1
|
Chiletti R, Fincher SH, Horton SB, Peek GJ, Checchia P, Butt W. The Role of Nitric Oxide in the Sweep Gas for Patients Receiving Extracorporeal Membrane Oxygenation or Cardiopulmonary Bypass. Can J Cardiol 2025; 41:621-629. [PMID: 39733940 DOI: 10.1016/j.cjca.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024] Open
Abstract
Nitric oxide (NO) was proclaimed the 1992 "molecule of the year" by Culotta in Science magazine because of its importance in neuroscience, physiology, and immunology. Inhaled NO has been in clinical use for over 35 years to decrease pulmonary hypertension and improve oxygenation. Over the past 20 years, there has been much research into understanding the role of NO on cell surface receptors, mitochondria, and intracellular processes that involve calcium and superoxide radicals. This research has shown that, irrespective of the cause, NO has a major role in the systemic inflammatory response syndrome and ischemia-reperfusion injury.1 More recent clinical research has focussed on NO use in patients undergoing cardiopulmonary bypass and receiving extracorporeal life support, with some centres incorporating NO into sweep gas as part of routine practice. In this article we review NO pathways in humans, the biologic effects of NO, the interplay between NO and red blood cells, and animal and human studies on the effects of exogenously administered NO.
Collapse
Affiliation(s)
- Roberto Chiletti
- Department of Paediatric Intensive Care, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Sophie H Fincher
- Department of Paediatric Intensive Care, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Critical Care, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen B Horton
- Department of Cardiac Surgery, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Melbourne, Victoria, Australia; Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Giles J Peek
- Congenital Heart Centre, University of Florida, Gainesville, Florida, USA
| | - Paul Checchia
- Division of Critical Care Medicine, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Warwick Butt
- Department of Paediatric Intensive Care, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Critical Care, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Melbourne, Victoria, Australia; ICU Research Clinical Sciences Theme MCRI, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Pan A, Liu X, Han H, Gao SQ, Lin YW. Disruption of a potential disulfide bond of Cys65-Cys141 on the structure and stability of globin X from zebrafish. Phys Chem Chem Phys 2025; 27:2828-2833. [PMID: 39817796 DOI: 10.1039/d4cp04253k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Globin X is a newly discovered member of the globin family, while its structure and function are not fully understood. In this study, we performed protein modelling studies using Alphafold3 and molecular dynamics simulations, which suggested that the protein adopts a typical globin fold, with the formation of a potential disulfide bond of Cys65 and Cys141. To elucidate the role of this unique disulfide in protein structure and stability, we constructed a double mutant of C65S/C141S by mutating the two cysteine residues to serine. As suggested by protein mass, ultraviolet-visible (UV-Vis) and circular dichroism (CD) spectroscopy analyses, the potential disulfide bond has minimal effect on the overall protein structure, but its absence reduces the protein stability. Electron paramagnetic resonance (EPR) analysis also revealed an increase in the proportion of high-spin state heme iron, which accelerates the rate of heme degradation in reaction with H2O2. This study highlights the critical role of the Cys65-Cys141 in maintaining the stability of globin X and the bis-His heme coordination state, providing insights into the structure-function relationship of the newly discovered globin.
Collapse
Affiliation(s)
- Aiqun Pan
- School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China.
| | - Xichun Liu
- School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China.
| | - Hui Han
- School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China.
| | - Shu-Qin Gao
- Laboratory of Protein Structure and Function, University of South China, Hengyang 421001, China
| | - Ying-Wu Lin
- School of Chemistry and Chemical Engineering, University of South China, Hengyang 421001, China.
- Laboratory of Protein Structure and Function, University of South China, Hengyang 421001, China
| |
Collapse
|
3
|
Simpson A, Johnston W, Carda-Diéguez M, Mira A, Easton C, Henriquez FL, Culshaw S, Rosier BT, Burleigh M. Periodontal treatment causes a longitudinal increase in nitrite-producing bacteria. Mol Oral Microbiol 2024; 39:491-506. [PMID: 39169836 DOI: 10.1111/omi.12479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/14/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND The oral microbiome-dependent nitrate (NO3 -)-nitrite (NO2 -)-nitric oxide (NO) pathway may help regulate blood pressure. NO2 --producing bacteria in subgingival plaque are reduced in relative abundance in patients with untreated periodontitis compared with periodontally healthy patients. In periodontitis patients, the NO2 --producing bacteria increase several months after periodontal treatment. The early effects of periodontal treatment on NO2 --producing bacteria and the NO3 --NO2 --NO pathway remain unknown. The aim of this study was to determine how periodontal treatment affects the oral NO2 --producing microbiome and salivary NO3 - and NO2 - levels over time. METHODS The subgingival microbiota of 38 periodontitis patients was analysed before (baseline [BL]) and 1, 7 and 90 days after periodontal treatment. Changes in NO2 --producing bacteria and periodontitis-associated bacteria were determined by 16s rRNA Illumina sequencing. Saliva samples were collected at all-time points to determine NO3 - and NO2 - levels using gas-phase chemiluminescence. RESULTS A significant increase was observed in the relative abundance of NO2 --producing species between BL and all subsequent timepoints (all p < 0.001). Periodontitis-associated species decreased at all timepoints, relative to BL (all p < 0.02). NO2 --producing species negatively correlated with periodontitis-associated species at all timepoints, with this relationship strongest 90 days post-treatment (ρ = -0.792, p < 0.001). Despite these findings, no significant changes were found in salivary NO3 - and NO2 - over time (all p > 0.05). CONCLUSIONS Periodontal treatment induced an immediate increase in the relative abundance of health-associated NO2 --producing bacteria. This increase persisted throughout periodontal healing. Future studies should test the effect of periodontal treatment combined with NO3 - intake on periodontal and cardiovascular health.
Collapse
Affiliation(s)
- Annabel Simpson
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, UK
| | - William Johnston
- School of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Miguel Carda-Diéguez
- Department of Health and Genomics, Centre for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Alex Mira
- Department of Health and Genomics, Centre for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Chris Easton
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, UK
| | - Fiona L Henriquez
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, UK
| | - Shauna Culshaw
- Oral Sciences, University of Glasgow Dental School, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Bob T Rosier
- Department of Health and Genomics, Centre for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Mia Burleigh
- Sport and Physical Activity Research Institute, University of the West of Scotland, Blantyre, UK
| |
Collapse
|
4
|
Margaritelis NV, Cobley JN, Nastos GG, Papanikolaou K, Bailey SJ, Kritsiligkou P, Nikolaidis MG. Evidence-based sports supplements: A redox analysis. Free Radic Biol Med 2024; 224:62-77. [PMID: 39147071 DOI: 10.1016/j.freeradbiomed.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Despite the overwhelming number of sports supplements on the market, only seven are currently recognized as effective. Biological functions are largely regulated through redox reactions, yet no comprehensive analysis of the redox properties of these supplements has been compiled. Here, we analyze the redox characteristics of these seven supplements: bicarbonates, beta-alanine, caffeine, creatine, nitrates, carbohydrates, and proteins. Our findings suggest that all sports supplements exhibit some degree of redox activity. However, the precise physiological implications of these redox properties remain unclear. Future research, employing unconventional perspectives and methodologies, will reveal new redox pixels of the exercise physiology and sports nutrition picture.
Collapse
Affiliation(s)
- Nikos V Margaritelis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Serres, Greece.
| | - James N Cobley
- School of Life Sciences, The University of Dundee, Dundee, Scotland, UK
| | - George G Nastos
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Serres, Greece
| | | | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Paraskevi Kritsiligkou
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Michalis G Nikolaidis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Serres, Greece
| |
Collapse
|
5
|
Lima L, Gaspar S, Rocha BS, Alves R, Almeida MG. Current clinical framework on nitric oxide role in periodontal disease and blood pressure. Clin Oral Investig 2024; 28:521. [PMID: 39264471 PMCID: PMC11392991 DOI: 10.1007/s00784-024-05913-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVES In this review, we explored potential associations between NO and its derivatives, nitrite and nitrate, with periodontal and cardiovascular diseases, with special emphasis on the former. By providing a state-of-the-art and integrative understanding of this topic, we aimed to shed light on the potential role of these three nitrogen oxides in the periodontitis-hypertension nexus, identify knowledge gaps, and point out critical aspects of the experimental methodologies. MATERIALS AND METHODS A comprehensive literature review was conducted on human salivary and plasma concentrations of nitrate and nitrite, and their impact on periodontal and cardiovascular health. RESULTS A nitrate-rich diet increases nitrate and nitrite levels in saliva and plasma, promoting oral health by favorably altering the oral microbiome. Chlorhexidine (CHX) mouthrinses disrupt the nitrate-nitrite-NO pathway, reducing NO bioavailability, and potentially affecting blood pressure. This is because CHX eliminates nitrate-reducing bacteria, which are essential for NO production. Although endogenous NO production may be insufficient, the nitrate-nitrite-NO pathway plays a critical role in maintaining appropriate endothelial function, which is balanced by the microbiome and dietary nitrate intake. Dietary nitrate supplementation may lead to beneficial changes in the oral microbiome, thereby increasing the NO bioavailability. However, NO bioavailability can be compromised by reactive oxygen species (ROS) and the uncoupling of endothelial nitric oxide synthase (eNOS), leading to further ROS generation and creating a detrimental cycle. Studies on NO and periodontal disease have shown increased nitrite concentrations in patients with periodontal disease, although these studies have some methodological limitations. In terms of blood pressure, literature suggests that CHX mouthrinses may reduce the capacity of nitrate-reducing bacteria, potentially leading to an increase in blood pressure. CONCLUSIONS Several studies have suggested an association between NO levels and the development of cardiovascular and periodontal diseases. However, the exact mechanisms linking these diseases remains to be fully elucidated. CLINICAL RELEVANCE Nitric oxide (NO) is a signaling molecule that plays a crucial role in several physiological processes such as vascular homeostasis, inflammation, immune cell activity, and pathologies such as hypertension and periodontitis.
Collapse
Affiliation(s)
- Leonel Lima
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Monte da Caparica, Almada, Portugal
| | - Sara Gaspar
- UCIBIO/i4HB- Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University of Lisbon, Caparica, Portugal
| | - Bárbara S Rocha
- Faculty of Pharmacy and Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ricardo Alves
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Monte da Caparica, Almada, Portugal
| | - M Gabriela Almeida
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Monte da Caparica, Almada, Portugal.
- UCIBIO/i4HB- Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University of Lisbon, Caparica, Portugal.
| |
Collapse
|
6
|
Jeddi S, Yousefzadeh N, Zarkesh M, Kashfi K, Ghasemi A. Effect of long-term inorganic nitrate administration on myocardial ischemia-reperfusion injury in ovariectomized rats. Front Pharmacol 2024; 15:1369379. [PMID: 38601460 PMCID: PMC11004245 DOI: 10.3389/fphar.2024.1369379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction: Menopause is associated with reduced nitric oxide (NO) bioavailability and lower tolerance against myocardial ischemia-reperfusion (IR) injury. This study investigated whether long-term nitrate administration provides resistance against myocardial IR injury in ovariectomized (OVX) rats. Method: After ovariectomy, female rats were assigned to the OVX and the OVX + nitrate groups (n = 14/group); the latter group consumed nitrate (100 mg/L) for 9 months. At month 9, each group was divided into two subgroups (n = 7/subgroup), of which one subgroup was exposed to myocardial IR (IR+ hearts) and the other was not exposed (IR- hearts). The hearts of rats were isolated, and NO metabolite (NOx), oxidative stress indices, and mRNA expressions of endothelial (eNOS), inducible (iNOS), and neuronal (nNOS) NO synthases, as well as markers of apoptosis, were measured in the IR- and IR+ hearts. In the IR+ hearts, cardiac function indices (CFI) and the infarct size were also measured. Results: Nitrate increased catalase activity (97%) and eNOS expression (2.94-fold) in the IR- hearts. In the IR+ hearts, nitrate reduced left ventricular (LV) end-diastolic pressure (11.6%) and infarct size (26.2%) and increased recovery of LV developed pressure (44.0%) and peak rate of positive (28.9%) and negative (15.4%) changes in LV pressure. In addition, in the IR+ hearts, nitrate increased eNOS and B-cell lymphoma-2 (Bcl-2) as well as decreased iNOS, Bcl-2 associated X protein (Bax), caspase-3, caspase-8, caspase-9, and tumor necrosis factor-α (TNF-α) expression. Nitrate increased total antioxidant capacity (TAC) and catalase (CAT) activity and decreased malondialdehyde (MDA) levels at month nine in serum and IR+ hearts. Conclusion: The favorable effects of nitrate against IR injury were associated with higher eNOS and Bcl-2 expression, CAT activity, TAC, and lower iNOS, Bax, caspase-3, caspase-8, caspase-9 and TNF-α expression, and MDA in the heart tissue. Nitrate preconditioning alleviated IR-induced myocardial injury in OVX rats; this effect was associated with eNOS upregulation before IR and the blunting of OVX-induced eNOS downregulation, iNOS upregulation, apoptosis, and oxidative stress in heart tissue after IR.
