51
|
Meat consumption in midlife and risk of cognitive impairment in old age: the Singapore Chinese Health Study. Eur J Nutr 2019; 59:1729-1738. [DOI: 10.1007/s00394-019-02031-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 06/15/2019] [Indexed: 12/23/2022]
|
52
|
Inhaled nitric oxide prevents systemic and pulmonary vasoconstriction due to hemoglobin-based oxygen carrier infusion: A case report. J Crit Care 2019; 51:213-216. [PMID: 30709560 PMCID: PMC10150649 DOI: 10.1016/j.jcrc.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/02/2018] [Accepted: 04/12/2018] [Indexed: 11/24/2022]
Abstract
Hemoglobin-based oxygen carriers (HBOCs) are used in extreme circumstances to increase hemoglobin concentration and improve oxygen delivery when allogenic red blood cell transfusions are contraindicated or not immediately available. However, HBOC-induced severe pulmonary and systemic vasoconstriction due to peripheral nitric oxide (NO) scavenging has stalled its implementation in clinical practice. We present a case of an 87 year-old patient with acute life-threatening anemia who received HBOC while breathing NO gas. This case shows that inhaled NO allows for the safe use of HBOC infusion by preventing HBOC-induced pulmonary and systemic vasoconstriction.
Collapse
|
53
|
Kosmachevskaya OV, Nasybullina EI, Blindar VN, Topunov AF. Binding of Erythrocyte Hemoglobin to the Membrane to Realize Signal-Regulatory Function (Review). APPL BIOCHEM MICRO+ 2019. [DOI: 10.1134/s0003683819020091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
54
|
Abstract
OBJECTIVE This research was aimed to investigate the correct dose of nitrite that would act as a protection against the ischemic effects induced by acute myocardial infarction (AMI). METHODS Mice were randomly divided into a sham-operation group (sham), an AMI operation group (AMI), and a nitrite pretreatment+AMI operation group (N+AMI). Seven days before the AMI operation, mice in the N+AMI group were pretreated with sodium nitrite in drinking water. RESULTS One week after the AMI operation, serum lactate dehydrogenase (LDH) and creatine kinase (CK) activities in both AMI and N+AMI group were significantly higher than those in the sham group, but there were no significant differences between AMI and N+AMI mice. Contents of inducible nitric oxide synthase (iNOS) in the noninfarct area of the left ventricle in the N+AMI mice were significantly higher than those in the AMI mice, with no difference in the infarct area. Coagulation necrosis in the cardiomyocytes was observed in both AMI and N+AMI mice; however, it was less severe in the N+AMI mice. Western blot analyses showed that nitrite pretreatment resulted in up-regulation of antiapoptotic factors Bcl-2 and p21waf1/cip1 signal proteins, but down-regulation of the proapoptotic factor Bax signal protein. Furthermore, nitrite pretreatment also showed significant alleviation of AMI-induced signal protein expressions of inflammatory factors of NF-K B and oxidative factors of Hsp 70 and HO-1. CONCLUSION These results suggest that nitrite show certain protective effects against the ischemic effects induced by AMI in mice, which might be attributed to the synthesis of NO induced by iNOS through up-regulation of antiapoptotic factors and down-regulation of proapoptotic and inflammatory factors.
Collapse
|
55
|
Kosmachevskaya OV, Topunov AF. Alternate and Additional Functions of Erythrocyte Hemoglobin. BIOCHEMISTRY (MOSCOW) 2019; 83:1575-1593. [PMID: 30878032 DOI: 10.1134/s0006297918120155] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The review discusses pleiotropic effects of erythrocytic hemoglobin (Hb) and their significance for human health. Hemoglobin is mostly known as an oxygen carrier, but its biochemical functions are not limited to this. The following aspects of Hb functioning are examined: (i) catalytic functions of the heme component (nitrite reductase, NO dioxygenase, monooxygenase, alkylhydroperoxidase) and of the apoprotein (esterase, lipoxygenase); (ii) participation in nitric oxide metabolism; (iii) formation of membrane-bound Hb and its role in the regulation of erythrocyte metabolism; (iv) physiological functions of Hb catabolic products (iron, CO, bilirubin, peptides). Special attention is given to Hb participation in signal transduction in erythrocytes. The relationships between various erythrocyte metabolic parameters, such as oxygen status, ATP formation, pH regulation, redox balance, and state of the cytoskeleton are discussed with regard to Hb. Hb polyfunctionality can be considered as a manifestation of the principle of biochemical economy.
Collapse
Affiliation(s)
- O V Kosmachevskaya
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - A F Topunov
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia.
| |
Collapse
|
56
|
Lim YJ, Foo TC, Yeung AWS, Tu X, Ma Y, Hawkins CL, Witting PK, Jameson GNL, Terentis AC, Thomas SR. Human Indoleamine 2,3-Dioxygenase 1 Is an Efficient Mammalian Nitrite Reductase. Biochemistry 2019; 58:974-986. [PMID: 30585477 DOI: 10.1021/acs.biochem.8b01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The heme enzyme indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the first reaction of l-tryptophan oxidation along the kynurenine pathway. IDO1 is a central immunoregulatory enzyme with important implications for inflammation, infectious disease, autoimmune disorders, and cancer. Here we demonstrate that IDO1 is a mammalian nitrite reductase capable of chemically reducing nitrite to nitric oxide (NO) under hypoxia. Ultraviolet-visible absorption and resonance Raman spectroscopy showed that incubation of dithionite-reduced, ferrous-IDO1 protein (FeII-IDO1) with nitrite under anaerobic conditions resulted in the time-dependent formation of an FeII-nitrosyl IDO1 species, which was inhibited by substrate l-tryptophan, dependent on the concentration of nitrite or IDO1, and independent of the concentration of the reductant, dithionite. The bimolecular rate constant for IDO1 nitrite reductase activity was determined as 5.4 M-1 s-1 (pH 7.4, 23 °C), which was comparable to that measured for myoglobin (3.6 M-1 s-1; pH 7.4, 23 °C), an efficient and biologically important mammalian heme-based nitrite reductase. IDO1 nitrite reductase activity was pH-dependent but differed with myoglobin in that it showed a reduced proton dependency at pH >7. Electron paramagnetic resonance studies measuring NO production showed that the conventional IDO1 dioxygenase reducing cofactors, ascorbate and methylene blue, enhanced IDO1's nitrite reductase activity and the time- and IDO1 concentration-dependent release of NO in a manner inhibited by l-tryptophan or the IDO inhibitor 1-methyl-l-tryptophan. These data identify IDO1 as an efficient mammalian nitrite reductase that is capable of generating NO under anaerobic conditions. IDO1's nitrite reductase activity may have important implications for the enzyme's biological actions when expressed within hypoxic tissues.
Collapse
Affiliation(s)
| | - Timothy C Foo
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | | | | | | - Clare L Hawkins
- Department of Biomedical Sciences , University of Copenhagen , Copenhagen N DK-2200 , Denmark
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Guy N L Jameson
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , VIC 3010 , Australia
| | - Andrew C Terentis
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | |
Collapse
|
57
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 PMCID: PMC6442925 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/30/2018] [Accepted: 05/06/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
58
|
Sierra-Campos E, Valdez-Solana MA, Campos-Almazan MI. Nitrate and nitrite in drinking water affect antioxidant enzymes in erythrocytes of rats. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.04.090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
59
|
Zhao Y, Wang X, Noviana M, Hou M. Nitric oxide in red blood cell adaptation to hypoxia. Acta Biochim Biophys Sin (Shanghai) 2018; 50:621-634. [PMID: 29860301 DOI: 10.1093/abbs/gmy055] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Indexed: 12/28/2022] Open
Abstract
Nitric oxide (NO) appears to be involved in virtually every aspect of cardiovascular biology. Most attention has been focused on the role of endothelial-derived NO in basal blood flow regulation by relaxing vascular smooth muscle; however, it is now known that NO derived from red blood cells (RBCs) plays a fundamental role in vascular homeostasis by enhancing oxygen (O2) release at the cellular and physiological level. Hypoxia is an often seen problem in diverse conditions; systemic adaptations to hypoxia permit people to adjust to the hypoxic environment at high altitudes and to disease processes. In addition to the cardiopulmonary and hematologic adaptations that support systemic O2 delivery in hypoxia, RBCs assist through newly described NO-based mechanisms, in line with their vital role in O2 transport and delivery. Furthermore, to increase the local blood flow in proportion to metabolic demand, NO regulates membrane mechanical properties thereby modulating RBC deformability and O2 carrying-releasing function. In this review article, we focus on the effect of NO bioactivity on RBC-based mechanisms that regulate blood flow and RBC deformability. RBC adaptations to hypoxia are summarized, with particular attention to NO-dependent S-nitrosylation of membrane proteins and hemoglobin (S-nitrosohemoglobin). The NO/S-nitrosylation/RBC vasoregulatory cascade contributes fundamentally to the molecular understanding of the role of NO in human adaptation to hypoxia and may inform novel therapeutic strategies.
Collapse
Affiliation(s)
- Yajin Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Xiang Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Milody Noviana
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Man Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
60
|
Results, meta-analysis and a first evaluation of U NOxR, the urinary nitrate-to-nitrite molar ratio, as a measure of nitrite reabsorption in experimental and clinical settings. Amino Acids 2018; 50:799-821. [PMID: 29728915 DOI: 10.1007/s00726-018-2573-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 04/19/2018] [Indexed: 02/07/2023]
Abstract
We recently found that renal carbonic anhydrase (CA) is involved in the reabsorption of inorganic nitrite (NO2-), an abundant reservoir of nitric oxide (NO) in tissues and cells. Impaired NO synthesis in the endothelium and decreased NO bioavailability in the circulation are considered major contributors to the development and progression of renal and cardiovascular diseases in different conditions including diabetes. Isolated human and bovine erythrocytic CAII and CAIV can convert nitrite to nitrous acid (HONO) and its anhydride N2O3 which, in the presence of thiols (RSH), are further converted to S-nitrosothiols (RSNO) and NO. Thus, CA may be responsible both for the homeostasis of nitrite and for its bioactivation to RSNO/NO. We hypothesized that enhanced excretion of nitrite in the urine may contribute to NO-related dysfunctions in the renal and cardiovascular systems, and proposed the urinary nitrate-to-nitrite molar ratio, i.e., UNOxR, as a measure of renal CA-dependent excretion of nitrite. Based on results from clinical and experimental animal studies, here, we report on a first evaluation of UNOxR. We determined UNOxR values in preterm neonates, healthy children, and adults, in children suffering from type 1 diabetes mellitus (T1DM) or Duchenne muscular dystrophy (DMD), in elderly subjects suffering from chronic rheumatic diseases, type 2 diabetes mellitus (T2DM), coronary artery disease (CAD), or peripheral arterial occlusive disease (PAOD). We also determined UNOxR values in healthy young men who ingested isosorbide dinitrate (ISDN), pentaerythrityl tetranitrate (PETN), or inorganic nitrate. In addition, we tested the utility of UNOxR in two animal models, i.e., the LEW.1AR1-iddm rat, an animal model of human T1DM, and the APOE*3-Leiden.CETP mice, a model of human dyslipidemia. Mean UNOxR values were lower in adult patients with rheumatic diseases (187) and in T2DM patients of the DALI study (74) as compared to healthy elderly adults (660) and healthy young men (1500). The intra- and inter-variabilities of UNOxR were of the order of 50% in young and elderly healthy subjects. UNOxR values were lower in black compared to white boys (314 vs. 483, P = 0.007), which is in line with reported lower NO bioavailability in black ethnicity. Mean UNOxR values were lower in DMD (424) compared to healthy (730) children, but they were higher in T1DM children (1192). ISDN (3 × 30 mg) decreased stronger UNOxR compared to PETN (3 × 80 mg) after 1 day (P = 0.046) and after 5 days (P = 0.0016) of oral administration of therapeutically equivalent doses. In healthy young men who ingested NaNO3 (0.1 mmol/kg/d), UNOxR was higher than in those who ingested the same dose of NaCl (1709 vs. 369). In LEW.1AR1-iddm rats, mean UNOxR values were lower than in healthy rats (198 vs. 308) and comparable to those in APOE*3-Leiden.CETP mice (151).
