1
|
Abolfazli S, Karav S, Johnston TP, Sahebkar A. Regulatory effects of resveratrol on nitric oxide signaling in cardiovascular diseases. Pharmacol Rep 2025; 77:355-374. [PMID: 39832074 DOI: 10.1007/s43440-025-00694-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular illnesses are multifactorial disorders and represent the primary reasons for death worldwide, according to the World Health Organization. As a signaling molecule, nitric oxide (NO) is extremely permeable across cellular membranes owing to its unique molecular features, like its small molecular size, lipophilicity, and free radical properties. Some of the biological effects of NO are vasodilation, inhibition in the growth of vascular smooth muscle cells, and functional regulation of cardiac cells. Several therapeutic approaches have been tested to increase the production of NO or some downstream NO signaling pathways. The health benefits of red wine are typically attributed to the polyphenolic phytoalexin, resveratrol (3,5,4'-trihydroxy-trans-stilbene), which is found in several plant species. Resveratrol has beneficial cardiovascular properties, some of which are mediated through endothelial nitric oxide synthase production (eNOS). Resveratrol promotes NO generation from eNOS through various methods, including upregulation of eNOS expression, activation in the enzymatic activity of eNOS, and reversal of eNOS uncoupling. Additionally, by reducing of oxidative stress, resveratrol inhibits the formation of superoxide and inactivation NO, increasing NO bioavailability. This review discusses the scientific literature on resveratrol's beneficial impact on NO signaling and how this effect improves the function of vascular endothelium.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Sanches-Lopes JM, Cássia-Barros A, Conde-Tella SO, Coelho EB, Kemp R, Lacchini R, Feelisch M, Salgado Júnior W, Tanus-Santos JE. Bariatric surgery blunts nitrate-mediated improvements in cardiovascular function of overweight women by interfering with gastric S-nitrosothiol formation. Redox Biol 2024; 78:103440. [PMID: 39580965 PMCID: PMC11625360 DOI: 10.1016/j.redox.2024.103440] [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: 11/07/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Inorganic nitrate (NO3-) and nitrate-rich foods have been shown to exert antioxidative effects and lower blood pressure in experimental animal models and human clinical studies. The specific handling of nitrate, including its enterosalivary recirculation, secretion into saliva, oral microbial reduction to nitrite (NO2-), and the pH-dependent nitrosative capacity in the stomach have all been recognized as being important for nitrate's beneficial effects. Obesity is of major health concern worldwide and associated with increased cardiovascular risk; whether nitrate lowers blood pressure and improves endothelial function in this setting has not been investigated. We here tested the hypotheses that i) nitrate elicits cardiovascular benefits in overweight women; and ii) these beneficial effects would be diminished in women who underwent bariatric Roux-en-Y gastric bypass (RYGB) surgery. Our controlled clinical trial included 15 women with prior RYGB surgery and 15 overweight female controls. All participants received a single dose of 0.1 mmol/kg/day nitrate in the form of a beetroot extract for 14 days. Blood collection, 24-h ambulatory blood pressure measurements and endothelial function tests were performed before and after nitrate treatment. Plasma nitrite, nitrate, and S-nitrosothiol (RSNO) concentrations were determined by ozone-based reductive chemiluminescence while thiobarbituric acid reactive substances (TBARS) and total antioxidant capacity (TAC) were measured using plate-reader based assays. Nitrate reduced blood pressure and improved endothelial function in controls, but not in women with prior bariatric surgery. Nitrate also increased circulating nitrate/nitrite and RSNO levels in controls, but the latter was blunted following RYGB surgery despite even larger increases in nitrite concentrations. Similarly, nitrate increased antioxidant responses in controls but not in women with prior bariatric surgery. This is the first study to show that nitrate exerts beneficial cardiovascular effects in obesity and that the morphological/functional modifications elicited by RYGB surgery abrogates nitrate's effectiveness by preventing gastric RSNO formation.