Collapse
Affiliation(s)
- Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, United States
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Yassaghi Y, Jeddi S, Kashfi K, Ghasemi A. Myocardial infarct size is reduced by nitrite and nitrate administration: a systematic review and meta-analysis of animal studies. EXCLI JOURNAL 2024; 23:18-33. [PMID: 38357094 PMCID: PMC10864704 DOI: 10.17179/excli2023-6740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/18/2023] [Indexed: 02/16/2024]
Abstract
Ischemic heart disease (IHD) is the leading cause of mortality worldwide and can be complicated by myocardial infarction (MI), leading to cardiac failure. Inorganic nitrite and nitrate, which release nitric oxide (NO), can protect the heart against myocardial injury. This animal systematic review and meta-analysis aims to assess whether the administration of nitrite/nitrate decreases myocardial infarct size. We systematically searched PubMed, Scopus, and Web of Science databases until October 2023; 15 eligible animal studies (35 study arms for in-vivo and 10 for in-vitro studies) published between 1989 and 2023 were included. In-vivo studies were conducted on rats, mice, cats, and dogs, and in-vitro studies on rats and mice with an overall exposure of 0.03 to 12713 mg/kg to nitrate/nitrite administrated before, after, or during ischemia mainly by intravenous single bolus or by oral over 270 days. All in-vitro studies used nitrite/nitrate before ischemia, with the concentration ranging between 0.34 to 201 μM. MI was induced by occlusion of the left anterior diagonal or left circumflex arteries in in-vitro studies and by isoproterenol in in-vivo studies. Infarct size was measured by direct staining of the sliced heart sections. In in-vivo studies, nitrite (overall effect size (ES)=-17.0 %, 95 % confidence interval (CI)=-21.3, -12.8, P<0.001) and nitrate (overall ES= -9.6 %, 95 % CI=-15.7, -3.4, P=0.002) reduced myocardial infarct size. In in-vitro studies, nitrite (overall ES=-15.8 %, 95 % CI=-25.5, -6.2, P=0.001) reduced the infarct size. Sensitivity analysis showed that the overall effect of nitrite on myocardial infarct size was unaffected by doses or health conditions in in-vivo and in-vitro studies. In conclusion, our meta-analysis showed that nitrite/nitrate administration can effectively reduce myocardial infarct size. However, these results should be approached with caution because of the limitations of animal studies and the existing high heterogeneity.
Collapse
Affiliation(s)
- Younes Yassaghi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Giv V, Aminaei M, Nikoei R. The effect of eight weeks beetroot juice supplement on aerobic, anaerobic power, and field performance of soccer players. Res Sports Med 2024; 32:132-144. [PMID: 35726852 DOI: 10.1080/15438627.2022.2090250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
The purpose of the study investigated the effect of eight weeks of soccer training with beetroot juice supplement on aerobic power, anaerobic power, and field performance of soccer players. This is experimental research, by the control group in pre and post-test. The statistical population was male soccer players. Forty subjects were randomly divided into four groups including the exercise group (EX) the exercise group with beetroot juice supplement (EX&BRJS) the beetroot juice supplement group (BRJS), and the control group (C). Aerobic power, anaerobic threshold, and respiratory exchange ratio, measured by the gas analyser (Cosmed), anaerobic power (peak, mean power), and fatigue index, by Wingate cycle test (Monark, 839), and field performance by (Bangsbo, field test performance). The statistical methods include the Kolmogorov-Smirnov, Levin, covariance (ANCOVA), and pair comparison with Bonferroni test. The results showed eight weeks' of soccer training with beetroot juice supplement, significantly changed aerobic power, respiratory exchange ratio, anaerobic threshold, anaerobic power, field performance, and fatigue index (P ≤ 0.05). Additionally, in all variables, the paired comparison showed that the EX&BRJS group progressed more than all other condition groups. The soccer athletes may use beetroot juice supplements along with soccer exercises to improve aerobic and anaerobic power and field performance.
Collapse
Affiliation(s)
- Vahid Giv
- Department of Sport Sciences, The Shahid Bahonar University of Kerman, Kerman, IRAN
| | - Mohsen Aminaei
- Department of Sport Sciences, The Shahid Bahonar University of Kerman, Kerman, IRAN
| | - Rohullah Nikoei
- Department of Sport Sciences, The Shahid Bahonar University of Kerman, Kerman, IRAN
| |
Collapse
|
9
|
Yang J, Sundqvist ML, Zheng X, Jiao T, Collado A, Tratsiakovich Y, Mahdi A, Tengbom J, Mergia E, Catrina SB, Zhou Z, Carlström M, Akaike T, Cortese-Krott MM, Weitzberg E, Lundberg JO, Pernow J. Hypoxic erythrocytes mediate cardioprotection through activation of soluble guanylate cyclase and release of cyclic GMP. J Clin Invest 2023; 133:e167693. [PMID: 37655658 PMCID: PMC10471167 DOI: 10.1172/jci167693] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 07/06/2023] [Indexed: 09/02/2023] Open
Abstract
Red blood cells (RBCs) mediate cardioprotection via nitric oxide-like bioactivity, but the signaling and the identity of any mediator released by the RBCs remains unknown. We investigated whether RBCs exposed to hypoxia release a cardioprotective mediator and explored the nature of this mediator. Perfusion of isolated hearts subjected to ischemia-reperfusion with extracellular supernatant from mouse RBCs exposed to hypoxia resulted in improved postischemic cardiac function and reduced infarct size. Hypoxia increased extracellular export of cyclic guanosine monophosphate (cGMP) from mouse RBCs, and exogenous cGMP mimicked the cardioprotection induced by the supernatant. The protection induced by hypoxic RBCs was dependent on RBC-soluble guanylate cyclase and cGMP transport and was sensitive to phosphodiesterase 5 and activated cardiomyocyte protein kinase G. Oral administration of nitrate to mice to increase nitric oxide bioactivity further enhanced the cardioprotective effect of hypoxic RBCs. In a placebo-controlled clinical trial, a clear cardioprotective, soluble guanylate cyclase-dependent effect was induced by RBCs collected from patients randomized to 5 weeks nitrate-rich diet. It is concluded that RBCs generate and export cGMP as a response to hypoxia, mediating cardioprotection via a paracrine effect. This effect can be further augmented by a simple dietary intervention, suggesting preventive and therapeutic opportunities in ischemic heart disease.
Collapse
Affiliation(s)
- Jiangning Yang
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Michaela L. Sundqvist
- Department of Physiology, Nutrition and Biomechanics, The Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Xiaowei Zheng
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Tong Jiao
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Aida Collado
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Yahor Tratsiakovich
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Ali Mahdi
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - John Tengbom
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Evanthia Mergia
- Institute for Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Zhichao Zhou
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Miriam M. Cortese-Krott
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Myocardial Infarction Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jon O. Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - John Pernow
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Yassaghi Y, Jeddi S, Yousefzadeh N, Kashfi K, Ghasemi A. Long-term inorganic nitrate administration protects against myocardial ischemia-reperfusion injury in female rats. BMC Cardiovasc Disord 2023; 23:411. [PMID: 37605135 PMCID: PMC10441752 DOI: 10.1186/s12872-023-03425-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND The favorable effects of nitrate against myocardial ischemia-reperfusion injury (MIRI) have primarily focused on male rats and in short term. Here we determine the impact of long-term nitrate intervention on baseline cardiac function and the resistance to MIRI in female rats. METHODS Female Wistar rats were randomly divided into untreated and nitrate-treated (100 mg/L sodium nitrate in drinking water for 9 months) groups (n = 14/group). At intervention end, levels of serum progesterone, nitric oxide metabolites (NOx), heart NOx concentration, and mRNA expressions of NO synthase isoforms (NOS), i.e., endothelial (eNOS), neuronal (nNOS), and inducible (iNOS), were measured. Isolated hearts were exposed to ischemia, and cardiac function indices (CFI) recorded. When the ischemia-reperfusion (IR) period ended, infarct size, NO metabolites, eNOS, nNOS, and iNOS expression were measured. RESULTS Nitrate-treated rats had higher serum progesterone (29.8%, P = 0.013), NOx (31.6%, P = 0.035), and higher heart NOx (60.2%, P = 0.067), nitrite (131%, P = 0.018), and eNOS expression (200%, P = 0.005). Nitrate had no significant effects on baseline CFI but it increased recovery of left ventricular developed pressure (LVDP, 19%, P = 0.020), peak rate of positive (+ dp/dt, 16%, P = 0.006) and negative (-dp/dt, 14%, P = 0.014) changes in left ventricular pressure and decreased left ventricular end-diastolic pressure (LVEDP, 17%, P < 0.001) and infarct size (34%, P < 0.001). After the IR, the two groups had significantly different heart nitrite, nitrate, NOx, and eNOS and iNOS mRNA expressions. CONCLUSIONS Long-term nitrate intervention increased the resistance to MIRI in female rats; this was associated with increased heart eNOS expression and circulating progesterone before ischemia and blunting ischemia-induced increased iNOS and decreased eNOS after MIRI.
Collapse
Affiliation(s)
- Younes Yassaghi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran
| | - Nasibeh Yousefzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Yaman Street, P.O. Box: 19395-4763, Velenjak, Tehran, Iran.
| |
Collapse
|
11
|
Maia LB. Bringing Nitric Oxide to the Molybdenum World-A Personal Perspective. Molecules 2023; 28:5819. [PMID: 37570788 PMCID: PMC10420851 DOI: 10.3390/molecules28155819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Molybdenum-containing enzymes of the xanthine oxidase (XO) family are well known to catalyse oxygen atom transfer reactions, with the great majority of the characterised enzymes catalysing the insertion of an oxygen atom into the substrate. Although some family members are known to catalyse the "reverse" reaction, the capability to abstract an oxygen atom from the substrate molecule is not generally recognised for these enzymes. Hence, it was with surprise and scepticism that the "molybdenum community" noticed the reports on the mammalian XO capability to catalyse the oxygen atom abstraction of nitrite to form nitric oxide (NO). The lack of precedent for a molybdenum- (or tungsten) containing nitrite reductase on the nitrogen biogeochemical cycle contributed also to the scepticism. It took several kinetic, spectroscopic and mechanistic studies on enzymes of the XO family and also of sulfite oxidase and DMSO reductase families to finally have wide recognition of the molybdoenzymes' ability to form NO from nitrite. Herein, integrated in a collection of "personal views" edited by Professor Ralf Mendel, is an overview of my personal journey on the XO and aldehyde oxidase-catalysed nitrite reduction to NO. The main research findings and the path followed to establish XO and AO as competent nitrite reductases are reviewed. The evidence suggesting that these enzymes are probable players of the mammalian NO metabolism is also discussed.
Collapse
Affiliation(s)
- Luisa B Maia
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology (FCT NOVA), 2829-516 Caparica, Portugal
| |
Collapse
|
12
|
Ghasemi A, Gheibi S, Kashfi K, Jeddi S. Anti-oxidant effect of nitrite in the pancreatic islets of type 2 diabetic male rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:420-428. [PMID: 37009002 PMCID: PMC10008387 DOI: 10.22038/ijbms.2023.68245.14900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/09/2023] [Indexed: 04/04/2023]
Abstract
Objectives Nitrite, a nitric oxide (NO) donor, increases insulin secretion from pancreatic islets and has positive metabolic effects in type 2 diabetes (T2D). Here, we test the hypothesis of whether nitrite-induced insulin secretion is due to blunting of diabetes-induced oxidative stress in the islets. Materials and Methods T2D was created in male rats using a combination of streptozotocin at 25 mg/kg and a high-fat diet. Wistar rats were assigned to 3 groups (n=6 in each group), including control, T2D, and T2D+nitrite; the latter group consumed drinking water containing sodium nitrite (50 mg/l) for eight weeks. At the end of the study, mRNA levels of NADPH oxidase (Nox1, 2, 3, and 4), superoxide dismutase (SOD1, 2, and 3), glutathione peroxides (GPX1 and 7), glutathione reductase (GR), catalase, thioredoxin (TXN1 and 2), and thioredoxin reductase (TXNRD1) were measured in the isolated pancreatic islets. Results In the islets of diabetic rats, mRNA expressions of Nox1, 2, and 4 were higher, whereas expressions of SOD1, 2, catalase, GPX1, 7, GR, and TXN1 were lower than controls. Nitrite significantly (all P-values<0.05) decreased gene expression of Nox1 (0.39-fold) and Nox4 (0.23-fold) and increased gene expression of SOD1 (2.2-fold), SOD2 (2.8-fold), catalase (2.7-fold), GPX1 (2.2-fold), GPX7 (6.0-fold), GR (3.0-fold), TXN1 (2.1-fold), and TXNRD1 (2.3-fold) in diabetic rats. Conclusion Nitrite decreased oxidative stress in isolated pancreatic islets of rats with T2D by suppressing oxidants and augmenting anti-oxidants. These findings favor the notion that nitrite-induced insulin secretion is partially due to decreased oxidative stress.
