51
|
Neuman RB, Hayek SS, Poole JC, Rahman A, Menon V, Kavtaradze N, Polhemus D, Veledar E, Lefer DJ, Quyyumi AA. Nitric Oxide Contributes to Vasomotor Tone in Hypertensive African Americans Treated With Nebivolol and Metoprolol. J Clin Hypertens (Greenwich) 2016; 18:223-31. [PMID: 26285691 PMCID: PMC4760906 DOI: 10.1111/jch.12649] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/30/2015] [Accepted: 07/05/2015] [Indexed: 11/29/2022]
Abstract
Endothelial dysfunction is more prevalent in African Americans (AAs) compared with whites. The authors hypothesized that nebivolol, a selective β1 -antagonist that stimulates nitric oxide (NO), will improve endothelial function in AAs with hypertension when compared with metoprolol. In a double-blind, randomized, crossover study, 19 AA hypertensive patients were randomized to a 12-week treatment period with either nebivolol 10 mg or metoprolol succinate 100 mg daily. Forearm blood flow (FBF) was measured using plethysmography at rest and after intra-arterial infusion of acetylcholine and sodium nitroprusside to estimate endothelium-dependent and independent vasodilation, respectively. Physiologic vasodilation was assessed during hand-grip exercise. Measurements were repeated after NO blockade with L-N(G) -monomethylarginine (L-NMMA) and after inhibition of endothelium-derived hyperpolarizing factor (EDHF) with tetraethylammonium chloride (TEA). NO blockade with L-NMMA produced a trend toward greater vasoconstriction during nebivolol compared with metoprolol treatment (21% vs 12% reduction in FBF, P=.06, respectively). This difference was more significant after combined administration of L-NMMA and TEA (P<.001). Similarly, there was a contribution of NO to exercise-induced vasodilation during nebivolol but not during metoprolol treatment. There were significantly greater contributions of NO and EDHF to resting vasodilator tone and of NO to exercise-induced vasodilation with nebivolol compared with metoprolol in AAs with hypertension.
Collapse
Affiliation(s)
- Robert B. Neuman
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Salim S. Hayek
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Joseph C. Poole
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Ayaz Rahman
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Vivek Menon
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Nino Kavtaradze
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - David Polhemus
- Department of PharmacologyLouisiana State University Health Sciences CenterNew OrleansLA
| | - Emir Veledar
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - David J. Lefer
- Department of PharmacologyLouisiana State University Health Sciences CenterNew OrleansLA
| | | |
Collapse
|
52
|
Hazim S, Curtis PJ, Schär MY, Ostertag LM, Kay CD, Minihane AM, Cassidy A. Acute benefits of the microbial-derived isoflavone metabolite equol on arterial stiffness in men prospectively recruited according to equol producer phenotype: a double-blind randomized controlled trial. Am J Clin Nutr 2016; 103:694-702. [PMID: 26843154 PMCID: PMC4763500 DOI: 10.3945/ajcn.115.125690] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/28/2015] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND There is much speculation with regard to the potential cardioprotective benefits of equol, a microbial-derived metabolite of the isoflavone daidzein, which is produced in the large intestine after soy intake in 30% of Western populations. Although cross-sectional and retrospective data support favorable associations between the equol producer (EP) phenotype and cardiometabolic health, few studies have prospectively recruited EPs to confirm this association. OBJECTIVE The aim was to determine whether the acute vascular benefits of isoflavones differ according to EP phenotype and subsequently investigate the effect of providing commercially produced S-(-)equol to non-EPs. DESIGN We prospectively recruited male EPs and non-EPs (n = 14/group) at moderate cardiovascular risk into a double-blind, placebo-controlled crossover study to examine the acute effects of soy isoflavones (80-mg aglycone equivalents) on arterial stiffness [carotid-femoral pulse-wave velocity (cfPWV)], blood pressure, endothelial function (measured by using the EndoPAT 2000; Itamar Medical), and nitric oxide at baseline (0 h) and 6 and 24 h after intake. In a separate assessment, non-EPs consumed 40 mg S-(-)equol with identical vascular measurements performed 2 h after intake. RESULTS After soy intake, cfPWV significantly improved in EPs at 24 h (cfPWV change from 0 h: isoflavone, -0.2 ± 0.2 m/s; placebo, 0.6 ± 0.2 m/s; P < 0.01), which was significantly associated with plasma equol concentrations (R = -0.36, P = 0.01). No vascular effects were observed in EPs at 6 h or in non-EPs at any time point. Similarly, no benefit of commercially produced S-(-)equol was observed in non-EPs despite mean plasma equol concentrations reaching 3.2 μmol/L. CONCLUSIONS Acute soy intake improved cfPWV in EPs, equating to an 11-12% reduced risk of cardiovascular disease if sustained. However, a single dose of commercially produced equol had no cardiovascular benefits in non-EPs. These data suggest that the EP phenotype is critical in unlocking the vascular benefits of equol in men, and long-term trials should focus on confirming the implications of EP phenotype on cardiovascular health. This trial was registered at clinicaltrials.gov as NCT01530893.
Collapse
Affiliation(s)
- Sara Hazim
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Peter J Curtis
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Manuel Y Schär
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Luisa M Ostertag
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Colin D Kay
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Anne-Marie Minihane
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Aedín Cassidy
- Department of Nutrition, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
53
|
Ren X, Ding Y, Lu N. Nitrite attenuated peroxynitrite and hypochlorite generation in activated neutrophils. Eur J Pharmacol 2016; 775:50-6. [DOI: 10.1016/j.ejphar.2016.02.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 01/15/2023]
|
54
|
DeVan AE, Johnson LC, Brooks FA, Evans TD, Justice JN, Cruickshank-Quinn C, Reisdorph N, Bryan NS, McQueen MB, Santos-Parker JR, Chonchol MB, Bassett CJ, Sindler AL, Giordano T, Seals DR. Effects of sodium nitrite supplementation on vascular function and related small metabolite signatures in middle-aged and older adults. J Appl Physiol (1985) 2015; 120:416-25. [PMID: 26607249 DOI: 10.1152/japplphysiol.00879.2015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/20/2015] [Indexed: 12/20/2022] Open
Abstract
Insufficient nitric oxide (NO) bioavailability plays an important role in endothelial dysfunction and arterial stiffening with aging. Supplementation with sodium nitrite, a precursor of NO, ameliorates age-related vascular endothelial dysfunction and arterial stiffness in mice, but effects on humans, including the metabolic pathways altered, are unknown. The purpose of this study was to determine the safety, feasibility, and efficacy of oral sodium nitrite supplementation for improving vascular function in middle-aged and older adults and to identify related circulating metabolites. Ten weeks of sodium nitrite (80 or 160 mg/day, capsules, TheraVasc; randomized, placebo control, double blind) increased plasma nitrite acutely (5- to 15-fold, P < 0.001 vs. placebo) and chronically (P < 0.10) and was well tolerated without symptomatic hypotension or clinically relevant elevations in blood methemoglobin. Endothelial function, measured by brachial artery flow-mediated dilation, increased 45-60% vs. baseline (P < 0.10) without changes in body mass or blood lipids. Measures of carotid artery elasticity (ultrasound and applanation tonometry) improved (decreased β-stiffness index, increased cross-sectional compliance, P < 0.05) without changes in brachial or carotid artery blood pressure. Aortic pulse wave velocity was unchanged. Nitrite-induced changes in vascular measures were significantly related to 11 plasma metabolites identified by untargeted analysis. Baseline abundance of multiple metabolites, including glycerophospholipids and fatty acyls, predicted vascular changes with nitrite. This study provides evidence that sodium nitrite supplementation is well tolerated, increases plasma nitrite concentrations, improves endothelial function, and lessens carotid artery stiffening in middle-aged and older adults, perhaps by altering multiple metabolic pathways, thereby warranting a larger clinical trial.
Collapse
Affiliation(s)
- Allison E DeVan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado;
| | - Lawrence C Johnson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Forrest A Brooks
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Trent D Evans
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Nichole Reisdorph
- Integrated Department of Immunology, National Jewish Health, Denver, Colorado
| | | | - Matthew B McQueen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Michel B Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, Colorado
| | - Candace J Bassett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Amy L Sindler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
55
|
Kautza B, Gomez H, Escobar D, Corey C, Ataya B, Luciano J, Botero AM, Gordon L, Brumfield J, Martinez S, Holder A, Ogundele O, Pinsky M, Shiva S, Zuckerbraun BS. Inhaled, nebulized sodium nitrite protects in murine and porcine experimental models of hemorrhagic shock and resuscitation by limiting mitochondrial injury. Nitric Oxide 2015; 51:7-18. [PMID: 26410351 DOI: 10.1016/j.niox.2015.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The cellular injury that occurs in the setting of hemorrhagic shock and resuscitation (HS/R) affects all tissue types and can drive altered inflammatory responses. Resuscitative adjuncts hold the promise of decreasing such injury. Here we test the hypothesis that sodium nitrite (NaNO2), delivered as a nebulized solution via an inhalational route, protects against injury and inflammation from HS/R. METHODS Mice underwent HS/R to a mean arterial pressure (MAP) of 20 or 25 mmHg. Mice were resuscitated with Lactated Ringers after 90-120 min of hypotension. Mice were randomized to receive nebulized NaNO2 via a flow through chamber (30 mg in 5 mL PBS). Pigs (30-35 kg) were anesthetized and bled to a MAP of 30-40 mmHg for 90 min, randomized to receive NaNO2 (11 mg in 2.5 mL PBS) nebulized into the ventilator circuit starting 60 min into the hypotensive period, followed by initial resuscitation with Hextend. Pigs had ongoing resuscitation and support for up to four hours. Hemodynamic data were collected continuously. RESULTS NaNO2 limited organ injury and inflammation in murine hemorrhagic shock. A nitrate/nitrite depleted diet exacerbated organ injury, as well as mortality, and inhaled NaNO2 significantly reversed this effect. Furthermore, NaNO2 limited mitochondrial oxidant injury. In porcine HS/R, NaNO2 had no significant influence on shock induced hemodynamics. NaNO2 limited hypoxia/reoxia or HS/R-induced mitochondrial injury and promoted mitochondrial fusion. CONCLUSION NaNO2 may be a useful adjunct to shock resuscitation based on its limitation of mitochondrial injury.
