry Section, Department of Botany, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Gurseen Rakhra
- Department of Nutrition & Dietetics, Faculty of Allied Health Sciences, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, 121004, India
| | - Parul Parihar
- Department of Biosciences and Biotechnology, Banasthali Vidyapith, Rajasthan, 304022, India
| | - M Nasir Khan
- Renewable Energy and Environmental Technology Center, University of Tabuk, Tabuk, 47913, Saudi Arabia
| | - Gurmeen Rakhra
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
3
|
Jia J, Lv P, Wei X, Qiu W. SNO-DCA: A model for predicting S-nitrosylation sites based on densely connected convolutional networks and attention mechanism. Heliyon 2024; 10:e23187. [PMID: 38148797 PMCID: PMC10750070 DOI: 10.1016/j.heliyon.2023.e23187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Abstract
Protein S-nitrosylation is a reversible oxidative reduction post-translational modification that is widely present in the biological community. S-nitrosylation can regulate protein function and is closely associated with a variety of diseases, thus identifying S-nitrosylation sites are crucial for revealing the function of proteins and related drug discovery. Traditional experimental methods are time-consuming and expensive; therefore, it is necessary to explore more efficient computational methods. Deep learning algorithms perform well in the field of bioinformatics sites prediction, and many studies show that they outperform existing machine learning algorithms. In this work, we proposed a deep learning algorithm-based predictor SNO-DCA for distinguishing between S-nitrosylated and non-S-nitrosylated sequences. First, one-hot encoding of protein sequences was performed. Second, the dense convolutional blocks were used to capture feature information, and an attention module was added to weigh different features to improve the prediction ability of the model. The 10-fold cross-validation and independent testing experimental results show that our SNO-DCA model outperforms existing S-nitrosylation sites prediction models under imbalanced data. In this paper, a web server prediction website: https://sno.cangmang.xyz/SNO-DCA/was established to provide an online prediction service for users. SNO-DCA can be available at https://github.com/peanono/SNO-DCA.
Collapse
Affiliation(s)
- Jianhua Jia
- Computer Department, Jingdezhen Ceramic University, Jingdezhen, 330403, China
| | - Peinuo Lv
- Computer Department, Jingdezhen Ceramic University, Jingdezhen, 330403, China
| | - Xin Wei
- Business School, Jiangxi Institute of Fashion Technology, Nanchang, 330201, China
| | - Wangren Qiu
- Computer Department, Jingdezhen Ceramic University, Jingdezhen, 330403, China
| |
Collapse
|
4
|
Omidkhah N, Hadizadeh F, Ghodsi R, Kesharwani P, Sahebkar A. In silico Evaluation of NO-Sartans against SARS-CoV-2. Curr Drug Discov Technol 2024; 21:e050324227669. [PMID: 38445698 DOI: 10.2174/0115701638279362240223070810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Numerous clinical trials are currently investigating the potential of nitric oxide (NO) as an antiviral agent against coronaviruses, including SARS-CoV-2. Additionally, some researchers have reported positive effects of certain Sartans against SARS-CoV-2. METHOD Considering the impact of NO-Sartans on the cardiovascular system, we have compiled information on the general structure, synthesis methods, and biological studies of synthesized NOSartans. In silico evaluation of all NO-Sartans and approved sartans against three key SARS-CoV- -2 targets, namely Mpro (PDB ID: 6LU7), NSP16 (PDB ID: 6WKQ), and ACE-2 (PDB ID: 1R4L), was performed using MOE. RESULTS Almost all NO-Sartans and approved sartans demonstrated promising results in inhibiting these SARS-CoV-2 targets. Compound 36 (CLC-1280) showed the best docking scores against the three evaluated targets and was further evaluated using molecular dynamics (MD) simulations. CONCLUSION Based on our in silico studies, CLC-1280 (a Valsartan dinitrate) has the potential to be considered as an inhibitor of the SARS-CoV-2 virus. However, further in vitro and in vivo evaluations are necessary for the drug development process.
Collapse
Affiliation(s)
- Negar Omidkhah
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, 110062, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Griswold-Prenner I, Kashyap AK, Mazhar S, Hall ZW, Fazelinia H, Ischiropoulos H. Unveiling the human nitroproteome: Protein tyrosine nitration in cell signaling and cancer. J Biol Chem 2023; 299:105038. [PMID: 37442231 PMCID: PMC10413360 DOI: 10.1016/j.jbc.2023.105038] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023] Open
Abstract
Covalent amino acid modification significantly expands protein functional capability in regulating biological processes. Tyrosine residues can undergo phosphorylation, sulfation, adenylation, halogenation, and nitration. These posttranslational modifications (PTMs) result from the actions of specific enzymes: tyrosine kinases, tyrosyl-protein sulfotransferase(s), adenylate transferase(s), oxidoreductases, peroxidases, and metal-heme containing proteins. Whereas phosphorylation, sulfation, and adenylation modify the hydroxyl group of tyrosine, tyrosine halogenation and nitration target the adjacent carbon residues. Because aberrant tyrosine nitration has been associated with human disorders and with animal models of disease, we have created an updated and curated database of 908 human nitrated proteins. We have also analyzed this new resource to provide insight into the role of tyrosine nitration in cancer biology, an area that has not previously been considered in detail. Unexpectedly, we have found that 879 of the 1971 known sites of tyrosine nitration are also sites of phosphorylation suggesting an extensive role for nitration in cell signaling. Overall, the review offers several forward-looking opportunities for future research and new perspectives for understanding the role of tyrosine nitration in cancer biology.
Collapse
Affiliation(s)
| | | | | | - Zach W Hall
- Nitrase Therapeutics, Brisbane, California, USA
| | - Hossein Fazelinia
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Münzel T, Daiber A. Vascular redox signaling, eNOS uncoupling and endothelial dysfunction in the setting of transportation noise exposure or chronic treatment with organic nitrates. Antioxid Redox Signal 2023; 38:1001-1021. [PMID: 36719770 PMCID: PMC10171967 DOI: 10.1089/ars.2023.0006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
SIGNIFICANCE Cardiovascular disease and drug-induced health side effects are frequently associated with - or even caused by - an imbalance between the concentrations of reactive oxygen and nitrogen species (RONS) and antioxidants respectively determining the metabolism of these harmful oxidants. RECENT ADVANCES According to the "kindling radical" hypothesis, initial formation of RONS may further trigger the additional activation of RONS formation under certain pathological conditions. The present review will specifically focus on a dysfunctional, uncoupled endothelial nitric oxide synthase (eNOS) caused by RONS in the setting of transportation noise exposure or chronic treatment with organic nitrates, especially nitroglycerin. We will further describe the various "redox switches" that are proposed to be involved in the uncoupling process of eNOS. CRITICAL ISSUES In particular, the oxidative depletion of tetrahydrobiopterin (BH4), and S-glutathionylation of the eNOS reductase domain will be highlighted as major pathways for eNOS uncoupling upon noise exposure or nitroglycerin treatment. In addition, oxidative disruption of the eNOS dimer, inhibitory phosphorylation of eNOS at threonine or tyrosine residues, redox-triggered accumulation of asymmetric dimethylarginine (ADMA) and L-arginine deficiency will be discussed as alternative mechanisms of eNOS uncoupling. FUTURE DIRECTIONS The clinical consequences of eNOS dysfunction due to uncoupling on cardiovascular disease will be summarized also providing a template for future clinical studies on endothelial dysfunction caused by pharmacological or environmental risk factors.
Collapse
Affiliation(s)
- Thomas Münzel
- University Medical Center of the Johannes Gutenberg University Mainz, 39068, Cardiology I, Mainz, Rheinland-Pfalz, Germany;
| | - Andreas Daiber
- University Medical Center of the Johannes Gutenberg University Mainz, 39068, Cardiology I, Mainz, Rheinland-Pfalz, Germany;
| |
Collapse
|
7
|
Lysenkov SP, Muzhenya DV, Tuguz AR, Urakova TU, Shumilov DS, Thakushinov IA. Participation of nitrogen oxide and its metabolites in the genesis of hyperimmune inflammation in COVID-19. CHINESE J PHYSIOL 2021; 64:167-176. [PMID: 34472447 DOI: 10.4103/cjp.cjp_38_21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Despite the success in the tactics of treating COVID-19, there are many unexplored issues related to the development and progression of the process in the lungs, brain, and other organs, as well as the role of individual elements, in particular, nitric oxide (NO), and in the pathogenesis of organ damage. Based on the analyzed literature data, we considered a possible pathophysiological mechanism of action of NO and its derivatives in COVID-19. It can be noted that hyperimmune systemic inflammation and "cytokine storm" are enhanced by the production of NO, products of its oxidation ("nitrosative stress"). It is noted in the work that as a result of the oxidation of NO, a large amount of the toxic compound peroxynitrite is formed, which is a powerful proinflammatory agent. Its presence significantly damages the endothelium of the vascular walls and also oxidizes lipids, hemoglobin, myoglobin, and cytochrome, binds SH-groups of proteins, and damages DNA in the target cells. This is confirmed by the picture of the vessels of the lungs on computed tomography and the data of biochemical studies. In case of peroxynitrite overproduction, inhibition of the synthesis of NO and its metabolic products seems to be justified. Another aspect considered in this work is the mechanism of damage by the virus to the central and peripheral nervous system, which remains poorly understood but may be important in understanding the consequences, as well as predicting brain functions in persons who have undergone COVID-19. According to the analyzed literature, it can be concluded that brain damage is possible due to the direct effect of the virus on the peripheral nerves and central structures, and indirectly through the effect on the endothelium of cerebral vessels. Disturbances in the central nervous regulation of immune responses may be associated with the insufficient function of the acetylcholine anti-inflammatory system. It is proposed to further study several approaches to influence various links of NO exchange, which are of interest for theoretical and practical medicine.
Collapse
Affiliation(s)
- Sergey Petrovich Lysenkov
- Department of Pathomorphology and Clinical Pathophysiology, Medical Institute, FSBEI HE "Maikop State Technological University", Maikop, Republic of Adygeya, Russia
| | - Dmitriy Vitalevich Muzhenya
- Department of Pathomorphology and Clinical Pathophysiology, Medical Institute, FSBEI HE "Maikop State Technological University", Maikop, Republic of Adygeya, Russia
| | - Aminat Ramazanovna Tuguz
- Immunogenetic Laboratory of the Research, Institute of Complex Problems, FSBEI HE "Adyghe State University", Maikop, Republic of Adygeya, Russia
| | - Tamara Ur'evna Urakova
- Department of Pathomorphology and Clinical Pathophysiology, Medical Institute, FSBEI HE "Maikop State Technological University", Maikop, Republic of Adygeya, Russia
| | - Dmitriy Sergeevich Shumilov
- Immunogenetic Laboratory of the Research, Institute of Complex Problems, FSBEI HE "Adyghe State University", Maikop, Republic of Adygeya, Russia
| | - Ibragim Askarbievich Thakushinov
- Department of Pathomorphology and Clinical Pathophysiology, Medical Institute, FSBEI HE "Maikop State Technological University", Maikop, Republic of Adygeya, Russia
| |
Collapse
|
8
|
Byrne NJ, Rajasekaran NS, Abel ED, Bugger H. Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy. Free Radic Biol Med 2021; 169:317-342. [PMID: 33910093 PMCID: PMC8285002 DOI: 10.1016/j.freeradbiomed.2021.03.046] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Even in the absence of coronary artery disease and hypertension, diabetes mellitus (DM) may increase the risk for heart failure development. This risk evolves from functional and structural alterations induced by diabetes in the heart, a cardiac entity termed diabetic cardiomyopathy (DbCM). Oxidative stress, defined as the imbalance of reactive oxygen species (ROS) has been increasingly proposed to contribute to the development of DbCM. There are several sources of ROS production including the mitochondria, NAD(P)H oxidase, xanthine oxidase, and uncoupled nitric oxide synthase. Overproduction of ROS in DbCM is thought to be counterbalanced by elevated antioxidant defense enzymes such as catalase and superoxide dismutase. Excess ROS in the cardiomyocyte results in further ROS production, mitochondrial DNA damage, lipid peroxidation, post-translational modifications of proteins and ultimately cell death and cardiac dysfunction. Furthermore, ROS modulates transcription factors responsible for expression of antioxidant enzymes. Lastly, evidence exists that several pharmacological agents may convey cardiovascular benefit by antioxidant mechanisms. As such, increasing our understanding of the pathways that lead to increased ROS production and impaired antioxidant defense may enable the development of therapeutic strategies against the progression of DbCM. Herein, we review the current knowledge about causes and consequences of ROS in DbCM, as well as the therapeutic potential and strategies of targeting oxidative stress in the diabetic heart.