Collapse
Affiliation(s)
- Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sevda Gheibi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Clinical Sciences in Malmö, Unit of Molecular Metabolism, Lund University Diabetes Center, Clinical Research Center, Lund University, Malmö, Sweden
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
- Corresponding authors: Khosrow Kashfi. Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA. ; Sajad Jeddi. Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Corresponding authors: Khosrow Kashfi. Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA. ; Sajad Jeddi. Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Bryan NS, Ahmed S, Lefer DJ, Hord N, von Schwarz ER. Dietary nitrate biochemistry and physiology. An update on clinical benefits and mechanisms of action. Nitric Oxide 2023; 132:1-7. [PMID: 36690137 DOI: 10.1016/j.niox.2023.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/08/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
It is now more than 35 years since endothelium derived relaxing factor was identified as nitric oxide (NO). The last few decades have seen an explosion around nitric oxide biochemistry, physiology and clinical translation. The science reveals that all chronic disease is associated with decreased blood flow to the affected organ which results in increased inflammation, oxidative stress and immune dysfunction. This is true for cardiovascular disease, neurological disease, kidney, lung, liver disorders and every other major disorder. Since nitric oxide controls and regulates blood flow, oxygen and nutrient delivery to every cell, tissue and organ in the body and also mitigates inflammation, oxidative stress and immune dysfunction, a focus on restoring nitric oxide production is an obvious therapeutic strategy for a number of poorly managed chronic diseases. Since dietary nitrate is a major contributor to endogenous nitric oxide production, it should be considered as a means of therapy and restoration of nitric oxide. This review will update on the current state of the science and effects of inorganic nitrate administered through the diet on several chronic conditions and reveal how much is needed. It is clear now that antiseptic mouthwash and use of antacids disrupt nitrate metabolism to nitric oxide leading to clinical symptoms of nitric oxide deficiency. Based on the science, nitrate should be considered an indispensable nutrient that should be accounted for in dietary guidelines.
Collapse
Affiliation(s)
| | | | - David J Lefer
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, United States
| | - Norman Hord
- OU Health, Harold Hamm Diabetes Center, Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | | |
Collapse
|
14
|
Distribution of dietary nitrate and its metabolites in rat tissues after 15N-labeled nitrate administration. Sci Rep 2023; 13:3499. [PMID: 36859526 PMCID: PMC9977953 DOI: 10.1038/s41598-023-28190-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/13/2023] [Indexed: 03/03/2023] Open
Abstract
The reduction pathway of nitrate (NO3-) and nitrite (NO2-) to nitric oxide (NO) contributes to regulating many physiological processes. To examine the rate and extent of dietary nitrate absorption and its reduction to nitrite, we supplemented rat diets with Na15NO3-containing water (1 g/L) and collected plasma, urine and several tissue samples. We found that plasma and urine showed 8.8- and 11.7-fold increases respectively in total nitrate concentrations in 1-day supplementation group compared to control. In tissue samples-gluteus, liver and eyes-we found 1.7-, 2.4- and 4.2-fold increases respectively in 1-day supplementation group. These increases remained similar in 3-day supplementation group. LC-MS/MS analysis showed that the augmented nitrate concentrations were primarily from the exogenously provided 15N-nitrate. Overall nitrite concentrations and percent of 15N-nitrite were also greatly increased in all samples after nitrate supplementation; eye homogenates showed larger increases compared to gluteus and liver. Moreover, genes related to nitrate transport and reduction (Sialin, CLC and XOR) were upregulated after nitrate supplementation for 3 days in muscle (Sialin 2.3-, CLC1 1.3-, CLC3 2.1-, XOR 2.4-fold) and eye (XOR 1.7-fold) homogenates. These results demonstrate that dietary nitrate is quickly absorbed into circulation and tissues, and it can be reduced to nitrite in tissues (and likely to NO) suggesting that nitrate-enriched diets can be an efficient intervention to enhance nitrite and NO bioavailability.
Collapse
|
15
|
Yamasaki H, Imai H, Tanaka A, Otaki JM. Pleiotropic Functions of Nitric Oxide Produced by Ascorbate for the Prevention and Mitigation of COVID-19: A Revaluation of Pauling's Vitamin C Therapy. Microorganisms 2023; 11:397. [PMID: 36838362 PMCID: PMC9963342 DOI: 10.3390/microorganisms11020397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Linus Pauling, who was awarded the Nobel Prize in Chemistry, suggested that a high dose of vitamin C (l-ascorbic acid) might work as a prevention or treatment for the common cold. Vitamin C therapy was tested in clinical trials, but clear evidence was not found at that time. Although Pauling's proposal has been strongly criticized for a long time, vitamin C therapy has continued to be tested as a treatment for a variety of diseases, including coronavirus infectious disease 2019 (COVID-19). The pathogen of COVID-19, SARS-CoV-2, belongs to the β-coronavirus lineage, which includes human coronavirus, severe acute respiratory syndrome (SARS), and Middle East respiratory syndrome (MERS). This review intends to shed new light on vitamin C antiviral activity that may prevent SARS-CoV-2 infection through the chemical production of nitric oxide (NO). NO is a gaseous free radical that is largely produced by the enzyme NO synthase (NOS) in cells. NO produced by upper epidermal cells contributes to the inactivation of viruses and bacteria contained in air or aerosols. In addition to enzymatic production, NO can be generated by the chemical reduction of inorganic nitrite (NO2-), an alternative mechanism for NO production in living organisms. Dietary vitamin C, largely contained in fruits and vegetables, can reduce the nitrite in saliva to produce NO in the oral cavity when chewing foods. In the stomach, salivary nitrite can also be reduced to NO by vitamin C secreted from the epidermal cells of the stomach. The strong acidic pH of gastric juice facilitates the chemical reduction of salivary nitrite to produce NO. Vitamin C contributes in multiple ways to the host innate immune system as a first-line defense mechanism against pathogens. Highlighting chemical NO production by vitamin C, we suggest that controversies on the therapeutic effects of vitamin C in previous clinical trials may partly be due to less appreciation of the pleiotropic functions of vitamin C as a universal bioreductant.
Collapse
Affiliation(s)
- Hideo Yamasaki
- Faculty of Science, University of the Ryukyus, Nishihara 903-0213, Okinawa, Japan
| | | | | | | |
Collapse
|
16
|
Quantitative aspects of nitric oxide production in the heart. Mol Biol Rep 2022; 49:11113-11122. [DOI: 10.1007/s11033-022-07889-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022]
|
17
|
Ouyang M, Ouyang X, Peng Z, Liu M, Xu G, Zou Z, Zhang M, Shang Q. Heart-targeted amelioration of sepsis-induced myocardial dysfunction by microenvironment responsive nitric oxide nanogenerators in situ. J Nanobiotechnology 2022; 20:263. [PMID: 35672697 PMCID: PMC9171488 DOI: 10.1186/s12951-022-01457-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/07/2022] [Indexed: 12/28/2022] Open
Abstract
Background A balanced endogenous level of bioavailable nitric oxide (NO) plays a key role in maintaining cardiovascular homeostasis. The bioactive NO level in the cardiomyocytes was much reduced during sepsis. However, it is clinically challenging for the NO gas therapy due to the lack of spatial and temporal release system with precise control. The purpose of this study is to design a NO-releasing biomaterial with heart-targeted capability responsive to the infectious microenvironment, thus ameliorating lipopolysaccharide (LPS)-induced cardiac dysfunction. Results The heart-targeted NO delivery and in situ releasing system, PCM-MSN@LA, was synthesized using hollow mesoporous silica nanoparticles (MSN) as the carrier, and L-arginine (LA) as the NO donor. The myocardial delivery was successfully directed to heart by specific peptide (PCM) combined with low-intensity focused ultrasound (LIFU) guidance. The myocardial system synthesized NO from the LA released from PCM-MSN@LA in the presence of increased endogenous nitric oxide synthase (NOS) activity induced by LPS. This targeted NO release in situ achieved extraordinary protective effects against LPS-challenged myocardial injury by reducing the recruitment of inflammatory cells, inhibiting oxidative stress and maintaining the mitochondria integrity. In particular, this protection was not compromised by simultaneous circulation collapse as an adverse event in the context. Conclusions PCM-MSN@LA + LIFU exhibited extraordinary cardioprotective effects against severe sepsis in the hearts of LPS-treated animals without the side effect of NO diffusion. This technology has great potential to be served as a novel therapeutic strategy for sepsis-induced myocardial injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01457-y.
Collapse
|
18
|
Cai Y, Zhang B, Shalamu A, Gao T, Ge J. Soluble guanylate cyclase (sGC) stimulator vericiguat alleviates myocardial ischemia-reperfusion injury by improving microcirculation. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:662. [PMID: 35845490 PMCID: PMC9279818 DOI: 10.21037/atm-22-2583] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/08/2022] [Indexed: 11/21/2022]
Abstract
Background This study aimed to verify the effect of soluble guanylate cyclase (sGC) stimulator vericiguat on myocardial ischemia-reperfusion injury and explore its mechanism. Methods A myocardial ischemia-reperfusion injury model of mice was established and intravenous administration was performed 2 minutes before reperfusion. Triphenyltetrazolium chloride (TTC) staining and echocardiography were used to verify the effect of vericiguat on myocardial ischemia-reperfusion injury in the infarct area, and immunofluorescence was used to observe myocardial pathological changes at different time points after reperfusion. Quantitative proteomics was conducted to analysis the main differentially expressed proteins after drug intervention. The distribution of endothelial cells and sGC after myocardial ischemia-reperfusion injury in mice was observed by immunofluorescence. RNA sequencing of endothelial cells was used to search for differentially expressed molecules. Thioflavin-S staining was used to observe the effect of vericiguat on improving the nonrecurrence phenomenon and reducing the infarct size after reperfusion. Results The effect of the sGC stimulator vericiguat on myocardial ischemia-reperfusion injury was verified, and myocardial microcirculation significantly increased after drug intervention. Quantitative proteomics found that the protein expression of myocardial tissue in the ischemia-reperfusion area was not significantly different in the drug intervention group, except for increased adenosine triphosphate (ATP) activity. Vericiguat, nitroglycerin, and nitrite did not directly affect apoptosis or cell viability. RNA sequencing of human umbilical vein endothelial cells screened the upregulated antioxidant response. Conclusions SGC stimulator vericiguat ameliorated myocardial ischemia-reperfusion injury through indirect pathways of improving microcirculation.
Collapse
Affiliation(s)
- Yun Cai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Beijian Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Adilan Shalamu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Tingwen Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Cardiovascular Diseases, Shanghai, China.,Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China.,Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
19
|
Bryan NS, Burleigh MC, Easton C. The oral microbiome, nitric oxide and exercise performance. Nitric Oxide 2022; 125-126:23-30. [PMID: 35636654 DOI: 10.1016/j.niox.2022.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 05/12/2022] [Accepted: 05/22/2022] [Indexed: 12/15/2022]
Abstract
The human microbiome comprises ∼1013-1014 microbial cells which form a symbiotic relationship with the host and play a critical role in the regulation of human metabolism. In the oral cavity, several species of bacteria are capable of reducing nitrate to nitrite; a key precursor of the signaling molecule nitric oxide. Nitric oxide has myriad physiological functions, which include the maintenance of cardiovascular homeostasis and the regulation of acute and chronic responses to exercise. This article provides a brief narrative review of the research that has explored how diversity and plasticity of the oral microbiome influences nitric oxide bioavailability and related physiological outcomes. There is unequivocal evidence that dysbiosis (e.g. through disease) or disruption (e.g. by use of antiseptic mouthwash or antibiotics) of the oral microbiota will suppress nitric oxide production via the nitrate-nitrite-nitric oxide pathway and negatively impact blood pressure. Conversely, there is preliminary evidence to suggest that proliferation of nitrate-reducing bacteria via the diet or targeted probiotics can augment nitric oxide production and improve markers of oral health. Despite this, it is yet to be established whether purposefully altering the oral microbiome can have a meaningful impact on exercise performance. Future research should determine whether alterations to the composition and metabolic activity of bacteria in the mouth influence the acute responses to exercise and the physiological adaptations to exercise training.