Collapse
Affiliation(s)
| | - Hernando Gomez
- Department of Critical Care Medicine, USA; The Center for Critical Care Nephrology, USA
| | | | | | | | | | | | | | | | | | | | | | - Michael Pinsky
- Department of Critical Care Medicine, USA; The Center for Critical Care Nephrology, USA
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, USA; Vascular Medicine Institute, University of Pittsburgh, USA.
| | - Brian S Zuckerbraun
- VA Pittsburgh Healthcare System, USA; Department of Surgery, USA; The Center for Critical Care Nephrology, USA; Vascular Medicine Institute, University of Pittsburgh, USA.
| |
Collapse
|
56
|
Bryan NS, Ivy JL. Inorganic nitrite and nitrate: evidence to support consideration as dietary nutrients. Nutr Res 2015; 35:643-54. [DOI: 10.1016/j.nutres.2015.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 06/02/2015] [Accepted: 06/05/2015] [Indexed: 01/29/2023]
|
57
|
de Lima Portella R, Lynn Bickta J, Shiva S. Nitrite Confers Preconditioning and Cytoprotection After Ischemia/Reperfusion Injury Through the Modulation of Mitochondrial Function. Antioxid Redox Signal 2015; 23:307-27. [PMID: 26094636 DOI: 10.1089/ars.2015.6260] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Nitrite is now recognized as an intrinsic signaling molecule that mediates a number of biological processes. One of the most reproducible effects of nitrite is its ability to mediate cytoprotection after ischemia/reperfusion (I/R). This robust phenomenon has been reproduced by a number of investigators in varying animal models focusing on different target organs. Furthermore, nitrite's cytoprotective versatility is highlighted by its ability to mediate delayed preconditioning and remote conditioning in addition to acute protection. RECENT ADVANCES In the last 10 years, significant progress has been made in elucidating the mechanisms underlying nitrite-mediated ischemic tolerance. CRITICAL ISSUES The mitochondrion, which is essential to both the progression of I/R injury and the protection afforded by preconditioning, has emerged as a major subcellular target for nitrite. This review will outline the role of the mitochondrion in I/R injury and preconditioning, review the accumulated preclinical studies demonstrating nitrite-mediated cytoprotection, and finally focus on the known interactions of nitrite with mitochondria and their role in the mechanism of nitrite-mediated ischemic tolerance. FUTURE DIRECTIONS These studies set the stage for current clinical trials testing the efficacy of nitrite to prevent warm and cold I/R injury.
Collapse
Affiliation(s)
- Rafael de Lima Portella
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Janelle Lynn Bickta
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
58
|
Kobayashi J, Ohtake K, Uchida H. NO-Rich Diet for Lifestyle-Related Diseases. Nutrients 2015; 7:4911-37. [PMID: 26091235 PMCID: PMC4488823 DOI: 10.3390/nu7064911] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 02/07/2023] Open
Abstract
Decreased nitric oxide (NO) availability due to obesity and endothelial dysfunction might be causally related to the development of lifestyle-related diseases such as insulin resistance, ischemic heart disease, and hypertension. In such situations, instead of impaired NO synthase (NOS)-dependent NO generation, the entero-salivary nitrate-nitrite-NO pathway might serve as a backup system for NO generation by transmitting NO activities in the various molecular forms including NO and protein S-nitrosothiols. Recently accumulated evidence has demonstrated that dietary intake of fruits and vegetables rich in nitrate/nitrite is an inexpensive and easily-practicable way to prevent insulin resistance and vascular endothelial dysfunction by increasing the NO availability; a NO-rich diet may also prevent other lifestyle-related diseases, including osteoporosis, chronic obstructive pulmonary disease (COPD), and cancer. This review provides an overview of our current knowledge of NO generation through the entero-salivary pathway and discusses its safety and preventive effects on lifestyle-related diseases.
Collapse
Affiliation(s)
- Jun Kobayashi
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Saitama 350-0295, Japan.
| | - Kazuo Ohtake
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Saitama 350-0295, Japan.
| | - Hiroyuki Uchida
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmaceutical Science, Josai University, Saitama 350-0295, Japan.
| |
Collapse
|
59
|
Bahadoran Z, Ghasemi A, Mirmiran P, Azizi F, Hadaegh F. Beneficial effects of inorganic nitrate/nitrite in type 2 diabetes and its complications. Nutr Metab (Lond) 2015; 12:16. [PMID: 25991919 PMCID: PMC4436104 DOI: 10.1186/s12986-015-0013-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/07/2015] [Indexed: 12/17/2022] Open
Abstract
Background and aim The ability of inorganic nitrate and nitrite to convert to nitric oxide (NO), and some of its properties e.g. regulation of glucose metabolism, vascular homeostasis, and insulin signaling pathway, have recently raised the hypothesis that inorganic nitrate and nitrite could be potential therapeutic agents in type 2 diabetes. In this review, we reviewed experimental and clinical studies investigating the effect of nitrate/nitrite administration on various aspects of type 2 diabetes. Findings Studies showed that an altered metabolism of nitrate/nitrite and impaired NO pathway occurs in diabetes which could contribute to its complications. Some important beneficial properties, including regulation of glucose homeostasis and insulin signaling pathway, improvement of insulin resistance and vascular function, hypotensive, hypolipidemic as well as anti-inflammatory and anti-oxidative effects have been observed following administration of inorganic nitrate/nitrite. Conclusion It seems that dietary nitrate/nitrite could be a compensatory fuel for a disrupted nitrate/nitrite/NO pathway and related disorders in diabetes. Although some beneficial properties of nitrate/nitrite have been reported by experimental investigations, long-term clinical studies with various doses of inorganic nitrate/nitrite supplementation, are recommended to confirm these effects.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, and Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran ; Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, 19395-4763 Tehran, Iran
| |
Collapse
|
60
|
Polhemus DJ, Bradley JM, Islam KN, Brewster LP, Calvert JW, Tao YX, Chang CC, Pipinos II, Goodchild TT, Lefer DJ. Therapeutic potential of sustained-release sodium nitrite for critical limb ischemia in the setting of metabolic syndrome. Am J Physiol Heart Circ Physiol 2015; 309:H82-92. [PMID: 25910804 DOI: 10.1152/ajpheart.00115.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/13/2015] [Indexed: 01/12/2023]
Abstract
Nitrite is a storage reservoir of nitric oxide that is readily reduced to nitric oxide under pathological conditions. Previous studies have demonstrated that nitrite levels are significantly reduced in cardiovascular disease states, including peripheral vascular disease. We investigated the cytoprotective and proangiogenic actions of a novel, sustained-release formulation of nitrite (SR-nitrite) in a clinically relevant in vivo swine model of critical limb ischemia (CLI) involving central obesity and metabolic syndrome. CLI was induced in obese Ossabaw swine (n = 18) by unilateral external iliac artery deployment of a full cross-sectional vessel occlusion device positioned within an endovascular expanded polytetrafluoroethylene-lined nitinol stent-graft. At post-CLI day 14, pigs were randomized to placebo (n = 9) or SR-nitrite (80 mg, n = 9) twice daily by mouth for 21 days. SR-nitrite therapy increased nitrite, nitrate, and S-nitrosothiol in plasma and ischemic skeletal muscle. Oxidative stress was reduced in ischemic limb tissue of SR-nitrite- compared with placebo-treated pigs. Ischemic limb tissue levels of proangiogenic growth factors were increased following SR-nitrite therapy compared with placebo. Despite the increases in cytoprotective and angiogenic signals with SR-nitrite therapy, new arterial vessel formation and enhancement of blood flow to the ischemic limb were not different from placebo. Our data clearly demonstrate cytoprotective and proangiogenic signaling in ischemic tissues following SR-nitrite therapy in a very severe model of CLI. Further studies evaluating longer-duration nitrite therapy and/or additional nitrite dosing strategies are warranted to more fully evaluate the therapeutic potential of nitrite therapy in peripheral vascular disease.
Collapse
Affiliation(s)
- David J Polhemus
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Jessica M Bradley
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Kazi N Islam
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - Luke P Brewster
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia; Surgery and Research Services, Atlanta Veterans Affairs Medical Center, Decatur, Georgia; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - John W Calvert
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, Alabama
| | | | - Iraklis I Pipinos
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Traci T Goodchild
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana
| | - David J Lefer
- Cardiovascular Center of Excellence and Department of Pharmacology, LSU Health Sciences Center, New Orleans, Louisiana;
| |
Collapse
|
61
|
Seals DR, Kaplon RE, Gioscia-Ryan RA, LaRocca TJ. You're only as old as your arteries: translational strategies for preserving vascular endothelial function with aging. Physiology (Bethesda) 2015; 29:250-64. [PMID: 24985329 DOI: 10.1152/physiol.00059.2013] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endothelial dysfunction develops with age and increases the risk of age-associated vascular disorders. Nitric oxide insufficiency, oxidative stress, and chronic low-grade inflammation, induced by upregulation of adverse cellular signaling processes and imbalances in stress resistance pathways, mediate endothelial dysfunction with aging. Healthy lifestyle behaviors preserve endothelial function with aging by inhibiting these mechanisms, and novel nutraceutical compounds that favorably modulate these pathways hold promise as a complementary approach for preserving endothelial health.
Collapse
Affiliation(s)
- Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Rachelle E Kaplon
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Rachel A Gioscia-Ryan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Thomas J LaRocca
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
62
|
Lira VA. Exercise-induced cardioprotection: more to k‘NO’w. Cardiology 2015; 130:172-174. [PMID: 25720747 DOI: 10.1159/000375399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 01/19/2015] [Indexed: 11/19/2022]
Affiliation(s)
- Vitor A Lira
- Department of Health and Human Physiology, Obesity Research and Education Initiative, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
63
|
Bryan NS. The potential use of salivary nitrite as a marker of NO status in humans. Nitric Oxide 2015; 45:4-6. [DOI: 10.1016/j.niox.2014.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 12/16/2014] [Accepted: 12/17/2014] [Indexed: 01/22/2023]
|
64
|
Zhao J, Wang J, Yang Y, Lu Y. The Determination of Nitrate and Nitrite in Human Urine and Blood by High-Performance Liquid Chromatography and Cloud-Point Extraction. J Chromatogr Sci 2015; 53:1169-77. [PMID: 25616990 DOI: 10.1093/chromsci/bmu212] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Indexed: 11/14/2022]
Abstract
A simple efficient and practical separation/preconcentration coupled with HPLC method for the determination nitrate and low concentrations of nitrite in human urine and blood was investigated. The method is based on precolumn derivatization using the Griess reaction and cloud-point extraction (CPE) of nitrite anion and direct determination of nitrate using its UV absorbance by ion-pair HPLC. The chromatographic process with detection at two wavelengths (510 and 220 nm) allows the determination of nitrite and nitrate. Decolorization and protein precipitation of urine and blood was applied to overcome the interference of matrix and enhance the sensitivity. The method was validated for linearity, accuracy and precision. Under the optimum conditions, the linear range of nitrite from 10 to 1,000 ng/mL and nitrate from 0.1 to 10 µg/mL. Product recoveries ranged from 92.4 to 99.9%. The limits of detection were 1 ng/mL and 0.1 µg/mL for nitrite and nitrate, respectively. Therefore, the technique was simple and reliable, with potential application in biological sample analysis of nitrate and nitrite.