Collapse
|
61
|
Imbrogno S, Filice M, Cerra MC, Gattuso A. NO, CO and H 2 S: What about gasotransmitters in fish and amphibian heart? Acta Physiol (Oxf) 2018; 223:e13035. [PMID: 29338122 DOI: 10.1111/apha.13035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 12/25/2022]
Abstract
The gasotransmitters nitric oxide (NO), carbon monoxide (CO), and hydrogen sulphide (H2 S), long considered only toxicant, are produced in vivo during the catabolism of common biological molecules and are crucial for a large variety of physiological processes. Mounting evidence is emerging that in poikilotherm vertebrates, as in mammals, they modulate the basal performance of the heart and the response to stress challenges. In this review, we will focus on teleost fish and amphibians to highlight the evolutionary importance in vertebrates of the cardiac control elicited by NO, CO and H2 S, and the conservation of the intracellular cascades they activate. Although many gaps are still present due to discontinuous information, we will use examples obtained by studies from our and other laboratories to illustrate the complexity of the mechanisms that, by involving gasotransmitters, allow beat-to-beat, short-, medium- and long-term cardiac homoeostasis. By presenting the latest data, we will also provide a framework in which the peculiar morpho-functional arrangement of the teleost and amphibian heart can be considered as a reference tool to decipher cardiac regulatory networks which are difficult to explore using more conventional vertebrates, such as mammals.
Collapse
Affiliation(s)
- S. Imbrogno
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - M. Filice
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - M. C. Cerra
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - A. Gattuso
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| |
Collapse
|
62
|
Gangwar A, Paul S, Ahmad Y, Bhargava K. Competing trends of ROS and RNS-mediated protein modifications during hypoxia as an alternate mechanism of NO benefits. Biochimie 2018; 148:127-138. [PMID: 29571702 DOI: 10.1016/j.biochi.2018.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/16/2018] [Indexed: 01/01/2023]
Abstract
Hypoxia, especially altitude associated hypoxia is known to cause severe physiological alterations and life-threatening conditions. Impaired redox balance along with oxidative stress, protein carbonylation and instigation of apoptotic events are common sub-cellular events that follow the hypoxic insult. The role of nitric oxide (NO) is very dynamic and versatile in preventing the ill effects of hypoxia vis-a-vis reacting with oxidative species and causing protein nitrosylation. Although several mechanisms of NO-mediated cytoprotection are known during hypoxic insult, limited pieces of evidence are available to support the relationship between two downstream events of oxidative stress, protein carbonylation (caused by carbonyl; CO radical) and protein nitrosylation/nitration (caused by NO/peroxynitrite; ONOO radical). In this study, we investigated an entirely new aspect of NO protection in hypoxia involving crosstalk between carbonylation and nitrosylation. Using standard NO inhibitor l-NAME and simulated hypoxic conditions in hypoxia-sensitive cell line H9c2, we evaluated the levels of radicals, cell death, mitochondrial membrane potential, levels of protein nitrosylation, protein nitration and carbonylation and glutathione content. The results were then carefully analyzed in light of NO bioavailability. Our study shows that reducing NO during hypoxia caused cell death via the increased degree of carbonylation in proteins. This provides a new aspect of NO benefits which furthers opens new possibilities to explore potential mechanisms and effects of cross-talk between nitrosylation, protein nitration and carbonylation, especially through some common antioxidant mediators such as glutathione and thioredoxin.
Collapse
Affiliation(s)
- Anamika Gangwar
- Peptide and Proteomics Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Subhojit Paul
- Peptide and Proteomics Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Yasmin Ahmad
- Peptide and Proteomics Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Kalpana Bhargava
- Peptide and Proteomics Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
63
|
Münzel T, Daiber A. Inorganic nitrite and nitrate in cardiovascular therapy: A better alternative to organic nitrates as nitric oxide donors? Vascul Pharmacol 2018; 102:1-10. [DOI: 10.1016/j.vph.2017.11.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/11/2017] [Accepted: 11/12/2017] [Indexed: 01/08/2023]
|
64
|
Liu T, Zhang M, Terry MH, Schroeder H, Wilson SM, Power GG, Li Q, Tipple TE, Borchardt D, Blood AB. Nitrite potentiates the vasodilatory signaling of S-nitrosothiols. Nitric Oxide 2018; 75:60-69. [PMID: 29428841 DOI: 10.1016/j.niox.2018.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 12/27/2022]
Abstract
Nitrite and S-nitrosothiols (SNOs) are both byproducts of nitric oxide (NO) metabolism and are proposed to cause vasodilation via activation of soluble guanylate cyclase (sGC). We have previously reported that while SNOs are potent vasodilators at physiological concentrations, nitrite itself only produces vasodilation at supraphysiological concentrations. Here, we tested the hypothesis that sub-vasoactive concentrations of nitrite potentiate the vasodilatory effects of SNOs. Multiple exposures of isolated sheep arteries to S-nitroso-glutathione (GSNO) resulted in a tachyphylactic decreased vasodilatory response to GSNO but not to NO, suggesting attenuation of signaling steps upstream from sGC. Exposure of arteries to 1 μM nitrite potentiated the vasodilatory effects of GSNO in naive arteries and abrogated the tachyphylactic response to GSNO in pre-exposed arteries, suggesting that nitrite facilitates GSNO-mediated activation of sGC. In intact anesthetized sheep and rats, inhibition of NO synthases to decrease plasma nitrite levels attenuated vasodilatory responses to exogenous infusions of GSNO, an effect that was reversed by exogenous infusion of nitrite at sub-vasodilating levels. This study suggests nitrite potentiates SNO-mediated vasodilation via a mechanism that lies upstream from activation of sGC.
Collapse
Affiliation(s)
- Taiming Liu
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Meijuan Zhang
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Michael H Terry
- Department of Respiratory Care, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Hobe Schroeder
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Sean M Wilson
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Gordon G Power
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Qian Li
- Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Trent E Tipple
- Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Dan Borchardt
- Department of Chemistry, University of California, Riverside, CA 92521, United States
| | - Arlin B Blood
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States; Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States.
| |
Collapse
|
65
|
Hathazi D, Scurtu F, Bischin C, Mot A, Attia AAA, Kongsted J, Silaghi-Dumitrescu R. The Reaction of Oxy Hemoglobin with Nitrite: Mechanism, Antioxidant-Modulated Effect, and Implications for Blood Substitute Evaluation. Molecules 2018; 23:molecules23020350. [PMID: 29414908 PMCID: PMC6017026 DOI: 10.3390/molecules23020350] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/27/2022] Open
Abstract
The autocatalytic reaction between nitrite and the oxy form of globins involves free radicals. For myoglobin (Mb), an initial binding of nitrite to the iron-coordinated oxygen molecule was proposed; the resulting ferrous-peroxynitrate species was not detected, but its decay product, the high-valent ferryl form, was demonstrated in stopped-flow experiments. Reported here are the stopped flow spectra recorded upon mixing oxy Hb (native, as well as chemically-derivatized in the form of several candidates of blood substitutes) with a supraphysiological concentration of nitrite. The data may be fitted to a simple kinetic model involving a transient met-aqua form, in contrast to the ferryl detected in the case of Mb in a similar reaction sequence. These data are in line with a previous observation of a transient accumulation of ferryl Hb under auto-catalytic conditions at much lower concentrations of nitrite (Grubina, R. et al. J. Biol. Chem. 2007, 282, 12916). The simple model for fitting the stopped-flow data leaves a small part of the absorbance changes unaccounted for, unless a fourth species is invoked displaying features similar to the oxy and tentatively assigned as ferrous-peroxynitrate. Density functional theory (DFT) calculations support this latter assignment. The reaction allows for differentiating between the reactivities of various chemically modified hemoglobins, including candidates for blood substitutes. Polymerization of hemoglobin slows the nitrite-induced oxidation, in sharp contrast to oxidative-stress type reactions which are generally accelerated, not inhibited. Sheep hemoglobin is found to be distinctly more resistant to reaction with nitrite compared to bovine Hb, at large nitrite concentrations (stopped-flow experiments directly observing the oxy + nitrite reaction) as well as under auto-catalytic conditions. Copolymerization of Hb with bovine serum albumin (BSA) using glutaraldehyde leads to a distinct increase of the lag time compared to native Hb as well as to any other form of derivatization examined in the present study. The Hb-BSA copolymer also displays a slower initial reaction with nitrite under stopped-flow conditions, compared to native Hb.
Collapse
Affiliation(s)
- Denisa Hathazi
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babeş-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania.
| | - Florina Scurtu
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babeş-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania.
| | - Cristina Bischin
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babeş-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania.
| | - Augustin Mot
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babeş-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania.
| | - Amr A A Attia
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babeş-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania.