Collapse
Affiliation(s)
- Nathan S Bryan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Mia C Burleigh
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Blantyre, UK
| | - Chris Easton
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Blantyre, UK.
| |
Collapse
|
20
|
Ghasemi A. Quantitative aspects of nitric oxide production from nitrate and nitrite. EXCLI JOURNAL 2022; 21:470-486. [PMID: 35391922 PMCID: PMC8983853 DOI: 10.17179/excli2022-4727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 11/05/2022]
Abstract
Nitric oxide (NO) is involved in many physiological and pathological processes in the human body. At least two major pathways produce NO: (1) the L-arginine-NO-oxidative pathway in which NO synthase (NOS) enzymes convert L-arginine to NO; (2) the nitrate-nitrite-NO reductive pathway in which NO is produced from the serial reduction of nitrate and nitrite. The deficiency of NO is involved in the pathophysiology of cardiometabolic disorders. Intervention with foods containing nitrate and nitrite can potentially prevent or treat some chronic diseases, including cardiovascular diseases and diabetes. A better understanding of the NO cycle would help develop effective strategies for preventing or treating the disorders in which NO homeostasis is disturbed. This review summarizes quantitative aspects of NO production, emphasizing the nitrate-nitrite-NO pathway. Available data indicates that total NO production by NOS-dependent L-arginine-NO pathway is about 1000 μmol.day-1. Of about 1700 μmol.day-1 ingested nitrate, ~25 % is extracted by the salivary glands and of which ~20 % is converted nitrite. It means that about 5 % of ingested nitrate is converted to nitrite in the oral cavity; assuming that all produced nitrite is reduced to NO in the stomach, it can be calculated that contribution of the nitrate-nitrite-NO pathway to the whole-body NO production is about 85 μmol.day-1 (1700 ×0.05=85) or approximately 100 μmol.day-1. The lower contribution of the nitrate-nitrite-NO pathway does not mean that this pathway has lower importance in the whole-body NO homeostasis. Even in the adequate L-arginine supply, NOS-dependent NO production is insufficient to meet all NO functions, and the nitrate-nitrite-NO pathway must provide the rest. In conclusion, the contribution of the nitrate-nitrite-NO pathway in the whole human body NO production is <10 %, and the nitrate-nitrite-NO pathway is complementary to the NOS-dependent NO production.
Collapse
Affiliation(s)
- Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Lehnert N, Kim E, Dong HT, Harland JB, Hunt AP, Manickas EC, Oakley KM, Pham J, Reed GC, Alfaro VS. The Biologically Relevant Coordination Chemistry of Iron and Nitric Oxide: Electronic Structure and Reactivity. Chem Rev 2021; 121:14682-14905. [PMID: 34902255 DOI: 10.1021/acs.chemrev.1c00253] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nitric oxide (NO) is an important signaling molecule that is involved in a wide range of physiological and pathological events in biology. Metal coordination chemistry, especially with iron, is at the heart of many biological transformations involving NO. A series of heme proteins, nitric oxide synthases (NOS), soluble guanylate cyclase (sGC), and nitrophorins, are responsible for the biosynthesis, sensing, and transport of NO. Alternatively, NO can be generated from nitrite by heme- and copper-containing nitrite reductases (NIRs). The NO-bearing small molecules such as nitrosothiols and dinitrosyl iron complexes (DNICs) can serve as an alternative vehicle for NO storage and transport. Once NO is formed, the rich reaction chemistry of NO leads to a wide variety of biological activities including reduction of NO by heme or non-heme iron-containing NO reductases and protein post-translational modifications by DNICs. Much of our understanding of the reactivity of metal sites in biology with NO and the mechanisms of these transformations has come from the elucidation of the geometric and electronic structures and chemical reactivity of synthetic model systems, in synergy with biochemical and biophysical studies on the relevant proteins themselves. This review focuses on recent advancements from studies on proteins and model complexes that not only have improved our understanding of the biological roles of NO but also have provided foundations for biomedical research and for bio-inspired catalyst design in energy science.
Collapse
Affiliation(s)
- Nicolai Lehnert
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Eunsuk Kim
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Hai T Dong
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Jill B Harland
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Andrew P Hunt
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Elizabeth C Manickas
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Kady M Oakley
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - John Pham
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Garrett C Reed
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Victor Sosa Alfaro
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| |
Collapse
|
22
|
Antioxidant tempol modulates the increases in tissue nitric oxide metabolites concentrations after oral nitrite administration. Chem Biol Interact 2021; 349:109658. [PMID: 34543659 DOI: 10.1016/j.cbi.2021.109658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 08/11/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022]
Abstract
Nitric oxide (NO) metabolites have physiological and pharmacological importance and increasing their tissue concentrations may result in beneficial effects. Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl) has antioxidant properties that may improve NO bioavailability. Moreover, tempol increases oral nitrite-derived gastric formation of S-nitrosothiols (RSNO). We hypothesized that pretreatment with tempol may further increase tissue concentrations of NO-related species after oral nitrite administration and therefore we carried out a time-dependent analysis of how tempol affects the concentrations of NO metabolites in different tissues after oral nitrite administration to rats. NO metabolites (nitrate, nitrite and RSNO) were assessed by ozone-based reductive chemiluminescence assays in plasma, stomach, aorta, heart and liver samples obtained from anesthetized rats at baseline conditions and 15 min, 30 min, 2 h or 24 h after oral nitrite (15 mg/kg) was administered to rats pretreated with tempol (18 mg/kg) or vehicle 15 min prior to nitrite administration. Aortic protein nitrosation was assessed by resin-assited capture (SNO-RAC) method. We found that pretreatment with tempol transiently enhanced nitrite-induced increases in nitrite, RSNO and nitrate concentrations in the stomach and in the plasma (all P < 0.05), particularly for 15-30 min, without affecting aortic protein nitrosation. Pretreatment with tempol enhanced nitrite-induced increases in nitrite (but not RSNO or nitrate) concentrations in the heart (P < 0.05). In contrast, tempol attenuated nitrite-induced increases in nitrite, RSNO or nitrate concentrations in the liver. These findings show that pretreatment with tempol affects oral nitrite-induced changes in tissue concentrations of NO metabolites depending on tissue type and does not increase nitrite-induced vascular nitrosation. These results may indicate that oral nitrite therapy aiming at achieving increased nitrosation of cardiovascular targets requires appropriate doses of nitrite and is not optimized by tempol.
Collapse
|
23
|
Griffiths K, Lee JJ, Frenneaux MP, Feelisch M, Madhani M. Nitrite and myocardial ischaemia reperfusion injury. Where are we now? Pharmacol Ther 2021; 223:107819. [PMID: 33600852 DOI: 10.1016/j.pharmthera.2021.107819] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite major advances in technology and treatment, with coronary heart disease (CHD) being a key contributor. Following an acute myocardial infarction (AMI), it is imperative that blood flow is rapidly restored to the ischaemic myocardium. However, this restoration is associated with an increased risk of additional complications and further cardiomyocyte death, termed myocardial ischaemia reperfusion injury (IRI). Endogenously produced nitric oxide (NO) plays an important role in protecting the myocardium from IRI. It is well established that NO mediates many of its downstream functions through the 'canonical' NO-sGC-cGMP pathway, which is vital for cardiovascular homeostasis; however, this pathway can become impaired in the face of inadequate delivery of necessary substrates, in particular L-arginine, oxygen and reducing equivalents. Recently, it has been shown that during conditions of ischaemia an alternative pathway for NO generation exists, which has become known as the 'nitrate-nitrite-NO pathway'. This pathway has been reported to improve endothelial dysfunction, protect against myocardial IRI and attenuate infarct size in various experimental models. Furthermore, emerging evidence suggests that nitrite itself provides multi-faceted protection, in an NO-independent fashion, against a myriad of pathophysiologies attributed to IRI. In this review, we explore the existing pre-clinical and clinical evidence for the role of nitrate and nitrite in cardioprotection and discuss the lessons learnt from the clinical trials for nitrite as a perconditioning agent. We also discuss the potential future for nitrite as a pre-conditioning intervention in man.
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jordan J Lee
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael P Frenneaux
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
24
|
Fernandes JO, Tella SOC, Ferraz IS, Ciampo LAD, Tanus-Santos JE. Assessment of nitric oxide metabolites concentrations in plasma, saliva, and breast milk and their relationship in lactating women. Mol Cell Biochem 2021; 476:1293-1302. [PMID: 33237454 DOI: 10.1007/s11010-020-03994-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022]
Abstract
Nitric oxide (NO) plays a role in many biological mechanisms. The amounts of physiologically produced NO are associated with the concentrations of its metabolites nitrate and nitrite. This study investigated whether there is any association between the concentrations of NO metabolites nitrate, nitrite, and nitrosylated species (RXNO) in mature breast milk, saliva, and plasma in healthy lactating women (N = 30). We hypothesized that the NO metabolites concentrations in plasma are associated with those found in saliva and in breast milk. NO metabolites concentrations were measured using chemiluminensce-based assays. Nitrate concentrations in breast milk are twice as much as plasma concentrations, whereas nitrate concentrations in saliva are about eightfold higher (both P < 0.001). Similar differences were found when nitrite concentrations were taken into consideration. RXNO concentrations in breast milk were negligible, and RXNO concentrations in saliva were approximately sixfold higher than those found in plasma samples (P < 0.0001). Nitrate concentrations in plasma are associated with nitrate concentrations in saliva (rs = 0.474, P = 0.004). However, no significant association was found between nitrate concentrations in breast milk and in plasma (P > 0.05). Our results show a significant association between nitrate concentrations in plasma with those found in saliva, whereas all other relationships were not significant. In conclusion, this report shows for the first time that the physiological concentrations of NO metabolites in human breast milk are probably independent of circulating NO metabolites concentrations and may depend mostly on endogenous NO synthesis in the breast. These findings may have clinical implications for newborns and lactating women.
Collapse
Affiliation(s)
- Juliana O Fernandes
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Sandra O C Tella
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Ivan S Ferraz
- Department of Puericulture and Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Luiz A D Ciampo
- Department of Puericulture and Pediatrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
25
|
Probiotic Properties of Lactiplantibacillus plantarum LB5 Isolated from Kimchi Based on Nitrate Reducing Capability. Foods 2020; 9:foods9121777. [PMID: 33266127 PMCID: PMC7760155 DOI: 10.3390/foods9121777] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 01/01/2023] Open
Abstract
The purpose of this study was to investigate the probiotic properties of lactic acid bacteria isolated from Korean radish water kimchi (dongchimi). A total of 800 isolates of lactic acid bacteria were isolated from kimchi, and the strain having reduction and tolerance capability for nitrate and nitrite was selected and identified as Lactiplantibacillus plantarum LB5 (LPLB5) by 16S rRNA sequencing. LPLB5 showed higher tolerance to acidic pH values (pH 2.5), 0.3% bile salts, and heat treatment (40, 50, and 60 °C). Antibacterial activity showed strong inhibition against four food-borne pathogenic bacteria (E. coli O157:H7 ATCC 35150, Pseudomonas aeruginosa KCCM 12539, Listeria monocytogenes KCCM 40307, and Staphylococcus aureus ATCC 25923). The strain did not show any antibiotic resistance, β-hemolytic activity, or ability to produce β-glucuronidase. LPLB5 also exhibited a 30% auto-aggregation ability and 33–60% co-aggregation ability with four pathogenic bacteria (E. coli O157: H7 ATCC 35150, E. coli KCTC 2571, L. monocytogenes ATCC 51776, and S. aureus ATCC 25923). Moreover, the strain showed approximately 40% 2,2-diphenyl-1-picryl-hydrazyl (DPPH) radical- and 10% 2-azino-bis-(3-ethylbenzothiazoline-6-sulfonic acid (ABTS) radical-scavenging activity. In cell culture studies, human colon epithelial cells (Caco-2) were treated with LPLB5 (106 and 107 CFU/mL); the bacteria showed more than 70% adherence onto and a 32% invasion rate into the Caco-2 cells. LPLB5 significantly decreased the mRNA expression levels of pro-inflammatory cytokines (interleukin-1 beta (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor-alpha (TNF-α)) and increased the mRNA expression levels of anti-inflammatory cytokines (interleukin-4 (IL-4), interleukin-10 (IL-10), and interferon-gamma (IFN-γ)) in lipopolysaccharide-stimulated Caco-2 cells. Our data suggest that LPLB5 is safe and possesses probiotic, antioxidant, and anti-inflammatory activities.