Collapse
Affiliation(s)
- Jiao Zhao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Jun Wang
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yaling Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Yunhui Lu
- The First People's Hospital of Yunnan Province, Kunming 650228, China
| |
Collapse
|
65
|
Hyde ER, Luk B, Cron S, Kusic L, McCue T, Bauch T, Kaplan H, Tribble G, Petrosino JF, Bryan NS. Characterization of the rat oral microbiome and the effects of dietary nitrate. Free Radic Biol Med 2014; 77:249-57. [PMID: 25305639 DOI: 10.1016/j.freeradbiomed.2014.09.017] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 09/10/2014] [Accepted: 09/11/2014] [Indexed: 01/09/2023]
Abstract
The nitrate-nitrite-NO pathway to nitric oxide (NO) production is a symbiotic pathway in mammals that is dependent on nitrate reducing oral commensal bacteria. Studies suggest that by contributing NO to the mammalian host, the oral microbiome helps maintain cardiovascular health. To begin to understand how changes in oral microbiota affect physiological functions such as blood pressure, we have characterized the Wistar rat nitrate reducing oral microbiome. Using 16S rRNA gene sequencing and analysis we compare the native Wistar rat tongue microbiome to that of healthy humans and to that of rats with sodium nitrate and chlorhexidine mouthwash treatments. We demonstrate that the rat tongue microbiome is less diverse than the human tongue microbiome, but that the physiological activity is comparable, as sodium nitrate supplementation significantly lowered diastolic blood pressure in Wistar rats and also lowers blood pressure (diastolic and systolic) in humans. We also show for the first time that sodium nitrate supplementation alters the abundance of specific bacterial species on the tongue. Our results suggest that the changes in oral nitrate reducing bacteria may affect nitric oxide availability and physiological functions such as blood pressure. Understanding individual changes in human oral microbiome may offer novel dietary approaches to restore NO availability and blood pressure.
Collapse
Affiliation(s)
- Embriette R Hyde
- Integrative Molecular and Biomedical Sciences Training Program, Baylor College of Medicine, Houston, TX 77030, USA; Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Berkley Luk
- Integrative Molecular and Biomedical Sciences Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stanley Cron
- Center for Nursing Research, School of Nursing, The University of Texas Health Science Center Houston, Houston, TX 77030, USA
| | - Lenka Kusic
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tyler McCue
- Integrative Molecular and Biomedical Sciences Training Program, Baylor College of Medicine, Houston, TX 77030, USA; Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tonya Bauch
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Heidi Kaplan
- Department of Microbiology and Molecular Genetics, Medical School, The University of Texas Health Science Center Houston, Houston, TX 77030, USA
| | - Gena Tribble
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX 77054, USA
| | - Joseph F Petrosino
- Integrative Molecular and Biomedical Sciences Training Program, Baylor College of Medicine, Houston, TX 77030, USA; Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Nathan S Bryan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center Houston, Houston, TX 77030, USA.
| |
Collapse
|
66
|
Neuman R, Hayek S, Rahman A, Poole JC, Menon V, Sher S, Newman JL, Karatela S, Polhemus D, Lefer DJ, De Staercke C, Hooper C, Quyyumi AA, Roback JD. Effects of storage-aged red blood cell transfusions on endothelial function in hospitalized patients. Transfusion 2014; 55:782-90. [PMID: 25393772 DOI: 10.1111/trf.12919] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Clinical and animal studies indicate that transfusions of older stored red blood cells (RBCs) impair clinical outcomes as compared to fresh RBC transfusions. It has been suggested that this effect is due to inhibition of nitric oxide (NO)-mediated vasodilation after transfusion of older RBC units. However, to date this effect has not been identified in human transfusion recipients. STUDY DESIGN AND METHODS Forty-three hospitalized patients with transfusion orders were randomly assigned to receive either fresh (<14 days) or older stored (>21 days) RBC units. Before transfusion, and at selected time points after the start of transfusion, endothelial function was assessed using noninvasive flow-mediated dilation assays. RESULTS After transfusion of older RBC units, there was a significant reduction in NO-mediated vasodilation at 24 hours after transfusion (p = 0.045), while fresh RBC transfusions had no effect (p = 0.231). CONCLUSIONS This study suggests for the first time a significant inhibitory effect of transfused RBC units stored more than 21 days on NO-mediated vasodilation in anemic hospitalized patients. This finding lends further support to the hypothesis that deranged NO signaling mediates adverse clinical effects of older RBC transfusions. Future investigations will be necessary to address possible confounding factors and confirm these results.
Collapse
Affiliation(s)
- Robert Neuman
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Salim Hayek
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Ayaz Rahman
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Joseph C Poole
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Vivek Menon
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Salman Sher
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - James L Newman
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Emory University School of Medicine, Atlanta, Georgia
| | - Sulaiman Karatela
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Emory University School of Medicine, Atlanta, Georgia
| | - David Polhemus
- Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - David J Lefer
- Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Christine De Staercke
- National Center on Birth Defects and Developmental Disabilities, Division of Blood Disorders, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Craig Hooper
- National Center on Birth Defects and Developmental Disabilities, Division of Blood Disorders, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Arshed A Quyyumi
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - John D Roback
- Department of Pathology and Laboratory Medicine, Center for Transfusion and Cellular Therapies, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
67
|
Justice JN, Gioscia-Ryan RA, Johnson LC, Battson ML, de Picciotto NE, Beck HJ, Jiang H, Sindler AL, Bryan NS, Enoka RM, Seals DR. Sodium nitrite supplementation improves motor function and skeletal muscle inflammatory profile in old male mice. J Appl Physiol (1985) 2014; 118:163-9. [PMID: 25377884 DOI: 10.1152/japplphysiol.00608.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Aging is associated with motor declines that lead to functional limitations and disability, necessitating the development of therapies to slow or reverse these events. We tested the hypothesis that sodium nitrite supplementation attenuates declines in motor function in older C57BL/6 mice. Motor function was assessed using a battery of tests (grip strength, open-field distance, rota-rod endurance) in old animals (age 20-24 mo) at baseline and after 8 wk of sodium nitrite (old nitrite, n = 22, 50 mg/liter) or no treatment (old control, n = 40), and in young reference animals (3 mo, n = 87). Eight weeks of sodium nitrite supplementation improved grip strength (old nitrite, +12.0 ± 14.9% vs. old control, +1.5 ± 15.2%, P < 0.05) and open field distance (old nitrite, +9.5 ± 7.7%, P < 0.01 vs. old control, -28.1 ± 2.0%) and completely restored rota-rod endurance-run time (old nitrite, +3.2 ± 7.1%, P < 0.01 vs. old control, -21.5 ± 7.2%; old nitrite after treatment P > 0.05 vs. young reference). Inflammatory cytokines were markedly increased in quadriceps of old compared with young reference animals (by ELISA, interleukin-1β [IL-1β] 3.86 ± 2.34 vs. 1.11 ± 0.74, P < 0.05; interferon-gamma [INF-γ] 8.31 ± 1.59 vs. 3.99 ± 2.59, P < 0.01; tumor necrosis factor-alpha [TNF-α] 1.69 ± 0.44 vs. 0.76 ± 0.30 pg/ml, P < 0.01), but were reduced to young reference levels after treatment (old nitrite, IL-1β 0.67 ± 0.95; INF-γ 5.22 ± 2.01, TNF-α 1.21 ± 0.39 pg/ml, P < 0.05 vs. old control, P > 0.05 vs. young reference). Cytokine expression and treatment (old nitrite vs. old control) predicted strength (R(2) = 0.822, P < 0.001, IL-1β, INF-γ, group), open field distance (R(2) = 0.574, P < 0.01, IL-1β, group) and endurance run time (R(2) = 0.477, P < 0.05, INF-γ). Our results suggest that sodium nitrite improves motor function in old mice, in part by reducing low-grade inflammation in muscle.
Collapse
Affiliation(s)
- Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Rachel A Gioscia-Ryan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Lawrence C Johnson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Micah L Battson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Natalie E de Picciotto
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Hannah J Beck
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Hong Jiang
- Texas Therapeutic Institute, Institute for Molecular Medicine, University of Texas Houston Health Sciences Center, Houston, Texas
| | - Amy L Sindler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Nathan S Bryan
- Texas Therapeutic Institute, Institute for Molecular Medicine, University of Texas Houston Health Sciences Center, Houston, Texas
| | - Roger M Enoka
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado; and
| |
Collapse
|
68
|
Dungel P, Hartinger J, Chaudary S, Slezak P, Hofmann A, Hausner T, Strassl M, Wintner E, Redl H, Mittermayr R. Low level light therapy by LED of different wavelength induces angiogenesis and improves ischemic wound healing. Lasers Surg Med 2014; 46:773-80. [PMID: 25363448 DOI: 10.1002/lsm.22299] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Low-level light therapy (LLLT) has been revealed as a potential means to improve wound healing. So far, most studies are being performed with irradiation in the red to near-infrared spectra. Recently, we showed that blue light (470 nm) can significantly influence biological systems such as nitric oxide (NO) metabolism and is able to release NO from nitrosyl-hemoglobin or mitochondrial protein complexes. Therefore, the aim of this study was to evaluate and compare the therapeutic value of blue or red light emitting diodes (LEDs) on wound healing in an ischemia disturbed rodent flap model. STUDY DESIGN/MATERIALS AND METHODS An abdominal flap was rendered ischemic by ligation of one epigastric bundle and subjected to LED illumination with a wavelength of 470 nm (blue, n = 8) or 629 nm (red, n = 8) each at 50 mW/cm(2) and compared to a non-treated control group (n = 8). Illumination was performed for 10 minutes on five consecutive days. RESULTS LED therapy with both wavelengths significantly increased angiogenesis in the sub-epidermal layer and intramuscularly (panniculus carnosus muscle) which was associated with significantly improved tissue perfusion 7 days after the ischemic insult. Accordingly, tissue necrosis was significantly reduced and shrinkage significantly less pronounced in the LED-treated groups of both wavelengths. CONCLUSIONS LED treatment of ischemia challenged tissue improved early wound healing by enhancing angiogenesis irrespective of the wavelength thus delineating this noninvasive means as a potential, cost effective tool in complicated wounds.