Collapse
Affiliation(s)
- Jéssica Maria Sanches-Lopes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | - Sandra Oliveira Conde-Tella
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Translational Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Eduardo Barbosa Coelho
- Department of Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rafael Kemp
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Brazil
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, UK; Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, UK
| | - Wilson Salgado Júnior
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jose Eduardo Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
3
|
Oliveira-Paula GH, I M Batista R, Stransky S, Tella SC, Ferreira GC, Portella RL, Pinheiro LC, Damacena-Angelis C, Riascos-Bernal DF, Sidoli S, Sibinga N, Tanus-Santos JE. Orally administered sodium nitrite prevents the increased α-1 adrenergic vasoconstriction induced by hypertension and promotes the S-nitrosylation of calcium/calmodulin-dependent protein kinase II. Biochem Pharmacol 2023; 212:115571. [PMID: 37127250 PMCID: PMC10198929 DOI: 10.1016/j.bcp.2023.115571] [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: 02/13/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
The unsatisfactory rates of adequate blood pressure control among patients receiving antihypertensive treatment calls for new therapeutic strategies to treat hypertension. Several studies have shown that oral sodium nitrite exerts significant antihypertensive effects, but the mechanisms underlying these effects remain unclear. While these mechanisms may involve nitrite-derived S-nitrosothiols, their implication in important alterations associated with hypertension, such as aberrant α1-adrenergic vasoconstriction, has not yet been investigated. Here, we examined the effects of oral nitrite treatment on vascular responses to the α1-adrenergic agonist phenylephrine in two-kidney, one clip (2K1C) hypertensive rats and investigated the potential underlying mechanisms. Our results show that treatment with oral sodium nitrite decreases blood pressure and prevents the increased α1-adrenergic vasoconstriction in 2K1C hypertensive rats. Interestingly, we found that these effects require vascular protein S-nitrosylation, and to investigate the specific S-nitrosylated proteins we performed an unbiased nitrosoproteomic analysis of vascular smooth muscle cells (VSMCs) treated with the nitrosylating compound S-nitrosoglutathione (GSNO). This analysis revealed that GSNO markedly increases the nitrosylation of calcium/calmodulin-dependent protein kinase II γ (CaMKIIγ), a multifunctional protein that mediates the α1-adrenergic receptor signaling. This result was associated with reduced α1-adrenergic receptor-mediated CaMKIIγ activity in VSMCs. We further tested the relevance of these findings in vivo and found that treatment with oral nitrite increases CaMKIIγ S-nitrosylation and blunts the increased CaMKIIγ activity induced by phenylephrine in rat aortas. Collectively, these results are consistent with the idea that oral sodium nitrite treatment increases vascular protein S-nitrosylation, including CaMKIIγ as a target, which may ultimately prevent the increased α1-adrenergic vasoconstriction induced by hypertension. These mechanisms may help to explain the antihypertensive effects of oral nitrite and hold potential implications in the therapy of hypertension and other cardiovascular diseases associated with abnormal α1-adrenergic vasoconstriction.
Collapse
Affiliation(s)
- Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Rose I M Batista
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Stephanie Stransky
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Sandra C Tella
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Graziele C Ferreira
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rafael L Portella
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Celio Damacena-Angelis
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil
| | - Dario F Riascos-Bernal
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Nicholas Sibinga
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
4
|
Tange Y, Sunakawa R, Yoshitake S. Renal replacement therapy removes a large number of nitric oxide donors responsible for the nitrate-nitrite-nitric oxide pathway. Int J Artif Organs 2023; 46:129-134. [PMID: 36825795 DOI: 10.1177/03913988231157427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
BACKGROUNDS Nitric oxide has a broad-spectrum antibacterial property promising as a new therapeutic agent for severe acute respiratory syndrome coronavirus-2 because nitric oxide donor (such as S-nitroso-N-acetylpenicillamine) reduces the replication of coronavirus-2. Patients with coronavirus disease 2019 undergoing dialysis generally have a higher mortality rate than the general population. Although the higher mortality rate in these patients may be related to their advanced age, it has been suggested that plasma nitrite and nitrate levels (products of nitric oxide metabolism) are significantly decreased after hemodialysis which may compromise the nitrate-nitrite-nitric oxide pathway and impair nitric oxide homeostasis. It results in increased cardiovascular mortality in patients undergoing dialysis. However, the profile of nitric oxide-producing substances is poorly understood during renal replacement therapy. METHODS We simulated continuous hemodialysis and hemodiafiltration to measure the amount of nitric oxide (nitric oxide-producing substance) clearance in vitro. RESULTS The results demonstrated increased nitric oxide clearance and higher clearance than creatinine (molecular weight: 113) and vitamin B12 (molecular weight: 1355) using highly efficient renal replacement therapy modes. CONCLUSION The high nitric oxide clearance may have partly contributed to the high cardiovascular and coronavirus-2 mortality risk in patients on dialysis.