Collapse
|
26
|
Sanches-Lopes JM, Ferreira GC, Pinheiro LC, Kemp R, Tanus-Santos JE. Consistent gastric pH-dependent effects of suppressors of gastric acid secretion on the antihypertensive responses to oral nitrite. Biochem Pharmacol 2020; 177:113940. [DOI: 10.1016/j.bcp.2020.113940] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/24/2020] [Indexed: 01/24/2023]
|
27
|
Tropea T, Renshall LJ, Nihlen C, Weitzberg E, Lundberg JO, David AL, Tsatsaris V, Stuckey DJ, Wareing M, Greenwood SL, Sibley CP, Cottrell EC. Beetroot juice lowers blood pressure and improves endothelial function in pregnant eNOS -/- mice: importance of nitrate-independent effects. J Physiol 2020; 598:4079-4092. [PMID: 32368787 DOI: 10.1113/jp279655] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Maternal hypertension is associated with increased rates of pregnancy pathologies, including fetal growth restriction, due at least in part to reductions in nitric oxide (NO) bioavailability and associated vascular dysfunction. Dietary nitrate supplementation, from beetroot juice (BRJ), has been shown to increase NO bioavailability and improve cardiovascular function in both preclinical and clinical studies. This study is the first to investigate effects of dietary nitrate supplementation in a pregnant animal model. Importantly, the effects of nitrate-containing BRJ were compared with both 'placebo' (nitrate-depleted) BRJ as well as water to control for potential nitrate-independent effects. Our data show novel, nitrate-independent effects of BRJ to lower blood pressure and improve vascular function in endothelial nitric oxide synthase knockout (eNOS-/- ) mice. These findings suggest potential beneficial effects of BRJ supplementation in pregnancy, and emphasize the importance of accounting for nitrate-independent effects of BRJ in study design and interpretation. ABSTRACT Maternal hypertension is associated with adverse pregnancy outcomes, including fetal growth restriction (FGR), due in part to reductions in nitric oxide (NO) bioavailability. We hypothesized that maternal dietary nitrate administration would increase NO bioavailability to reduce systolic blood pressure (SBP), improve vascular function and increase fetal growth in pregnant endothelial NO synthase knockout (eNOS-/- ) mice, which exhibit hypertension, endothelial dysfunction and FGR. Pregnant wildtype (WT) and eNOS-/- mice were supplemented with nitrate-containing beetroot juice (BRJ+) from gestational day (GD) 12.5. Control mice received an equivalent dose of nitrate-depleted BRJ (BRJ-) or normal drinking water. At GD17.5, maternal SBP was measured; at GD18.5, maternal nitrate/nitrite concentrations, uterine artery (UtA) blood flow and endothelial function were assessed, and pregnancy outcomes were determined. Plasma nitrate concentrations were increased in both WT and eNOS-/- mice supplemented with BRJ+ (P < 0.001), whereas nitrite concentrations were increased only in eNOS-/- mice (P < 0.001). BRJ- did not alter nitrate/nitrite concentrations. SBP was lowered and UtA endothelial function was enhanced in eNOS-/- mice supplemented with either BRJ+ or BRJ-, indicating nitrate-independent effects of BRJ. Improvements in endothelial function in eNOS-/- mice were abrogated in the presence of 25 mm KCl, implicating enhanced EDH signalling in BRJ- treated animals. At GD18.5, eNOS-/- fetuses were significantly smaller than WT animals (P < 0.001), but BRJ supplementation did not affect fetal weight. BRJ may be a beneficial intervention in pregnancies associated with hypertension, endothelial dysfunction and reduced NO bioavailability. Our data showing biological effects of non-nitrate components of BRJ have implications for both interpretation of previous findings and in the design of future clinical trials.
Collapse
Affiliation(s)
- Teresa Tropea
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, UK
| | - Lewis J Renshall
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, UK
| | - Carina Nihlen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, SE-171 77, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, SE-171 77, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, SE-171 77, Sweden
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
| | - Vassilis Tsatsaris
- Obstetrics and Gynecology Unit, Maternité Port-Royal, APHP, Paris V, Paris, France
| | - Daniel J Stuckey
- Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Mark Wareing
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, UK
| | - Susan L Greenwood
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, UK
| | - Colin P Sibley
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, UK
| | - Elizabeth C Cottrell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, St. Mary's Hospital, Manchester, UK
| |
Collapse
|
28
|
Pinheiro LC, Ferreira GC, Damacena de Angelis C, Toledo JC, Tanus-Santos JE. A comprehensive time course study of tissue nitric oxide metabolites concentrations after oral nitrite administration. Free Radic Biol Med 2020; 152:43-51. [PMID: 32151744 DOI: 10.1016/j.freeradbiomed.2020.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 02/18/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022]
Abstract
Nitrite and nitrate are considered nitric oxide (NO) storage pools. The assessment of their tissue concentrations may improve our understanding of how they attenuate pathophysiological mechanisms promoting disease. We hypothesized that significant differences exist when the tissue concentrations of nitrite, nitrate, and nitrosylated species (RXNO) are compared among different tissues, particularly when nitrite is administered orally because nitrite generates various NO-related species in the stomach. We studied the different time-dependent changes in plasma and tissue concentrations of nitrite, nitrate, and RXNO after oral nitrite 15 mg/kg was administered rats, which were euthanized 15, 30, 60, 120, 240, 480 or 1440 min after nitrite administration. A control group received water. Arterial blood samples were collected and the rats were perfused with a PBS solution containing NEM/DTPA to prevent the destruction of RXNO. After perfusion, heart, aorta, mesenteric artery, brain, stomach, liver and femoral muscle were harvested and immediately stored at -70°C until analyzed for their nitrite, nitrate and RXNO contents using an ozone-based reductive chemiluminescence assay. While nitrite administration did not increase aortic nitrite or nitrate concentrations for at least 60 min, both aorta and mesenteric vessels stored nitrite from 8 to 24 h after its administration and their tissue concentrations increased from 10 to 40-fold those found in plasma. In contrast, the other studied tissues showed only transient increases in the concentrations of these NO metabolites, including RXNO. The differences among tissues may reflect differences in mechanisms regulating cellular influx of nitrite. These findings have important pharmacological and clinical implications.
Collapse
Affiliation(s)
- Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Graziele C Ferreira
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Célio Damacena de Angelis
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, 13083-887, Campinas, Sao Paulo, Brazil
| | - Jose Carlos Toledo
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of Sao Paulo, 14040-901, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
29
|
Amaral JH, Rizzi ES, Alves-Lopes R, Pinheiro LC, Tostes RC, Tanus-Santos JE. Antioxidant and antihypertensive responses to oral nitrite involves activation of the Nrf2 pathway. Free Radic Biol Med 2019; 141:261-268. [PMID: 31251976 DOI: 10.1016/j.freeradbiomed.2019.06.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/14/2019] [Accepted: 06/24/2019] [Indexed: 12/27/2022]
Abstract
Impaired redox balance contributes to the cardiovascular alterations of hypertension and activation of nuclear factor erythroid 2-related factor 2 (Nrf2) pathway may counteract these alterations. While nitrite recycles back to NO and exerts antioxidant and antihypertensive effects, the mechanisms involved in these responses are not fully understood. We hypothesized that nitrite treatment of two-kidney, one-clip (2K1C) hypertensive rats activates the Nrf2 pathway, promotes the transcription of antioxidant genes, and improves the vascular redox imbalance and dysfunction in this model. Two doses of oral nitrite were studied: 15 mg/kg and the sub-antihypertensive dose of 1 mg/kg. Nitrite 15 mg/kg (but not 1 mg/kg) decreased blood pressure and increased circulating plasma nitrite and nitrate. Both doses blunted hypertension-induced increases in mesenteric artery reactive oxygen species concentrations assessed by DHE technique and restored the impaired mesenteric artery responses to acetylcholine. While 2K1C hypertension decreased nuclear Nrf2 accumulation, both doses of nitrite increased nuclear Nrf2 accumulation and mRNA expression of Nrf2-regulated genes including superoxide dismutase-1 (SOD1), catalase (CAT), glutathione peroxidase (GPX), thioredoxin-1(TRDX-1) and -2 (TRDX-2). To further confirm nitrite-mediated antioxidant effects, we measured vascular SOD and GPX activity and we found that nitrite at 1 or 15 mg/kg increased the activity of both enzymes (P < 0.05). These results suggest that activation of the Nrf2 pathway promotes antioxidant effects of nitrite, which may improve the vascular dysfunction in hypertension, even when nitrite is given at a sub-antihypertensive dose. These findings may have many clinical implications, particularly in the therapy of hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Jefferson H Amaral
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Elen S Rizzi
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Rhéure Alves-Lopes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
30
|
Mechanism of nitrite-dependent NO synthesis by human sulfite oxidase. Biochem J 2019; 476:1805-1815. [DOI: 10.1042/bcj20190143] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/21/2019] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
AbstractIn addition to nitric oxide (NO) synthases, molybdenum-dependent enzymes have been reported to reduce nitrite to produce NO. Here, we report the stoichiometric reduction in nitrite to NO by human sulfite oxidase (SO), a mitochondrial intermembrane space enzyme primarily involved in cysteine catabolism. Kinetic and spectroscopic studies provide evidence for direct nitrite coordination at the molybdenum center followed by an inner shell electron transfer mechanism. In the presence of the physiological electron acceptor cytochrome c, we were able to close the catalytic cycle of sulfite-dependent nitrite reduction thus leading to steady-state NO synthesis, a finding that strongly supports a physiological relevance of SO-dependent NO formation. By engineering SO variants with reduced intramolecular electron transfer rate, we were able to increase NO generation efficacy by one order of magnitude, providing a mechanistic tool to tune NO synthesis by SO.
Collapse
|
31
|
NADPH oxidase is a primary target for antioxidant effects by inorganic nitrite in lipopolysaccharide-induced oxidative stress in mice and in macrophage cells. Nitric Oxide 2019; 89:46-53. [PMID: 31063820 DOI: 10.1016/j.niox.2019.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/23/2019] [Accepted: 05/02/2019] [Indexed: 12/24/2022]
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and oxidative stress is usually considered as an important factor to the pathogenesis of various diseases. Inorganic nitrite, previously viewed as a harmful substance in our diet or inert metabolites of endogenous NO, is recently identified as an important biological NO reservoir in vasculature and tissues. Stimulation of a nitrite-NO pathway shows organ-protective effects on oxidative stress and inflammation, but the mechanisms or target are not clear. In this study, the hypothesis that inorganic nitrite attenuated lipopolysaccharide (LPS)-induced oxidative stress in mice and in macrophage cells by modulating NADPH oxidase activity and NO bioavailability were investigated. We showed that nitrite treatment, in sharp contrast with the worsening effect of NO synthases inhibition, significantly attenuated aortic oxidative stress, endothelial dysfunction and mortality in LPS-induced shock in mice. Mechanistically, protective effects of nitrite were abolished by NO scavenger and xanthine oxidase inhibitor, and inhibition of NADPH oxidase with apocynin attenuated LPS-induced oxidative stress similar to that of nitrite. In the presence of nitrite, no further effect of apocynin was observed, suggesting NADPH oxidase as a possible target. In LPS-activated macrophage cells, nitrite reduced NADPH oxidase activity and oxidative stress and these effects of nitrite were also abolished by NO scavenger and xanthine oxidase inhibitor, where xanthine oxidase-mediated reduction of nitrite attenuated NADPH oxidase activity in activated macrophages via a NO-dependent mechanism. In conclusion, these novel findings position NADPH oxidase in the inflammatory vasculature as a prime target for the antioxidant effects of inorganic nitrite, and open a new direction to modulate the inflammatory response.
Collapse
|
32
|
Bahadoran Z, Mirmiran P, Jeddi S, Carlström M, Azizi F, Ghasemi A. Circulating markers of nitric oxide homeostasis and cardiometabolic diseases: insights from population-based studies. Free Radic Res 2019; 53:359-376. [PMID: 30821533 DOI: 10.1080/10715762.2019.1587168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Emerging data suggest that impaired nitric oxide (NO) homeostasis has a key role in development of cardiometabolic disorders. The association between circulating levels of NO metabolites, i.e. nitrate and nitrite (NOx), and risk of chronic diseases has not yet been fully clarified. This work aims to address epidemiologic aspects of NO metabolism and discusses different physiologic and pathophysiologic conditions influencing circulating NOx. Further, cross-sectional associations of serum NOx with metabolic disorders are described and along the way, potential short-term and long-term power of serum NOx for predicting cardiometabolic outcomes are reviewed. Results from population-based studies show that circulating NOx is affected by aging, smoking habits, pregnancy, menopause status, thyroid hormones, and various pathologic conditions including type 2 diabetes, insulin resistance, hypertension, and renal dysfunction. Lifestyle factors, especially dietary habits, but also smoking habits and the degree of physical activity influence NO homeostasis and the circulating levels of NOx. Elevated serum NOx, due to increased iNOS activity, is associated with increased incidence of metabolic syndrome, different obesity phenotypes, and cardiovascular events.