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| | - Jacob Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| | - Radu Silaghi-Dumitrescu
- Department of Chemistry, Faculty of Chemistry and Chemical Engineering, Babeş-Bolyai University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania.
| |
Collapse
|
66
|
Ferrer-Sueta G, Campolo N, Trujillo M, Bartesaghi S, Carballal S, Romero N, Alvarez B, Radi R. Biochemistry of Peroxynitrite and Protein Tyrosine Nitration. Chem Rev 2018; 118:1338-1408. [DOI: 10.1021/acs.chemrev.7b00568] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Gerardo Ferrer-Sueta
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Nicolás Campolo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Silvina Bartesaghi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Sebastián Carballal
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Romero
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Beatriz Alvarez
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Laboratorio
de Fisicoquímica Biológica, Facultad de
Ciencias, ‡Center for Free Radical and Biomedical Research, §Departamento de Bioquímica,
Facultad de Medicina, ∥Laboratorio de Enzimología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
67
|
Milanese C, Tapias V, Gabriels S, Cerri S, Levandis G, Blandini F, Tresini M, Shiva S, Greenamyre JT, Gladwin MT, Mastroberardino PG. Mitochondrial Complex I Reversible S-Nitrosation Improves Bioenergetics and Is Protective in Parkinson's Disease. Antioxid Redox Signal 2018; 28:44-61. [PMID: 28816057 PMCID: PMC5749586 DOI: 10.1089/ars.2017.6992] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS This study was designed to explore the neuroprotective potential of inorganic nitrite as a new therapeutic avenue in Parkinson's disease (PD). RESULTS Administration of inorganic nitrite ameliorates neuropathology in phylogenetically distinct animal models of PD. Beneficial effects are not confined to prophylactic treatment and also occur if nitrite is administered when the pathogenic cascade is already active. Mechanistically, the effect is mediated by both complex I S-nitrosation, which under nitrite administration is favored over formation of other forms of oxidation, and down-stream activation of the antioxidant Nrf2 pathway. Nitrite also rescues respiratory reserve capacity and increases proton leakage in LRRK2 PD patients' dermal fibroblasts. INNOVATION The study proposes an unprecedented approach based on the administration of the nitrosonium donor nitrite to contrast complex I and redox anomalies in PD. Dysfunctional mitochondrial complex I propagates oxidative stress in PD, and treatments mitigating this defect may, therefore, limit disease progression. Therapeutic complex I targeting has been successfully achieved in ischemia/reperfusion by using nitrosonium donors such as nitrite to reversibly modify its subunits and protect from oxidative damage after reperfusion. This evidence led to the innovative hypothesis that nitrite could exert protective effects also in pathological conditions where complex I dysfunction occurs in normoxia, such as in PD. CONCLUSIONS Overall, these results demonstrate that administration of inorganic nitrite improves mitochondrial function in PD, and it, therefore, represents an amenable intervention to hamper disease progression. Antioxid. Redox Signal. 28, 44-61.
Collapse
Affiliation(s)
- Chiara Milanese
- 1 Department of Molecular Genetics, Erasmus MC , Rotterdam, The Netherlands .,2 Ri.MED Foundation, Palermo , Italy
| | - Victor Tapias
- 3 Department of Neurology, University of Pittsburgh , Pittsburgh, Pennsylvania.,4 Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sylvia Gabriels
- 1 Department of Molecular Genetics, Erasmus MC , Rotterdam, The Netherlands
| | - Silvia Cerri
- 5 Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute , Pavia, Italy
| | - Giovanna Levandis
- 5 Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute , Pavia, Italy
| | - Fabio Blandini
- 5 Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, C. Mondino National Neurological Institute , Pavia, Italy
| | - Maria Tresini
- 1 Department of Molecular Genetics, Erasmus MC , Rotterdam, The Netherlands
| | - Sruti Shiva
- 6 Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,7 Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - John Timothy Greenamyre
- 3 Department of Neurology, University of Pittsburgh , Pittsburgh, Pennsylvania.,4 Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- 8 Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,9 Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | |
Collapse
|
68
|
Ho XL, Loke WM. Dietary Plant Sterols Supplementation Increases In Vivo Nitrite and Nitrate Production in Healthy Adults: A Randomized, Controlled Study. J Food Sci 2017; 82:1750-1756. [PMID: 28708316 PMCID: PMC5601184 DOI: 10.1111/1750-3841.13752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 11/29/2022]
Abstract
A randomized, double-blinded, placebo-controlled and crossover study was conducted to simultaneously measure the effects, 3 h after consumption and after 4-wk daily exposure to plant sterols-enriched food product, on in vivo nitrite and nitrate production in healthy adults. Eighteen healthy participants (67% female, 35.3 [mean] ± 9.5 [SD] years, mean body mass index 22.8 kg/m2 ) received 2 soy milk (20 g) treatments daily: placebo and one containing 2.0 g free plant sterols equivalent of their palmityl esters (β-sitosterol, 55%; campesterol, 29%; and stigmasterol, 23%). Nitrite and nitrate concentrations were measured in the blood plasma and urine, using stable isotope-labeled gas chromatography-mass spectrometry. L-arginine and asymmetric dimethylarginine concentrations in blood serum were measured using commercially available enzyme immunoassays. Nitrite and nitrate concentrations in blood plasma (nitrite 5.83 ± 0.50 vs. 4.52 ± 0.27; nitrate 15.78 ± 0.96 vs. 13.43 ± 0.81 μmol/L) and urine (nitrite 1.12 ± 0.22 vs. 0.92 ± 0.36, nitrate 12.23 ± 1.15 vs. 9.71 ± 2.04 μmol/L) were significantly elevated after 4-wk plant sterols supplementation Placebo and 3-h treatments did not affect the blood plasma and urinary concentrations of nitrite and nitrate. Circulating levels of L-arginine and asymmetric dimethylarginine were unchanged in the placebo and treatment arms. Total plant sterols, β-Sitosterol, campesterol, and stigmasterol concentrations were significantly elevated after 4-wk treatments compared to the placebo and 3-h treatments. Blood plasma nitrite and nitrate concentrations correlated significantly with the plasma total and specific plant sterol concentrations. Our results suggest that dietary plant sterols, in the combination used, can upregulate nitrite, and nitrate production in vivo.
Collapse
Affiliation(s)
- Xing Lin Ho
- Centre for Functional Foods & Human Nutrition, School of Chemical & Life Sciences, Nanyang Polytechnic, 180 Ang Mo Kio Ave 8, Singapore, 569830, Singapore.,Food Science & Nutrition Group, School of Chemical & Life Sciences, Nanyang Polytechnic, 180 Ang Mo Kio Ave 8, Singapore, 569830, Singapore
| | - Wai Mun Loke
- Centre for Functional Foods & Human Nutrition, School of Chemical & Life Sciences, Nanyang Polytechnic, 180 Ang Mo Kio Ave 8, Singapore, 569830, Singapore.,Food Science & Nutrition Group, School of Chemical & Life Sciences, Nanyang Polytechnic, 180 Ang Mo Kio Ave 8, Singapore, 569830, Singapore
| |
Collapse
|
69
|
Adams PS, Zahid M, Khalifa O, Feingold B, Lo CW. Low Nasal NO in Congenital Heart Disease With Systemic Right Ventricle and Postcardiac Transplantation. J Am Heart Assoc 2017; 6:JAHA.117.007447. [PMID: 29212650 PMCID: PMC5779050 DOI: 10.1161/jaha.117.007447] [Citation(s) in RCA: 6] [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: 12/18/2022]
Abstract
Background NO bioavailability has not been systematically examined in congenital heart disease (CHD). To assess NO in patients with CHD, we measured nasal NO (nNO) generated by the nasal epithelia, given blood NO is difficult to measure (half‐life, <2 ms). Given NO's role in hemodynamic regulation and the association of NO bioavailability with heart failure risk, we hypothesized NO levels may differ with varying severity of CHD physiologic characteristics. Methods and Results Six‐hundred eighteen subjects, 483 with CHD and 135 controls, had nNO measured noninvasively via the nares using American Thoracic Society/European Respiratory Society guidelines. Subjects were dichotomized as having low or normal nNO based on age‐specific cutoff values. Prevalence of low nNO was examined by various CHD physiologic feature types. Low nNO was more prevalent with CHD than controls (odds ratio, 2.28; P=0.001). A logistic regression model showed overall significance (P=0.035) for single ventricle, systemic right ventricle, ventricular dysfunction, oxygen desaturation, and heterotaxy predicting low nNO, with systemic right ventricle independently having twice the odds of low nNO (odds ratio, 2.04; P=0.014). Patients with low nNO had a higher risk of experiencing heart transplant or death (hazard ratio, 2.75; P=0.048), and heart transplant recipients (N=16) exhibited 5 times the odds of low nNO (69% versus 30%; odds ratio, 5.1; P=0.001). Conclusions Patients with CHD have increased prevalence of low nNO, with highest odds seen with systemic right ventricle and heart transplant. Further studies are needed to investigate heart failure risks in patients with CHD with left versus right systemic ventricle physiologic characteristics and utility of low nNO for predicting heart failure risk.
Collapse
Affiliation(s)
- Phillip S Adams
- Division of Pediatric Anesthesiology, Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Maliha Zahid
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Omar Khalifa
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Brian Feingold
- Division of Pediatric Cardiology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
70
|
Notay K, Incognito AV, Millar PJ. Acute beetroot juice supplementation on sympathetic nerve activity: a randomized, double-blind, placebo-controlled proof-of-concept study. Am J Physiol Heart Circ Physiol 2017; 313:H59-H65. [DOI: 10.1152/ajpheart.00163.2017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/03/2017] [Accepted: 05/03/2017] [Indexed: 01/09/2023]
Abstract
Acute dietary nitrate ([Formula: see text]) supplementation reduces resting blood pressure in healthy normotensives. This response has been attributed to increased nitric oxide bioavailability and peripheral vasodilation, although nitric oxide also tonically inhibits central sympathetic outflow. We hypothesized that acute dietary [Formula: see text] supplementation using beetroot (BR) juice would reduce blood pressure and muscle sympathetic nerve activity (MSNA) at rest and during exercise. Fourteen participants (7 men and 7 women, age: 25 ± 10 yr) underwent blood pressure and MSNA measurements before and after (165–180 min) ingestion of 70ml high-[Formula: see text] (~6.4 mmol [Formula: see text]) BR or [Formula: see text]-depleted BR placebo (PL; ~0.0055 mmol [Formula: see text]) in a double-blind, randomized, crossover design. Blood pressure and MSNA were also collected during 2 min of static handgrip (30% maximal voluntary contraction). The changes in resting MSNA burst frequency (−3 ± 5 vs. 3 ± 4 bursts/min, P = 0.001) and burst incidence (−4 ± 7 vs. 4 ± 5 bursts/100 heart beats, P = 0.002) were lower after BR versus PL, whereas systolic blood pressure (−1 ± 5 vs. 2 ± 5 mmHg, P = 0.30) and diastolic blood pressure (4 ± 5 vs. 5 ± 7 mmHg, P = 0.68) as well as spontaneous arterial sympathetic baroreflex sensitivity ( P = 0.95) were not different. During static handgrip, the change in MSNA burst incidence (1 ± 8 vs. 8 ± 9 bursts/100 heart beats, P = 0.04) was lower after BR versus PL, whereas MSNA burst frequency (6 ± 6 vs. 11 ± 10 bursts/min, P = 0.11) as well as systolic blood pressure (11 ± 7 vs. 12 ± 8 mmHg, P = 0.94) and diastolic blood pressure (11 ± 4 vs. 11 ± 4 mmHg, P = 0.60) were not different. Collectively, these data provide proof of principle that acute BR supplementation can decrease central sympathetic outflow at rest and during exercise. Dietary [Formula: see text] supplementation may represent a novel intervention to target exaggerated sympathetic outflow in clinical populations. NEW & NOTEWORTHY The hemodynamic benefits of dietary nitrate supplementation have been attributed to nitric oxide-mediated peripheral vasodilation. Here, we provide proof of concept that acute dietary nitrate supplementation using beetroot juice can decrease muscle sympathetic outflow at rest and during exercise in a normotensive population. These results have applications for targeting central sympathetic overactivation in disease.