Collapse
Affiliation(s)
- Nikole J Byrne
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Namakkal S Rajasekaran
- Cardiac Aging & Redox Signaling Laboratory, Molecular and Cellular Pathology, Department of Pathology, Birmingham, AL, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
9
|
Rentz T, Wanschel ACBA, de Carvalho Moi L, Lorza-Gil E, de Souza JC, Dos Santos RR, Oliveira HCF. The Anti-atherogenic Role of Exercise Is Associated With the Attenuation of Bone Marrow-Derived Macrophage Activation and Migration in Hypercholesterolemic Mice. Front Physiol 2020; 11:599379. [PMID: 33329050 PMCID: PMC7719785 DOI: 10.3389/fphys.2020.599379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
An early event in atherogenesis is the recruitment and infiltration of circulating monocytes and macrophage activation in the subendothelial space. Atherosclerosis subsequently progresses as a unresolved inflammatory disease, particularly in hypercholesterolemic conditions. Although physical exercise training has been a widely accepted strategy to inhibit atherosclerosis, its impact on arterial wall inflammation and macrophage phenotype and function has not yet been directly evaluated. Thus, the aim of this study was to investigate the effects of aerobic exercise training on the inflammatory state of atherosclerotic lesions with a focus on macrophages. Hypercholesterolemic LDL-receptor-deficient male mice were subjected to treadmill training for 8 weeks and fed a high-fat diet. Analyses included plasma lipoprotein and cytokine levels; aortic root staining for lipids (oil red O); macrophages (CD68, MCP1 and IL1β); oxidative (nitrotyrosine and, DHE) and endoplasmic reticulum (GADD) stress markers. Primary bone marrow-derived macrophages (BMDM) were assayed for migration activity, motility phenotype (Rac1 and F-actin) and inflammation-related gene expression. Plasma levels of HDL cholesterol were increased, while levels of proinflammatory cytokines (TNFa, IL1b, and IL6) were markedly reduced in the exercised mice. The exercised mice developed lower levels of lipid content and inflammation in atherosclerotic plaques. Additionally, lesions in the exercised mice had lower levels of oxidative and ER stress markers. BMDM isolated from the exercised mice showed a marked reduction in proinflammatory cytokine gene expression and migratory activity and a disrupted motility phenotype. More importantly, bone marrow from exercised mice transplanted into sedentary mice led to reduced atherosclerosis in the recipient sedentary mice, thus suggesting that epigenetic mechanisms are associated with exercise. Collectively, the presented data indicate that exercise training prevents atherosclerosis by inhibiting bone marrow-derived macrophage recruitment and activation.
Collapse
Affiliation(s)
- Thiago Rentz
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Amarylis C B A Wanschel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Leonardo de Carvalho Moi
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Estela Lorza-Gil
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Jane C de Souza
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Renata R Dos Santos
- Division of Radiotherapy, Faculty of Medical Sciences, Medical School Hospital, State University of Campinas, Campinas, Brazil
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| |
Collapse
|
10
|
Ziegler K, Kunert AT, Reinmuth-Selzle K, Leifke AL, Widera D, Weller MG, Schuppan D, Fröhlich-Nowoisky J, Lucas K, Pöschl U. Chemical modification of pro-inflammatory proteins by peroxynitrite increases activation of TLR4 and NF-κB: Implications for the health effects of air pollution and oxidative stress. Redox Biol 2020; 37:101581. [PMID: 32739154 PMCID: PMC7767743 DOI: 10.1016/j.redox.2020.101581] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 01/05/2023] Open
Abstract
Environmental pollutants like fine particulate matter can cause adverse health effects through oxidative stress and inflammation. Reactive oxygen and nitrogen species (ROS/RNS) such as peroxynitrite can chemically modify proteins, but the effects of such modifications on the immune system and human health are not well understood. In the course of inflammatory processes, the Toll-like receptor 4 (TLR4) can sense damage-associated molecular patterns (DAMPs). Here, we investigate how the TLR4 response and pro-inflammatory potential of the proteinous DAMPs α-Synuclein (α-Syn), heat shock protein 60 (HSP60), and high-mobility-group box 1 protein (HMGB1), which are relevant in neurodegenerative and cardiovascular diseases, changes upon chemical modification with peroxynitrite. For the peroxynitrite-modified proteins, we found a strongly enhanced activation of TLR4 and the pro-inflammatory transcription factor NF-κB in stable reporter cell lines as well as increased mRNA expression and secretion of the pro-inflammatory cytokines TNF-α, IL-1β, and IL-8 in human monocytes (THP-1). This enhanced activation of innate immunity via TLR4 is mediated by covalent chemical modifications of the studied DAMPs. Our results show that proteinous DAMPs modified by peroxynitrite more potently amplify inflammation via TLR4 activation than the native DAMPs, and provide first evidence that such modifications can directly enhance innate immune responses via a defined receptor. These findings suggest that environmental pollutants and related ROS/RNS may play a role in promoting acute and chronic inflammatory disorders by structurally modifying the body's own DAMPs. This may have important consequences for chronic neurodegenerative, cardiovascular or gastrointestinal diseases that are prevalent in modern societies, and calls for action, to improve air quality and climate in the Anthropocene.
Pollutants and oxidative stress can cause protein nitration and oligomerization. Peroxynitrite amplifies inflammatory potential of disease-related proteins in vitro. Chemical modification of damage-associated molecular patterns (DAMPs). Positive feedback of modified DAMPs via pattern recognition receptor (TLR4). Air pollution may promote inflammatory disorders in the Anthropocene.
Collapse
Affiliation(s)
- Kira Ziegler
- Max Planck Institute for Chemistry, Multiphase Chemistry Department, 55128, Mainz, Germany
| | - Anna T Kunert
- Max Planck Institute for Chemistry, Multiphase Chemistry Department, 55128, Mainz, Germany
| | | | - Anna Lena Leifke
- Max Planck Institute for Chemistry, Multiphase Chemistry Department, 55128, Mainz, Germany
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, RG6 6AP, Reading, UK
| | - Michael G Weller
- Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center of the Johannes Gutenberg University, 55131, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, MA, 02215, USA
| | | | - Kurt Lucas
- Max Planck Institute for Chemistry, Multiphase Chemistry Department, 55128, Mainz, Germany.
| | - Ulrich Pöschl
- Max Planck Institute for Chemistry, Multiphase Chemistry Department, 55128, Mainz, Germany.
| |
Collapse
|
11
|
Wang CH, Chang WT, Tsai MS, Huang CH, Chen WJ. Synergistic Effects of Moderate Therapeutic Hypothermia and Levosimendan on Cardiac Function and Survival After Asphyxia-Induced Cardiac Arrest in Rats. J Am Heart Assoc 2020; 9:e016139. [PMID: 32476598 PMCID: PMC7429058 DOI: 10.1161/jaha.120.016139] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background This study investigated whether levosimendan, an inotropic calcium sensitizer, when combined with moderate therapeutic hypothermia, may exert synergistic benefits on post–cardiac arrest myocardial dysfunction and improve outcomes. Methods and Results After 9.5‐minute asphyxia‐induced cardiac arrest and resuscitation, 48 rats were randomized equally into 4 groups following return of spontaneous circulation (ROSC), including normothermia, hypothermia, normothermia–levosimendan, and hypothermia–levosimendan groups. For the normothermia group, the target temperature was 37°C while for the hypothermia group, the target temperature was 32°C, both of which were to be maintained for 4 hours after ROSC. Levosimendan was administered after ROSC with a loading dose of 10 μg/kg and then infused at 0.1 μg/kg per min for 4 hours. In the hypothermia–levosimendan group, left ventricular systolic function and cardiac output increased significantly, whereas the heart rate and systemic vascular resistance decreased significantly compared with the normothermia group. Also, the concentrations of interleukin 1β at 4 hours post‐ROSC and the production of NO between 1 hour and 4 hours post‐ROSC were reduced significantly in the hypothermia–levosimendan group compared with the normothermia group. The 72‐hour post‐ROSC survival and neurological recovery were also significantly better in the hypothermia–levosimendan group compared with the normothermia group (survival, 100% versus 50%, χ2 test, P=0.006). Conclusions Compared with normothermia, only combined moderate therapeutic hypothermia and levosimendan treatment could consistently improve post–cardiac arrest myocardial dysfunction and decrease the release of pro‐inflammatory molecules, thereby improving survival and neurological outcomes. These findings suggest synergistic benefits between moderate therapeutic hypothermia and levosimendan.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Wei-Tien Chang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Min-Shan Tsai
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Chien-Hua Huang
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan
| | - Wen-Jone Chen
- Department of Emergency Medicine National Taiwan University Hospital Taipei Taiwan.,Department of Emergency Medicine College of Medicine National Taiwan University Taipei Taiwan.,Division of Cardiology Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| |
Collapse
|
12
|
Vujacic-Mirski K, Bruns K, Kalinovic S, Oelze M, Kröller-Schön S, Steven S, Mojovic M, Korac B, Münzel T, Daiber A. Development of an Analytical Assay for Electrochemical Detection and Quantification of Protein-Bound 3-Nitrotyrosine in Biological Samples and Comparison with Classical, Antibody-Based Methods. Antioxidants (Basel) 2020; 9:E388. [PMID: 32384768 PMCID: PMC7278855 DOI: 10.3390/antiox9050388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 01/12/2023] Open
Abstract
Reactive oxygen and nitrogen species (RONS) cause oxidative damage, which is associated with endothelial dysfunction and cardiovascular disease, but may also contribute to redox signaling. Therefore, their precise detection is important for the evaluation of disease mechanisms. Here, we compared three different methods for the detection of 3-nitrotyrosine (3-NT), a marker of nitro-oxidative stress, in biological samples. Nitrated proteins were generated by incubation with peroxynitrite or 3-morpholino sydnonimine (Sin-1) and subjected to total hydrolysis using pronase, a mixture of different proteases. The 3-NT was then separated by high performance liquid chromatography (HPLC) and quantified by electrochemical detection (ECD, CoulArray) and compared to classical methods, namely enzyme-linked immunosorbent assay (ELISA) and dot blot analysis using specific 3-NT antibodies. Calibration curves for authentic 3-NT (detection limit 10 nM) and a concentration-response pattern for 3-NT obtained from digested nitrated bovine serum albumin (BSA) were highly linear over a wide 3-NT concentration range. Also, ex vivo nitration of protein from heart, isolated mitochondria, and serum/plasma could be quantified using the HPLC/ECD method and was confirmed by LC-MS/MS. Of note, nitro-oxidative damage of mitochondria results in increased superoxide (O2•-) formation rates (measured by dihydroethidium-based HPLC assay), pointing to a self-amplification mechanism of oxidative stress. Based on our ex vivo data, the CoulArray quantification method for 3-NT seems to have some advantages regarding sensitivity and selectivity. Establishing a reliable automated HPLC assay for the routine quantification of 3-NT in biological samples of cell culture, of animal and human origin seems to be more sophisticated than expected.
Collapse
Affiliation(s)
- Ksenija Vujacic-Mirski
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (K.V.-M.); (S.K.); (M.O.); (S.K.-S.); (S.S.); (T.M.)
| | - Kai Bruns
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Sanela Kalinovic
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (K.V.-M.); (S.K.); (M.O.); (S.K.-S.); (S.S.); (T.M.)
| | - Matthias Oelze
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (K.V.-M.); (S.K.); (M.O.); (S.K.-S.); (S.S.); (T.M.)
| | - Swenja Kröller-Schön
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (K.V.-M.); (S.K.); (M.O.); (S.K.-S.); (S.S.); (T.M.)
| | - Sebastian Steven
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (K.V.-M.); (S.K.); (M.O.); (S.K.-S.); (S.S.); (T.M.)
| | - Milos Mojovic
- Faculty of Physical Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia;
| | - Bato Korac
- Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Thomas Münzel
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (K.V.-M.); (S.K.); (M.O.); (S.K.-S.); (S.S.); (T.M.)
- Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andreas Daiber
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (K.V.-M.); (S.K.); (M.O.); (S.K.-S.); (S.S.); (T.M.)
- Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
13
|
Brodbelt JS, Morrison LJ, Santos I. Ultraviolet Photodissociation Mass Spectrometry for Analysis of Biological Molecules. Chem Rev 2020; 120:3328-3380. [PMID: 31851501 PMCID: PMC7145764 DOI: 10.1021/acs.chemrev.9b00440] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of new ion-activation/dissociation methods continues to be one of the most active areas of mass spectrometry owing to the broad applications of tandem mass spectrometry in the identification and structural characterization of molecules. This Review will showcase the impact of ultraviolet photodissociation (UVPD) as a frontier strategy for generating informative fragmentation patterns of ions, especially for biological molecules whose complicated structures, subtle modifications, and large sizes often impede molecular characterization. UVPD energizes ions via absorption of high-energy photons, which allows access to new dissociation pathways relative to more conventional ion-activation methods. Applications of UVPD for the analysis of peptides, proteins, lipids, and other classes of biologically relevant molecules are emphasized in this Review.
Collapse
Affiliation(s)
- Jennifer S. Brodbelt
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Lindsay J. Morrison
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Inês Santos
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
14
|
Malik HI, Mir AR, Abidi M, Habib S, Khan FH, Moinuddin. Preferential recognition of epitopes on peroxynitrite-modified alpha-2-macroglobulin by circulating autoantibodies in rheumatoid arthritis patients. J Biomol Struct Dyn 2020; 38:1984-1994. [PMID: 31179888 DOI: 10.1080/07391102.2019.1623073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 05/19/2019] [Indexed: 10/26/2022]
Abstract
Autoimmune responses against post-translationally modified antigens are a hallmark of several autoimmune diseases. In this work, we have studied the changes in alpha-2-macroglobulin (α2M) upon modification by peroxynitrite. Furthermore, we have evaluated the immunogenicity of modified α2M in experimental rabbits and rheumatoid arthritis (RA) patients. Peroxynitrite-modified α2M showed disturbed microenvironment and altered aromatic residues under UV and fluorescence studies. Aggregation, reduction in β-sheet content, production of nitrotyrosine and shift in amide I and II bands were observed in the modified α2M by polyacrylamide gel electrophoresis besides CD and FTIR spectroscopic analysis. The exposure of hydrophobic clusters and changes in contact positions were observed in ANS and ThT binding assays. Immunological studies using ELISA showed peroxynitrite-modified α2M as highly immunogenic producing high titre of specific antibodies in immunized rabbits. Cross-reactivity studies revealed the polyspecificity of the elicited antibodies. Direct binding ELISA and competitive inhibition studies confirmed the presence of circulating antibodies in the sera of RA patients having high specificity towards the peroxynitrite-modified α2M as compared to the native α2M. Sera from healthy (normal) human subjects showed lower binding with the native and modified protein. This study confirms that peroxynitrite induces structural modifications in α2M and makes it immunogenic. The presence of neo-antigenic determinants on modified α2M with enhanced binding for circulating autoantibodies in RA patients could offer new possibilities for diagnosis and etiopathology of the disease. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Heena Imtiaz Malik
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Abdul Rouf Mir
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Minhal Abidi
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Safia Habib
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| | - Fahim Halim Khan
- bDepartment of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP, India
| | - Moinuddin
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, UP, India
| |
Collapse
|
15
|
Begara-Morales JC, Sánchez-Calvo B, Gómez-Rodríguez MV, Chaki M, Valderrama R, Mata-Pérez C, López-Jaramillo J, Corpas FJ, Barroso JB. Short-Term Low Temperature Induces Nitro-Oxidative Stress that Deregulates the NADP-Malic Enzyme Function by Tyrosine Nitration in Arabidopsis thaliana. Antioxidants (Basel) 2019; 8:antiox8100448. [PMID: 31581524 PMCID: PMC6827146 DOI: 10.3390/antiox8100448] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/20/2019] [Indexed: 12/31/2022] Open
Abstract
Low temperature (LT) negatively affects plant growth and development via the alteration of the metabolism of reactive oxygen and nitrogen species (ROS and RNS). Among RNS, tyrosine nitration, the addition of an NO2 group to a tyrosine residue, can modulate reduced nicotinamide-dinucleotide phosphate (NADPH)-generating systems and, therefore, can alter the levels of NADPH, a key cofactor in cellular redox homeostasis. NADPH also acts as an indispensable electron donor within a wide range of enzymatic reactions, biosynthetic pathways, and detoxification processes, which could affect plant viability. To extend our knowledge about the regulation of this key cofactor by this nitric oxide (NO)-related post-translational modification, we analyzed the effect of tyrosine nitration on another NADPH-generating enzyme, the NADP-malic enzyme (NADP-ME), under LT stress. In Arabidopsis thaliana seedlings exposed to short-term LT (4 °C for 48 h), a 50% growth reduction accompanied by an increase in the content of superoxide, nitric oxide, and peroxynitrite, in addition to diminished cytosolic NADP-ME activity, were found. In vitro assays confirmed that peroxynitrite inhibits cytosolic NADP-ME2 activity due to tyrosine nitration. The mass spectrometric analysis of nitrated NADP-ME2 enabled us to determine that Tyr-73 was exclusively nitrated to 3-nitrotyrosine by peroxynitrite. The in silico analysis of the Arabidopsis NADP-ME2 protein sequence suggests that Tyr73 nitration could disrupt the interactions between the specific amino acids responsible for protein structure stability. In conclusion, the present data show that short-term LT stress affects the metabolism of ROS and RNS, which appears to negatively modulate the activity of cytosolic NADP-ME through the tyrosine nitration process.
Collapse
Affiliation(s)
- Juan C Begara-Morales
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Campus "Las Lagunillas", s/n, E-23071 Jaén, Spain.
| | - Beatriz Sánchez-Calvo
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Campus "Las Lagunillas", s/n, E-23071 Jaén, Spain.
| | - María V Gómez-Rodríguez
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Campus "Las Lagunillas", s/n, E-23071 Jaén, Spain.
| | - Mounira Chaki
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Campus "Las Lagunillas", s/n, E-23071 Jaén, Spain.
| | - Raquel Valderrama
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Campus "Las Lagunillas", s/n, E-23071 Jaén, Spain.
| | - Capilla Mata-Pérez
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Campus "Las Lagunillas", s/n, E-23071 Jaén, Spain.
| | - Javier López-Jaramillo
- Institute of Biotechnology, Department of Organic Chemistry, Faculty of Sciences, University of Granada, E-18071 Granada, Spain.
| | - Francisco J Corpas
- Group of Antioxidants, Free Radicals, and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, C/ Profesor Albareda 1, E-18080 Granada, Spain.
| | - Juan B Barroso
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of Jaén, Campus "Las Lagunillas", s/n, E-23071 Jaén, Spain.
| |
Collapse
|
16
|
Structure effect of water-soluble iron porphyrins on catalyzing protein tyrosine nitration in the presence of nitrite and hydrogen peroxide. Nitric Oxide 2019; 91:42-51. [PMID: 31351146 DOI: 10.1016/j.niox.2019.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022]
Abstract
Water-soluble iron porphyrins, such as FeTPPS (5,10,15,20-tetrakis (4-sulfonatophenyl) porphyrinato iron (III)), FeTMPyP (5,10,15,20-tetrakis (N-methyl-4'-pyridyl) porphyrinato iron (III) chloride) and FeTBAP (5,10,15,20-tetrakis (4-benzoic acid) porphyrinato iron (III)), are highly active catalysts for peroxynitrite decomposition and thereby have been suggested as therapeutic agent for inflammatory diseases that implicate the involvement of nitrotyrosine formation. Here, we systemically investigated catalytic properties of FeTPPS, FeTMPyP and FeTBAP on protein nitration in the presence of hydrogen peroxide and nitrite. We showed that FeTPPS, FeTBAP and FeTMPyP all exhibited higher peroxidase activity in compared with hemin. As to protein nitration, the catalytic effect of FeTPPS and FeTBAP are effective in the presence of hydrogen peroxide and nitrite, while negligible BSA nitration was observed in the case of FeTMPyP. Moreover, the underlying mechanism of the oxidation of FeTPPS, FeTBAP and FeTMPyP was further studied. Collectively, our results suggest that, compound I and II species are involved in as the key intermediates in FeTMPyP/H2O2 system as similar as those in FeTPPS/H2O2 and FeTBAP/H2O2 system. As compared to weak antioxidants, TPPS and TBAP, however, TMPyP scavenges oxo-Fe (IV) intermediates of FeTMPyP at a faster rate by significant self-degradation; results in the shortest lifetimes of OFeIV-TMPyP and the lowest catalytic activity on oxidizing tyrosine and nitrite; and therefore, attributes to inactivation of FeTMPyP in protein nitration. In addition, association of FeTMPyP to BSA was found weak, while strong binding of FeTPPS and FeTBAP were observed. The weak binding keeps away of target residue of BSA from the center of FeTMPyP where the RNS is generated, which might be attributed as additional factors to the inactivation of FeTMPyP in protein nitration.
Collapse
|
17
|
Tang L, Fang C. Nitration of Tyrosine Channels Photoenergy through a Conical Intersection in Water. J Phys Chem B 2019; 123:4915-4928. [PMID: 31094198 DOI: 10.1021/acs.jpcb.9b03464] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nitration of tyrosine occurs under oxidative stress in vivo. The product, 3-nitrotyrosine (3NY), has a dramatically decreased quantum yield and can be used as a molecular ruler. In this study, femtosecond transient absorption spectroscopy and quantum calculations were implemented to elucidate the photoinduced relaxation processes of anionic 3NY in water. Upon 400 nm excitation into an excited electronic state with notable charge-transfer (CT) character, a barrierless nitro-twisting motion rapidly (<100 fs) guides the chromophore into an adjacent twisted intramolecular CT state, therein reaching a sloped S1/S0 conical intersection on the ∼100 fs time scale. Once in the hot ground state, excess energy is further released through vibrational cooling with biexponential time constants of ∼140 and 680 fs in water. Nitro back-twisting occurs on longer time scales (∼1.1 and 9 ps in water), returning the system to original ground state. Systematic evaluations of excited-state potential energies of anionic 3NY were performed by density functional theory (DFT) and time-dependent DFT calculations, showing that intersystem crossing (ISC) from the first singlet state (S1) to the first or second triplet state (T1 or T2) is unlikely. Inclusion of an explicit water molecule in calculations leads to improved mapping of the excited-state energy ordering of the second singlet state (S2) and T2, further diminishing ISC probability from S1 and favoring an ultrafast internal conversion to S0. These results provide deep insights into the highly efficient nonradiative decay of anionic 3NY in aqueous solution, with nitro-site-specific information that can help infer the characterization and potential optogenetic control of 3NY in protein environment.
Collapse
Affiliation(s)
- Longteng Tang
- Department of Chemistry , Oregon State University , 153 Gilbert Hall , Corvallis , Oregon 97331-4003 , United States
| | - Chong Fang
- Department of Chemistry , Oregon State University , 153 Gilbert Hall , Corvallis , Oregon 97331-4003 , United States
| |
Collapse
|
18
|
Stiff Substrates Enhance Endothelial Oxidative Stress in Response to Protein Kinase C Activation. Appl Bionics Biomech 2019; 2019:6578492. [PMID: 31110559 PMCID: PMC6487160 DOI: 10.1155/2019/6578492] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/28/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
Abstract
Arterial stiffness, which increases with aging and hypertension, is an independent cardiovascular risk factor. While stiffer substrates are known to affect single endothelial cell morphology and migration, the effect of substrate stiffness on endothelial monolayer function is less understood. The objective of this study was to determine if substrate stiffness increased endothelial monolayer reactive oxygen species (ROS) in response to protein kinase C (PKC) activation and if this oxidative stress then impacted adherens junction integrity. Porcine aortic endothelial cells were cultured on varied stiffness polyacrylamide gels and treated with phorbol 12-myristate 13-acetate (PMA), which stimulates PKC and ROS without increasing actinomyosin contractility. PMA-treated endothelial cells on stiffer substrates increased ROS and adherens junction loss without increased contractility. ROS scavengers abrogated PMA effects on cell-cell junctions, with a more profound effect in cells on stiffer substrates. Finally, endothelial cells in aortae from elastin haploinsufficient mice (Eln+/-), which were stiffer than aortae from wild-type mice, showed decreased VE-cadherin colocalization with peripheral actin following PMA treatment. These data suggest that oxidative stress may be enhanced in endothelial cells in stiffer vessels, which could contribute to the association between arterial stiffness and cardiovascular disease.