Collapse
Affiliation(s)
- Yoshihiro Tange
- Department of Advanced Medical Sciences, Faculty of Medicine, Oita University, Oita, Japan
| | - Reo Sunakawa
- Department of Medical Engineering, Kyushu University of Health and Welfare, Miyazaki, Japan
| | - Shigenori Yoshitake
- Department of Clinical Psychology, Kyushu University of Health and Welfare, Miyazaki, Japan
| |
Collapse
|
5
|
LaPenna KB, Li Z, Doiron JE, Sharp TE, Xia H, Moles K, Koul K, Wang JS, Polhemus DJ, Goodchild TT, Patel RB, Shah SJ, Lefer DJ. Combination Sodium Nitrite and Hydralazine Therapy Attenuates Heart Failure With Preserved Ejection Fraction Severity in a "2-Hit" Murine Model. J Am Heart Assoc 2023; 12:e028480. [PMID: 36752224 PMCID: PMC10111505 DOI: 10.1161/jaha.122.028480] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2023]
Abstract
Background Recent studies have suggested that cardiac nitrosative stress mediated by pathological overproduction of nitric oxide (NO) via inducible NO synthase (iNOS) contributes to the pathogenesis of heart failure with preserved ejection fraction (HFpEF). Other studies have suggested that endothelial NO synthase (eNOS) dysfunction and attenuated NO bioavailability contribute to HFpEF morbidity and mortality. We sought to further investigate dysregulated NO signaling and to examine the effects of a NO-based dual therapy (sodium nitrite+hydralazine) following the onset of HFpEF using a "2-hit" murine model. Methods and Results Nine-week-old male C57BL/6 N mice (n=15 per group) were treated concurrently with high-fat diet and N(ω)-nitro-L-arginine methyl ester (L-NAME) (0.5 g/L per day) via drinking water for 10 weeks. At week 5, mice were randomized into either vehicle (normal saline) or combination treatment with sodium nitrite (75 mg/L in the drinking water) and hydralazine (2.0 mg/kg IP, BID). Cardiac structure and function were monitored with echocardiography and invasive hemodynamic measurements. Cardiac mitochondrial respiration, aortic vascular function, and exercise performance were also evaluated. Circulating and myocardial nitrite were measured to determine the bioavailability of NO. Circulating markers of oxidative or nitrosative stress as well as systemic inflammation were also determined. Severe HFpEF was evident by significantly elevated E/E', LVEDP, and Tau in mice treated with L-NAME and HFD, which was associated with impaired NO bioavailability, mitochondrial respiration, aortic vascular function, and exercise capacity. Treatment with sodium nitrite and hydralazine restored NO bioavailability, reduced oxidative and nitrosative stress, preserved endothelial function and mitochondrial respiration, limited the fibrotic response, and improved exercise capacity, ultimately attenuating the severity of "two-hit" HFpEF. Conclusions Our data demonstrate that nitrite, a well-established biomarker of NO bioavailability and a physiological source of NO, is significantly reduced in the heart and circulation in the "2-hit" mouse HFpEF model. Furthermore, sodium nitrite+hydralazine combined therapy significantly attenuated the severity of HFpEF in the "2-hit" cardiometabolic HFpEF. These data suggest that supplementing NO-based therapeutics with a potent antioxidant and vasodilator agent may result in synergistic benefits for the treatment of HFpEF.