Collapse
Affiliation(s)
- Zahra Bahadoran
- a Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Parvin Mirmiran
- b Department of Clinical Nutrition and Diet Therapy, Faculty of Nutrition Sciences and Food Technology , National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Sajad Jeddi
- c Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mattias Carlström
- d Department of Physiology and Pharmacology , Karolinska Institutet , Stockholm , Sweden
| | - Fereidoun Azizi
- e Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Asghar Ghasemi
- c Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
33
|
Wang L, Almeida LEF, Kamimura S, van der Meulen JH, Nagaraju K, Quezado M, Wakim P, Quezado ZMN. The role of nitrite in muscle function, susceptibility to contraction injury, and fatigability in sickle cell mice. Nitric Oxide 2018; 80:70-81. [PMID: 30114530 PMCID: PMC6186197 DOI: 10.1016/j.niox.2018.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 07/05/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
Abstract
Sickle cell disease (SCD) patients can have limited exercise capacity and muscle dysfunction characterized by decreased force, atrophy, microvascular abnormalities, fiber distribution changes, and skeletal muscle energetics abnormalities. Growing evidence suggests that in SCD there is alteration in nitric oxide (NO) availability/signaling and that nitrate/nitrite can serve as a NO reservoir and enhance muscle performance. Here, we examined effects of nitrite on muscle strength, exercise capacity, and on contractile properties of fast-(extensor digitorum longus, EDL) and slow-twitch (soleus) muscles in SCD mice. Compared to controls, homozygotes (sickling) had decreased grip strength, impaired wheel running performance, and decreased muscle mass of fast-twitch, but not slow-twitch muscle. Nitrite treatment yielded increases in nitrite plasma levels in controls, heterozygotes, and homozygotes but decreases in muscle nitrite levels in heterozygotes and homozygotes. Regardless of genotype, nitrite yielded increases in grip strength, which were coupled with increases in specific force in EDL, but not in soleus muscle. Further, nitrite increased EDL, but not soleus, fatigability in all genotypes. Conversely, in controls, nitrite decreased, whereas in homozygotes, it increased EDL susceptibility to contraction-induced injury. Interestingly, nitrite yielded no changes in distances ran on the running wheel. These differential effects of nitrite in fast- and slow-twitch muscles suggest that its ergogenic effects would be observed in high-intensity/short exercises as found with grip force increases but no changes on wheel running distances. Further, the differential effects of nitrite in homozygotes and control animals suggests that sickling mice, which have altered NO availability/signaling, handle nitrite differently than do control animals.
Collapse
Affiliation(s)
- Li Wang
- The Sheikh Zayed Institute for Pediatric Surgical Innovation and Center for Neuroscience Research, Children's Research Institute, Washington, DC, 20010, USA
| | - Luis E F Almeida
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sayuri Kamimura
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jack H van der Meulen
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Health System, Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Health System, Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20010, USA
| | - Martha Quezado
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Paul Wakim
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health Clinical Center, Bethesda, MD, 20892, USA
| | - Zenaide M N Quezado
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
34
|
Maia LB, Moura JJG. Putting xanthine oxidoreductase and aldehyde oxidase on the NO metabolism map: Nitrite reduction by molybdoenzymes. Redox Biol 2018; 19:274-289. [PMID: 30196191 PMCID: PMC6129670 DOI: 10.1016/j.redox.2018.08.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide radical (NO) is a signaling molecule involved in several physiological and pathological processes and a new nitrate-nitrite-NO pathway has emerged as a physiological alternative to the "classic" pathway of NO formation from L-arginine. Since the late 1990s, it has become clear that nitrite can be reduced back to NO under hypoxic/anoxic conditions and exert a significant cytoprotective action in vivo under challenging conditions. To reduce nitrite to NO, mammalian cells can use different metalloproteins that are present in cells to perform other functions, including several heme proteins and molybdoenzymes, comprising what we denominated as the "non-dedicated nitrite reductases". Herein, we will review the current knowledge on two of those "non-dedicated nitrite reductases", the molybdoenzymes xanthine oxidoreductase and aldehyde oxidase, discussing the in vitro and in vivo studies to provide the current picture of the role of these enzymes on the NO metabolism in humans.
Collapse
Affiliation(s)
- Luisa B Maia
- LAQV, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal.
| | - José J G Moura
- LAQV, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
35
|
Kina-Tanada M, Sakanashi M, Arasaki A, Tsutsui M. [Long-term dietary nitrite and nitrate deficiency causes metabolic syndrome, endothelial dysfunction, and cardiovascular death in mice]. Nihon Yakurigaku Zasshi 2018; 151:148-154. [PMID: 29628462 DOI: 10.1254/fpj.151.148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Nitric oxide (NO) is synthesized not only from L-arginine by NO synthases (NOSs), but also from its inert metabolites, nitrite and nitrate. Green leafy vegetables are abundant in nitrate, however whether or not a deficiency in dietary nitrite/nitrate spontaneously causes disease remains to be clarified. In this study, we tested our hypothesis that long-term dietary nitrite/nitrate deficiency induces metabolic syndrome (MetS) in mice. To this end, we prepared a low nitrite/nitrate diet (LND) consisting of an amino acid-based low nitrite/nitrate chow in which the contents of L-arginine, fat, carbohydrates, protein, and energy were identical with a regular chow, and potable ultrapure water. Nitrite and nitrate were undetectable in both the chow and the water. Intriguingly, in comparison with a regular diet, 3 months of the LND significantly elicited visceral adiposity, dyslipidaemia, and glucose intolerance; 18 months of the LND significantly provoked increased body weight, hypertension, insulin resistance, and impaired endothelium-dependent relaxations to acetylcholine; and 22 months of the LND significantly led to death due to cardiovascular disease, including acute myocardial infarction. These abnormalities were reversed by simultaneous treatment with sodium nitrate, and were significantly associated with endothelial NOS down-regulation, adiponectin insufficiency, and gut microbiota dysbiosis. These results provide the first evidence that long-term dietary nitrite/nitrate deficiency gives rise to MetS, endothelial dysfunction, and cardiovascular death in mice, indicating a novel pathogenetic role of the exogenous NO production system in MetS and its vascular complications.
Collapse
Affiliation(s)
- Mika Kina-Tanada
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus.,Department of Oral and Maxillofacial Functional Rehabilitation, Graduate School of Medicine, University of the Ryukyus
| | - Mayuko Sakanashi
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus
| | - Akira Arasaki
- Department of Oral and Maxillofacial Functional Rehabilitation, Graduate School of Medicine, University of the Ryukyus
| | - Masato Tsutsui
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus
| |
Collapse
|
36
|
Waldron M, Waldron L, Lawlor C, Gray A, Highton J. Beetroot supplementation improves the physiological responses to incline walking. Eur J Appl Physiol 2018; 118:1131-1141. [PMID: 29546639 DOI: 10.1007/s00421-018-3843-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/08/2018] [Indexed: 12/23/2022]
Abstract
PURPOSE We investigated the effects of an acute 24-h nitrate-rich beetroot juice supplement (BR) on the energy cost, exercise efficiency and blood pressure responses to intermittent walking at different gradients. METHODS In a double-blind, cross-over design, eight participants were provided with a total of 350 ml of nitrate-rich (~ 20.5 mmol nitrate) BR or placebo (PLA) across 24 h before completing intermittent walking at 3 km/h on treadmill at gradients of 1, 5, 10, 15 and 20%. RESULTS Resting mean arterial pressure (MAP) was ~ 4.1% lower after BR (93 vs. 89 mmHg; P = 0.001), as well as during exercise (102 vs. 99 mmHg; P = 0.011) and recovery (97 vs. 94 mmHg; P = 0.001). Exercising (1227 vs. 1129 ml/min P < 0.001) and end-stage (1404 vs. 1249 ml/min; P = 0.002) oxygen uptake ([Formula: see text]O2) was lower in BR compared to PLA, which was accompanied by an average reduction in phase II [Formula: see text]O2 amplitude (1067 vs. 940 ml/min; P = 0.025). Similarly, recovery [Formula: see text]O2 (509 vs. 458 ml/min; P = 0.001) was lower in BR. Whole blood potassium concentration increased from pre-post exercise in PLA (4.1 ± 0.3 vs. 4.5 ± 0.3 mmol/L; P = 0.013) but not BR (4.1 ± 0.31 vs. 4.3 ± 0.2 mmol/L; P = 0.188). CONCLUSIONS Energy cost of exercise, recovery of [Formula: see text]O2, MAP and blood markers were ameliorated after BR. Previously-reported mechanisms explain these findings, which are more noticeable during less-efficient walking at steep gradients (15-20%). These findings have practical implications for hill-walkers.
Collapse
Affiliation(s)
- Mark Waldron
- School of Sport, Health and Applied Science, St Mary's University, Waldegrave Road, Twickenham, London, TW1 4SX, UK. .,School of Science and Technology, University of New England, Armidale, NSW, 2350, Australia.
| | - Luke Waldron
- Medical Education Centre, Royal Cornwall Hospitals NHS Trust, Truro, TR1 3LJ, UK
| | - Craig Lawlor
- School of Science and Technology, University of New England, Armidale, NSW, 2350, Australia
| | - Adrian Gray
- School of Science and Technology, University of New England, Armidale, NSW, 2350, Australia
| | - Jamie Highton
- Department of Sports and Exercise Sciences, University of Chester, Parkgate Road, Chester, CH14BJ, UK
| |
Collapse
|
37
|
McDonagh STJ, Wylie LJ, Thompson C, Vanhatalo A, Jones AM. Potential benefits of dietary nitrate ingestion in healthy and clinical populations: A brief review. Eur J Sport Sci 2018. [PMID: 29529987 DOI: 10.1080/17461391.2018.1445298] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This article provides an overview of the current literature relating to the efficacy of dietary nitrate (NO3-) ingestion in altering aspects of cardiovascular and metabolic health and exercise capacity in healthy and diseased individuals. The consumption of NO3--rich vegetables, such as spinach and beetroot, have been variously shown to promote nitric oxide bioavailability, reduce systemic blood pressure, enhance tissue blood flow, modulate muscle O2 utilisation and improve exercise tolerance both in normoxia and in hypoxia, as is commonly observed in a number of disease states. NO3- ingestion may, therefore, act as a natural means for augmenting performance and attenuating complications associated with limited O2 availability or transport, hypertension and the metabolic syndrome. Recent studies indicate that dietary NO3- might also augment intrinsic skeletal muscle contractility and improve the speed and power of muscle contraction. Moreover, several investigations suggest that NO3- supplementation may improve aspects of cognitive performance both at rest and during exercise. Collectively, these observations position NO3- as more than a putative ergogenic aid and suggest that increasing natural dietary NO3- intake may act as a prophylactic in countering the predations of senescence and certain cardiovascular-metabolic diseases.
Collapse
Affiliation(s)
- Sinead T J McDonagh
- a Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus , University of Exeter , Exeter , Devon , UK
| | - Lee J Wylie
- a Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus , University of Exeter , Exeter , Devon , UK
| | - Christopher Thompson
- a Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus , University of Exeter , Exeter , Devon , UK
| | - Anni Vanhatalo
- a Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus , University of Exeter , Exeter , Devon , UK
| | - Andrew M Jones
- a Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus , University of Exeter , Exeter , Devon , UK
| |
Collapse
|
38
|
Gilliard CN, Lam JK, Cassel KS, Park JW, Schechter AN, Piknova B. Effect of dietary nitrate levels on nitrate fluxes in rat skeletal muscle and liver. Nitric Oxide 2018; 75:1-7. [PMID: 29378248 DOI: 10.1016/j.niox.2018.01.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/17/2018] [Accepted: 01/23/2018] [Indexed: 01/28/2023]
Abstract
Rodent skeletal muscle has high levels of nitrate ions and this endogenous nitrate reservoir can supply nitrite/nitric oxide (NO) for functional hyperemia and/or for other physiological processes in muscle during exercise. Mice with a NOS1 knockout have markedly reduced muscle nitrate levels, suggesting NO production by NOS and its reaction with oxymyoglobin as a source of nitrate. However, oxygen levels are normally low in most internal organs, which raises the possibility that nitrate-derived NO pathway is physiologically important even at "normoxia", and muscle nitrate reservoir is the main endogenous NO backup when exogeneous (dietary) nitrate intake is low. Using dietary nitrate manipulations, we explore the importance of diet for maintaining and renewal of muscle nitrate reservoir and its levels in other tissues. We found that skeletal muscle nitrate is extensively used when nitrate in diet is low. One week of nitrate starvation leads to dramatic nitrate depletion in skeletal muscle and a substantial decrease in liver. Nitrate depleted from skeletal muscle during starvation is quickly recovered from new dietary sources, with an unexpected significant "overload" compared with animals not subjected to nitrate starvation. Our results suggest the importance of dietary nitrate for nitrate reserves in muscle and in other tissues, when compared with endogenous NOS-derived sources. This requires an active transport mechanism for sequestering nitrate into cells, stimulated by lack of dietary nitrate or other enzymatic changes. These results confirm the hypothesis that muscle is a major storage site for nitrate in mammals.