Collapse
|
19
|
Poly(ADP-ribose) Polymerase (PARP) and PARP Inhibitors: Mechanisms of Action and Role in Cardiovascular Disorders. Cardiovasc Toxicol 2019; 18:493-506. [PMID: 29968072 DOI: 10.1007/s12012-018-9462-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Poly(ADP-ribosyl)ation is an immediate cellular repair response to DNA damage and is catalyzed primarily by poly(ADP-ribose)polymerase-1 (PARP1), which is the most abundant of the 18 different PARP isoforms and accounts for more than 90% of the catalytic activity of PARP in the cell nucleus. Upon detection of a DNA strand break, PARP1 binds to the DNA, cleaves nicotinamide adenine dinucleotide between nicotinamide and ribose and then modifies the DNA nuclear acceptor proteins by formation of a bond between the protein and the ADP-ribose residue. This generates ribosyl-ribosyl linkages that act as a signal for other DNA-repairing enzymes and DNA base repair. Extensive DNA breakage in cells results in excessive activation of PARP with resultant depletion of the cellular stores of nicotinamide adenine dinucleotide (NAD+) which slows the rate of glycolysis, mitochondrial electron transport, and ultimately ATP formation in these cells. This paper focuses on PARP in DNA repair in atherosclerosis, acute myocardial infarction/reperfusion injury, and congestive heart failure and the role of PARP inhibitors in combating the effects of excessive PARP activation in these diseases. Free oxygen radicals and nitrogen radicals in arteries contribute to disruption of the vascular endothelial glycocalyx, which increase the permeability of the endothelium to inflammatory cells and also low-density lipoproteins and the accumulation of lipid in the vascular intima. Mild inflammation and DNA damage within vascular cells promote PARP1 activation and DNA repair. Moderate DNA damage induces caspase-dependent PARP cleavage and vascular cell apoptosis. Severe DNA damage due to vascular inflammation causes excessive activation of PARP1. This causes endothelial cell depletion of NAD+ and ATP, downregulation of atheroprotective SIRT1, necrotic cell death, and ultimately atherosclerotic plaque disruption. Inhibition of PARP decreases vascular endothelial cell adhesion P-selectin and ICAM-1 molecules, inflammatory cells, pro-death caspase-3, and c-Jun N-terminal kinase (JNK) activation and upregulates prosurvival extracellular signal-regulated kinases and AKT, which decrease vascular cell apoptosis and necrosis and limit atherosclerosis and plaque disruption. In myocardial infarction with coronary occlusion then reperfusion, which occurs with coronary angioplasty or thrombolytic therapy, reperfusion injury occurs in as many as 31% of patients and is caused by inflammatory cells, free oxygen and nitrogen radicals, the rapid transcriptional activation of inflammatory cytokines, and the activation of PARP1. Inhibition of PARP attenuates neutrophil infiltration and inflammatory cytokine expression in the reperfused myocardium and preserves myocardial NAD+ and ATP. In addition, PARP inhibition increases the activation of myocyte survival enzymes protein kinase B (Akt) and protein kinase C epsilon (PKCε), and decreases the activity of myocardial ventricular remodeling enzymes PKCα/β, PKCζ/λ, and PKCδ. As a consequence, cardiomyocyte and vascular endothelial cell necrosis is decreased and myocardial contractility is preserved. In heart failure and circulatory shock in animal models, PARP inhibition significantly attenuates decreases in left ventricular systolic pressure, ventricular contractility and relaxation, stroke volume, and increases survival by limiting or preventing upregulation of adhesion molecules, proinflammatory cytokines, myocardial mononuclear cell infiltration, and PKCα/β and PKC λ/ζ. In this manner, PARP inhibition partially restores the myocardial concentrations of NAD+, limits ventricular remodeling and fibrosis, and prevents significant decreases in myocardial contractility. Based primarily on investigations in preclinical models of atherosclerosis, myocardial infarction, and heart failure, PARP inhibition appears to be beneficial in limiting or inhibiting cardiovascular dysfunction. These studies indicate that investigations of acute and chronic PARP inhibition are warranted in patients with atherosclerotic coronary artery disease.
Collapse
|
20
|
Ahmad R, Hussain A, Ahsan H. Peroxynitrite: cellular pathology and implications in autoimmunity. J Immunoassay Immunochem 2019; 40:123-138. [PMID: 30843753 DOI: 10.1080/15321819.2019.1583109] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In inflamed tissues, the reaction of nitric oxide and superoxide leads to the formation of an extremely reactive peroxynitrite (ONOO-), which is a well known oxidizing and nitrating agent that exhibits high reactivity at physiological pH. The peroxynitrite formed can attack a wide range of biomolecules via direct oxidative reactions or indirect radical-mediated mechanisms thus triggering cellular responses leading to cell signaling, oxidative injury, committing cells to necrosis or apoptosis. Cellular DNA is an important target for ONOO- attack, and can react with deoxyribose, nucleobases or induces single strand breaks. The free radical-mediated damage to proteins results in the modification of amino acid residues, cross-linking of side chains and fragmentation. Free/protein-bound tyrosines are attacked by various reactive nitrogen species (RNS), including peroxynitrite, to form free/protein-bound nitrotyrosine (NT). The formation of NT represents a specific peroxynitrite-mediated protein modification, and the detection of NT in proteins is considered as a biomarker for endogenous peroxynitrite activity. The peroxynitrite-driven oxidation and nitration of biomolecules may lead to autoimmunity and age-related neurodegenerative diseases. Hence, peroxynitrite modified DNA and nitrated proteins can act as neoantigens and lead to the generation of autoantibodies against self-components in autoimmune disorders.
Collapse
Affiliation(s)
- Rizwan Ahmad
- a Department of Academic Affairs, College of Medicine , Imam Abdulrahman bin Faisal University , Dammam , KSA
| | - Ahtesham Hussain
- b Lee's Biotech , Korean Institute of Bioscience and Biotechnology , Daejeon , South Korea
| | - Haseeb Ahsan
- c Department of Biochemistry, Faculty of Dentistry , Jamia Millia Islamia , New Delhi , India
| |
Collapse
|
21
|
Xu DD, Li WT, Jiang D, Wu HG, Ren MS, Chen MQ, Wu YB. 3-N-Butylphthalide mitigates high glucose-induced injury to Schwann cells: association with nitrosation and apoptosis. Neural Regen Res 2019; 14:513-518. [PMID: 30539821 PMCID: PMC6334601 DOI: 10.4103/1673-5374.245590] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A high glucose state readily causes peripheral axon atrophy, demyelination, loss of nerve fiber function, and delayed regeneration. However, few studies have examined whether nitration is also critical for diabetic peripheral neuropathy. Therefore, this study investigated the effects of high glucose on proliferation, apoptosis, and 3-nitrotyrosine levels of Schwann cells treated with butylphthalide. In addition, we explored potential protective mechanisms of butylphthalide on peripheral nerves. Schwann cells were cultured in vitro with high glucose then stimulated with the peroxynitrite anion inhibitors uric acid and 3-n-butylphthalide for 48 hours. Cell Counting Kit-8 and flow cytometry were used to investigate the effects of uric acid and 3-n-butylphthalide on proliferation and apoptosis of Schwann cells exposed to a high glucose environment. Effects of uric acid and 3-n-butylphthalide on levels of 3-nitrotyrosine in Schwann cells were detected by enzyme-linked immunosorbent assay. The results indicated that Schwann cells cultured in high glucose showed decreased proliferation, but increased apoptosis and intracellular 3-nitrotyrosine levels. However, intervention with uric acid or 3-n-butylphthalide could increase proliferation of Schwann cells cultured in high glucose, and inhibited apoptosis and intracellular 3-nitrotyrosine levels. According to our data, 3-n-butylphthalide may inhibit cell nitrification and apoptosis, and promote cell proliferation, thereby reducing damage to Schwann cells caused by high glucose.
Collapse
Affiliation(s)
- Dan-Dan Xu
- Department of Neurology, First Affiliated Hospital of University of Science and Technology of China; Department of Neurology, Anhui Second People's Hospital, Hefei, Anhui Province, China
| | - Wen-Ting Li
- Department of Infection, First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui Province, China
| | - Dan Jiang
- Department of Neurology, Anhui Second People's Hospital, Hefei, Anhui Province, China
| | - Huai-Guo Wu
- Department of Neurology, Anhui Second People's Hospital, Hefei, Anhui Province, China
| | - Ming-Shan Ren
- Department of Neurology, First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui Province, China
| | - Mei-Qiao Chen
- Department of Neurology, Affiliated Anhui Provincial Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yuan-Bo Wu
- Department of Neurology, First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
22
|
Everson F, Genis A, Ogundipe T, De Boever P, Goswami N, Lochner A, Blackhurst D, Strijdom H. Treatment with a fixed dose combination antiretroviral therapy drug containing tenofovir, emtricitabine and efavirenz is associated with cardioprotection in high calorie diet-induced obese rats. PLoS One 2018; 13:e0208537. [PMID: 30517206 PMCID: PMC6281242 DOI: 10.1371/journal.pone.0208537] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 11/18/2018] [Indexed: 12/31/2022] Open
Abstract
HIV-infection, certain antiretroviral drug classes, especially protease inhibitors (PI), and obesity are associated with increased ischaemic heart disease (IHD) risk. However, the effect of PI-free fixed dose combination (FDC) antiretroviral therapy (ART) on hearts exposed to ischaemia-reperfusion injury (I/R) is unknown, particularly in obesity. This is becoming relevant as World Health Organisation guidelines recommend a FDC ART containing (non-) nucleoside reverse transcriptase inhibitors (tenofovir (TDF), emtricitabine (FTC) and efavirenz (EFV)) as first-line HIV treatment. Additionally, obesity rates are rising in HIV-infected populations, not only in ART-experienced individuals, but also at the time of ART initiation, which may further increase the risk of IHD. Therefore, we investigated the effects of PI-free FDC ART in myocardial I/R-exposed hearts from obese rats. Obesity was induced in male wistar rats via a 16-week high calorie diet. At week 10, treatment with a FDC ART drug containing TDF/FTC/EFV was initiated. Biometric and metabolic parameters, as well as myocardial functional recovery and infract size (IS), and myocardial signalling proteins following I/R were assessed after 16 weeks. Obese rats presented with increased body and intraperitoneal fat mass, elevated triglyceride and TBARS levels, whilst the hearts responded to I/R with impaired functional performance and increased IS. The FDC ART treatment did not alter biometric and metabolic parameters in obese rats. In a novel finding, ART protected obese hearts against I/R as shown by improved functional performance and smaller IS vs. untreated obese hearts. Cardioprotection was underscored by increased myocardial phosphorylated endothelial nitric oxide synthase (eNOS) and reduced AMP-kinase levels. In conclusion, these results demonstrate for the first time, that 6-weeks treatment of obese rats with a FDC ART drug specifically containing TDF/FTC/EFV conferred cardioprotection against I/R. The FDC ART-induced cardioprotection was seemingly unrelated to metabolic changes, but rather due to direct cardiac mechanisms including the up-regulation of myocardial eNOS.