Collapse
Affiliation(s)
- Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Rassaf T, Ferdinandy P, Schulz R. Nitrite in organ protection. Br J Pharmacol 2014; 171:1-11. [PMID: 23826831 DOI: 10.1111/bph.12291] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 06/09/2013] [Accepted: 06/21/2013] [Indexed: 12/15/2022] Open
Abstract
In the last decade, the nitrate-nitrite-nitric oxide pathway has emerged to therapeutical importance. Modulation of endogenous nitrate and nitrite levels with the subsequent S-nitros(yl)ation of the downstream signalling cascade open the way for novel cytoprotective strategies. In the following, we summarize the actual literature and give a short overview on the potential of nitrite in organ protection.
Collapse
Affiliation(s)
- Tienush Rassaf
- Department of Medicine, Division of Cardiology, Pulmonary and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|
70
|
Inhibitory effect of nitrite on coagulation processes demonstrated by thrombelastography. Nitric Oxide 2014; 40:45-51. [PMID: 24858214 DOI: 10.1016/j.niox.2014.05.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/13/2014] [Accepted: 05/15/2014] [Indexed: 02/08/2023]
Abstract
Nitric oxide (NO) can be generated by two-step reduction pathway in which nitrate is converted first into nitrite and then into NO via several mechanisms, as well as from arginine by endogenous nitric oxide synthase (NOS). We have recently shown that nitrite ions in the presence of erythrocytes inhibit platelet aggregation and activation, as measured by aggregometry and flow cytometric analysis of P-selectin, through its reduction to NO under partially deoxygenated conditions. In the current study, we investigated how nitrite may affect overall clotting processes via modulating platelet function using thrombelastography (TEG). We measured three major TEG parameters, reaction time (R, time to initial fibrin formation), α angle (velocity of clot growth) and maximum amplitude (MA, maximum clot strength) using blood from healthy volunteers. An NO donor (DEANONOate) showed inhibitory effects on all TEG parameters in platelet rich plasma (PRP) and whole blood, resulting in delayed R, decreased angle, and reduced MA in a dose dependent manner. Nitrite ions also exhibited inhibitory effects in whole blood at 20% hematocrit, and this was greatly enhanced under hypoxic conditions, being demonstrable at 0.1 μM concentration. Neither compound changed any TEG parameters in plasma. Our results suggest that nitrite affects overall blood clotting and that TEG may be used to follow this process. Further the physiological effects of factors which determine NO bioavailability, such as endogenous levels of blood and tissue nitrite, may be useful as biomarkers for predicting hemostatic potential.
Collapse
|
71
|
Affiliation(s)
- Luisa B. Maia
- REQUIMTE/CQFB, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - José J. G. Moura
- REQUIMTE/CQFB, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
72
|
Metagenomic analysis of nitrate-reducing bacteria in the oral cavity: implications for nitric oxide homeostasis. PLoS One 2014; 9:e88645. [PMID: 24670812 PMCID: PMC3966736 DOI: 10.1371/journal.pone.0088645] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/08/2014] [Indexed: 01/09/2023] Open
Abstract
The microbiota of the human lower intestinal tract helps maintain healthy host physiology, for example through nutrient acquisition and bile acid recycling, but specific positive contributions of the oral microbiota to host health are not well established. Nitric oxide (NO) homeostasis is crucial to mammalian physiology. The recently described entero-salivary nitrate-nitrite-nitric oxide pathway has been shown to provide bioactive NO from dietary nitrate sources. Interestingly, this pathway is dependent upon oral nitrate-reducing bacteria, since humans lack this enzyme activity. This pathway appears to represent a newly recognized symbiosis between oral nitrate-reducing bacteria and their human hosts in which the bacteria provide nitrite and nitric oxide from nitrate reduction. Here we measure the nitrate-reducing capacity of tongue-scraping samples from six healthy human volunteers, and analyze metagenomes of the bacterial communities to identify bacteria contributing to nitrate reduction. We identified 14 candidate species, seven of which were not previously believed to contribute to nitrate reduction. We cultivated isolates of four candidate species in single- and mixed-species biofilms, revealing that they have substantial nitrate- and nitrite-reduction capabilities. Colonization by specific oral bacteria may thus contribute to host NO homeostasis by providing nitrite and nitric oxide. Conversely, the lack of specific nitrate-reducing communities may disrupt the nitrate-nitrite-nitric oxide pathway and lead to a state of NO insufficiency. These findings may also provide mechanistic evidence for the oral systemic link. Our results provide a possible new therapeutic target and paradigm for NO restoration in humans by specific oral bacteria.
Collapse
|
73
|
Li J, Jia W, Zhao Q. Excessive nitrite affects zebrafish valvulogenesis through yielding too much NO signaling. PLoS One 2014; 9:e92728. [PMID: 24658539 PMCID: PMC3962429 DOI: 10.1371/journal.pone.0092728] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/25/2014] [Indexed: 11/19/2022] Open
Abstract
Sodium nitrite, a common food additive, exists widely not only in the environment but also in our body. Excessive nitrite causes toxicological effects on human health; however, whether it affects vertebrate heart valve development remains unknown. In vertebrates, developmental defects of cardiac valves usually lead to congenital heart disease. To understand the toxic effects of nitrite on valvulogenesis, we exposed zebrafish embryos with different concentrations of sodium nitrite. Our results showed that sodium nitrite caused developmental defects of zebrafish heart dose dependently. It affected zebrafish heart development starting from 36 hpf (hour post fertilization) when heart initiates looping process. Comprehensive analysis on the embryos at 24 hpf and 48 hpf showed that excessive nitrite did not affect blood circulation, vascular network, myocardium and endocardium development. But development of endocardial cells in atrioventricular canal (AVC) of the embryos at 48 hpf was disrupted by too much nitrite, leading to defective formation of primitive valve leaflets at 76 hpf. Consistently, excessive nitrite diminished expressions of valve progenitor markers including bmp4, has2, vcana and notch1b at 48 hpf. Furthermore, 3', 5'-cyclic guanosine monophosphate (cGMP), downstream of nitric oxide (NO) signaling, was increased its level significantly in the embryos exposed with excessive nitrite and microinjection of soluble guanylate cyclase inhibitor ODQ (1H-[1], [2], [4]Oxadiazolo[4,3-a] quinoxalin-1-one), an antagonist of NO signaling, into nitrite-exposed embryos could partly rescue the cardiac valve malformation. Taken together, our results show that excessive nitrite affects early valve leaflet formation by producing too much NO signaling.
Collapse
Affiliation(s)
- Junbo Li
- Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Wenshuang Jia
- Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Qingshun Zhao
- Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| |
Collapse
|
74
|
Sindler AL, Devan AE, Fleenor BS, Seals DR. Inorganic nitrite supplementation for healthy arterial aging. J Appl Physiol (1985) 2014; 116:463-77. [PMID: 24408999 PMCID: PMC3949212 DOI: 10.1152/japplphysiol.01100.2013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 01/03/2014] [Indexed: 12/12/2022] Open
Abstract
Aging is the major risk factor for cardiovascular diseases (CVD). This is attributable primarily to adverse changes in arteries, notably, increases in large elastic artery stiffness and endothelial dysfunction mediated by inadequate concentrations of the vascular-protective molecule, nitric oxide (NO), and higher levels of oxidative stress and inflammation. Inorganic nitrite is a promising precursor molecule for augmenting circulating and tissue NO bioavailability because it requires only a one-step reduction to NO. Nitrite also acts as an independent signaling molecule, exerting many of the effects previously attributed to NO. Results of recent studies indicate that nitrite may be effective in the treatment of vascular aging. In old mice, short-term oral sodium nitrite supplementation reduces aortic pulse wave velocity, the gold-standard measure of large elastic artery stiffness, and ameliorates endothelial dysfunction, as indicated by normalization of NO-mediated endothelium-dependent dilation. These improvements in age-related vascular dysfunction with nitrite are mediated by reductions in oxidative stress and inflammation, and may be linked to increases in mitochondrial biogenesis and health. Increasing nitrite levels via dietary intake of nitrate appears to have similarly beneficial effects in many of the same physiological and clinical settings. Several clinical trials are being performed to determine the broad therapeutic potential of increasing nitrite bioavailability on human health and disease, including studies related to vascular aging. In summary, inorganic nitrite, as well as dietary nitrate supplementation, represents a promising therapy for treatment of arterial aging and prevention of age-associated CVD in humans.
Collapse
Affiliation(s)
- Amy L Sindler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | | | | | | |
Collapse
|
75
|
Jiang H, Torregrossa AC, Potts A, Pierini D, Aranke M, Garg HK, Bryan NS. Dietary nitrite improves insulin signaling through GLUT4 translocation. Free Radic Biol Med 2014; 67:51-7. [PMID: 24157451 DOI: 10.1016/j.freeradbiomed.2013.10.809] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 01/31/2023]
Abstract
Diabetes mellitus type 2 is a syndrome of disordered metabolism with inappropriate hyperglycemia owing to a reduction in the biological effectiveness of insulin. Type 2 diabetes is associated with an impaired nitric oxide (NO) pathway that probably serves as the key link between metabolic disorders and cardiovascular disease. Insulin-mediated translocation of GLUT4 involves the PI3K/Akt kinase signal cascade that results in activation of endothelial NO synthase (eNOS). eNOS is dysfunctional during diabetes. We hypothesize that loss of eNOS-derived NO terminates the signaling cascade and therefore cannot activate GLUT4 translocation and that dietary nitrite may repair this pathway. In this study, we administered 50mg/L sodium nitrite to db/db diabetic mice for 4 weeks. After 4 weeks treatment, the db/db mice experienced less weight gain, improved fasting glucose levels, and reduced insulin levels. Cell culture experiments using CHO-HIRc-myc-GLUT4eGFP cell lines stably expressing insulin receptor and myc-GLUT4eGFP protein, as well as L6 skeletal muscle cells stably expressing rat GLUT4 with a Myc epitope (L6-GLUT4myc), showed that NO, nitrite, and GSNO stimulate GLUT4 translocation independent of insulin, which is inhibited by NEM. Collectively our data suggest that nitrite improves insulin signaling through restoration of NO-dependent nitrosation of GLUT4 signaling translocation. These data suggest that NO-mediated nitrosation of GLUT4 by nitrite or other nitrosating agents is necessary and sufficient for GLUT4 translocation in target tissue. Description of this pathway may justify a high-nitrate/nitrite diet along with the glycemic index to provide a safe and nutritional regimen for the management and treatment of diabetes.