Collapse
Affiliation(s)
- Cameron N Gilliard
- Molecular Medicine Branch, NIDDK, NIH, Bethesda, MD, United States; Penn State Health Milton S. Hershey Medical Center, Department of Anesthesiology, Hershey, PA, United States
| | - Jeff K Lam
- Molecular Medicine Branch, NIDDK, NIH, Bethesda, MD, United States; Icahn School of Medicine, Mt. Sinai, New York, NY, United States
| | - Katelyn S Cassel
- Molecular Medicine Branch, NIDDK, NIH, Bethesda, MD, United States
| | - Ji Won Park
- Molecular Medicine Branch, NIDDK, NIH, Bethesda, MD, United States
| | - Alan N Schechter
- Molecular Medicine Branch, NIDDK, NIH, Bethesda, MD, United States
| | - Barbora Piknova
- Molecular Medicine Branch, NIDDK, NIH, Bethesda, MD, United States.
| |
Collapse
|
39
|
Abstract
Dietary triggers are commonly reported by patients with a variety of headaches, particularly those with migraines. The presence of any specific dietary trigger in migraine patients varies from 10 to 64 % depending on study population and methodology. Some foods trigger headache within an hour while others develop within 12 h post ingestion. Alcohol (especially red wine and beer), chocolate, caffeine, dairy products such as aged cheese, food preservatives with nitrates and nitrites, monosodium glutamate (MSG), and artificial sweeteners such as aspartame have all been studied as migraine triggers in the past. This review focuses the evidence linking these compounds to headache and examines the prevalence of these triggers from prior population-based studies. Recent literature surrounding headache related to fasting and weight loss as well as elimination diets based on serum food antibody testing will also be summarized to help physicians recommend low-risk, non-pharmacological adjunctive therapies for patients with debilitating headaches.
Collapse
Affiliation(s)
- Zoya Zaeem
- Division of Neurology, University of British Columbia, 8219-2775 Laurel Street, Vancouver, BC, V5Z 1M9, Canada
| | - Lily Zhou
- Division of Neurology, University of British Columbia, 8219-2775 Laurel Street, Vancouver, BC, V5Z 1M9, Canada
| | - Esma Dilli
- Division of Neurology, University of British Columbia, 8219-2775 Laurel Street, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
40
|
Almeida LEF, Kamimura S, Nettleton MY, de Souza Batista CM, Walek E, Khaibullina A, Wang L, Quezado ZMN. Blood collection vials and clinically used intravenous fluids contain significant amounts of nitrite. Free Radic Biol Med 2017; 108:533-541. [PMID: 28416347 DOI: 10.1016/j.freeradbiomed.2017.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 12/21/2022]
Abstract
The biology of the inorganic anion nitrite is linked to nitric oxide (NO) as nitrite can be reduced to NO and mediate its biological activities. Thus, studies of nitrite biology require sensitive and selective chemical assays. The acetic and ascorbic acids method is selective for nitrite and measures it in biological matrices. However, one of the pitfalls of nitrite measurements is its ubiquitous presence in sample collection tubes. Here, we showed high levels of nitrite in collection tubes containing EDTA, sodium citrate or sodium heparin and smaller amounts in tubes containing lithium heparin or serum clot activator. We also showed the presence of nitrite in colloid and crystalloid solutions frequently administered to patients and found variable levels of nitrite in 5% albumin, 0.9% sodium chloride, lactated ringer's, and dextrose-plus-sodium chloride solutions. These levels of nitrite varied across lots and manufacturers of the same type of fluid. Because these fluids are administered intravenously to patients (including those in shock), sometimes in large volumes (liters), it is possible that infusions of these nitrite-containing fluids may have clinical implications. A protocol for blood collection free of nitrite contamination was developed and used to examine nitrite levels in whole blood, red blood cells, plasma and urine from normal volunteers. Nitrite measurements were reproducible, had minimal variability, and did not indicate sex-differences. These findings validated a method and protocol for selective nitrite assay in biological fluids free of nitrite contamination which can be applied for study of diseases where dysfunctional NO signaling has been implicated.
Collapse
Affiliation(s)
- Luis E F Almeida
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, School of Medicine and Health Sciences, George Washington University, Washington, DC 20010, USA; Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sayuri Kamimura
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, School of Medicine and Health Sciences, George Washington University, Washington, DC 20010, USA; Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Margaret Y Nettleton
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, School of Medicine and Health Sciences, George Washington University, Washington, DC 20010, USA; Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Elizabeth Walek
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, School of Medicine and Health Sciences, George Washington University, Washington, DC 20010, USA
| | - Alfia Khaibullina
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, School of Medicine and Health Sciences, George Washington University, Washington, DC 20010, USA
| | - Li Wang
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, School of Medicine and Health Sciences, George Washington University, Washington, DC 20010, USA
| | - Zenaide M N Quezado
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, School of Medicine and Health Sciences, George Washington University, Washington, DC 20010, USA; Department of Perioperative Medicine, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
41
|
Kina-Tanada M, Sakanashi M, Tanimoto A, Kaname T, Matsuzaki T, Noguchi K, Uchida T, Nakasone J, Kozuka C, Ishida M, Kubota H, Taira Y, Totsuka Y, Kina SI, Sunakawa H, Omura J, Satoh K, Shimokawa H, Yanagihara N, Maeda S, Ohya Y, Matsushita M, Masuzaki H, Arasaki A, Tsutsui M. Long-term dietary nitrite and nitrate deficiency causes the metabolic syndrome, endothelial dysfunction and cardiovascular death in mice. Diabetologia 2017; 60:1138-1151. [PMID: 28352942 DOI: 10.1007/s00125-017-4259-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/27/2017] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Nitric oxide (NO) is synthesised not only from L-arginine by NO synthases (NOSs), but also from its inert metabolites, nitrite and nitrate. Green leafy vegetables are abundant in nitrate, but whether or not a deficiency in dietary nitrite/nitrate spontaneously causes disease remains to be clarified. In this study, we tested our hypothesis that long-term dietary nitrite/nitrate deficiency would induce the metabolic syndrome in mice. METHODS To this end, we prepared a low-nitrite/nitrate diet (LND) consisting of an amino acid-based low-nitrite/nitrate chow, in which the contents of L-arginine, fat, carbohydrates, protein and energy were identical with a regular chow, and potable ultrapure water. Nitrite and nitrate were undetectable in both the chow and the water. RESULTS Three months of the LND did not affect food or water intake in wild-type C57BL/6J mice compared with a regular diet (RD). However, in comparison with the RD, 3 months of the LND significantly elicited visceral adiposity, dyslipidaemia and glucose intolerance. Eighteen months of the LND significantly provoked increased body weight, hypertension, insulin resistance and impaired endothelium-dependent relaxations to acetylcholine, while 22 months of the LND significantly led to death mainly due to cardiovascular disease, including acute myocardial infarction. These abnormalities were reversed by simultaneous treatment with sodium nitrate, and were significantly associated with endothelial NOS downregulation, adiponectin insufficiency and dysbiosis of the gut microbiota. CONCLUSIONS/INTERPRETATION These results provide the first evidence that long-term dietary nitrite/nitrate deficiency gives rise to the metabolic syndrome, endothelial dysfunction and cardiovascular death in mice, indicating a novel pathogenetic role of the exogenous NO production system in the metabolic syndrome and its vascular complications.
Collapse
Affiliation(s)
- Mika Kina-Tanada
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
- Department of Oral and Maxillofacial Functional Rehabilitation, University of the Ryukyus, Okinawa, Japan
| | - Mayuko Sakanashi
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Akihide Tanimoto
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tadashi Kaname
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Toshihiro Matsuzaki
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Katsuhiko Noguchi
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Taro Uchida
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Junko Nakasone
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Chisayo Kozuka
- Second Department of Internal Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masayoshi Ishida
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
- Regenerative Medicine Research Center, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Haruaki Kubota
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Yuji Taira
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Yuichi Totsuka
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| | - Shin-Ichiro Kina
- Department of Oral and Maxillofacial Functional Rehabilitation, University of the Ryukyus, Okinawa, Japan
| | - Hajime Sunakawa
- Department of Oral and Maxillofacial Functional Rehabilitation, University of the Ryukyus, Okinawa, Japan
| | - Junichi Omura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Yanagihara
- Department of Pharmacology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yusuke Ohya
- Third Department of Internal Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masayuki Matsushita
- Department of Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Hiroaki Masuzaki
- Second Department of Internal Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Akira Arasaki
- Department of Oral and Maxillofacial Functional Rehabilitation, University of the Ryukyus, Okinawa, Japan
| | - Masato Tsutsui
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan.
| |
Collapse
|
42
|
Cirino G, Vellecco V, Bucci M. Nitric oxide and hydrogen sulfide: the gasotransmitter paradigm of the vascular system. Br J Pharmacol 2017; 174:4021-4031. [PMID: 28407204 DOI: 10.1111/bph.13815] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/06/2017] [Accepted: 03/19/2017] [Indexed: 01/20/2023] Open
Abstract
There are several reviews on NO and hydrogen sulfide (H2 S) and their role in vascular diseases in the current relevant literature. The aim of this review is to discuss, within the limits of present knowledge, the interconnection between these two gasotransmitters in vascular function. In particular, the review focuses on the role played by the balance between the NO and H2 S pathways in either physiological or pathological conditions. The distinction between physiology and pathology has been made in order to dissect the molecular basis of this crosstalk, highlighting how and if this balance varies, depending upon the vascular status. Perspectives and possible novel therapeutic approaches are also discussed. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
43
|
Bryan NS, Tribble G, Angelov N. Oral Microbiome and Nitric Oxide: the Missing Link in the Management of Blood Pressure. Curr Hypertens Rep 2017; 19:33. [DOI: 10.1007/s11906-017-0725-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
44
|
Van Hecke T, Van Camp J, De Smet S. Oxidation During Digestion of Meat: Interactions with the Diet andHelicobacter pyloriGastritis, and Implications on Human Health. Compr Rev Food Sci Food Saf 2017; 16:214-233. [DOI: 10.1111/1541-4337.12248] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Thomas Van Hecke
- the Laboratory for Animal Nutrition and Animal Product Quality; Ghent Univ.; Ghent Belgium
| | - John Van Camp
- the Unit of Food Chemistry and Human Nutrition; Ghent Univ.; Ghent Belgium
| | - Stefaan De Smet
- the Laboratory for Animal Nutrition and Animal Product Quality; Ghent Univ.; Ghent Belgium
| |
Collapse
|
45
|
Bailey SJ, Blackwell JR, Wylie LJ, Emery A, Taylor E, Winyard PG, Jones AM. Influence of iodide ingestion on nitrate metabolism and blood pressure following short-term dietary nitrate supplementation in healthy normotensive adults. Nitric Oxide 2016; 63:13-20. [PMID: 28024935 DOI: 10.1016/j.niox.2016.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/08/2016] [Accepted: 12/14/2016] [Indexed: 02/07/2023]
Abstract
Uptake of inorganic nitrate (NO3-) into the salivary circulation is a rate-limiting step for dietary NO3- metabolism in mammals. It has been suggested that salivary NO3- uptake occurs in competition with inorganic iodide (I-). Therefore, this study tested the hypothesis that I- supplementation would interfere with NO3- metabolism and blunt blood pressure reductions after dietary NO3- supplementation. Nine healthy adults (4 male, mean ± SD, age 20 ± 1 yr) reported to the laboratory for initial baseline assessment (control) and following six day supplementation periods with 140 mL·day-1 NO3--rich beetroot juice (8.4 mmol NO3-·day-1) and 198 mg potassium gluconate·day-1 (nitrate), and 140 mL·day-1 NO3--rich beetroot juice and 450 μg potassium iodide·day-1 (nitrate + iodide) in a randomized, cross-over experiment. Salivary [I-] was higher in the nitrate + iodide compared to the control and NIT trials (P < 0.05). Salivary and plasma [NO3-] and [NO2-] were higher in the nitrate and nitrate + iodide trials compared to the control trial (P < 0.05). Plasma [NO3-] was higher (474 ± 127 vs. 438 ± 117 μM) and the salivary-plasma [NO3-] ratio was lower (14 ± 6 vs. 20 ± 6 μM), indicative of a lower salivary NO3- uptake, in the nitrate + iodide trial compared to the nitrate trial (P < 0.05). Plasma and salivary [NO2-] were not different between the nitrate and nitrate + iodide trials (P > 0.05). Systolic blood pressure was lower than control (112 ± 13 mmHg) in the nitrate (106 ± 13 mmHg) and nitrate + iodide (106 ± 11 mmHg) trials (P < 0.05), with no differences between the nitrate and nitrate + iodide trials (P > 0.05). In conclusion, co-ingesting NO3- and I- perturbed salivary NO3- uptake, but the increase in salivary and plasma [NO2-] and the lowering of blood pressure were similar compared to NO3- ingestion alone. Therefore, increased dietary I- intake, which is recommended in several countries worldwide as an initiative to offset hypothyroidism, does not appear to compromise the blood pressure reduction afforded by increased dietary NO3- intake.