Collapse
Affiliation(s)
- Frans Everson
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Republic of South Africa
| | - Amanda Genis
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Republic of South Africa
| | - Temitope Ogundipe
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Republic of South Africa
| | - Patrick De Boever
- Environmental Risk and Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Nandu Goswami
- Department of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Amanda Lochner
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Republic of South Africa
| | - Dee Blackhurst
- Division of Chemical Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hans Strijdom
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Republic of South Africa
| |
Collapse
|
23
|
DeepNitro: Prediction of Protein Nitration and Nitrosylation Sites by Deep Learning. GENOMICS PROTEOMICS & BIOINFORMATICS 2018; 16:294-306. [PMID: 30268931 PMCID: PMC6205083 DOI: 10.1016/j.gpb.2018.04.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/12/2018] [Accepted: 04/27/2018] [Indexed: 11/24/2022]
Abstract
Protein nitration and nitrosylation are essential post-translational modifications (PTMs) involved in many fundamental cellular processes. Recent studies have revealed that excessive levels of nitration and nitrosylation in some critical proteins are linked to numerous chronic diseases. Therefore, the identification of substrates that undergo such modifications in a site-specific manner is an important research topic in the community and will provide candidates for targeted therapy. In this study, we aimed to develop a computational tool for predicting nitration and nitrosylation sites in proteins. We first constructed four types of encoding features, including positional amino acid distributions, sequence contextual dependencies, physicochemical properties, and position-specific scoring features, to represent the modified residues. Based on these encoding features, we established a predictor called DeepNitro using deep learning methods for predicting protein nitration and nitrosylation. Using n-fold cross-validation, our evaluation shows great AUC values for DeepNitro, 0.65 for tyrosine nitration, 0.80 for tryptophan nitration, and 0.70 for cysteine nitrosylation, respectively, demonstrating the robustness and reliability of our tool. Also, when tested in the independent dataset, DeepNitro is substantially superior to other similar tools with a 7%−42% improvement in the prediction performance. Taken together, the application of deep learning method and novel encoding schemes, especially the position-specific scoring feature, greatly improves the accuracy of nitration and nitrosylation site prediction and may facilitate the prediction of other PTM sites. DeepNitro is implemented in JAVA and PHP and is freely available for academic research at http://deepnitro.renlab.org.
Collapse
|
24
|
Oxidative Stress in Preeclampsia and Placental Diseases. Int J Mol Sci 2018; 19:ijms19051496. [PMID: 29772777 PMCID: PMC5983711 DOI: 10.3390/ijms19051496] [Citation(s) in RCA: 378] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/09/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia is a persistent hypertensive gestational disease characterized by high blood pressure and proteinuria, which presents from the second trimester of pregnancy. At the cellular level, preeclampsia has largely been associated with the release of free radicals by the placenta. Placenta-borne oxidative and nitrosative stresses are even sometimes considered as the major molecular determinants of the maternal disease. In this review, we present the recent literature evaluating free radical production in both normal and pathological placentas (including preeclampsia and other major pregnancy diseases), in humans and animal models. We then assess the putative effects of these free radicals on the placenta and maternal endothelium. This analysis was conducted with regard to recent papers and possible therapeutic avenues.
Collapse
|
25
|
Tilson MD. Autoimmunity in the Abdominal Aortic Aneurysm and its Association with Smoking. AORTA : OFFICIAL JOURNAL OF THE AORTIC INSTITUTE AT YALE-NEW HAVEN HOSPITAL 2018; 5:159-167. [PMID: 29766007 DOI: 10.12945/j.aorta.2017.17.693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 12/05/2017] [Indexed: 11/18/2022]
Abstract
Smoking increases the risk of abdominal aortic aneurysm (AAA) in both humans and mice, although the underlying mechanisms are not completely understood. An adventitial aortic antigen, AAAP-40, has been partially sequenced. It has motifs with similarities to all three fibrinogen chains and appears to be connected in evolution to a large family of proteins called fibrinogen-related proteins. Fibrinogen may undergo non-enzymatic nitration, which may result from exposure to nitric oxide in cigarette smoke. Nitration of proteins renders them more immunogenic. It has recently been reported that anti-fibrinogen antibody promotes AAA development in mice. Also, anti-fibrinogen antibodies are present in patients with AAA. These matters are reviewed in the overall context of autoimmunity in AAA. The evidence suggests that smoking amplifies an auto-immune reaction that is critical to the pathogenesis of AAA.
Collapse
Affiliation(s)
- M David Tilson
- Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
26
|
Exposure of fibrinogen and thrombin to nitric oxide donor ProliNONOate affects fibrin clot properties. Blood Coagul Fibrinolysis 2018; 28:356-364. [PMID: 27755019 DOI: 10.1097/mbc.0000000000000602] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
: Fibrin fibers form the structural backbone of blood clots. The structural properties of fibrin clots are highly dependent on formation kinetics. Environmental factors such as protein concentration, pH, salt, and protein modification, to name a few, can affect fiber kinetics through altered fibrinopeptide release, monomer association, and/or lateral aggregation. The objective of our study was to determine the effect of thrombin and fibrinogen exposed to nitric oxide on fibrin clot properties. ProliNONOate (5 μmol/l) was added to fibrinogen and thrombin before clot initiation and immediately following the addition of thrombin to the fibrinogen solution. Resulting fibrin fibers were probed with an atomic force microscope to determine their diameter and extensibility and fibrin clots were analyzed for clot density using confocal microscopy. Fiber diameters were also determined by confocal microscopy and the rate of clot formation was recorded using UV-vis spectrophotometry. Protein oxidation and S-nitrosation was determined by UV-vis, ELISA, and chemiluminescence. The addition of ProliNONOate to fibrinogen or thrombin resulted in a change in clot structure. ProliNONOate exposure produced clots with lower fiber density, thicker fibers, and increased time to maximum turbidity. The effect of the exposure of nitric oxide to thrombin and fibrinogen were measured independently and indicated that each plays a role in altering clot properties. We detected thrombin S-nitrosation and protein carbonyl formation after nitric oxide exposure. Our study reveals a regulation of fibrin clot properties by nitric oxide exposure and suggests a role of peroxynitrite in oxidative modifications of the proteins. These results relate nitric oxide bioavailability and oxidative stress to altered clot properties.
Collapse
|
27
|
Lin YW. Structure and function of heme proteins regulated by diverse post-translational modifications. Arch Biochem Biophys 2018; 641:1-30. [PMID: 29407792 DOI: 10.1016/j.abb.2018.01.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/10/2018] [Accepted: 01/13/2018] [Indexed: 01/08/2023]
|
28
|
Jain P, Bhatla SC. Molecular mechanisms accompanying nitric oxide signalling through tyrosine nitration and S-nitrosylation of proteins in plants. FUNCTIONAL PLANT BIOLOGY : FPB 2018; 45:70-82. [PMID: 32291022 DOI: 10.1071/fp16279] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 02/01/2017] [Indexed: 05/08/2023]
Abstract
Nitric oxide (NO) signalling in plants is responsible for modulation of a variety of plant developmental processes. Depending on the tissue system, the signalling of NO-modulated biochemical responses majorly involves the processes of tyrosine nitration or S-nitrosylation of specific proteins/enzymes. It has further been observed that there is a significant impact of various biotic/abiotic stress conditions on the extent of tyrosine nitration and S-nitrosylation of various metabolic enzymes, which may act as a positive or negative modulator of the specific routes associated with adaptive mechanisms employed by plants under the said stress conditions. In addition to recent findings on the modulation of enzymes of primary metabolism by NO through these two biochemical mechanisms, a major mechanism for regulating the levels of reactive oxygen species (ROS) under stress conditions has also been found to be through tyrosine nitration or S-nitrosylation of ROS-scavenging enzymes. Recent investigations have further highlighted the differential manner in which the ROS-scavenging enzymes may be S-nitrosylated and tyrosine nitrated, with reference to their tissue distribution. Keeping in mind the very recent findings on these aspects, the present review has been prepared to provide an analytical view on the significance of protein tyrosine nitration and S-nitrosylation in plant development.
Collapse
Affiliation(s)
- Prachi Jain
- Laboratory of Plant Physiology and Biochemistry, Department of Botany, University of Delhi, Delhi, India
| | - Satish C Bhatla
- Laboratory of Plant Physiology and Biochemistry, Department of Botany, University of Delhi, Delhi, India
| |
Collapse
|
29
|
A boronate-based ratiometric fluorescent probe for fast selective detection of peroxynitrite. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2017.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Thompson S, Martínez-Burgo B, Sepuru KM, Rajarathnam K, Kirby JA, Sheerin NS, Ali S. Regulation of Chemokine Function: The Roles of GAG-Binding and Post-Translational Nitration. Int J Mol Sci 2017; 18:ijms18081692. [PMID: 28771176 PMCID: PMC5578082 DOI: 10.3390/ijms18081692] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/28/2017] [Accepted: 07/30/2017] [Indexed: 12/12/2022] Open
Abstract
The primary function of chemokines is to direct the migration of leukocytes to the site of injury during inflammation. The effects of chemokines are modulated by several means, including binding to G-protein coupled receptors (GPCRs), binding to glycosaminoglycans (GAGs), and through post-translational modifications (PTMs). GAGs, present on cell surfaces, bind chemokines released in response to injury. Chemokines bind leukocytes via their GPCRs, which directs migration and contributes to local inflammation. Studies have shown that GAGs or GAG-binding peptides can be used to interfere with chemokine binding and reduce leukocyte recruitment. Post-translational modifications of chemokines, such as nitration, which occurs due to the production of reactive species during oxidative stress, can also alter their biological activity. This review describes the regulation of chemokine function by GAG-binding ability and by post-translational nitration. These are both aspects of chemokine biology that could be targeted if the therapeutic potential of chemokines, like CXCL8, to modulate inflammation is to be realised.
Collapse
Affiliation(s)
- Sarah Thompson
- Applied Immunobiology and Transplantation Group, Institute of Cellular Medicine, Medical School, University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH, UK.
| | - Beatriz Martínez-Burgo
- Applied Immunobiology and Transplantation Group, Institute of Cellular Medicine, Medical School, University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH, UK.
| | - Krishna Mohan Sepuru
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| | - Krishna Rajarathnam
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| | - John A Kirby
- Applied Immunobiology and Transplantation Group, Institute of Cellular Medicine, Medical School, University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH, UK.
| | - Neil S Sheerin
- Applied Immunobiology and Transplantation Group, Institute of Cellular Medicine, Medical School, University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH, UK.
| | - Simi Ali
- Applied Immunobiology and Transplantation Group, Institute of Cellular Medicine, Medical School, University of Newcastle upon Tyne, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
31
|
Reinmuth-Selzle K, Kampf CJ, Lucas K, Lang-Yona N, Fröhlich-Nowoisky J, Shiraiwa M, Lakey PSJ, Lai S, Liu F, Kunert AT, Ziegler K, Shen F, Sgarbanti R, Weber B, Bellinghausen I, Saloga J, Weller MG, Duschl A, Schuppan D, Pöschl U. Air Pollution and Climate Change Effects on Allergies in the Anthropocene: Abundance, Interaction, and Modification of Allergens and Adjuvants. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:4119-4141. [PMID: 28326768 PMCID: PMC5453620 DOI: 10.1021/acs.est.6b04908] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/07/2017] [Accepted: 03/22/2017] [Indexed: 05/13/2023]
Abstract
Air pollution and climate change are potential drivers for the increasing burden of allergic diseases. The molecular mechanisms by which air pollutants and climate parameters may influence allergic diseases, however, are complex and elusive. This article provides an overview of physical, chemical and biological interactions between air pollution, climate change, allergens, adjuvants and the immune system, addressing how these interactions may promote the development of allergies. We reviewed and synthesized key findings from atmospheric, climate, and biomedical research. The current state of knowledge, open questions, and future research perspectives are outlined and discussed. The Anthropocene, as the present era of globally pervasive anthropogenic influence on planet Earth and, thus, on the human environment, is characterized by a strong increase of carbon dioxide, ozone, nitrogen oxides, and combustion- or traffic-related particulate matter in the atmosphere. These environmental factors can enhance the abundance and induce chemical modifications of allergens, increase oxidative stress in the human body, and skew the immune system toward allergic reactions. In particular, air pollutants can act as adjuvants and alter the immunogenicity of allergenic proteins, while climate change affects the atmospheric abundance and human exposure to bioaerosols and aeroallergens. To fully understand and effectively mitigate the adverse effects of air pollution and climate change on allergic diseases, several challenges remain to be resolved. Among these are the identification and quantification of immunochemical reaction pathways involving allergens and adjuvants under relevant environmental and physiological conditions.