Collapse
Affiliation(s)
- Karambir Notay
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| | - Anthony V. Incognito
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
| | - Philip J. Millar
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada; and
- Toronto General Research Institute, Toronto General Hospital, Toronto, Ontario, Canada
| |
Collapse
|
71
|
Abstract
AbstractCVD is the leading cause of death worldwide, a consequence of mostly poor lifestyle and dietary behaviours. Although whole fruit and vegetable consumption has been consistently shown to reduce CVD risk, the exact protective constituents of these foods are yet to be clearly identified. A recent and biologically plausible hypothesis supporting the cardioprotective effects of vegetables has been linked to their inorganic nitrate content. Approximately 60–80 % inorganic nitrate exposure in the human diet is contributed by vegetable consumption. Although inorganic nitrate is a relatively stable molecule, under specific conditions it can be metabolised in the body to produce NO via the newly discovered nitrate–nitrite–NO pathway. NO is a major signalling molecule in the human body, and has a key role in maintaining vascular tone, smooth muscle cell proliferation, platelet activity and inflammation. Currently, there is accumulating evidence demonstrating that inorganic nitrate can lead to lower blood pressure and improved vascular compliance in humans. The aim of this review is to present an informative, balanced and critical review of the current evidence investigating the role of inorganic nitrate and nitrite in the development, prevention and/or treatment of CVD. Although there is evidence supporting short-term inorganic nitrate intakes for reduced blood pressure, there is a severe lack of research examining the role of long-term nitrate intakes in the treatment and/or prevention of hard CVD outcomes, such as myocardial infarction and cardiovascular mortality. Epidemiological evidence is needed in this field to justify continued research efforts.
Collapse
|
72
|
Cabrales P, Caroen S, Oronsky A, Carter C, Trepel J, Summers T, Reid T, Oronsky N, Lybeck M, Oronsky B. The macrophage stimulating anti-cancer agent, RRx-001, protects against ischemia-reperfusion injury. Expert Rev Hematol 2017; 10:575-582. [PMID: 28448172 DOI: 10.1080/17474086.2017.1324779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND RRx-001, a clinical macrophage-stimulating anti-cancer agent that also produces nitric oxide (NO) was studied in a model of ischemia-reperfusion injury. METHODS The production of NO is dependent on the oxygen tension because nitric oxide synthases convert l-arginine to NO and l-citrulline in the presence of O2. Since the P450 enzymes, which metabolize nitrate esters such as nitroglycerin are dependent on oxygen, the generation of 'exogenous' NO is also sensitive to alterations in tissue PO2. I/R injury was studied in a hamster chamber window, with compression of the periphery of the window for 1 h to induce ischemia. Animals received RRx-001 (5 mg/kg) 24 h before ischemia and sodium nitrite (10 nmols/kg) was supplemented 10 min after the start of reperfusion. Vessel diameter, blood flow, adherent leukocytes, and functional capillary density were assessed by intravital microscopy at 0.5, 2, and 24 h following the release of the ischemia. RESULTS The results demonstrated that, compared to control, RRx-001 preconditioning increased blood flow and functional capillary density, and preserved tissue viability in the absence of side effects over a sustained time period. CONCLUSION Thus, RRx-001 may serve as a long-lived protective agent during postsurgical restoration of flow and other ischemia-reperfusion associated conditions, increasing blood flow and functional capillary density as well as preserving tissue viability in the absence of side effects.
Collapse
Affiliation(s)
- Pedro Cabrales
- a Department of Bioengineering , University of California San Diego (UCSD) , La Jolla , CA , USA
| | | | | | - Corey Carter
- d Walter Reed Military Medical Center , Murtha Cancer Center , Bethesda , MD , USA
| | - Jane Trepel
- e Moores Cancer Center , University of California San Diego (UCSD) , La Jolla , CA , USA
| | - Thomas Summers
- d Walter Reed Military Medical Center , Murtha Cancer Center , Bethesda , MD , USA
| | | | | | | | | |
Collapse
|
73
|
Reeder BJ. Redox and Peroxidase Activities of the Hemoglobin Superfamily: Relevance to Health and Disease. Antioxid Redox Signal 2017; 26:763-776. [PMID: 27637274 DOI: 10.1089/ars.2016.6803] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
SIGNIFICANCE Erythrocyte hemoglobin (Hb) and myocyte myoglobin, although primarily oxygen-carrying proteins, have the capacity to do redox chemistry. Such redox activity in the wider family of globins now appears to have important associations with the mechanisms of cell stress response. In turn, an understanding of such mechanisms in vivo may have a potential in the understanding of cancer therapy resistance and neurodegenerative disorders such as Alzheimer's. Recent Advances: There has been an enhanced understanding of the redox chemistry of the globin superfamily in recent years, leading to advances in development of Hb-based blood substitutes and in hypotheses relating to specific disease mechanisms. Neuroglobin (Ngb) and cytoglobin (Cygb) have been linked to cell protection mechanisms against hypoxia and oxidative stress, with implications in the onset and progression of neurodegenerative diseases for Ngb and cancer for Cygb. CRITICAL ISSUES Despite advances in the understanding of redox chemistry of globins, the physiological roles of many of these proteins still remain ambiguous at best. Confusion over potential physiological roles may relate to multifunctional roles for globins, which may be modulated by surface-exposed cysteine pairs in some globins. Such roles may be critical in deciphering the relationships of these globins in human diseases. FUTURE DIRECTIONS Further studies are required to connect the considerable knowledge on the mechanisms of globin redox chemistry in vitro with the physiological and pathological roles of globins in vivo. In doing so, new therapies for neurodegenerative disorders and cancer therapy resistance may be targeted. Antioxid. Redox Signal. 26, 763-776.
Collapse
Affiliation(s)
- Brandon J Reeder
- School of Biological Sciences, University of Essex , Essex, United Kingdom
| |
Collapse
|
74
|
Hirsch RE, Sibmooh N, Fucharoen S, Friedman JM. HbE/β-Thalassemia and Oxidative Stress: The Key to Pathophysiological Mechanisms and Novel Therapeutics. Antioxid Redox Signal 2017; 26:794-813. [PMID: 27650096 PMCID: PMC5421591 DOI: 10.1089/ars.2016.6806] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/16/2016] [Indexed: 01/19/2023]
Abstract
SIGNIFICANCE Oxidative stress and generation of free radicals are fundamental in initiating pathophysiological mechanisms leading to an inflammatory cascade resulting in high rates of morbidity and death from many inherited point mutation-derived hemoglobinopathies. Hemoglobin (Hb)E is the most common point mutation worldwide. The βE-globin gene is found in greatest frequency in Southeast Asia, including Thailand, Malaysia, Indonesia, Vietnam, Cambodia, and Laos. With the wave of worldwide migration, it is entering the gene pool of diverse populations with greater consequences than expected. CRITICAL ISSUES While HbE by itself presents as a mild anemia and a single gene for β-thalassemia is not serious, it remains unexplained why HbE/β-thalassemia (HbE/β-thal) is a grave disease with high morbidity and mortality. Patients often exhibit defective physical development, severe chronic anemia, and often die of cardiovascular disease and severe infections. Recent Advances: This article presents an overview of HbE/β-thal disease with an emphasis on new findings pointing to pathophysiological mechanisms derived from and initiated by the dysfunctional property of HbE as a reduced nitrite reductase concomitant with excess α-chains exacerbating unstable HbE, leading to a combination of nitric oxide imbalance, oxidative stress, and proinflammatory events. FUTURE DIRECTIONS Additionally, we present new therapeutic strategies that are based on the emerging molecular-level understanding of the pathophysiology of this and other hemoglobinopathies. These strategies are designed to short-circuit the inflammatory cascade leading to devastating chronic morbidity and fatal consequences. Antioxid. Redox Signal. 26, 794-813.
Collapse
Affiliation(s)
- Rhoda Elison Hirsch
- Department of Medicine (Hematology), Albert Einstein College of Medicine, Bronx, New York
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Nathawut Sibmooh
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakornpathom, Thailand
| | - Joel M. Friedman
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
75
|
Zhang X, Zhao Q, Li Y, Duan X, Tang Y. Multifunctional Probe Based on Cationic Conjugated Polymers for Nitroreductase-Related Analysis: Sensing, Hypoxia Diagnosis, and Imaging. Anal Chem 2017; 89:5503-5510. [DOI: 10.1021/acs.analchem.7b00477] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Xiaoqian Zhang
- Key Laboratory of Analytical
Chemistry for Life Science of Shaanxi Province, Key Laboratory of
Applied Surface and Colloid Chemistry, Ministry of Education, School
of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an, Shaanxi 710062, P. R. China
| | - Qi Zhao
- Key Laboratory of Analytical
Chemistry for Life Science of Shaanxi Province, Key Laboratory of
Applied Surface and Colloid Chemistry, Ministry of Education, School
of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an, Shaanxi 710062, P. R. China
| | - Yanru Li
- Key Laboratory of Analytical
Chemistry for Life Science of Shaanxi Province, Key Laboratory of
Applied Surface and Colloid Chemistry, Ministry of Education, School
of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an, Shaanxi 710062, P. R. China
| | - Xinrui Duan
- Key Laboratory of Analytical
Chemistry for Life Science of Shaanxi Province, Key Laboratory of
Applied Surface and Colloid Chemistry, Ministry of Education, School
of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an, Shaanxi 710062, P. R. China
| | - Yanli Tang
- Key Laboratory of Analytical
Chemistry for Life Science of Shaanxi Province, Key Laboratory of
Applied Surface and Colloid Chemistry, Ministry of Education, School
of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi’an, Shaanxi 710062, P. R. China
| |
Collapse
|
76
|
Koch CD, Gladwin MT, Freeman BA, Lundberg JO, Weitzberg E, Morris A. Enterosalivary nitrate metabolism and the microbiome: Intersection of microbial metabolism, nitric oxide and diet in cardiac and pulmonary vascular health. Free Radic Biol Med 2017; 105:48-67. [PMID: 27989792 PMCID: PMC5401802 DOI: 10.1016/j.freeradbiomed.2016.12.015] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/18/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Recent insights into the bioactivation and signaling actions of inorganic, dietary nitrate and nitrite now suggest a critical role for the microbiome in the development of cardiac and pulmonary vascular diseases. Once thought to be the inert, end-products of endothelial-derived nitric oxide (NO) heme-oxidation, nitrate and nitrite are now considered major sources of exogenous NO that exhibit enhanced vasoactive signaling activity under conditions of hypoxia and stress. The bioavailability of nitrate and nitrite depend on the enzymatic reduction of nitrate to nitrite by a unique set of bacterial nitrate reductase enzymes possessed by specific bacterial populations in the mammalian mouth and gut. The pathogenesis of pulmonary hypertension (PH), obesity, hypertension and CVD are linked to defects in NO signaling, suggesting a role for commensal oral bacteria to shape the development of PH through the formation of nitrite, NO and other bioactive nitrogen oxides. Oral supplementation with inorganic nitrate or nitrate-containing foods exert pleiotropic, beneficial vascular effects in the setting of inflammation, endothelial dysfunction, ischemia-reperfusion injury and in pre-clinical models of PH, while traditional high-nitrate dietary patterns are associated with beneficial outcomes in hypertension, obesity and CVD. These observations highlight the potential of the microbiome in the development of novel nitrate- and nitrite-based therapeutics for PH, CVD and their risk factors.