Collapse
Affiliation(s)
- Hong Jiang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Ashley C Torregrossa
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Amy Potts
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Dan Pierini
- California State University at Fullerton, Fullerton, CA 92831, USA
| | - Mayank Aranke
- The University of Texas at Austin, Austin, TX 78712, USA
| | - Harsha K Garg
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Nathan S Bryan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA.
| |
Collapse
|
76
|
Umbrello M, Dyson A, Feelisch M, Singer M. The key role of nitric oxide in hypoxia: hypoxic vasodilation and energy supply-demand matching. Antioxid Redox Signal 2013; 19:1690-710. [PMID: 23311950 DOI: 10.1089/ars.2012.4979] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
SIGNIFICANCE A mismatch between energy supply and demand induces tissue hypoxia with the potential to cause cell death and organ failure. Whenever arterial oxygen concentration is reduced, increases in blood flow--hypoxic vasodilation--occur in an attempt to restore oxygen supply. Nitric oxide (NO) is a major signaling and effector molecule mediating the body's response to hypoxia, given its unique characteristics of vasodilation (improving blood flow and oxygen supply) and modulation of energetic metabolism (reducing oxygen consumption and promoting utilization of alternative pathways). RECENT ADVANCES This review covers the role of oxygen in metabolism and responses to hypoxia, the hemodynamic and metabolic effects of NO, and mechanisms underlying the involvement of NO in hypoxic vasodilation. Recent insights into NO metabolism will be discussed, including the role for dietary intake of nitrate, endogenous nitrite (NO₂⁻) reductases, and release of NO from storage pools. The processes through which NO levels are elevated during hypoxia are presented, namely, (i) increased synthesis from NO synthases, increased reduction of NO₂⁻ to NO by heme- or pterin-based enzymes and increased release from NO stores, and (ii) reduced deactivation by mitochondrial cytochrome c oxidase. CRITICAL ISSUES Several reviews covered modulation of energetic metabolism by NO, while here we highlight the crucial role NO plays in achieving cardiocirculatory homeostasis during acute hypoxia through both vasodilation and metabolic suppression. FUTURE DIRECTIONS We identify a key position for NO in the body's adaptation to an acute energy supply-demand mismatch.
Collapse
Affiliation(s)
- Michele Umbrello
- 1 Department of Medicine, Bloomsbury Institute of Intensive Care Medicine, University College London , London, United Kingdom
| | | | | | | |
Collapse
|
77
|
Nicholson CK, Lambert JP, Chow CW, Lefer DJ, Calvert JW. Chronic exercise downregulates myocardial myoglobin and attenuates nitrite reductase capacity during ischemia-reperfusion. J Mol Cell Cardiol 2013; 64:1-10. [PMID: 23962643 PMCID: PMC3800246 DOI: 10.1016/j.yjmcc.2013.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/22/2013] [Accepted: 08/07/2013] [Indexed: 01/31/2023]
Abstract
The infarct sparing effects of exercise are evident following both long-term and short-term training regimens. Here we compared the infarct-lowering effects of nitrite therapy, voluntary exercise, and the combination of both following myocardial ischemia-reperfusion (MI/R) injury. We also compared the degree to which each strategy increased cardiac nitrite levels, as well as the effects of each strategy on the nitrite reductase activity of the heart. Mice subjected to voluntary wheel running (VE) for 4weeks displayed an 18% reduction in infarct size when compared to sedentary mice, whereas mice administered nitrite therapy (25mg/L in drinking water) showed a 53% decrease. However, the combination of VE and nitrite exhibited no further protection than VE alone. Although the VE and nitrite therapy mice showed similar nitrite levels in the heart, cardiac nitrite reductase activity was significantly reduced in the VE mice. Additionally, the cardiac protein expression of myoglobin, a known nitrite reductase, was also reduced after VE. Further studies revealed that cardiac NFAT activity was lower after VE due to a decrease in calcineurin activity and an increase in GSK3β activity. These data suggest that VE downregulates cardiac myoglobin levels by inhibiting calcineurin/NFAT signaling. Additionally, these results suggest that the modest infarct sparing effects of VE are the result of a decrease in the hearts ability to reduce nitrite to nitric oxide during MI/R.
Collapse
Affiliation(s)
- Chad K. Nicholson
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA 30308
| | - Jonathan P. Lambert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA 30308
| | - Chi-Wing Chow
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David J. Lefer
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA 30308
| | - John W. Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA 30308
| |
Collapse
|
78
|
Krenz M, Baines C, Kalogeris T, Korthuis R. Cell Survival Programs and Ischemia/Reperfusion: Hormesis, Preconditioning, and Cardioprotection. ACTA ACUST UNITED AC 2013. [DOI: 10.4199/c00090ed1v01y201309isp044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
79
|
Bailey J, Arif S, Mascaro J, Bonser RS, Frenneaux MP, Madhani M. Effect of nitrite treatment on endothelial nitric oxide synthase in human left ventricle biopsy. BMC Pharmacol Toxicol 2013. [PMCID: PMC3765628 DOI: 10.1186/2050-6511-14-s1-p3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
80
|
Jones JA, Ninnis JR, Hopper AO, Ibrahim Y, Merritt TA, Wan KW, Power GG, Blood AB. Nitrite and nitrate concentrations and metabolism in breast milk, infant formula, and parenteral nutrition. JPEN J Parenter Enteral Nutr 2013; 38:856-66. [PMID: 23894175 DOI: 10.1177/0148607113496118] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Dietary nitrate and nitrite are sources of gastric NO, which modulates blood flow, mucus production, and microbial flora. However, the intake and importance of these anions in infants is largely unknown. Nitrate and nitrite levels were measured in breast milk of mothers of preterm and term infants, infant formulas, and parenteral nutrition. Nitrite metabolism in breast milk was measured after freeze-thawing, at different temperatures, varying oxygen tensions, and after inhibition of potential nitrite-metabolizing enzymes. Nitrite concentrations averaged 0.07 ± 0.01 μM in milk of mothers of preterm infants, less than that of term infants (0.13 ± 0.02 μM) (P < .01). Nitrate concentrations averaged 13.6 ± 3.7 μM and 12.7 ± 4.9 μM, respectively. Nitrite and nitrate concentrations in infant formulas varied from undetectable to many-fold more than breast milk. Concentrations in parenteral nutrition were equivalent to or lower than those of breast milk. Freeze-thawing decreased nitrite concentration ~64%, falling with a half-life of 32 minutes at 37°C. The disappearance of nitrite was oxygen-dependent and prevented by ferricyanide and 3 inhibitors of lactoperoxidase. Nitrite concentrations in breast milk decrease with storage and freeze-thawing, a decline likely mediated by lactoperoxidase. Compared to adults, infants ingest relatively little nitrite and nitrate, which may be of importance in the modulation of blood flow and the bacterial flora of the infant GI tract, especially given the protective effects of swallowed nitrite.
Collapse
Affiliation(s)
| | - Janet R Ninnis
- Department of Pediatrics, Division of Neonatology Posthumous
| | | | | | - T Allen Merritt
- Department of Pediatrics, Division of Neonatology Poznan University of Medical Sciences, Poznan, Poland
| | - Kim-Wah Wan
- Neonatal Intensive Care Pharmacy, Loma Linda University, Loma Linda, California
| | | | - Arlin B Blood
- Department of Pediatrics, Division of Neonatology Center for Perinatal Biology
| |
Collapse
|
81
|
Dungel P, Teuschl AH, Banerjee A, Paier-Pourani J, Redl H, Kozlov AV. Impact of mitochondria on nitrite metabolism in HL-1 cardiomyocytes. Front Physiol 2013; 4:101. [PMID: 23730288 PMCID: PMC3657631 DOI: 10.3389/fphys.2013.00101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/23/2013] [Indexed: 12/17/2022] Open
Abstract
Apart from ATP synthesis mitochondria have many other functions, one being nitrite reductase activity. Nitric oxide (NO) released from nitrite has been shown to protect the heart from ischemia/reperfusion (I/R) injury in a cGMP-dependent manner. However, the exact impact of mitochondria on the release of NO from nitrite in cardiomyocytes is not completely understood. Besides mitochondria, a number of non-mitochondrial metalloproteins have been suggested to facilitate this process. The aim of this study was to investigate the impact of mitochondria on the bioactivation of nitrite in HL-1 cardiomyocytes. The levels of nitrosyl complexes of hemoglobin (NO-Hb) and cGMP levels were measured by electron spin resonance spectroscopy and enzyme immunoassay. In addition the formation of free NO was determined by confocal microscopy as well as intracellular nitrite and S-nitrosothiols by chemoluminescence analysis. NO was released from nitrite in cell culture in an oxygen-dependent manner. Application of specific inhibitors of the respiratory chain, p450, NO synthases (NOS) and xanthine oxidoreductase (XOR) showed that all four enzymatic systems are involved in the release of NO, but more than 50% of NO is released via the mitochondrial pathway. Only NO released by mitochondria activated cGMP synthesis. Cardiomyocytes co-cultured with red blood cells (RBC) competed with RBC for nitrite, but free NO was detected only in HL-1 cells suggesting that RBC are not a source of NO in this model. Apart from activation of cGMP synthesis, NO formed in HL-1 cells diffused out of the cells and formed NO-Hb complexes. In addition nitrite was converted by HL-1 cells to S-nitrosyl complexes. In HL-1 cardiomyocytes, several enzymatic systems are involved in nitrite reduction to NO but only the mitochondrial pathway of NO release activates cGMP synthesis. Our data suggest that this pathway may be a key regulator of myocardial contractility especially under hypoxic conditions.