Collapse
Affiliation(s)
| | - Christopher J. Kampf
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- Institute
of Inorganic and Analytical Chemistry, Johannes
Gutenberg University, Mainz, 55128, Germany
| | - Kurt Lucas
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Naama Lang-Yona
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | | | - Manabu Shiraiwa
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- Department
of Chemistry, University of California, Irvine, California 92697-2025, United States
| | - Pascale S. J. Lakey
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Senchao Lai
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
- South
China University of Technology, School of
Environment and Energy, Guangzhou, 510006, China
| | - Fobang Liu
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Anna T. Kunert
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Kira Ziegler
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Fangxia Shen
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Rossella Sgarbanti
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Bettina Weber
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| | - Iris Bellinghausen
- Department
of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, 55131, Germany
| | - Joachim Saloga
- Department
of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, 55131, Germany
| | - Michael G. Weller
- Division
1.5 Protein Analysis, Federal Institute
for Materials Research and Testing (BAM), Berlin, 12489, Germany
| | - Albert Duschl
- Department
of Molecular Biology, University of Salzburg, 5020 Salzburg, Austria
| | - Detlef Schuppan
- Institute
of Translational Immunology and Research Center for Immunotherapy,
Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, 55131 Germany
- Division
of Gastroenterology, Beth Israel Deaconess
Medical Center and Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Ulrich Pöschl
- Multiphase
Chemistry Department, Max Planck Institute
for Chemistry, Mainz, 55128, Germany
| |
Collapse
|
32
|
Akolkar G, Bagchi AK, Ayyappan P, Jassal DS, Singal PK. Doxorubicin-induced nitrosative stress is mitigated by vitamin C via the modulation of nitric oxide synthases. Am J Physiol Cell Physiol 2017; 312:C418-C427. [PMID: 28100487 DOI: 10.1152/ajpcell.00356.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/12/2017] [Accepted: 01/12/2017] [Indexed: 12/14/2022]
Abstract
An increase in oxidative stress is suggested to be the main cause in Doxorubicin (Dox)–induced cardiotoxicity. However, there is now evidence that activation of inducible nitric oxide synthase (iNOS) and nitrosative stress are also involved. The role of vitamin C (Vit C) in the regulation of nitric oxide synthase (NOS) and reduction of nitrosative stress in Dox-induced cardiotoxicity is unknown. The present study investigated the effects of Vit C in the mitigation of Dox-induced changes in the levels of nitric oxide (NO), NOS activity, protein expression of NOS isoforms, and nitrosative stress as well as cytokines TNF-α and IL-10 in isolated cardiomyocytes. Cardiomyocytes isolated from adult Sprague-Dawley rats were segregated into four groups: 1) control, 2) Vit C (25 µM), 3) Dox (10 µM), and 4) Vit C + Dox. Dox caused a significant increase in the generation of superoxide radical (O2·−), peroxynitrite, and NO, and these effects of Dox were blunted by Vit C. Dox increased the expression of iNOS and altered protein expression as well as activation of endothelial NOS (eNOS). These changes were prevented by Vit C. Dox induced an increase in the ratio of monomeric/dimeric eNOS, promoting the production of O2·−, which was prevented by Vit C by increasing the stability of the dimeric form of eNOS. Vit C protected against the Dox-induced increase in TNFα as well as a reduction in IL-10. These results suggest that Vit C provides cardioprotection by reducing oxidative/nitrosative stress and inflammation via a modulation of Dox-induced increase in the NO levels and NOS activity.
Collapse
Affiliation(s)
- Gauri Akolkar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ashim K. Bagchi
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Prathapan Ayyappan
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder S. Jassal
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan K. Singal
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
33
|
|
34
|
Multiple Forms of Glutamate Dehydrogenase in Animals: Structural Determinants and Physiological Implications. BIOLOGY 2016; 5:biology5040053. [PMID: 27983623 PMCID: PMC5192433 DOI: 10.3390/biology5040053] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/26/2016] [Accepted: 12/07/2016] [Indexed: 11/17/2022]
Abstract
Glutamate dehydrogenase (GDH) of animal cells is usually considered to be a mitochondrial enzyme. However, this enzyme has recently been reported to be also present in nucleus, endoplasmic reticulum and lysosomes. These extramitochondrial localizations are associated with moonlighting functions of GDH, which include acting as a serine protease or an ATP-dependent tubulin-binding protein. Here, we review the published data on kinetics and localization of multiple forms of animal GDH taking into account the splice variants, post-translational modifications and GDH isoenzymes, found in humans and apes. The kinetic properties of human GLUD1 and GLUD2 isoenzymes are shown to be similar to those published for GDH1 and GDH2 from bovine brain. Increased functional diversity and specific regulation of GDH isoforms due to alternative splicing and post-translational modifications are also considered. In particular, these structural differences may affect the well-known regulation of GDH by nucleotides which is related to recent identification of thiamine derivatives as novel GDH modulators. The thiamine-dependent regulation of GDH is in good agreement with the fact that the non-coenzyme forms of thiamine, i.e., thiamine triphosphate and its adenylated form are generated in response to amino acid and carbon starvation.
Collapse
|
35
|
Zhang G, Li X, Sheng C, Chen X, Chen Y, Zhu D, Gao P. Macrophages activate iNOS signaling in adventitial fibroblasts and contribute to adventitia fibrosis. Nitric Oxide 2016; 61:20-28. [PMID: 27664590 DOI: 10.1016/j.niox.2016.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/20/2016] [Indexed: 12/31/2022]
Abstract
A large amount of NO is generated through the inducible nitric oxide synthase (iNOS) pathway from the vascular adventitia in various vascular diseases. However, it is currently not fully understood how the iNOS signaling pathway is activated. In the present study, this question was addressed in the context of adventitial cellular interactions. A rat model of acute hypertension in the contralateral carotid arteries was established through transverse aortic constriction (TAC) surgery. In this model, activated macrophages were found surrounded by a large quantity of iNOS-expressing adventitial fibroblasts (AFs), suggesting a possible causal relationship between macrophages and iNOS activation of the neighboring AFs. In an in vitro model, a macrophage-like cell line RAW 264.7 was first activated by LPS treatment. The supernatant was then harvested and applied to treat primary rat AFs. iNOS in AFs was activated robustly by the supernatant treatment but not by LPS itself. Treating AFs with interleukin-1β (IL-1β) also activated iNOS signaling, suggesting that the IL-1β pathway might be a possible mediator. As a consequence of the iNOS activation, total protein nitration and S-nitrosylation significantly increased in those AFs. Additionally, increased deposition of type I and type III collagens was observed in both in vitro and in vivo models. The collagen deposition was partially restored by an iNOS inhibitor, 1400 W. These findings highlight the importance of iNOS signaling during vascular inflammation, and advance our understanding of its activation through a cellular interaction perspective.
Collapse
Affiliation(s)
- Guannan Zhang
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Li
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chengyu Sheng
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohui Chen
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Chen
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dingliang Zhu
- Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pingjin Gao
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
36
|
Cardioprotective effects of Viscum album L. ssp. album (Loranthaceae) on isoproterenol-induced heart failure via regulation of the nitric oxide pathway in rats. Anatol J Cardiol 2016; 16:923-930. [PMID: 27443473 PMCID: PMC5324911 DOI: 10.14744/anatoljcardiol.2016.6780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Viscum album L. has favorable cardiovascular effects including antihypertensive and vasorelaxant activity, and the nitric oxide (NO) pathway upregulation has been proposed to be the underlying mechanism. NO also plays an important role in the pathophysiology of heart failure. However, its effects on cardiac systolic function are unclear. METHODS A total of 30 male Wistar albino rats at 12 weeks of age were randomly divided into three groups: control, isoproterenol-induced heart failure group (ISO), and isoproterenol-induced heart failure + V. album treatment group (VA) groups (n=10 in each group). V. album was orally given at a dose of 250 mg/kg/day by gavage. Parameters of heart failure were compared among the groups. Tamhane's T2 test, paired sample t-test, and Bonferroni methods were used for statistical analysis. RESULTS V. album resulted in an improvement in all parameters of heart failure including left ventricular diameters (6.34±0.23 mm, 6.98±0.35 mm, and 6.71±0.10 mm for left ventricular end-diastolic diameter in control, ISO, and VA groups, respectively, p<0.05), ejection fraction (73.3±3.1%, 56.7±2.6%, and 65.2±1.5% for control, ISO, and VA groups, respectively, p<0.05), serum NT-proBNP levels, and histopathological changes. V. album treatment resulted in a statistically significant attenuation of increased levels of NO and iNOS (p<0.0001). The levels of hs-CRP were also found to be lower in the VA group compared with the controls and ISO groups (p<0.01). CONCLUSION V. album exerted favorable effects on left ventricular function in isoproterenol-induced heart failure rats. Upregulation of the NO pathway seems to be the possible pathophysiological mechanism. Favorable vascular outcomes can also be speculated considering the reduction in serum hs-CRP levels.
Collapse
|
37
|
Omar SA, Webb AJ, Lundberg JO, Weitzberg E. Therapeutic effects of inorganic nitrate and nitrite in cardiovascular and metabolic diseases. J Intern Med 2016; 279:315-36. [PMID: 26522443 DOI: 10.1111/joim.12441] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nitric oxide (NO) is generated endogenously by NO synthases to regulate a number of physiological processes including cardiovascular and metabolic functions. A decrease in the production and bioavailability of NO is a hallmark of many major chronic diseases including hypertension, ischaemia-reperfusion injury, atherosclerosis and diabetes. This NO deficiency is mainly caused by dysfunctional NO synthases and increased scavenging of NO by the formation of reactive oxygen species. Inorganic nitrate and nitrite are emerging as substrates for in vivo NO synthase-independent formation of NO bioactivity. These anions are oxidation products of endogenous NO generation and are also present in the diet, with green leafy vegetables having a high nitrate content. The effects of nitrate and nitrite are diverse and include vasodilatation, improved endothelial function, enhanced mitochondrial efficiency and reduced generation of reactive oxygen species. Administration of nitrate or nitrite in animal models of cardiovascular disease shows promising results, and clinical trials are currently ongoing to investigate the therapeutic potential of nitrate and nitrite in hypertension, pulmonary hypertension, peripheral artery disease and myocardial infarction. In addition, the nutritional aspects of the nitrate-nitrite-NO pathway are interesting as diets suggested to protect against cardiovascular disease, such as the Mediterranean diet, are especially high in nitrate. Here, we discuss the potential therapeutic opportunities for nitrate and nitrite in prevention and treatment of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- S A Omar
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - A J Webb
- Cardiovascular Division, Department of Clinical Pharmacology, King's College London British Heart Foundation Centre, London, UK
| | - J O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - E Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
38
|
Nuriel T, Whitehouse J, Ma Y, Mercer EJ, Brown N, Gross SS. ANSID: A Solid-Phase Proteomic Approach for Identification and Relative Quantification of Aromatic Nitration Sites. Front Chem 2016; 3:70. [PMID: 26779476 PMCID: PMC4703760 DOI: 10.3389/fchem.2015.00070] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/11/2015] [Indexed: 12/21/2022] Open
Abstract
Nitration of tyrosine and other aromatic amino acid residues in proteins occurs in the setting of inflammatory, neurodegenerative, and cardiovascular diseases—importantly, this modification has been implicated in the pathogenesis of diverse diseases and the physiological process of aging. To understand the biological consequences of aromatic nitration in both health and disease, it is critical to molecularly identify the proteins that undergo nitration, specify their cognate modification sites and quantify their extent of nitration. To date, unbiased identification of nitrated proteins has often involved painstaking 2D-gel electrophoresis followed by Western Blotting with an anti-nitrotyrosine antibody for detection. Apart from being relatively slow and laborious, this method suffers from limited coverage, the potential for false-positive identifications, and failure to reveal specific amino acid modification sites. To overcome these shortcomings, we have developed a solid-phase, chemical-capture approach for unbiased and high-throughput discovery of nitrotyrosine and nitrotryptophan sites in proteins. Utilizing this method, we have successfully identified several endogenously nitrated proteins in rat brain and a total of 244 nitrated peptides from 145 proteins following in vitro exposure of rat brain homogenates to the nitrating agent peroxynitrite (1 mM). As expected, Tyr residues constituted the great majority of peroxynitrite-mediated protein nitration sites; however, we were surprised to discover several brain proteins that contain nitrated Trp residues. By incorporating a stable-isotope labeling step, this new Aromatic Nitration Site IDentification (ANSID) method was also adapted for relative quantification of nitration site abundances in proteins. Application of the ANSID method offers great potential to advance our understanding of the role of protein nitration in disease pathogenesis and normal physiology.