Collapse
Affiliation(s)
- Stephen J Bailey
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK.
| | - Jamie R Blackwell
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Lee J Wylie
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Annabelle Emery
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Ellie Taylor
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Paul G Winyard
- University of Exeter Medical School, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Andrew M Jones
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| |
Collapse
|
46
|
Bailey SJ, Blackwell JR, Wylie LJ, Holland T, Winyard PG, Jones AM. Improvement in blood pressure after short-term inorganic nitrate supplementation is attenuated in cigarette smokers compared to non-smoking controls. Nitric Oxide 2016; 61:29-37. [PMID: 27744007 DOI: 10.1016/j.niox.2016.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 01/22/2023]
Abstract
Dietary supplementation with inorganic nitrate (NO3-) has been reported to improve cardiovascular health indices in healthy adults. Cigarette smoking increases circulating thiocyanate (SCN-), which has been suggested to competitively inhibit salivary nitrate (NO3-) uptake, a rate-limiting step in dietary NO3- metabolism. Therefore, this study tested the hypothesis that dietary NO3- supplementation would be less effective at increasing the circulating plasma nitrite concentration ([NO2-]) and lowering blood pressure in smokers (S) compared to non-smokers (NS). Nine healthy smokers and eight healthy non-smoking controls reported to the laboratory at baseline (CON) and following six day supplementation periods with 140 mL day-1 NO3--rich (8.4 mmol NO3- day-1; NIT) and NO3--depleted (0.08 mmol NO3- day-1; PLA) beetroot juice in a cross-over experiment. Plasma and salivary [SCN-] were elevated in smokers compared to non-smokers in all experimental conditions (P < 0.05). Plasma and salivary [NO3-] and [NO2-] were elevated in the NIT condition compared to CON and PLA conditions in smokers and non-smokers (P < 0.05). However, the change in salivary [NO3-] (S: 3.5 ± 2.1 vs. NS: 7.5 ± 4.4 mM), plasma [NO3-] (S: 484 ± 198 vs. NS: 802 ± 199 μM) and plasma [NO2-] (S: 218 ± 128 vs. NS: 559 ± 419 nM) between the CON and NIT conditions was lower in the smokers compared to the non-smokers (P < 0.05). Salivary [NO2-] increased above CON to a similar extent with NIT in smokers and non-smokers (P > 0.05). Systolic blood pressure was lowered compared to PLA with NIT in non-smokers (P < 0.05), but not smokers (P > 0.05). These findings suggest that dietary NO3- metabolism is compromised in smokers leading to an attenuated blood pressure reduction compared to non-smokers after NO3- supplementation. These observations may provide novel insights into the cardiovascular risks associated with cigarette smoking and suggest that this population may be less likely to benefit from improved cardiovascular health if they increase dietary NO3- intake.
Collapse
Affiliation(s)
- Stephen J Bailey
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK.
| | - Jamie R Blackwell
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Lee J Wylie
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Terezia Holland
- University of Exeter Medical School, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Paul G Winyard
- University of Exeter Medical School, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| | - Andrew M Jones
- Sport and Health Sciences, College of Life and Environmental Sciences, St. Luke's Campus, University of Exeter, Heavitree Road, Exeter, UK
| |
Collapse
|
47
|
Peleli M, Zollbrecht C, Montenegro MF, Hezel M, Zhong J, Persson EG, Holmdahl R, Weitzberg E, Lundberg JO, Carlström M. Enhanced XOR activity in eNOS-deficient mice: Effects on the nitrate-nitrite-NO pathway and ROS homeostasis. Free Radic Biol Med 2016; 99:472-484. [PMID: 27609225 DOI: 10.1016/j.freeradbiomed.2016.09.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/16/2016] [Accepted: 09/02/2016] [Indexed: 01/19/2023]
Abstract
Xanthine oxidoreductase (XOR) is generally known as the final enzyme in purine metabolism and as a source of reactive oxygen species (ROS). In addition, this enzyme has been suggested to mediate nitric oxide (NO) formation via reduction of inorganic nitrate and nitrite. This NO synthase (NOS)-independent pathway for NO generation is of particular importance during certain conditions when NO bioavailability is diminished due to reduced activity of endothelial NOS (eNOS) or increased oxidative stress, including aging and cardiovascular disease. The exact interplay between NOS- and XOR-derived NO generation is not fully elucidated yet. The aim of the present study was to investigate if eNOS deficiency is associated with changes in XOR expression and activity and the possible impact on nitrite, NO and ROS homeostasis. Plasma levels of nitrate and nitrite were similar between eNOS deficient (eNOS-/-) and wildtype (wt) mice. XOR activity was upregulated in eNOS-/- compared with wt, but not in nNOS-/-, iNOS-/- or wt mice treated with the non-selective NOS inhibitor L-NAME. Following an acute dose of nitrate, plasma nitrite increased more in eNOS-/- compared with wt, and this augmented response was abolished by the selective XOR inhibitor febuxostat. Livers from eNOS-/- displayed higher nitrite reducing capacity compared with wt, and this effect was attenuated by febuxostat. Dietary supplementation with nitrate increased XOR expression and activity, but concomitantly reduced superoxide generation. The latter effect was also seen in vitro after nitrite administration. Treatment with febuxostat elevated blood pressure in eNOS-/-, but not in wt mice. A high dose of dietary nitrate reduced blood pressure in naïve eNOS-/- mice, and again this effect was abolished by febuxostat. In conclusion, eNOS deficiency is associated with an upregulation of XOR facilitating the nitrate-nitrite-NO pathway and decreasing the generation of ROS. This interplay between XOR and eNOS is proposed to play a significant role in NO homeostasis and blood pressure regulation.
Collapse
Affiliation(s)
- Maria Peleli
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Christa Zollbrecht
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marcelo F Montenegro
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Hezel
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jianghong Zhong
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Erik G Persson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Rikard Holmdahl
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
48
|
Globin X is a six-coordinate globin that reduces nitrite to nitric oxide in fish red blood cells. Proc Natl Acad Sci U S A 2016; 113:8538-43. [PMID: 27407144 DOI: 10.1073/pnas.1522670113] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The discovery of novel globins in diverse organisms has stimulated intense interest in their evolved function, beyond oxygen binding. Globin X (GbX) is a protein found in fish, amphibians, and reptiles that diverged from a common ancestor of mammalian hemoglobins and myoglobins. Like mammalian neuroglobin, GbX was first designated as a neuronal globin in fish and exhibits six-coordinate heme geometry, suggesting a role in intracellular electron transfer reactions rather than oxygen binding. Here, we report that GbX to our knowledge is the first six-coordinate globin and the first globin protein apart from hemoglobin, found in vertebrate RBCs. GbX is present in fish erythrocytes and exhibits a nitrite reduction rate up to 200-fold faster than human hemoglobin and up to 50-fold higher than neuroglobin or cytoglobin. Deoxygenated GbX reduces nitrite to form nitric oxide (NO) and potently inhibits platelet activation in vitro, to a greater extent than hemoglobin. Fish RBCs also reduce nitrite to NO and inhibit platelet activation to a greater extent than human RBCs, whereas GbX knockdown inhibits this nitrite-dependent NO signaling. The description of a novel, six-coordinate globin in RBCs with dominant electron transfer and nitrite reduction functionality provides new insights into the evolved signaling properties of ancestral heme-globins.
Collapse
|
49
|
Dal S, Sigrist S. The Protective Effect of Antioxidants Consumption on Diabetes and Vascular Complications. Diseases 2016; 4:E24. [PMID: 28933404 PMCID: PMC5456287 DOI: 10.3390/diseases4030024] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/20/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022] Open
Abstract
Obesity and diabetes is generally accompanied by a chronic state of oxidative stress, disequilibrium in the redox balance, implicated in the development and progression of complications such as micro- and macro-angiopathies. Disorders in the inner layer of blood vessels, the endothelium, play an early and critical role in the development of these complications. Blunted endothelium-dependent relaxation and/or contractions are quietly associated to oxidative stress. Thus, preserving endothelial function and oxidative stress seems to be an optimization strategy in the prevention of vascular complications associated with diabetes. Diet is a major lifestyle factor that can greatly influence the incidence and the progression of type 2 diabetes and cardiovascular complications. The notion that foods not only provide basic nutrition but can also prevent diseases and ensure good health and longevity is now attained greater prominence. Some dietary and lifestyle modifications associated to antioxidative supply could be an effective prophylactic means to fight against oxidative stress in diabesity and complications. A significant benefit of phytochemicals (polyphenols in wine, grape, teas), vitamins (ascorbate, tocopherol), minerals (selenium, magnesium), and fruits and vegetables in foods is thought to be capable of scavenging free radicals, lowering the incidence of chronic diseases. In this review, we discuss the role of oxidative stress in diabetes and complications, highlight the endothelial dysfunction, and examine the impact of antioxidant foods, plants, fruits, and vegetables, currently used medication with antioxidant properties, in relation to the development and progression of diabetes and cardiovascular complications.
Collapse
Affiliation(s)
- Stéphanie Dal
- DIATHEC EA 7294 UMR Centre Européen d'Etude du Diabète (CeeD), Université de Strasbourg (UdS), boulevard René Leriche, Strasbourg 67200, France.
| | - Séverine Sigrist
- DIATHEC EA 7294 UMR Centre Européen d'Etude du Diabète (CeeD), Université de Strasbourg (UdS), boulevard René Leriche, Strasbourg 67200, France.
| |
Collapse
|
50
|
Jiang H, Polhemus DJ, Islam KN, Torregrossa AC, Li Z, Potts A, Lefer DJ, Bryan NS. Nebivolol Acts as a S-Nitrosoglutathione Reductase Inhibitor. J Cardiovasc Pharmacol Ther 2016; 21:478-85. [DOI: 10.1177/1074248415626300] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/12/2015] [Indexed: 11/15/2022]
Abstract
Background and Purpose: Published data on nebivolol reveal selective β1 adrenergic selectively along with novel nitric oxide (NO)-dependent vasodilatory properties. However, the exact molecular mechanism is unknown. Protein S-nitrosylation constitutes a large part of the ubiquitous influence of NO on cellular signal transduction and is involved in a number of human diseases. More recently, protein denitrosylation has been shown to play a major role in controlling cellular S-nitrosylation (SNO). Several enzymes have been reported to catalyze the reduction of SNOs and are viewed as candidate denitrosylases. One of the first described is known as S-nitrosoglutathione reductase (GSNOR). Importantly, GSNOR has been shown to play a role in regulating SNO signaling downstream of the β-adrenergic receptor and is therefore operative in cellular signal transduction. Pharmacological inhibition or genetic deletion of GSNOR leads to enhanced vasodilation and characteristic of known effects of nebivolol. Structurally, nebivolol is similar to known inhibitors of GSNOR. Therefore, we hypothesize that some of the known effects of nebivolol may occur through this mechanism. Experimental Approach: Using cell culture systems, tissue organ bath, and intact animal models, we report that nebivolol treatment leads to a dose-dependent accumulation of nitrosothiols in cells, and this is associated with an enhanced vasodilation by S-nitrosoglutathione. Key Results: These data suggest a new mechanism of action of nebivolol that may explain in part the reported NO activity. Conclusions and Implications: Because exogenous mediators of protein SNO or denitrosylation can substantially affect the development or progression of disease, this may call for new utility of nebivolol.
Collapse
Affiliation(s)
- Hong Jiang
- Texas Therapeutics Institute at Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - David J. Polhemus
- LSU Health Science Center, Cardiovascular Center of Excellence, New Orleans, LA, USA
| | - Kazi N. Islam
- LSU Health Science Center, Cardiovascular Center of Excellence, New Orleans, LA, USA
| | - Ashley C. Torregrossa
- Texas Therapeutics Institute at Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Zhen Li
- LSU Health Science Center, Cardiovascular Center of Excellence, New Orleans, LA, USA
| | - Amy Potts
- Texas Therapeutics Institute at Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - David J. Lefer
- LSU Health Science Center, Cardiovascular Center of Excellence, New Orleans, LA, USA
| | - Nathan S. Bryan
- Texas Therapeutics Institute at Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|