Collapse
Affiliation(s)
- Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA research centre Vienna, Austria
| | | | | | | | | | | |
Collapse
|
82
|
Low-dose sodium nitrite attenuates myocardial ischemia and vascular ischemia-reperfusion injury in human models. J Am Coll Cardiol 2013; 61:2534-41. [PMID: 23623914 DOI: 10.1016/j.jacc.2013.03.050] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 03/09/2013] [Accepted: 03/13/2013] [Indexed: 01/24/2023]
Abstract
OBJECTIVES The aim of this study was to assess the potential benefits of inorganic nitrite in 2 clinical models: stress-induced myocardial ischemia and whole-arm ischemia-reperfusion. BACKGROUND Inorganic nitrite, traditionally considered a relatively inert metabolite of nitric oxide, may exert vasomodulatory and vasoprotective effects. Despite promising results from animal models, few have shown effectiveness in human model systems, and none have fully translated to the clinical setting. METHODS In 10 patients with inducible myocardial ischemia, saline and low-dose sodium nitrite (NaNO₂) (1.5 μmol/min for 20 min) were administered in a double-blind fashion during dobutamine stress echocardiography, at separate visits and in a random order; long-axis myocardial function was quantified by peak systolic velocity (Vs) and strain rate (SR) responses. In 19 healthy subjects, flow-mediated dilation was assessed before and after whole-arm ischemia-reperfusion; nitrite was given before ischemia or during reperfusion. RESULTS Comparing saline and nitrite infusions, Vs and SR at peak dobutamine increased in regions exhibiting ischemia (Vs from 9.5 ± 0.5 cm/s to 12.4 ± 0.6 cm/s, SR from -2.0 ± 0.2 s(-1) to -2.8 ± 0.3 s(-1)), whereas they did not change in normally functioning regions (Vs from 12.6 ± 0.4 cm/s to 12.6 ± 0.6 cm/s, SR from -2.6 ± 0.3 s(-1) to -2.3 ± 0.1 s(-1)) (p < 0.001, analysis of variance). With NaNO2, the increment of Vs (normalized for increase in heart rate) increased only in poorly functioning myocardial regions (+122%, p < 0.001). Peak flow-mediated dilation decreased by 43% after ischemia-reperfusion when subjects received only saline (6.8 ± 0.7% vs. 3.9 ± 0.7%, p < 0.01); administration of NaNO2 before ischemia prevented this decrease in flow-mediated dilation (5.9 ± 0.7% vs. 5.2 ± 0.5%, p = NS), whereas administration during reperfusion did not. CONCLUSIONS Low-dose NaNO₂ improves functional responses in ischemic myocardium but has no effect on normal regions. Low-dose NaNO₂ protects against vascular ischemia-reperfusion injury only when it is given before the onset of ischemia.
Collapse
|
83
|
Alexander JT, El-Ali AM, Newman JL, Karatela S, Predmore BL, Lefer DJ, Sutliff RL, Roback JD. Red blood cells stored for increasing periods produce progressive impairments in nitric oxide-mediated vasodilation. Transfusion 2013; 53:2619-2628. [PMID: 23480490 DOI: 10.1111/trf.12111] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 11/20/2012] [Accepted: 12/01/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND Clinical outcomes in transfused patients may be affected by the duration of blood storage, possibly due to red blood cell (RBC)-mediated disruption of nitric oxide (NO) signaling, a key regulator of vascular tone and blood flow. STUDY DESIGN AND METHODS AS-1 RBC units stored up to 42 days were sampled at selected storage times. Samples were added to aortic rings ex vivo, a system where NO-mediated vasodilation could be experimentally controlled. RESULTS RBC units showed storage-dependent changes in plasma hemoglobin (Hb), RBC 2,3-diphosphoglycerate acid, and RBC adenosine triphosphate conforming to expected profiles. When freshly collected (Day 0) blood was added to rat aortic rings, methacholine (MCh) stimulated substantial NO-mediated vasodilation. In contrast, MCh produced no vasodilation in the presence of blood stored for 42 days. Surprisingly, the vasoinhibitory effects of stored RBCs were almost totally mediated by RBCs themselves: removal of the supernatant did not attenuate the inhibitory effects, while addition of supernatant alone to the aortic rings only minimally inhibited MCh-stimulated relaxation. Stored RBCs did not inhibit vasodilation by a direct NO donor, demonstrating that the RBC-mediated vasoinhibitory mechanism did not work by NO scavenging. CONCLUSIONS These studies have revealed a previously unrecognized vasoinhibitory activity of stored RBCs, which is more potent than the described effects of free Hb and works through a different mechanism that does not involve NO scavenging but may function by reducing endothelial NO production. Through this novel mechanism, transfusion of small volumes of stored blood may be able to disrupt physiologic vasodilatory responses and thereby possibly cause adverse clinical outcomes.
Collapse
Affiliation(s)
- Jason T Alexander
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Alexander M El-Ali
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - James L Newman
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Sulaiman Karatela
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Benjamin L Predmore
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - David J Lefer
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Roy L Sutliff
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - John D Roback
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| |
Collapse
|
84
|
Alexander JT, El-Ali AM, Newman JL, Karatela S, Predmore BL, Lefer DJ, Sutliff RL, Roback JD. Red blood cells stored for increasing periods produce progressive impairments in nitric oxide-mediated vasodilation. Transfusion 2013. [PMID: 23480490 DOI: 10.1111/trf.1211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Clinical outcomes in transfused patients may be affected by the duration of blood storage, possibly due to red blood cell (RBC)-mediated disruption of nitric oxide (NO) signaling, a key regulator of vascular tone and blood flow. STUDY DESIGN AND METHODS AS-1 RBC units stored up to 42 days were sampled at selected storage times. Samples were added to aortic rings ex vivo, a system where NO-mediated vasodilation could be experimentally controlled. RESULTS RBC units showed storage-dependent changes in plasma hemoglobin (Hb), RBC 2,3-diphosphoglycerate acid, and RBC adenosine triphosphate conforming to expected profiles. When freshly collected (Day 0) blood was added to rat aortic rings, methacholine (MCh) stimulated substantial NO-mediated vasodilation. In contrast, MCh produced no vasodilation in the presence of blood stored for 42 days. Surprisingly, the vasoinhibitory effects of stored RBCs were almost totally mediated by RBCs themselves: removal of the supernatant did not attenuate the inhibitory effects, while addition of supernatant alone to the aortic rings only minimally inhibited MCh-stimulated relaxation. Stored RBCs did not inhibit vasodilation by a direct NO donor, demonstrating that the RBC-mediated vasoinhibitory mechanism did not work by NO scavenging. CONCLUSIONS These studies have revealed a previously unrecognized vasoinhibitory activity of stored RBCs, which is more potent than the described effects of free Hb and works through a different mechanism that does not involve NO scavenging but may function by reducing endothelial NO production. Through this novel mechanism, transfusion of small volumes of stored blood may be able to disrupt physiologic vasodilatory responses and thereby possibly cause adverse clinical outcomes.
Collapse
Affiliation(s)
- Jason T Alexander
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Alexander M El-Ali
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - James L Newman
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Sulaiman Karatela
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Benjamin L Predmore
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - David J Lefer
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Roy L Sutliff
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - John D Roback
- Division of Pulmonary Medicine, Department of Medicine, the Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, and the Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine and the Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| |
Collapse
|
85
|
Park JW, Piknova B, Huang PL, Noguchi CT, Schechter AN. Effect of blood nitrite and nitrate levels on murine platelet function. PLoS One 2013; 8:e55699. [PMID: 23383344 PMCID: PMC3562242 DOI: 10.1371/journal.pone.0055699] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/02/2013] [Indexed: 11/18/2022] Open
Abstract
Nitric oxide (NO) appears to play an important role in the regulation of thrombosis and hemostasis by inhibiting platelet function. The discovery of NO generation by reduction of nitrite (NO2−) and nitrate (NO3−) in mammals has led to increased attention to these anions with respect to potential beneficial effects in cardiovascular diseases. We have previously shown that nitrite anions at 0.1 µM inhibit aggregation and activation of human platelet preparations in vitro in the presence of red blood cells and this effect was enhanced by deoxygenation, an effect likely due to NO generation. In the present study, we hypothesized that nitrite and nitrate derived from the diet could also alter platelet function upon their conversion to NO in vivo. To manipulate the levels of nitrite and nitrate in mouse blood, we used antibiotics, NOS inhibitors, low nitrite/nitrate (NOx) diets, endothelial NOS knock-out mice and also supplementation with high levels of nitrite or nitrate in the drinking water. We found that all of these perturbations affected nitrite and nitrate levels but that the lowest whole blood values were obtained by dietary restriction. Platelet aggregation and ATP release were measured in whole blood and the results show an inverse correlation between nitrite/nitrate levels and platelet activity in aggregation and ATP release. Furthermore, we demonstrated that nitrite-supplemented group has a prolonged bleeding time compared with control or low NOx diet group. These results show that diet restriction contributes greatly to blood nitrite and nitrate levels and that platelet reactivity can be significantly affected by these manipulations. Our study suggests that endogenous levels of nitrite and nitrate may be used as a biomarker for predicting platelet function and that dietary manipulation may affect thrombotic processes.
Collapse
Affiliation(s)
- Ji Won Park
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | | | | | | |
Collapse
|
86
|
Caplin B, Wang Z, Slaviero A, Tomlinson J, Dowsett L, Delahaye M, Salama A, Wheeler DC, Leiper J. Alanine-glyoxylate aminotransferase-2 metabolizes endogenous methylarginines, regulates NO, and controls blood pressure. Arterioscler Thromb Vasc Biol 2012; 32:2892-900. [PMID: 23023372 DOI: 10.1161/atvbaha.112.254078] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Asymmetric dimethylarginine is an endogenous inhibitor of NO synthesis that may mediate cardiovascular disease. Alanine-glyoxylate aminotransferase-2 (AGXT2) has been proposed to degrade asymmetric dimethylarginine. We investigated the significance of AGXT2 in methylarginine metabolism in vivo and examined the effect of this enzyme on blood pressure. METHODS AND RESULTS In isolated mouse kidney mitochondria, we show asymmetric dimethylarginine deamination under physiological conditions. We demonstrate increased asymmetric dimethylarginine, reduced NO, and hypertension in an AGXT2 knockout mouse. We provide evidence for a role of AGXT2 in methylarginine metabolism in humans by demonstrating an inverse relationship between renal (allograft) gene expression and circulating substrate levels and an association between expression and urinary concentrations of the product. Finally, we examined data from a meta-analysis of blood pressure genome-wide association studies. No genome-wide significance was observed, but taking a hypothesis-driven approach, there was a suggestive association between the T allele at rs37369 (which causes a valine-isoleucine substitution and altered levels of AGXT2 substrate) and a modest increase in diastolic blood pressure (P=0.0052). CONCLUSIONS Although the effect of variation at rs37369 needs further study, these findings suggest that AGXT2 is an important regulator of methylarginines and represents a novel mechanism through which the kidney regulates blood pressure.