Collapse
Affiliation(s)
- Tal Nuriel
- Department of Pharmacology, Weill Cornell Medical CollegeNew York, NY, USA; Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical CollegeNew York, NY, USA
| | - Julia Whitehouse
- Department of Pharmacology, Weill Cornell Medical College New York, NY, USA
| | - Yuliang Ma
- Department of Pharmacology, Weill Cornell Medical College New York, NY, USA
| | - Emily J Mercer
- Department of Pharmacology, Weill Cornell Medical CollegeNew York, NY, USA; Department of Surgery, Weill Cornell Medical CollegeNew York, NY, USA
| | - Neil Brown
- Department of Pharmacology, Weill Cornell Medical College New York, NY, USA
| | - Steven S Gross
- Department of Pharmacology, Weill Cornell Medical College New York, NY, USA
| |
Collapse
|
39
|
Begara-Morales JC, Sánchez-Calvo B, Chaki M, Valderrama R, Mata-Pérez C, Padilla MN, Corpas FJ, Barroso JB. Antioxidant Systems are Regulated by Nitric Oxide-Mediated Post-translational Modifications (NO-PTMs). FRONTIERS IN PLANT SCIENCE 2016; 7:152. [PMID: 26909095 PMCID: PMC4754464 DOI: 10.3389/fpls.2016.00152] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/29/2016] [Indexed: 05/18/2023]
Abstract
Nitric oxide (NO) is a biological messenger that orchestrates a plethora of plant functions, mainly through post-translational modifications (PTMs) such as S-nitrosylation or tyrosine nitration. In plants, hundreds of proteins have been identified as potential targets of these NO-PTMs under physiological and stress conditions indicating the relevance of NO in plant-signaling mechanisms. Among these NO protein targets, there are different antioxidant enzymes involved in the control of reactive oxygen species (ROS), such as H2O2, which is also a signal molecule. This highlights the close relationship between ROS/NO signaling pathways. The major plant antioxidant enzymes, including catalase, superoxide dismutases (SODs) peroxiredoxins (Prx) and all the enzymatic components of the ascorbate-glutathione (Asa-GSH) cycle, have been shown to be modulated to different degrees by NO-PTMs. This mini-review will update the recent knowledge concerning the interaction of NO with these antioxidant enzymes, with a special focus on the components of the Asa-GSH cycle and their physiological relevance.
Collapse
Affiliation(s)
- Juan C. Begara-Morales
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of JaénJaén, Spain
| | - Beatriz Sánchez-Calvo
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of JaénJaén, Spain
| | - Mounira Chaki
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of JaénJaén, Spain
| | - Raquel Valderrama
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of JaénJaén, Spain
| | - Capilla Mata-Pérez
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of JaénJaén, Spain
| | - María N. Padilla
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of JaénJaén, Spain
| | - Francisco J. Corpas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cellular and Molecular Biology of Plants, Estación Experimental del Zaidín, Consejo Superior de Investigaciones CientíficasGranada, Spain
| | - Juan B. Barroso
- Group of Biochemistry and Cell Signaling in Nitric Oxide, Department of Experimental Biology, Center for Advanced Studies in Olive Grove and Olive Oils, Faculty of Experimental Sciences, University of JaénJaén, Spain
- *Correspondence: Juan B. Barroso,
| |
Collapse
|
40
|
Mikhed Y, Bruns K, Schildknecht S, Jörg M, Dib M, Oelze M, Lackner KJ, Münzel T, Ullrich V, Daiber A. Formation of 2-nitrophenol from salicylaldehyde as a suitable test for low peroxynitrite fluxes. Redox Biol 2015; 7:39-47. [PMID: 26629950 PMCID: PMC4683390 DOI: 10.1016/j.redox.2015.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 01/13/2023] Open
Abstract
There has been some dispute regarding reaction products formed at physiological peroxynitrite fluxes in the nanomolar range with phenolic molecules, when used to predict the behavior of protein-bound aromatic amino acids like tyrosine. Previous data showed that at nanomolar fluxes of peroxynitrite, nitration of these phenolic compounds was outcompeted by dimerization (e.g. biphenols or dityrosine). Using 3-morpholino sydnonimine (Sin-1), we created low fluxes of peroxynitrite in our reaction set-up to demonstrate that salicylaldehyde displays unique features in the detection of physiological fluxes of peroxynitrite, yielding detectable nitration but only minor dimerization products. By means of HPLC analysis and detection at 380 nm we could identify the expected nitration products 3- and 5-nitrosalicylaldehyde, but also novel nitrated products. Using mass spectrometry, we also identified 2-nitrophenol and a not fully characterized nitrated dimerization product. The formation of 2-nitrophenol could proceed either by primary generation of a phenoxy radical, followed by addition of the NO2-radical to the various resonance structures, or by addition of the peroxynitrite anion to the polarized carbonyl group with subsequent fragmentation of the adduct (as seen with carbon dioxide). Interestingly, we observed almost no 3- and 5-nitrosalicylic acid products and only minor dimerization reaction. Our results disagree with the previous general assumption that nitration of low molecular weight phenolic compounds is always outcompeted by dimerization at nanomolar peroxynitrite fluxes and highlight unique features of salicylaldehyde as a probe for physiological concentrations of peroxynitrite.
There are no specific probes for peroxynitrite formation in vivo. Salicylaldehyde reacts with peroxynitrite to form the product 2-nitrophenol. Only high/supraphysiological •NO or peroxidase/H2O2/NO2─ levels yield 2-nitrophenol. Salicylaldehyde is suitable for detection of nanomolar fluxes of peroxynitrite.
Collapse
Affiliation(s)
- Yuliya Mikhed
- Center of Cardiology, Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Kai Bruns
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Michael Jörg
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Mobin Dib
- Center of Cardiology, Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Oelze
- Center of Cardiology, Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Münzel
- Center of Cardiology, Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Volker Ullrich
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Andreas Daiber
- Center of Cardiology, Laboratory of Molecular Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
41
|
Li X, Tao RR, Hong LJ, Cheng J, Jiang Q, Lu YM, Liao MH, Ye WF, Lu NN, Han F, Hu YZ, Hu YH. Visualizing peroxynitrite fluxes in endothelial cells reveals the dynamic progression of brain vascular injury. J Am Chem Soc 2015; 137:12296-303. [PMID: 26352914 DOI: 10.1021/jacs.5b06865] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Accumulating evidence suggests that formation of peroxynitrite (ONOO(-)) in the cerebral vasculature contributes to the progression of ischemic damage, while the underlying molecular mechanisms remain elusive. To fully understand ONOO(-) biology, efficient tools that can realize the real-time tracing of endogenous ONOO(-) fluxes are indispensable. While a few ONOO(-) fluorescent probes have been reported, direct visualization of ONOO(-) fluxes in the cerebral vasculature of live mice remains a challenge. Herein, we present a fluorescent switch-on probe (NP3) for ONOO(-) imaging. NP3 exhibits good specificity, fast response, and high sensitivity toward ONOO(-) both in vitro and in vivo. Moreover, NP3 is two-photon excitable and readily blood-brain barrier penetrable. These desired photophysical and pharmacokinetic properties endow NP3 with the capability to monitor brain vascular ONOO(-) generation after injury with excellent temporal and spatial resolution. As a proof of concept, NP3 has enabled the direct visualization of neurovascular ONOO(-) formation in ischemia progression in live mouse brain by use of two-photon laser scanning microscopy. Due to these favorable properties, NP3 holds great promise for visualizing endogenous peroxynitrite fluxes in a variety of pathophysiological progressions in vitro and in vivo.
Collapse
Affiliation(s)
- Xin Li
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Rong-Rong Tao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Ling-Juan Hong
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Juan Cheng
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Quan Jiang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Ying-Mei Lu
- School of Medicine, Zhejiang University City College , Hangzhou 310015, China
| | - Mei-Hua Liao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Wei-Feng Ye
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Nan-Nan Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Feng Han
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - Yong-Zhou Hu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| | - You-Hong Hu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou 310058, China
| |
Collapse
|
42
|
Kim CS, Park S, Chun Y, Song W, Kim HJ, Kim J. Treadmill Exercise Attenuates Retinal Oxidative Stress in Naturally-Aged Mice: An Immunohistochemical Study. Int J Mol Sci 2015; 16:21008-20. [PMID: 26404251 PMCID: PMC4613238 DOI: 10.3390/ijms160921008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 08/21/2015] [Accepted: 08/26/2015] [Indexed: 01/30/2023] Open
Abstract
In the retina, a number of degenerative diseases, including glaucoma, diabetic retinopathy, and age-related macular degeneration, may occur as a result of aging. Oxidative damage is believed to contribute to the pathogenesis of aging as well as to age-related retinal disease. Although physiological exercise has been shown to reduce oxidative stress in rats and mice, it is not known whether it has a similar effect in retinal tissues. The aim of this study was to evaluate retinal oxidative stress in naturally-aged mice. In addition, we evaluated the effects of aerobic training on retinal oxidative stress by immunohistochemically evaluating oxidative stress markers. A group of twelve-week-old male mice were not exercised (young control). Two groups of twenty-two-month-old male mice were created: an old control group and a treadmill exercise group. The old control group mice were not exercised. The treadmill exercise group mice ran on a treadmill (5 to 12 m/min, 30 to 60 min/day, 3 days/week for 12 weeks). The retinal thickness and number of cells in the ganglion cell layer of the naturally-aged mice were reduced compared to those in the young control mice. However, treadmill exercise reversed these morphological changes in the retinas. We evaluated retinal expression of carboxymethyllysine (CML), 8-hydroxy-2'-deoxyguanosine (8-OHdG) and nitrotyrosine. The retinas from the aged mice showed increased CML, 8-OHdG, and nitrotyrosine immunostaining intensities compared to young control mice. The exercise group exhibited significantly lower CML levels and nitro-oxidative stress than the old control group. These results suggest that regular exercise can reduce retinal oxidative stress and that physiological exercise may be distinctly advantageous in reducing retinal oxidative stress.
Collapse
Affiliation(s)
- Chan-Sik Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.
| | - Sok Park
- Department of Sports and Health Management, Mokwon University, Daejeon 35349, Korea.
| | - Yoonseok Chun
- Sports Wellness Center, Yong In University, Gyeonggi-do 17092, Korea.
| | - Wook Song
- Health and Exercise Science Laboratory, Seoul National University, Seoul 08826, Korea.
| | - Hee-Jae Kim
- Health and Exercise Science Laboratory, Seoul National University, Seoul 08826, Korea.
| | - Junghyun Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.
| |
Collapse
|
43
|
Chaki M, Álvarez de Morales P, Ruiz C, Begara-Morales JC, Barroso JB, Corpas FJ, Palma JM. Ripening of pepper (Capsicum annuum) fruit is characterized by an enhancement of protein tyrosine nitration. ANNALS OF BOTANY 2015; 116:637-47. [PMID: 25814060 PMCID: PMC4577987 DOI: 10.1093/aob/mcv016] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/12/2014] [Accepted: 01/05/2015] [Indexed: 05/18/2023]
Abstract
BACKGROUND AND AIMS Pepper (Capsicum annuum, Solanaceae) fruits are consumed worldwide and are of great economic importance. In most species ripening is characterized by important visual and metabolic changes, the latter including emission of volatile organic compounds associated with respiration, destruction of chlorophylls, synthesis of new pigments (red/yellow carotenoids plus xanthophylls and anthocyanins), formation of pectins and protein synthesis. The involvement of nitric oxide (NO) in fruit ripening has been established, but more work is needed to detail the metabolic networks involving NO and other reactive nitrogen species (RNS) in the process. It has been reported that RNS can mediate post-translational modifications of proteins, which can modulate physiological processes through mechanisms of cellular signalling. This study therefore examined the potential role of NO in nitration of tyrosine during the ripening of California sweet pepper. METHODS The NO content of green and red pepper fruit was determined spectrofluorometrically. Fruits at the breaking point between green and red coloration were incubated in the presence of NO for 1 h and then left to ripen for 3 d. Profiles of nitrated proteins were determined using an antibody against nitro-tyrosine (NO2-Tyr), and profiles of nitrosothiols were determined by confocal laser scanning microscopy. Nitrated proteins were identified by 2-D electrophoresis and MALDI-TOF/TOF analysis. KEY RESULTS Treatment with NO delayed the ripening of fruit. An enhancement of nitrosothiols and nitroproteins was observed in fruit during ripening, and this was reversed by the addition of exogenous NO gas. Six nitrated proteins were identified and were characterized as being involved in redox, protein, carbohydrate and oxidative metabolism, and in glutamate biosynthesis. Catalase was the most abundant nitrated protein found in both green and red fruit. CONCLUSIONS The RNS profile reported here indicates that ripening of pepper fruit is characterized by an enhancement of S-nitrosothiols and protein tyrosine nitration. The nitrated proteins identified have important functions in photosynthesis, generation of NADPH, proteolysis, amino acid biosynthesis and oxidative metabolism. The decrease of catalase in red fruit implies a lower capacity to scavenge H2O2, which would promote lipid peroxidation, as has already been reported in ripe pepper fruit.