Collapse
Affiliation(s)
- Carl D Koch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA.
| | - Mark T Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA
| | - Bruce A Freeman
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh PA 15261, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Physiology and Pharmacology, Karolinska Institutet, S-17177 Stockholm, Sweden
| |
Collapse
|
77
|
Dezfulian C, Taft M, Corey C, Hill G, Krehel N, Rittenberger JC, Guyette FX, Shiva S. Biochemical signaling by remote ischemic conditioning of the arm versus thigh: Is one raise of the cuff enough? Redox Biol 2017; 12:491-498. [PMID: 28334684 PMCID: PMC5362138 DOI: 10.1016/j.redox.2017.03.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 03/08/2017] [Indexed: 01/23/2023] Open
Abstract
Remote Ischemic Conditioning (RIC), induced by brief cycles of ischemia and reperfusion, protects vital organs from a prolonged ischemic insult. While several biochemical mediators have been implicated in RIC's mechanism of action, it remains unclear whether the localization or “dose” of RIC affects the extent of protective signaling. In this randomized crossover study of healthy individuals, we tested whether the number of cycles of RIC and its localization (arm versus thigh) determines biochemical signaling and cytoprotection. Subjects received either arm or thigh RIC and then were crossed over to receive RIC in the other extremity. Blood flow, tissue perfusion, concentrations of the circulating protective mediator nitrite, and platelet mitochondrial function were measured after each RIC cycle. We found that plasma nitrite concentration peaked after the first RIC cycle and remained elevated throughout RIC. This plasma nitrite conferred cytoprotection in an in vitro myocyte model of hypoxia/reoxygenation. Notably, though plasma nitrite returned to baseline at 24 h, RIC conditioned plasma still mediated protection. Additionally, no difference in endpoints between RIC in thigh versus arm was found. These data demonstrate that localization and “dose” of RIC does not affect cytoprotection and further elucidate the mechanisms by which nitrite contributes to RIC-dependent protection.
Collapse
Affiliation(s)
- Cameron Dezfulian
- Safar Center for Resuscitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Maia Taft
- Safar Center for Resuscitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Catherine Corey
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Gabrielle Hill
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Nicholas Krehel
- Safar Center for Resuscitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Jon C Rittenberger
- Applied Physiology Laboratory, Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Frank X Guyette
- Applied Physiology Laboratory, Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Applied Physiology Laboratory, Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
78
|
Parikh J, Kapela A, Tsoukias NM. Can endothelial hemoglobin-α regulate nitric oxide vasodilatory signaling? Am J Physiol Heart Circ Physiol 2017; 312:H854-H866. [PMID: 28130333 DOI: 10.1152/ajpheart.00315.2016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 11/22/2022]
Abstract
We used mathematical modeling to investigate nitric oxide (NO)-dependent vasodilatory signaling in the arteriolar wall. Detailed continuum cellular models of calcium (Ca2+) dynamics and membrane electrophysiology in smooth muscle and endothelial cells (EC) were coupled with models of NO signaling and biotransport in an arteriole. We used this theoretical approach to examine the role of endothelial hemoglobin-α (Hbα) as a modulator of NO-mediated myoendothelial feedback, as previously suggested in Straub et al. (Nature 491: 473-477, 2012). The model considers enriched expression of inositol 1,4,5-triphosphate receptors (IP3Rs), endothelial nitric oxide synthase (eNOS) enzyme, Ca2+-activated potassium (KCa) channels and Hbα in myoendothelial projections (MPs) between the two cell layers. The model suggests that NO-mediated myoendothelial feedback is plausible if a significant percentage of eNOS is localized within or near the myoendothelial projection. Model results show that the ability of Hbα to regulate the myoendothelial feedback is conditional to its colocalization with eNOS near MPs at concentrations in the high nanomolar range (>0.2 μM or 24,000 molecules). Simulations also show that the effect of Hbα observed in in vitro experimental studies may overestimate its contribution in vivo, in the presence of blood perfusion. Thus, additional experimentation is required to quantify the presence and spatial distribution of Hbα in the EC, as well as to test that the strong effect of Hbα on NO signaling seen in vitro, translates also into a physiologically relevant response in vivo.NEW & NOTEWORTHY Mathematical modeling shows that although regulation of nitric oxide signaling by hemoglobin-α (Hbα) is plausible, it is conditional to its presence in significant concentrations colocalized with endothelial nitric oxide synthase in myoendothelial projections. Additional experimentation is required to test that the strong effect of Hbα seen in vitro translates into a physiologically relevant response in vivo.
Collapse
Affiliation(s)
- Jaimit Parikh
- Department of Biomedical Engineering, Florida International University, Miami, Florida; and
| | - Adam Kapela
- Department of Biomedical Engineering, Florida International University, Miami, Florida; and
| | - Nikolaos M Tsoukias
- Department of Biomedical Engineering, Florida International University, Miami, Florida; and .,School of Chemical Engineering, National Technical University of Athens, Athens, Greece
| |
Collapse
|
79
|
Dulce RA, Kulandavelu S, Schulman IH, Fritsch J, Hare JM. Nitric Oxide Regulation of Cardiovascular Physiology and Pathophysiology. Nitric Oxide 2017. [DOI: 10.1016/b978-0-12-804273-1.00024-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
80
|
Liu Y, Buerk DG, Barbee KA, Jaron D. A mathematical model for the role of N 2O 3 in enhancing nitric oxide bioavailability following nitrite infusion. Nitric Oxide 2016; 60:1-9. [PMID: 27565833 PMCID: PMC5343674 DOI: 10.1016/j.niox.2016.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/23/2016] [Accepted: 08/22/2016] [Indexed: 12/18/2022]
Abstract
Nitrite infusion into the bloodstream has been shown to elicit vasodilation and protect against ischemia-reperfusion injury through nitric oxide (NO) release in hypoxic conditions. However, the mechanism by which nitrite-derived NO escapes scavenging by hemoglobin in the erythrocyte has not been fully elucidated, owing in part to the difficulty in measuring the reactions and transport on NO in vivo. We developed a mathematical model for an arteriole and surrounding tissue to examine the hypothesis that dinitrogen trioxide (N2O3) acts as a stable intermediate for preserving NO. Our simulations predict that with hypoxia and moderate nitrite concentrations, the N2O3 pathway can significantly preserve the NO produced by hemoglobin nitrite reductase in the erythrocyte and elevate NO reaching the smooth muscle cells. Nitrite retains its ability to increase NO bioavailability even at varying flow conditions, but there is minimal effect under normoxia or very low nitrite concentrations. Our model demonstrates a viable pathway for reconciling experimental findings of potentially beneficial effects of nitrite infusions despite previous models showing negligible NO elevation associated with hemoglobin nitrite reductase. Our results suggest that additional mechanisms may be needed to explain the efficacy of nitrite-induced vasodilation at low infusion concentrations.
Collapse
Affiliation(s)
- Yien Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA.
| |
Collapse
|
81
|
Chirinos JA, Zamani P. The Nitrate-Nitrite-NO Pathway and Its Implications for Heart Failure and Preserved Ejection Fraction. Curr Heart Fail Rep 2016; 13:47-59. [PMID: 26792295 DOI: 10.1007/s11897-016-0277-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathogenesis of exercise intolerance in patients with heart failure and preserved ejection fraction (HFpEF) is likely multifactorial. In addition to cardiac abnormalities (diastolic dysfunction, abnormal contractile reserve, chronotropic incompetence), several peripheral abnormalities are likely to be involved. These include abnormal pulsatile hemodynamics, abnormal arterial vasodilatory responses to exercise, and abnormal peripheral O2 delivery, extraction, and utilization. The nitrate-nitrite-NO pathway is emerging as a potential target to modify key physiologic abnormalities, including late systolic left ventricular (LV) load from arterial wave reflections (which has deleterious short- and long-term consequences for the LV), arterial vasodilatory reserve, muscle O2 delivery, and skeletal muscle mitochondrial function. In a recently completed randomized trial, the administration of a single dose of exogenous inorganic nitrate has been shown to exert various salutary arterial hemodynamic effects, ultimately leading to enhanced aerobic capacity in patients with HFpEF. These effects have the potential for both immediate improvements in exercise tolerance and for long-term "disease-modifying" effects. In this review, we provide an overview of key mechanistic contributors to exercise intolerance in HFpEF, and of the potential therapeutic role of drugs that target the nitrate-nitrite-NO pathway.
Collapse
Affiliation(s)
- Julio A Chirinos
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Hospital of the University of Pennsylvania, Philadelphia, PA, USA. .,Ghent University, Ghent, Belgium.
| | - Payman Zamani
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
82
|
Wang A, Kluger R. Enhanced Nitrite Reductase Activity and Its Correlation with Oxygen Affinity in Hemoglobin Bis-Tetramers. Biochemistry 2016; 55:4688-96. [PMID: 27454142 DOI: 10.1021/acs.biochem.6b00542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The vasoactivity of circulating cross-linked hemoglobin is consistent with the acellular protein penetrating the endothelial lining of blood vessels where hemoglobin can bind nitric oxide, the signal for relaxation of the muscles that surround blood vessels. In an important contrast, derivatives of bis-tetramers that are produced from hemoglobin by chemical coupling do not cause vasoconstriction in animal models. Presumably, they are unable to enter the endothelia where hemoglobin tetramers bind to nitric oxide. In addition, hemoglobin bis-tetramers can produce nitric oxide in circulation through their intrinsic nitrite reductase activity. Examination of this activity for hemoglobin-derived bis-tetramers that are acetylated at lysyl amino groups in their α subunits reveals enhanced activity (k = 2.21 M(-1) s(-1)) compared to that of nonacetylated bis-tetramers (k = 0.70 M(-1) s(-1)). Plots of nitrite reductase activities as a function of the corresponding oxygen affinities of certain allosteric-state-stabilized derivatives reveal a significant correlation, providing a basis for interpretation of the correlated functions.