Collapse
Affiliation(s)
- Ben Caplin
- Nitric Oxide Signalling Group, Medical Research Council Clinical Sciences Center, Imperial College London, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Bailey SJ, Vanhatalo A, Winyard PG, Jones AM. The nitrate-nitrite-nitric oxide pathway: Its role in human exercise physiology. Eur J Sport Sci 2012. [DOI: 10.1080/17461391.2011.635705] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
88
|
Bhushan S, Kondo K, Predmore BL, Zlatopolsky M, King AL, Pearce C, Huang H, Tao YX, Condit ME, Lefer DJ. Selective β2-adrenoreceptor stimulation attenuates myocardial cell death and preserves cardiac function after ischemia-reperfusion injury. Arterioscler Thromb Vasc Biol 2012; 32:1865-74. [PMID: 22652602 DOI: 10.1161/atvbaha.112.251769] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE β(2)-adrenoreceptor activation has been shown to protect cardiac myocytes from cell death. We hypothesized that acute β(2)-adrenoreceptor stimulation, using arformoterol (ARF), would attenuate myocardial ischemia/reperfusion (R) injury via NO synthase activation and cause a subsequent increase in NO bioavailability. METHODS AND RESULTS Male C57BL/6J and endothelial NO synthase (eNOS) knockout mice were subjected to 45 minutes of myocardial ischemia and 24 hours of R. ARF or vehicle was administered 5 minutes before R. Serum troponin-I was measured, and infarct size per area-at-risk was evaluated at 24 hours of R. Echocardiography was performed at baseline and 2 weeks after R. Myocardial cAMP, protein kinase A, eNOS/Akt phosphorylation status, and NO metabolite levels were assayed. ARF (1 µg/kg) reduced infarct size per area-at-risk by 53.1% (P<0.001 versus vehicle) and significantly reduced troponin-I levels (P<0.001 versus vehicle). Ejection fraction was significantly preserved in ARF-treated hearts compared with vehicle hearts at 2 weeks of R. Serum cAMP and nuclear protein kinase A C-α increased 5 and 15 minutes after ARF injection, respectively (P<0.01). ARF increased Akt phosphorylation at Thr(308) (P<0.001) and Ser(473) (P<0.01), and eNOS phosphorylation at Ser(1177) (P<0.01). ARF treatment increased heart nitrosothiol levels (P<0.001) at 15 min after injection. ARF failed to reduce infarct size in eNOS(-/-) mice. CONCLUSIONS Our results indicate that β(2)-adrenoreceptor stimulation activates cAMP, protein kinase A, Akt, and eNOS and augments NO bioavailability. Activation of this prosurvival signaling pathway attenuates myocardial cell death and preserves cardiac function after ischemia/reperfusion.
Collapse
Affiliation(s)
- Shashi Bhushan
- Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine, 550 Peachtree St NE, Atlanta, GA 30308, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Aragón JP, Condit ME, Bhushan S, Predmore BL, Patel SS, Grinsfelder DB, Gundewar S, Jha S, Calvert JW, Barouch LA, Lavu M, Wright HM, Lefer DJ. Beta3-adrenoreceptor stimulation ameliorates myocardial ischemia-reperfusion injury via endothelial nitric oxide synthase and neuronal nitric oxide synthase activation. J Am Coll Cardiol 2012; 58:2683-91. [PMID: 22152956 DOI: 10.1016/j.jacc.2011.09.033] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Revised: 09/09/2011] [Accepted: 09/13/2011] [Indexed: 11/18/2022]
Abstract
OBJECTIVES This paper examined whether nebivolol protects the heart via nitric oxide (NO) synthase and NO-dependent signaling in an in vivo model of acute myocardial infarction. BACKGROUND Beta(3)-adrenergic receptor (AR) activation promotes endothelial nitric oxide synthase (eNOS) activity and NO bioavailability. We hypothesized that specific beta(3)-AR agonists would attenuate myocardial ischemia-reperfusion (MI/R) injury via eNOS activation and increased NO bioavailability. METHODS Mice were subjected to 45 min of myocardial ischemia in vivo followed by 24 h of reperfusion (R). Nebivolol (500 ng/kg), CL 316243 (1 μg/kg), BRL-37344 (1 μg/kg), or vehicle (VEH) was administered at the time of R. Myocardial area-at-risk (AAR) and infarct size (INF)/AAR was measured at 24 h of R. Cardiac tissue and plasma were collected to evaluate eNOS phosphorylation, neuronal nitric oxide synthase (nNOS), inducible nitric oxide synthase expression, and nitrite and nitrosothiol levels. RESULTS Nebivolol (500 ng/kg) reduced INF/AAR by 37% (p < 0.001 vs. VEH) and serum troponin-I levels from 41 ± 4 ng/ml to 25 ± 4 ng/ml (p < 0.05 vs. VEH). CL 316243 and BRL-37344 reduced INF by 39% and 42%, respectively (p < 0.001 vs. VEH). Nebivolol and CL 316243 increased eNOS phosphorylation at Ser-1177 (p < 0.05 vs. VEH) and increased nitrite and total nitrosylated protein levels. Nebivolol and CL 316243 significantly increased myocardial nNOS expression. Nebivolol failed to reduce INF after MI/R in beta(3)-AR (-/-), eNOS(-/-), and in nNOS(-/-) mice. CONCLUSIONS Our results indicate that beta(3)-AR agonists protect against MI/R injury. Furthermore, the cardioprotective effects of beta(3)-AR agonists are mediated by rapid eNOS and nNOS activation and increased NO bioavailability.
Collapse
Affiliation(s)
- Juan P Aragón
- Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia 30308, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Baliga RS, Milsom AB, Ghosh SM, Trinder SL, Macallister RJ, Ahluwalia A, Hobbs AJ. Dietary nitrate ameliorates pulmonary hypertension: cytoprotective role for endothelial nitric oxide synthase and xanthine oxidoreductase. Circulation 2012; 125:2922-32. [PMID: 22572914 DOI: 10.1161/circulationaha.112.100586] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a multifactorial disease characterized by increased pulmonary vascular resistance and right ventricular failure; morbidity and mortality remain unacceptably high. Loss of nitric oxide (NO) bioactivity is thought to contribute to the pathogenesis of PH, and agents that augment pulmonary NO signaling are clinically effective in the disease. Inorganic nitrate (NO(3)(-)) and nitrite (NO(2)(-)) elicit a reduction in systemic blood pressure in healthy individuals; this effect is underpinned by endogenous and sequential reduction to NO. Herein, we determined whether dietary nitrate and nitrite might be preferentially reduced to NO by the hypoxia associated with PH, and thereby offer a convenient, inexpensive method of supplementing NO functionality to reduce disease severity. METHODS AND RESULTS Dietary nitrate reduced the right ventricular pressure and hypertrophy, and pulmonary vascular remodeling in wild-type mice exposed to 3 weeks of hypoxia; this beneficial activity was mirrored largely by dietary nitrite. The cytoprotective effects of dietary nitrate were associated with increased plasma and lung concentrations of nitrite and cGMP. The beneficial effects of dietary nitrate and nitrite were reduced in mice lacking endothelial NO synthase or treated with the xanthine oxidoreductase inhibitor allopurinol. CONCLUSIONS These data demonstrate that dietary nitrate, and to a lesser extent dietary nitrite, elicit pulmonary dilatation, prevent pulmonary vascular remodeling, and reduce the right ventricular hypertrophy characteristic of PH. This favorable pharmacodynamic profile depends on endothelial NO synthase and xanthine oxidoreductase -catalyzed reduction of nitrite to NO. Exploitation of this mechanism (ie, dietary nitrate/nitrite supplementation) represents a viable, orally active therapy for PH.
Collapse
Affiliation(s)
- Reshma S Baliga
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
91
|
Concentration- and stage-specific effects of nitrite on colon cancer cell lines. Nitric Oxide 2012; 26:267-73. [DOI: 10.1016/j.niox.2012.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 03/10/2012] [Accepted: 03/24/2012] [Indexed: 01/22/2023]
|
92
|
Natural product nitric oxide chemistry: new activity of old medicines. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:873210. [PMID: 22548122 PMCID: PMC3324039 DOI: 10.1155/2012/873210] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 01/19/2012] [Indexed: 01/19/2023]
Abstract
The use of complementary and alternative medicine (CAM) as a therapy and preventative care measure for cardiovascular diseases (CVD) may prove to be beneficial when used in conjunction with or in place of conventional medicine. However, the lack of understanding of a mechanism of action of many CAMs limits their use and acceptance in western medicine. We have recently recognized and characterized specific nitric oxide (NO) activity of select alternative and herbal medicines that may account for many of their reported health benefits. The ability of certain CAM to restore NO homeostasis both through enhancing endothelial production of NO and by providing a system for reducing nitrate and nitrite to NO as a compensatory pathway for repleting NO bioavailability may prove to be a safe and cost-effective strategy for combating CVD. We will review the current state of science behind NO activity of herbal medicines and their effects on CVD.