Collapse
Affiliation(s)
- Mounira Chaki
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Apartado 419, 18008 Granada, Spain and
| | - Paz Álvarez de Morales
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Apartado 419, 18008 Granada, Spain and
| | - Carmelo Ruiz
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Apartado 419, 18008 Granada, Spain and
| | - Juan C Begara-Morales
- Group of Biochemistry and Cell Signaling in Nitric Oxide. Department of Biochemistry and Molecular Biology, University of Jaén, 23071 Jaén, Spain
| | - Juan B Barroso
- Group of Biochemistry and Cell Signaling in Nitric Oxide. Department of Biochemistry and Molecular Biology, University of Jaén, 23071 Jaén, Spain
| | - Francisco J Corpas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Apartado 419, 18008 Granada, Spain and
| | - José M Palma
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Biochemistry, Cell and Molecular Biology of Plants, Estación Experimental del Zaidín, CSIC, Apartado 419, 18008 Granada, Spain and
| |
Collapse
|
44
|
Inducible NO synthase is constitutively expressed in porcine myocardium and its level decreases along with tachycardia-induced heart failure. Cardiovasc Pathol 2015; 25:3-11. [PMID: 26361649 DOI: 10.1016/j.carpath.2015.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 07/24/2015] [Accepted: 08/09/2015] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The adverse effects of oxidative stress and the presence of proinflammatory factors in the heart have been widely demonstrated mainly on rodent models. However, larger clinical trials focusing on inflammation or oxidative stress in heart failure (HF) have not been carried out. This may be due to differences in the anatomy and physiology of the cardiovascular system between small rodents and large mammals. Thus, we investigated myocardial inflammatory factors, such as inducible NO synthase (iNOS) and oxidative stress indices in female pigs with chronic tachycardia-induced cardiomyopathy. METHODS Homogenous female siblings of Large White breed swine (n=15) underwent continuous right ventricular (RV) pacing at 170bpm, whereas five sham-operated subjects served as controls. In the course of RV pacing, animals developed a clinical picture of HF and were euthanized at subsequent stages of the disease: mild, moderate and severe HF. Left ventricle (LV) sections were examined with electron microscopy. The relative expression of iNOS in LV was determined by quantitative PCR. The protein level of iNOS was determined by Western blotting and immunohistochemistry. The level of the S-nitrosylated (S-NO) protein in LV was determined after S-NO moieties were substituted by biotin, followed by a colorimetrical detection with streptavidin. Malondialdehyde (MDA), a marker of lipid peroxidation, was evaluated in the LV and serum using thiobarbituric acid. The aconitase activity (based on measurement of the concomitant formation of NADPH from NADP(+)), a marker of oxidative stress, was analyzed in mitochondrial and cytosolic LV fractions. The concentration of interleukin-1β (IL-1β) was measured in LV homogenates using enzyme-linked immunosorbent assay. RESULTS RV pacing resulted in an impairment of LV systolic function, LV dilatation and neurohormonal activation. The electron microscopy revealed abnormalities within the cardiomyocytes of failing hearts, i.e. swollen mitochondria and myofibril derangement. iNOS was expressed in the control LV myocardium. The development of HF was accompanied by a decrease in iNOS mRNA (P<.05), which was also reflected at a protein level, and a decrease in the protein S-nitrosylation (P<.05). Both iNOS mRNA and S-NO relative moiety levels were inversely related to the dilatation of the LV (P<.05). There was no difference in the concentration of MDA in the LV and serum. Similarly, no differences in the concentration of IL-1β LV were found between diseased and healthy animals. Aconitase activity was decreased only in the LV mitochondrial fraction of pigs with severe HF. CONCLUSIONS iNOS was shown to be constitutively expressed within porcine LV. Its level decreases during the progression of systolic nonischemic HF in the pig model. Thus, it can be assumed that an up-regulation of proinflammatory factors is not involved in porcine tachycardia-induced cardiomyopathy and that the impact of oxidative stress may be restricted to the mitochondria in this HF model.
Collapse
|
45
|
Lundberg JO, Gladwin MT, Weitzberg E. Strategies to increase nitric oxide signalling in cardiovascular disease. Nat Rev Drug Discov 2015; 14:623-41. [PMID: 26265312 DOI: 10.1038/nrd4623] [Citation(s) in RCA: 416] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is a key signalling molecule in the cardiovascular, immune and central nervous systems, and crucial steps in the regulation of NO bioavailability in health and disease are well characterized. Although early approaches to therapeutically modulate NO bioavailability failed in clinical trials, an enhanced understanding of fundamental subcellular signalling has enabled a range of novel therapeutic approaches to be identified. These include the identification of: new pathways for enhancing NO synthase activity; ways to amplify the nitrate-nitrite-NO pathway; novel classes of NO-donating drugs; drugs that limit NO metabolism through effects on reactive oxygen species; and ways to modulate downstream phosphodiesterases and soluble guanylyl cyclases. In this Review, we discuss these latest developments, with a focus on cardiovascular disease.
Collapse
Affiliation(s)
- Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Mark T Gladwin
- Vascular Medicine Institute, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pennsylvania 15213, USA
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| |
Collapse
|
46
|
Penna C, Angotti C, Pagliaro P. Protein S-nitrosylation in preconditioning and postconditioning. Exp Biol Med (Maywood) 2015; 239:647-62. [PMID: 24668550 DOI: 10.1177/1535370214522935] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The coronary artery disease is a leading cause of death and morbidity worldwide. This disease has a complex pathophysiology that includes multiple mechanisms. Among these is the oxidative/nitrosative stress. Paradoxically, oxidative/nitrosative signaling plays a major role in cardioprotection against ischemia/reperfusion injury. In this context, the gas transmitter nitric oxide may act through several mechanisms, such as guanylyl cyclase activation and via S-nitrosylation of proteins. The latter is a covalent modification of a protein cysteine thiol by a nitric oxide-group that generates an S-nitrosothiol. Here, we report data showing that nitric oxide and S-nitrosylation of proteins play a pivotal role not only in preconditioning but also in postconditioning cardioprotection.
Collapse
|
47
|
Münzel T, Gori T, Keaney JF, Maack C, Daiber A. Pathophysiological role of oxidative stress in systolic and diastolic heart failure and its therapeutic implications. Eur Heart J 2015; 36:2555-64. [PMID: 26142467 DOI: 10.1093/eurheartj/ehv305] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/15/2015] [Indexed: 02/07/2023] Open
Abstract
Systolic and diastolic myocardial dysfunction has been demonstrated to be associated with an activation of the circulating and local renin-angiotensin-aldosterone system (RAAS), and with a subsequent inappropriately increased production of reactive oxygen species (ROS). While, at low concentrations, ROS modulate important physiological functions through changes in cellular signalling and gene expression, overproduction of ROS may adversely alter cardiac mechanics, leading to further worsening of systolic and diastolic function. In addition, vascular endothelial dysfunction due to uncoupling of the nitric oxide synthase, activation of vascular and phagocytic membrane oxidases or mitochondrial oxidative stress may lead to increased vascular stiffness, further compromising cardiac performance in afterload-dependent hearts. In the present review, we address the potential role of ROS in the pathophysiology of myocardial and vascular dysfunction in heart failure (HF) and their therapeutic targeting. We discuss possible mechanisms underlying the failure of antioxidant vitamins in improving patients' prognosis, the impact of angiotensin-converting enzyme inhibitors or AT1 receptor blockers on oxidative stress, and the mechanism of the benefit of combination of hydralazine/isosorbide dinitrate. Further, we provide evidence supporting the existence of differences in the pathophysiology of HF with preserved vs. reduced ejection fraction and whether targeting mitochondrial ROS might be a particularly interesting therapeutic option for patients with preserved ejection fraction.
Collapse
Affiliation(s)
- Thomas Münzel
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Tommaso Gori
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - John F Keaney
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Christoph Maack
- Klinik für Innere Medizin III Universitätsklinikum des Saarlandes, Homburg/Saar, Germany
| | - Andreas Daiber
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Langenbeckstrasse 1, Mainz 55131, Germany
| |
Collapse
|
48
|
Label-free in-situ monitoring of protein tyrosine nitration in blood by surface-enhanced Raman spectroscopy. Biosens Bioelectron 2015; 69:1-7. [DOI: 10.1016/j.bios.2015.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/26/2014] [Accepted: 01/02/2015] [Indexed: 12/26/2022]
|
49
|
Bults P, van de Merbel NC, Bischoff R. Quantification of biopharmaceuticals and biomarkers in complex biological matrices: a comparison of liquid chromatography coupled to tandem mass spectrometry and ligand binding assays. Expert Rev Proteomics 2015; 12:355-74. [DOI: 10.1586/14789450.2015.1050384] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
50
|
Szuba A, Kasprowicz-Maluśki A, Wojtaszek P. Nitration of plant apoplastic proteins from cell suspension cultures. J Proteomics 2015; 120:158-68. [PMID: 25805245 DOI: 10.1016/j.jprot.2015.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/20/2015] [Accepted: 03/03/2015] [Indexed: 12/27/2022]
Abstract
Nitric oxide causes numerous protein modifications including nitration of tyrosine residues. This modification, though one of the greatest biological importance, is poorly recognized in plants and is usually associated with stress conditions. In this study we analyzed nitrotyrosines from suspension cultures of Arabidopsis thaliana and Nicotiana tabacum, treated with NO modulators and exposed to osmotic stress, as well as of BY2 cells long-term adapted to osmotic stress conditions. Using confocal microscopy, we showed that the cell wall area is one of the compartments most enriched in nitrotyrosines within a plant cell. Subsequently, we analyzed nitration of ionically-bound cell-wall proteins and identified selected proteins with MALDI-TOF spectrometry. Proteomic analysis indicated that there was no significant increase in the amount of nitrated proteins under the influence of NO modulators, among them 3-morpholinosydnonimine (SIN-1), considered a donor of nitrating agent, peroxynitrite. Moreover, osmotic stress conditions did not increase the level of nitration in cell wall proteins isolated from suspension cells, and in cultures long-term adapted to stress conditions; that level was even reduced in comparison with control samples. Among identified nitrotyrosine-containing proteins dominated the ones associated with carbon circulation as well as the numerous proteins responding to stress conditions, mainly peroxidases. BIOLOGICAL SIGNIFICANCE High concentrations of nitric oxide found in the cell wall and the ability to produce large amounts of ROS make the apoplast a site highly enriched in nitrotyrosines, as presented in this paper. Analysis of ionically bound fraction of the cell wall proteins indicating generally unchanged amounts of nitrotyrosines under influence of NO modulators and osmotic stress, is noticeably different from literature data concerning, however, the total plant proteins analysis. This observation is supplemented by further nitroproteome analysis, for cells long-term adapted to stressful conditions, and results showing that such conditions did not always cause an increase in nitrotyrosine content. These findings may be interpreted as characteristic features of apoplastic protein nitration.
Collapse
Affiliation(s)
- Agnieszka Szuba
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznań, Poland; Institute of Dendrology, Polish Academy of Sciences, Parkowa 5, 62-035 Kórnik Poland.
| | - Anna Kasprowicz-Maluśki
- Department of Molecular and Cellular Biology, Adam Mickiewicz University, Umultowska 89, 61-613 Poznań, Poland
| | - Przemysław Wojtaszek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznań, Poland; Department of Molecular and Cellular Biology, Adam Mickiewicz University, Umultowska 89, 61-613 Poznań, Poland
| |
Collapse
|