Collapse
Affiliation(s)
- Aizhou Wang
- Davenport Chemical Laboratories, Department of Chemistry, University of Toronto , Toronto, ON, Canada M5S 3H6
| | - Ronald Kluger
- Davenport Chemical Laboratories, Department of Chemistry, University of Toronto , Toronto, ON, Canada M5S 3H6
| |
Collapse
|
83
|
Abstract
Background Hypoxia causes secondary headaches such as high-altitude headache (HAH) and headache due to acute mountain sickness. These secondary headaches mimic primary headaches such as migraine, which suggests a common link. We review and discuss the possible role of hypoxia in migraine and cluster headache. Methods This narrative review investigates the current level of knowledge on the relation of hypoxia in migraine and cluster headache based on epidemiological and experimental studies. Findings Epidemiological studies suggest that living in high-altitude areas increases the risk of migraine and especially migraine with aura. Human provocation models show that hypoxia provokes migraine with and without aura, whereas cluster headache has not been reliably induced by hypoxia. Possible pathophysiological mechanisms include hypoxia-induced release of nitric oxide and calcitonin gene-related peptide, cortical spreading depression and leakage of the blood-brain barrier. Conclusion There is a possible link between hypoxia and migraine and maybe cluster headache, but the exact mechanism is currently unknown. Provocation models of hypoxia have yielded interesting results suggesting a novel approach to study in depth the mechanism underlying hypoxia and primary headaches.
Collapse
Affiliation(s)
- Josefine Britze
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Nanna Arngrim
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Henrik Winther Schytz
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Messoud Ashina
- Danish Headache Center and Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
84
|
Hanff E, Böhmer A, Zinke M, Gambaryan S, Schwarz A, Supuran CT, Tsikas D. Carbonic anhydrases are producers of S-nitrosothiols from inorganic nitrite and modulators of soluble guanylyl cyclase in human platelets. Amino Acids 2016; 48:1695-706. [PMID: 27129464 DOI: 10.1007/s00726-016-2234-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 04/08/2016] [Indexed: 12/29/2022]
Abstract
Nitric oxide (NO), S-nitrosoglutathione (GSNO) and S-nitrosocysteine are highly potent signaling molecules, acting both by cGMP-dependent and cGMP-independent mechanisms. The NO metabolite nitrite (NO2 (-)) is a major NO reservoir. Hemoglobin, xanthine oxidoreductase and carbonic anhydrase (CA) have been reported to reduce/convert nitrite to NO. We evaluated the role and the physiological importance of CA for an extra-platelet CA/nitrite/NO/cGMP pathway in human platelets. Authentic NO was analyzed by an NO-sensitive electrode. GSNO and GS(15)NO were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS). cGMP was determined by LC-MS/MS or RIA. In reduced glutathione (GSH) containing aqueous buffer (pH 7.4), human and bovine erythrocytic CAII-mediated formation of GSNO from nitrite and GS(15)NO from (15)N-nitrite. In the presence of L-cysteine and GSH, this reaction was accompanied by NO release. Incubation of nitrite with bovine erythrocytic CAII and recombinant soluble guanylyl cyclase resulted in cGMP formation. Upon incubation of nitrite with bovine erythrocytic CAII and washed human platelets, cGMP and P-VASP(S239) were formed in the platelets. This study provides the first evidence that extra-platelet nitrite and erythrocytic CAII may modulate platelet function in a cGMP-dependent manner. The new nitrite-dependent CA activity may be a general principle and explain the cardioprotective effects of inorganic nitrite in the vasculature. We propose that nitrous acid (ONOH) is the primary CA-catalyzed reaction product of nitrite.
Collapse
Affiliation(s)
- Erik Hanff
- Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Anke Böhmer
- Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Maximilian Zinke
- Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Stepan Gambaryan
- Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.,Department of Cytology and Histology, S. Petersburg State University, Universitetskaya Nab 7/9, 199034, S. Petersburg, Russia
| | - Alexandra Schwarz
- Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Claudiu T Supuran
- Dipartimento Neurofarba, Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy
| | - Dimitrios Tsikas
- Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
85
|
Tsikas D, Böhmer A, Gros G, Endeward V. Evidence of the chemical reaction of (18)O-labelled nitrite with CO2 in aqueous buffer of neutral pH and the formation of (18)OCO by isotope ratio mass spectrometry. Nitric Oxide 2016; 55-56:25-35. [PMID: 26976364 DOI: 10.1016/j.niox.2016.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/06/2016] [Accepted: 03/09/2016] [Indexed: 11/19/2022]
Abstract
Inorganic nitrite (NO2(-), ON-O(-) ←→ (-)O-NO) is the autoxidation product of nitric oxide (NO). Nitrite can also be formed from inorganic nitrate (ONO2(-)), the major oxidation product of NO in erythrocytes, by the catalytic action of bacterial nitrate reductase in gut and oral microflora. Nitrite can be reduced to NO by certain cellular proteins and enzymes, as well as in the gastric juice under acidic conditions. Hemoglobin, xanthine oxidoreductase and carbonic anhydrase (CA) have been reported to convert nitrite to NO. Renal CA isoforms are involved in the reabsorption of nitrite and may, therefore, play an important role in NO homeostasis. Yet, the mechanisms underlying the action of CA on nitrite are incompletely understood. The nitrate/nitrite system is regarded as a reservoir of NO. We have recently shown that nitrite reacts chemically with carbon dioxide (CO2), the regular substrate of CA. The present communication reports a stable isotope ratio mass spectrometry (IRMS) study on the reaction of NO2(-) and CO2 performed in 50 mM HEPES buffer of pH 7.4 at 37 °C. By using (18)O-labelled nitrite ((18)ON-O(-)/(-18)O-NO) and CO2 we observed formation of (18)O-labelled CO2. This finding is an unequivocal evidence of the chemical reaction of (18)ON-O(-)/(-18)O-NO with CO2. The reaction is rapid and involves nucleophilic attack of the negatively charged nitrite via one of its oxygen atoms on the partially positively charged CO2 molecule to form the putative intermediate (18)ON-O-CO2(-)/(-)O2C-(18)O-NO. The by far largest fraction of this intermediate decomposes back to (18)ON-O(-)/(-18)O-NO and CO2. A very small fraction of the intermediate, however, rearranges and finally decomposes to form (18)OCO and nitrite. This reaction is slower in the presence of an isolated erythrocytic CA isoform II. In summary, NO2(-), CO2 and CA are ubiquitous. The chemical reaction of NO2(-) with CO2 and its modulation by CA isoforms may play important roles in the transport of nitrite in red blood cells, the kidney and other cells and organs.
Collapse
Affiliation(s)
- Dimitrios Tsikas
- Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Anke Böhmer
- Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Gerolf Gros
- Institute of Neurophysiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Volker Endeward
- Institute of Neurophysiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
86
|
Basudhar D, Ridnour LA, Cheng R, Kesarwala AH, Heinecke J, Wink DA. Biological signaling by small inorganic molecules. Coord Chem Rev 2016; 306:708-723. [PMID: 26688591 PMCID: PMC4680994 DOI: 10.1016/j.ccr.2015.06.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Small redox active molecules such as reactive nitrogen and oxygen species and hydrogen sulfide have emerged as important biological mediators that are involved in various physiological and pathophysiological processes. Advancement in understanding of cellular mechanisms that tightly regulate both generation and reactivity of these molecules is central to improved management of various disease states including cancer and cardiovascular dysfunction. Imbalance in the production of redox active molecules can lead to damage of critical cellular components such as cell membranes, proteins and DNA and thus may trigger the onset of disease. These small inorganic molecules react independently as well as in a concerted manner to mediate physiological responses. This review provides a general overview of the redox biology of these key molecules, their diverse chemistry relevant to physiological processes and their interrelated nature in cellular signaling.
Collapse
Affiliation(s)
- Debashree Basudhar
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Lisa A. Ridnour
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Robert Cheng
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Aparna H. Kesarwala
- Radiation Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - Julie Heinecke
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | - David A. Wink
- Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| |
Collapse
|
87
|
Salgado MT, Cao Z, Nagababu E, Mohanty JG, Rifkind JM. Red Blood Cell Membrane-Facilitated Release of Nitrite-Derived Nitric Oxide Bioactivity. Biochemistry 2015; 54:6712-23. [DOI: 10.1021/acs.biochem.5b00643] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Maria T. Salgado
- Molecular Dynamics Section,
National Institute on Aging, National Institutes of Health, 251 Bayview
Boulevard, Baltimore, Maryland 21224, United States
| | - Zeling Cao
- Molecular Dynamics Section,
National Institute on Aging, National Institutes of Health, 251 Bayview
Boulevard, Baltimore, Maryland 21224, United States
| | - Enika Nagababu
- Molecular Dynamics Section,
National Institute on Aging, National Institutes of Health, 251 Bayview
Boulevard, Baltimore, Maryland 21224, United States
| | - Joy G. Mohanty
- Molecular Dynamics Section,
National Institute on Aging, National Institutes of Health, 251 Bayview
Boulevard, Baltimore, Maryland 21224, United States
| | - Joseph M. Rifkind
- Molecular Dynamics Section,
National Institute on Aging, National Institutes of Health, 251 Bayview
Boulevard, Baltimore, Maryland 21224, United States
| |
Collapse
|
88
|
Galinato MGI, Fogle RS, Stetz A, Galan JF. Modulating the nitrite reductase activity of globins by varying the heme substituents: Utilizing myoglobin as a model system. J Inorg Biochem 2015; 154:7-20. [PMID: 26544504 DOI: 10.1016/j.jinorgbio.2015.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/08/2015] [Accepted: 10/19/2015] [Indexed: 11/18/2022]
Abstract
Globins, such as hemoglobin (Hb) and myoglobin (Mb), have gained attention for their ability to reduce nitrite (NO2(-)) to nitric oxide (NO). The molecular interactions that regulate this chemistry are not fully elucidated, therefore we address this issue by investigating one part of the active site that may control this reaction. Here, the effects of the 2,4-heme substituents on the nitrite reductase (NiR) reaction, and on the structures and energies of the ferrous nitrite intermediates, are investigated using Mb as a model system. This is accomplished by studying Mbs with hemes that have different 2,4-R groups, namely diacetyldeuteroMb (-acetyl), protoMb (wild-type (wt) Mb, -vinyl), deuteroMb (-H), and mesoMb (-ethyl). While trends on the natural charge on Fe and O-atom of bound nitrite are observed among the series of Mbs, the Fe(II)-NPyr (Pyr=pyrrole) and Fe(II)-NHis93 (His=histidine) bond lengths do not significantly change. Kinetic analysis shows increasing NiR activity as follows: diacetyldeuteroMb<wt Mb<deuteroMb<mesoMb. Nitrite binding energy calculations of the different Mb(II)-nitrite conformations demonstrate the N-bound complexes to be more stable than the O-bound complexes for all the different types of heme structures, with diacetyldeuteroMb having the greatest nitrite binding affinity. Spectral deconvolution on the final product generated from the reaction between Mb(II) and NO2(-) for the reconstituted Mbs indicates the formation of 1:1 Mb(III) and Mb(II)-NO. The electronic changes induced by the -R groups on the 2,4-positions do not alter the stoichiometric ratio of the products, resembling wt Mb.