Collapse
|
93
|
Jädert C, Petersson J, Massena S, Ahl D, Grapensparr L, Holm L, Lundberg JO, Phillipson M. Decreased leukocyte recruitment by inorganic nitrate and nitrite in microvascular inflammation and NSAID-induced intestinal injury. Free Radic Biol Med 2012; 52:683-692. [PMID: 22178413 DOI: 10.1016/j.freeradbiomed.2011.11.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 11/09/2011] [Accepted: 11/12/2011] [Indexed: 01/13/2023]
Abstract
Nitric oxide (NO) generated by vascular NO synthases can exert anti-inflammatory effects, partly through its ability to decrease leukocyte recruitment. Inorganic nitrate and nitrite, from endogenous or dietary sources, have emerged as alternative substrates for NO formation in mammals. Bioactivation of nitrate is believed to require initial reduction to nitrite by oral commensal bacteria. Here we investigated the effects of inorganic nitrate and nitrite on leukocyte recruitment in microvascular inflammation and in NSAID-induced small-intestinal injury. We show that leukocyte emigration in response to the proinflammatory chemokine MIP-2 is reduced by 70% after 7 days of dietary nitrate supplementation as well as by acute intravenous nitrite administration. Nitrite also reduced leukocyte adhesion to a similar extent and this effect was inhibited by the soluble guanylyl cyclase inhibitor ODQ, whereas the effect on emigrated leukocytes was not altered by this treatment. Further studies in TNF-α-stimulated endothelial cells revealed that nitrite dose-dependently reduced the expression of ICAM-1. In rats and mice subjected to a challenge with diclofenac, dietary nitrate prevented the increase in myeloperoxidase and P-selectin levels in small-intestinal tissue. Antiseptic mouthwash, which eliminates oral nitrate reduction, markedly blunted the protective effect of dietary nitrate on P-selectin levels. Despite attenuation of the acute immune response, the overall ability to clear an infection with Staphylococcus aureus was not suppressed by dietary nitrate as revealed by noninvasive IVIS imaging. We conclude that dietary nitrate markedly reduces leukocyte recruitment to inflammation in a process involving attenuation of P-selectin and ICAM-1 upregulation. Bioactivation of dietary nitrate requires intermediate formation of nitrite by oral nitrate-reducing bacteria and then probably further reduction to NO and other bioactive nitrogen oxides in the tissues.
Collapse
Affiliation(s)
- Cecilia Jädert
- Department of Physiology and Pharmacology, Karolinska Institute, S-171 77 Stockholm, Sweden
| | - Joel Petersson
- Department of Medical Cell Biology, Uppsala University, S-751 23 Uppsala, Sweden
| | - Sara Massena
- Department of Medical Cell Biology, Uppsala University, S-751 23 Uppsala, Sweden
| | - David Ahl
- Department of Medical Cell Biology, Uppsala University, S-751 23 Uppsala, Sweden
| | - Liza Grapensparr
- Department of Medical Cell Biology, Uppsala University, S-751 23 Uppsala, Sweden
| | - Lena Holm
- Department of Medical Cell Biology, Uppsala University, S-751 23 Uppsala, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institute, S-171 77 Stockholm, Sweden.
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, S-751 23 Uppsala, Sweden.
| |
Collapse
|
94
|
Lu N, Chen W, Zhu J, Peng YY. Enhancement of nitrite on heme-induced oxidative reactions: A potential toxicological implication. Toxicol In Vitro 2012; 26:81-5. [DOI: 10.1016/j.tiv.2011.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 07/17/2011] [Accepted: 10/20/2011] [Indexed: 10/15/2022]
|
95
|
Dietary nitrite attenuates oxidative stress and activates antioxidant genes in rat heart during hypobaric hypoxia. Nitric Oxide 2011; 26:61-73. [PMID: 22197744 DOI: 10.1016/j.niox.2011.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 12/02/2011] [Accepted: 12/08/2011] [Indexed: 01/19/2023]
Abstract
The nitrite anion represents the circulatory and tissue storage form of nitric oxide (NO) and a signaling molecule, capable of conferring cardioprotection and many other health benefits. However, molecular mechanisms for observed cardioprotective properties of nitrite remain largely unknown. We have evaluated the NO-like bioactivity and cardioprotective efficacies of sodium nitrite supplemented in drinking water in rats exposed to short-term chronic hypobaric hypoxia. We observed that, nitrite significantly attenuates hypoxia-induced oxidative stress, modulates HIF-1α stability and promotes NO-cGMP signaling in hypoxic heart. To elucidate potential downstream targets of nitrite during hypoxia, we performed a microarray analysis of nitrite supplemented hypoxic hearts and compared with both hypoxic and nitrite supplemented normoxic hearts respectively. The analysis revealed a significant increase in the expression of many antioxidant genes, transcription factors and cardioprotective signaling pathways which was subsequently confirmed by qRT-PCR and Western blotting. Conversely, hypoxia exposure increased oxidative stress, activated inflammatory cytokines, downregulated ion channels and altered expression of both pro- and anti-oxidant genes. Our results illustrate the physiological function of nitrite as an eNOS-independent source of NO in heart profoundly modulating the oxidative status and cardiac transcriptome during hypoxia.
Collapse
|
96
|
|
97
|
Vitturi DA, Patel RP. Current perspectives and challenges in understanding the role of nitrite as an integral player in nitric oxide biology and therapy. Free Radic Biol Med 2011; 51:805-12. [PMID: 21683783 PMCID: PMC3148353 DOI: 10.1016/j.freeradbiomed.2011.05.037] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 05/19/2011] [Accepted: 05/26/2011] [Indexed: 12/20/2022]
Abstract
Beyond an inert oxidation product of nitric oxide (NO) metabolism, current thinking posits a key role for nitrite as a mediator of NO signaling, especially during hypoxia. This concept has been discussed in the context of nitrite serving a role as an endogenous modulator of NO homeostasis, but also from a novel clinical perspective whereby nitrite therapy may replenish NO signaling and prevent ischemic tissue injury. Indeed, the relatively rapid translation of studies delineating mechanisms of action to ongoing and planned clinical trials has been critical in fuelling interest in nitrite biology, and several excellent reviews have been written on this topic. In this article we limit our discussions to current concepts and what we feel are questions that remain unanswered within the paradigm of nitrite being a mediator of NO biology.
Collapse
Affiliation(s)
- Dario A Vitturi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
98
|
Zhu SG, Kukreja RC, Das A, Chen Q, Lesnefsky EJ, Xi L. Dietary nitrate supplementation protects against Doxorubicin-induced cardiomyopathy by improving mitochondrial function. J Am Coll Cardiol 2011; 57:2181-9. [PMID: 21596234 DOI: 10.1016/j.jacc.2011.01.024] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 01/07/2011] [Accepted: 01/11/2011] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The aim of this study was to test the hypothesis that long-term dietary nitrate supplementation protects against doxorubicin-induced cardiomyopathy by improving ventricular function and reducing mitochondrial respiratory chain damage. BACKGROUND Doxorubicin is a powerful anthracycline antibiotic used to treat divergent human neoplasms. Its clinical use is limited because of severe cardiotoxic side effects. Dietary nitrate and nitrite are essential nutrients for maintenance of steady-state tissue levels of nitric oxide and may play a therapeutic role in diseases associated with nitric oxide insufficiency or dysregulation. Dietary nitrate and nitrite supplementation alleviates myocardial injury caused by ischemia-reperfusion and cardiac arrest-resuscitation. METHODS Adult male CF-1 mice were given a single dose of doxorubicin (15 mg/kg intraperitoneally), and left ventricular contractile function was assessed 5 days later using both echocardiography and pressure-volume Millar catheterization. A nitrate supplementation regimen (1 g/l sodium nitrate in drinking water) was started 7 days before doxorubicin injection and continued thereafter. Cardiomyocyte necrosis and apoptosis, tissue lipid peroxidation, and plasma nitrate and nitrite levels were assessed. In addition, mitochondrial complex I activity, oxidative phosphorylation capacity, and hydrogen peroxide generation were determined in parallel experiments. RESULTS Doxorubicin caused impairment of ventricular contractility and cell death, which were significantly reduced by nitrate supplementation (p < 0.05). These cardioprotective effects were associated with a significant decrease in tissue lipid peroxidation. Nitrate supplementation significantly preserved mitochondrial complex I activity and oxidative phosphorylation and attenuated hydrogen peroxide generation after doxorubicin treatment. CONCLUSIONS Long-term oral intake of inorganic nitrate attenuates doxorubicin-induced ventricular dysfunction, cell death, oxidative stress, and mitochondrial respiratory chain damage. Nitrate could be a promising therapeutic agent against doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shu-Guang Zhu
- VCU Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia 23298-0204, USA
| | | | | | | | | | | |
Collapse
|
99
|
Machha A, Schechter AN. Dietary nitrite and nitrate: a review of potential mechanisms of cardiovascular benefits. Eur J Nutr 2011; 50:293-303. [PMID: 21626413 PMCID: PMC3489477 DOI: 10.1007/s00394-011-0192-5] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 03/21/2011] [Indexed: 12/20/2022]
Abstract
PURPOSE In the last decade, a growing scientific and medical interest has emerged toward cardiovascular effects of dietary nitrite and nitrate; however, many questions concerning their mode of action(s) remain unanswered. In this review, we focus on multiple mechanisms that might account for potential cardiovascular beneficial effects of dietary nitrite and nitrate. RESULTS Beneficial changes to cardiovascular health from dietary nitrite and nitrate might result from several mechanism(s) including their reduction into nitric oxide, improvement in endothelial function, vascular relaxation, and/or inhibition of the platelet aggregation. From recently obtained evidence, it appears that the longstanding concerns about the toxicity of oral nitrite or nitrate are overstated. CONCLUSION Dietary nitrite and nitrate may have cardiovascular protective effects in both healthy individuals and also those with cardiovascular disease conditions. A role for nitrite and nitrate in nitric oxide biosynthesis and/or in improving nitric oxide bioavailability may eventually provide a rationale for using dietary nitrite and nitrate supplementation in the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Ajay Machha
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg 10, Room 9N314B, 10 Center Drive, Bethesda, MD 20892, USA
| | - Alan N. Schechter
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg 10, Room 9N314B, 10 Center Drive, Bethesda, MD 20892, USA
| |
Collapse
|
100
|
Marozkina NV, Gaston B. S-Nitrosylation signaling regulates cellular protein interactions. Biochim Biophys Acta Gen Subj 2011; 1820:722-9. [PMID: 21745537 DOI: 10.1016/j.bbagen.2011.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/13/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND S-Nitrosothiols are made by nitric oxide synthases and other metalloproteins. Unlike nitric oxide, S-nitrosothiols are involved in localized, covalent signaling reactions in specific cellular compartments. These reactions are enzymatically regulated. SCOPE S-Nitrosylation affects interactions involved in virtually every aspect of normal cell biology. This article is part of a Special Issue entitled Regulation of Cellular Processes by S-nitrosylation. MAJOR CONCLUSIONS AND SIGNIFICANCE S-Nitrosylation is a regulated signaling reaction.
Collapse
Affiliation(s)
- Nadzeya V Marozkina
- University of Virginia School of Medicine, Division of Pediatric Respiratory Medicine, PO Box 800386, Charlottesville, VA 22908, USA.
| | | |
Collapse
|