Collapse
Affiliation(s)
- Mary Grace I Galinato
- School of Science-Chemistry, Penn State Erie, The Behrend College, Erie, PA 16563, United States.
| | - Robert S Fogle
- School of Science-Chemistry, Penn State Erie, The Behrend College, Erie, PA 16563, United States
| | - Amanda Stetz
- School of Science-Chemistry, Penn State Erie, The Behrend College, Erie, PA 16563, United States
| | - Jhenny F Galan
- Dept. of Marine Sciences, Texas A&M at Galveston, 200 Seawolf Parkway, Galveston, TX 77553, United States.
| |
Collapse
|
89
|
Zinke M, Hanff E, Böhmer A, Supuran CT, Tsikas D. Discovery and microassay of a nitrite-dependent carbonic anhydrase activity by stable-isotope dilution gas chromatography-mass spectrometry. Amino Acids 2015; 48:245-55. [PMID: 26334347 DOI: 10.1007/s00726-015-2081-3] [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: 08/17/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022]
Abstract
The intrinsic activity of carbonic anhydrase (CA) is the hydration of CO2 to carbonic acid and its dehydration to CO2. CA may also function as esterase and phosphatase. Recently, we demonstrated that renal CA is mainly responsible for the reabsorption of nitrite (NO2(-)) which is the most abundant reservoir of the biologically highly potent nitric oxide (NO). By means of a stable-isotope dilution GC-MS method, we discovered a novel CA activity which strictly depends upon nitrite. We found that bovine erythrocytic CAII (beCAII) catalyses the incorporation of (18)O from H2 (18)O into nitrite at pH 7.4. After derivatization with pentafluorobenzyl bromide, gas chromatographic separation and mass spectrometric analysis, we detected ions at m/z 48 for singly (18)O-labelled nitrite ((16)O=N-(18)O(-)/(18)O=N-(16)O(-)) and at m/z 50 for doubly (18)O-labelled nitrite ((18)O=N-(18)O(-)) in addition to m/z 46 for unlabelled nitrite. Using (15)N-labelled nitrite ((15)NO2 (-), m/z 47) as an internal standard and selected-ion monitoring of m/z 46, m/z 48, m/z 50 and m/z 47, we developed a GC-MS microassay for the quantitative determination of the nitrite-dependent beCAII activity. The CA inhibitors acetazolamide and FC5 207A did not alter beCAII-catalysed formation of singly and doubly (18)O-labelled nitrite. Cysteine and the experimental CA inhibitor DIDS (a diisothiocyanate) increased several fold the beCAII-catalysed formation of the (18)O-labelled nitrite species. Cysteine, acetazolamide, FC5 207A, and DIDS by themselves had no effect on the incorporation of (18)O from H2 (18)O into nitrite. We conclude that erythrocytic CA possesses a nitrite-dependent activity which can only be detected when nitrite is used as the substrate and the reaction is performed in buffers of neutral pH values prepared in H2 (18)O. This novel CA activity, i.e., the nitrous acid anhydrase activity, represents a bioactivation of nitrite and may have both beneficial (via S-nitrosylation and subsequent NO release) and possibly adverse (via C- and N-nitrosylation) effects in living organisms.
Collapse
Affiliation(s)
- Maximilian Zinke
- Bioanalytical Research Laboratory for NO, Oxidative Stress and Eicosanoids (BIOFORNOX20), Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Erik Hanff
- Bioanalytical Research Laboratory for NO, Oxidative Stress and Eicosanoids (BIOFORNOX20), Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Anke Böhmer
- Bioanalytical Research Laboratory for NO, Oxidative Stress and Eicosanoids (BIOFORNOX20), Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Claudiu T Supuran
- Dipartmento di Chimica Ugo Schiff, Università degli Studi di Firenze, Sesto Fiorentino, Florence, Italy
| | - Dimitrios Tsikas
- Bioanalytical Research Laboratory for NO, Oxidative Stress and Eicosanoids (BIOFORNOX20), Centre of Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany.
| |
Collapse
|
90
|
Abstract
The year 2008 witnessed the first report on the increase in the concentration of L-homoarginine (hArg) in the maternal plasma during human pregnancy. This observation, along with a well-known function of hArg, the methylene homologue of L-arginine (Arg), as a substrate for nitric oxide (NO) synthase, was the ignition for the start of intense research on the physiology and pathology of hArg. The circulating concentration of hArg was found to be lower in patients suffering from various diseases, and hArg emerged within only very few years as a novel cardiovascular risk factor. The compendium in hand comprises original and review articles covering several aspects of hArg, Arg and its symmetrically and asymmetrically guanidine (N (G))-dimethylated derivatives SDMA and ADMA, respectively. In contrast to ADMA and SDMA, low hArg concentrations in plasma or serum and in urine are associated with high risks for morbidity and mortality, notably in the renal and cardiovascular systems. Acutely and chronically administered Arg as a nutritional supplement or in the form of dietary proteins is safe in animals and humans and leads to concomitant formation of hArg and ADMA, albeit in a different hArg/ADMA ratio. Despite the close but opposite associations of hArg and ADMA with disease in adults, children and adolescents, the underlying biochemical processes are largely unknown, presumably not restricted to NO, and warrant deeper investigation. As the common substrate for hArg and ADMA, Arg may play a key role in the biosynthesis and homeostasis of hArg and ADMA, two putative antagonists. In animal models of stroke and obesity, hArg has beneficial effects. The potential utility of hArg as a therapeutic drug or nutritional supplement in humans and animals remains to be elaborated.
Collapse
|
91
|
Effects of chronic oral l-arginine administration on the l-arginine/NO pathway in patients with peripheral arterial occlusive disease or coronary artery disease: l-Arginine prevents renal loss of nitrite, the major NO reservoir. Amino Acids 2015; 47:1961-74. [DOI: 10.1007/s00726-015-2031-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/13/2015] [Indexed: 12/20/2022]
|
92
|
Kelley EE. Dispelling dogma and misconceptions regarding the most pharmacologically targetable source of reactive species in inflammatory disease, xanthine oxidoreductase. Arch Toxicol 2015; 89:1193-207. [PMID: 25995007 DOI: 10.1007/s00204-015-1523-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 04/27/2015] [Indexed: 01/04/2023]
Abstract
Xanthine oxidoreductase (XOR), the molybdoflavin enzyme responsible for the terminal steps of purine degradation in humans, is also recognized as a significant source of reactive species contributory to inflammatory disease. In animal models and clinical studies, inhibition of XOR has resulted in diminution of symptoms and enhancement of function in a number of pathologies including heart failure, diabetes, sickle cell anemia, hypertension and ischemia-reperfusion injury. For decades, XOR involvement in pathologic processes has been established by salutary outcomes attained from treatment with the XOR inhibitor allopurinol. This has served to frame a working dogma that elevation of XOR-specific activity is associated with enhanced rates of reactive species generation that mediate negative outcomes. While adherence to this narrowly focused practice of designating elevated XOR activity to be "bad" has produced some benefit, it has also led to significant underdevelopment of the processes mediating XOR regulation, identification of alternative reactants and products as well as micro-environmental factors that alter enzymatic activity. This is exemplified by recent reports: (1) identifying XOR as a nitrite reductase and thus a source of beneficial nitric oxide ((•)NO) under in vivo conditions similar to those where XOR inhibition has been assumed an optimal treatment choice, (2) describing XOR-derived uric acid (UA) as a critical pro-inflammatory mediator in vascular and metabolic disease and (3) ascribing an antioxidant/protective role for XOR-derived UA. When taken together, these proposed and countervailing functions of XOR affirm the need for a more comprehensive evaluation of product formation as well as the factors that govern product identity. As such, this review will critically evaluate XOR-catalyzed oxidant, (•)NO and UA formation as well as identify factors that mediate their production, inhibition and the resultant impact on inflammatory disease.
Collapse
Affiliation(s)
- Eric E Kelley
- Department of Anesthesiology and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, W1357 BST, 200 Lothrop Street, Pittsburgh, PA, 15213, USA,
| |
Collapse
|
93
|
Regulation of oxygen delivery to the body via hypoxic vasodilation. Proc Natl Acad Sci U S A 2015; 112:6254-5. [PMID: 25944936 DOI: 10.1073/pnas.1506523112] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
94
|
Haugaa H, Gómez H, Maberry DR, Holder A, Ogundele O, Quintero AMB, Escobar D, Tønnessen TI, Airgood H, Dezfulian C, Kenny E, Shiva S, Zuckerbraun B, Pinsky MR. Effects of inhalation of low-dose nitrite or carbon monoxide on post-reperfusion mitochondrial function and tissue injury in hemorrhagic shock swine. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:184. [PMID: 25899004 PMCID: PMC4422303 DOI: 10.1186/s13054-015-0903-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/30/2015] [Indexed: 01/29/2023]
Abstract
Introduction Tissue reperfusion following hemorrhagic shock may paradoxically cause tissue injury and organ dysfunction by mitochondrial free radical expression. Both nitrite and carbon monoxide (CO) may protect from this reperfusion injury by limiting mitochondrial free radial production. We explored the effects of very small doses of inhaled nitrite and CO on tissue injury in a porcine model of hemorrhagic shock. Methods Twenty pigs (mean wt. 30.6 kg, range 27.2 to 36.4 kg) had microdialysis catheters inserted in muscle, peritoneum, and liver to measure lactate, pyruvate, glucose, glycerol, and nitrite. Nineteen of the pigs were bled at a rate of 20 ml/min to a mean arterial pressure of 30 mmHg and kept between 30 and 40 mmHg for 90 minutes and then resuscitated. One pig was instrumented but not bled (sham). Hemorrhaged animals were randomized to inhale nothing (control, n = 7), 11 mg nitrite (nitrite, n = 7) or 250 ppm CO (CO, n = 5) over 30 minutes before fluid resuscitation. Mitochondrial respiratory control ratio was measured in muscle biopsies. Repeated measures from microdialysis catheters were analyzed in a random effects mixed model. Results Neither nitrite nor CO had any effects on the measured hemodynamic variables. Following inhalation of nitrite, plasma, but not tissue, nitrite increased. Following reperfusion, plasma nitrite only increased in the control and CO groups. Thereafter, nitrite decreased only in the nitrite group. Inhalation of nitrite was associated with decreases in blood lactate, whereas both nitrite and CO were associated with decreases in glycerol release into peritoneal fluid. Following resuscitation, the muscular mitochondrial respiratory control ratio was reduced in the control group but preserved in the nitrite and CO groups. Conclusions We conclude that small doses of nebulized sodium nitrite or inhaled CO may be associated with intestinal protection during resuscitation from severe hemorrhagic shock. Electronic supplementary material The online version of this article (doi:10.1186/s13054-015-0903-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Håkon Haugaa
- Department of Critical Care Medicine, Cardiopulmonary Research Laboratory, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA. .,Department of Emergencies and Critical Care, Oslo University Hospital, Sognsvannsveien 27 0424, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Sognsvannsveien 20 0424, Oslo, Norway.
| | - Hernando Gómez
- Department of Critical Care Medicine, Cardiopulmonary Research Laboratory, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA. .,Center for Critical Care Nephrology, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - Donald R Maberry
- Department of Critical Care Medicine, Cardiopulmonary Research Laboratory, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA.
| | - Andre Holder
- Department of Critical Care Medicine, Cardiopulmonary Research Laboratory, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA.
| | - Olufunmilayo Ogundele
- Department of Critical Care Medicine, Cardiopulmonary Research Laboratory, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA.
| | - Ana Maria B Quintero
- Institute of Clinical Medicine, University of Oslo, Sognsvannsveien 20 0424, Oslo, Norway.
| | - Daniel Escobar
- Institute of Clinical Medicine, University of Oslo, Sognsvannsveien 20 0424, Oslo, Norway.
| | - Tor Inge Tønnessen
- Department of Emergencies and Critical Care, Oslo University Hospital, Sognsvannsveien 27 0424, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Sognsvannsveien 20 0424, Oslo, Norway.
| | - Hannah Airgood
- Department of Critical Care Medicine, Safar Center for Resuscitation Research University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - Cameron Dezfulian
- Department of Critical Care Medicine, Safar Center for Resuscitation Research University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - Elizabeth Kenny
- Department of Critical Care Medicine, Safar Center for Resuscitation Research University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA.
| | - Brian Zuckerbraun
- Department of Surgery, University of Pittsburgh, 3380 Boulevard of the Allies 390, Pittsburgh, PA, 15213, USA.
| | - Michael R Pinsky
- Department of Critical Care Medicine, Cardiopulmonary Research Laboratory, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
95
|
Tejero J, Gladwin MT. The globin superfamily: functions in nitric oxide formation and decay. Biol Chem 2014; 395:631-9. [PMID: 24477516 DOI: 10.1515/hsz-2013-0289] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/27/2014] [Indexed: 11/15/2022]
Abstract
Globin proteins are ubiquitous in living organisms and carry out a variety of functions related to the ability of their prosthetic heme group to bind gaseous ligands, such as oxygen, nitric oxide (NO), and CO. Moreover, they catalyze important reactions with nitrogen oxide species, such as NO dioxygenation and nitrite reduction. The formation of NO from nitrite is a reaction catalyzed by globins that has received increasing attention due to its potential as a hypoxic NO signaling mechanism. In this review, we revisit the current knowledge about the role of globins in NO formation and its physiological implications.
Collapse
|
96
|
Computational analysis of nitric oxide biotransport to red blood cell in the presence of free hemoglobin and NO donor. Microvasc Res 2014; 95:15-25. [DOI: 10.1016/j.mvr.2014.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 05/16/2014] [Accepted: 06/09/2014] [Indexed: 02/06/2023]
|
97
|
Keszler A, Brandal G, Baumgardt S, Ge ZD, Pratt PF, Riess ML, Bienengraeber M. Far red/near infrared light-induced protection against cardiac ischemia and reperfusion injury remains intact under diabetic conditions and is independent of nitric oxide synthase. Front Physiol 2014; 5:305. [PMID: 25202275 PMCID: PMC4141548 DOI: 10.3389/fphys.2014.00305] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/26/2014] [Indexed: 12/27/2022] Open
Abstract
Far red/near-infrared light (NIR) promotes a wide range of biological effects including tissue protection but whether and how NIR is capable of acutely protecting myocardium against ischemia and reperfusion injury in vivo is not fully elucidated. Our previous work indicates that NIR exposure immediately before and during early reperfusion protects the myocardium against infarction through mechanisms that are nitric oxide (NO)-dependent. Here we tested the hypothesis that NIR elicits protection in a diabetic mouse model where other cardioprotective interventions such as pre- and postconditioning fail, and that the protection is independent of nitric oxide synthase (NOS). NIR reduced infarct size dose dependently. Importantly, NIR-induced protection was preserved in a diabetic mouse model (db/db) and during acute hyperglycemia, as well as in endothelial NOS(-/-) mice and in wild type mice treated with NOS inhibitor L-NAME. In in vitro experiments NIR light liberates NO from nitrosyl hemoglobin (HbNO) and nitrosyl myoglobin (MbNO) in a wavelength-(660-830 nm) and dose-dependent manner. Irradiation at 660 nm yields the highest release of NO, while at longer wavelengths a dramatic decrease of NO release can be observed. Similar wavelength dependence was observed for the protection of mice against cardiac ischemia and reperfusion injury in vivo. NIR-induced NO release from deoxymyoglobin in the presence of nitrite mildly inhibits respiration of isolated mitochondria after hypoxia. In summary, NIR applied during reperfusion protects the myocardium against infarction in an NO-dependent, but NOS-independent mechanisms, whereby mitochondria may be a target of NO released by NIR, leading to reduced reactive oxygen species generation during reperfusion. This unique mechanism preserves protection even during diabetes where other protective strategies fail.
Collapse
Affiliation(s)
- Agnes Keszler
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Garth Brandal
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Shelley Baumgardt
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Zhi-Dong Ge
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Phillip F. Pratt
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Matthias L. Riess
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Anesthesiology, Clement J. Zablocki VA Medical CenterMilwaukee, WI, USA
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Martin Bienengraeber
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukee, WI, USA
| |
Collapse
|
98
|
Crosstalk between nitrite, myoglobin and reactive oxygen species to regulate vasodilation under hypoxia. PLoS One 2014; 9:e105951. [PMID: 25148388 PMCID: PMC4141839 DOI: 10.1371/journal.pone.0105951] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/28/2014] [Indexed: 11/19/2022] Open
Abstract
The systemic response to decreasing oxygen levels is hypoxic vasodilation. While this mechanism has been known for more than a century, the underlying cellular events have remained incompletely understood. Nitrite signaling is critically involved in vessel relaxation under hypoxia. This can be attributed to the presence of myoglobin in the vessel wall together with other potential nitrite reductases, which generate nitric oxide, one of the most potent vasodilatory signaling molecules. Questions remain relating to the precise concentration of nitrite and the exact dose-response relations between nitrite and myoglobin under hypoxia. It is furthermore unclear whether regulatory mechanisms exist which balance this interaction. Nitrite tissue levels were similar across all species investigated. We then investigated the exact fractional myoglobin desaturation in an ex vivo approach when gassing with 1% oxygen. Within a short time frame myoglobin desaturated to 58±12%. Given that myoglobin significantly contributes to nitrite reduction under hypoxia, dose-response experiments using physiological to pharmacological nitrite concentrations were conducted. Along all concentrations, abrogation of myoglobin in mice impaired vasodilation. As reactive oxygen species may counteract the vasodilatory response, we used superoxide dismutase and its mimic tempol as well as catalase and ebselen to reduce the levels of reactive oxygen species during hypoxic vasodilation. Incubation of tempol in conjunction with catalase alone and catalase/ebselen increased the vasodilatory response to nitrite. Our study shows that modest hypoxia leads to a significant nitrite-dependent vessel relaxation. This requires the presence of vascular myoglobin for both physiological and pharmacological nitrite levels. Reactive oxygen species, in turn, modulate this vasodilation response.
Collapse
|
99
|
Liu T, Schroeder HJ, Barcelo L, Bragg SL, Terry MH, Wilson SM, Power GG, Blood AB. Role of blood and vascular smooth muscle in the vasoactivity of nitrite. Am J Physiol Heart Circ Physiol 2014; 307:H976-86. [PMID: 25108012 DOI: 10.1152/ajpheart.00138.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent evidence from humans and rats indicates that nitrite is a vasodilator under hypoxic conditions by reacting with metal-containing proteins to produce nitric oxide (NO). We tested the hypothesis that near-physiological concentrations of nitrite would produce vasodilation in a hypoxia- and concentration-dependent manner in the hind limb of sheep. Anesthetized sheep were instrumented to measure arterial blood pressure and femoral blood flows continuously in both hind limbs. Nitrite was infused into one femoral artery to raise the nitrite concentration in the femoral vein by 10 to 15-fold while the sheep breathed 50%, 14% or 12% oxygen in inspired air. In contrast to reports in humans and rats, the nitrite infusion had no measurable effect on mean femoral blood flows or vascular conductances, regardless of inspired O2 levels. In vitro experiments showed no significant difference in the release of NO from nitrite in sheep and human red blood cells. Further experiments demonstrated nitrite is converted to NO in rat artery homogenates faster than sheep arteries, and that this source of NO production is attenuated in the presence of a heme oxidizer. Finally, western blots indicate that concentrations of the heme-containing protein cytoglobin, but not myoglobin, are markedly lower in sheep arteries compared with rats. Overall, the results demonstrate that nitrite is not a physiological vasodilator in sheep. This is likely due to a lack of conversion of nitrite to NO within the vascular smooth muscle, perhaps due to deficient amounts of the heme-containing protein cytoglobin.
Collapse
Affiliation(s)
- Taiming Liu
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, California
| | - Hobe J Schroeder
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; and
| | - Lisa Barcelo
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, California
| | - Shannon L Bragg
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; and
| | - Michael H Terry
- Department of Respiratory Care, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean M Wilson
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; and
| | - Gordon G Power
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; and
| | - Arlin B Blood
- Department of Pediatrics, Division of Neonatology, Loma Linda University School of Medicine, Loma Linda, California; Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California; and
| |
Collapse
|
100
|
The effect of intermittent pneumatic compression of legs on the levels of nitric oxide related species in blood and on arterial function in the arm. Nitric Oxide 2014; 40:117-22. [DOI: 10.1016/j.niox.2014.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/20/2014] [Accepted: 06/19/2014] [Indexed: 11/18/2022]
|