Collapse
Affiliation(s)
- Kyle B. LaPenna
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA
| | - Zhen Li
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| | - Jake E. Doiron
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLA
| | - Thomas E. Sharp
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
- Department of Medicine, Section of CardiologyLouisiana State University Health Sciences CenterNew OrleansLA
| | - Huijing Xia
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - Karl Moles
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - Kashyap Koul
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | - John S. Wang
- Cardiovascular Center of ExcellenceLouisiana State University Health Sciences CenterNew OrleansLA
| | | | - Traci T. Goodchild
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| | - Ravi B. Patel
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoIL
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoIL
| | - David J. Lefer
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCA
| |
Collapse
|
6
|
Williams XM, Bossert AT, Devalance E, Lewis SE, Gunther MR, Kelley EE. Indirect Antioxidant Effects of the Nitrite Anion: Focus on Xanthine Oxidase. ADVANCES IN REDOX RESEARCH 2022; 7. [PMID: 37063462 PMCID: PMC10100591 DOI: 10.1016/j.arres.2022.100058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
One electron reduction of nitrite (NO2 -) has been determined to be a significant, noncanonical source of nitric oxide (NO) with molybdopterin enzymes being identified as critical to this process. Of the molybdopterin enzymes identified as NO2 - reductases, xanthine oxidoreductase (XOR) is the most extensively studied. Paradoxically, XOR generates oxidants and thus can contribute to oxidative stress under inflammatory conditions when the oxidase form (XO) of XOR is abundant. However, under similar inflammatory conditions XO has been associated with NO generation, especially when NO2 - levels are elevated which begs the question: if reaction of nitrite with XO consumes electrons, then does it subsequently reduce oxidant generation? To address this question, electron paramagnetic resonance (EPR) was used, under controlled O2 tensions, to assess superoxide (O2 •-) generation by endothelial-bound XO plus xanthine and the resultant impact of introducing NO2 -. Nitrite diminished XO-derived O2 •- under hypoxia (1% O2) whereas at 21% O2, it had no impact. To confirm these results and discount contributions from the reaction of NO with O2 •-, molecular O2 consumption was assessed. The presence of NO2 - decreased the rate of XO/xanthine-dependent O2 consumption in a concentration-dependent manner with greater impact under hypoxic conditions (1% O2) compared to 21% O2. In a more biologic setting, NO2 - also diminished XO-dependent H2O2 formation in murine liver homogenates supplemented with xanthine. Interestingly, nitrate (NO3 -) did not alter XO-dependent O2 consumption at either 21% or 1% O2; yet it did slightly impact nitrite-mediated effects when present at 2:1 ratio vs. NO2 -. When combined, these data: 1) show a significant indirect antioxidant function for NO2 - by decreasing oxidant generation from XO, 2) demonstrate that both XO-derived H2O2 and O2 •- production are diminished by the presence of NO2 - and 3) incentivize further exploration of the difference between XO reaction with NO2 - vs. NO3 -.
Collapse
|
7
|
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]
|
8
|
The C2H2 Zinc Finger Protein MaNCP1 Contributes to Conidiation through Governing the Nitrate Assimilation Pathway in the Entomopathogenic Fungus Metarhizium acridum. J Fungi (Basel) 2022; 8:jof8090942. [PMID: 36135667 PMCID: PMC9505000 DOI: 10.3390/jof8090942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/22/2022] Open
Abstract
Zinc finger proteins are an important class of multifunctional regulators. Here, the roles of a C2H2 zinc finger protein MaNCP1 (Metarhizium acridum nitrate-related conidiation pattern shift regulatory factor 1) in nitrogen utilization and conidiation were explored in the entomopathogenic fungus M. acridum. The results showed that MaNCP1-disruption mutant (ΔMaNCP1) impaired the ability to utilize nitrate, ammonium and glutamine and reduced the expression of nitrate assimilation-related genes, suggesting that MaNCP1 was involved in governing nitrogen utilization. In addition, the conidial yield of the ΔMaNCP1 strain, cultured on the microcycle conidiation medium (SYA), was significantly decreased, which could be restored or even enhanced than that of the WT strain through increasing the nitrate content in SYA medium. Further study showed that MaAreA, a core regulator in the nitrogen catabolism repression (NCR) pathway, was a downstream target gene of MaNCP1. Screening the differential expression genes between WT and ΔMaNCP1 strains revealed that the conidial yield of M. acridum regulated by nitrate might be related to NCR pathway on SYA medium. It could be concluded that MaNCP1 contributes to the nitrate assimilation and conidiation, which will provide further insights into the relationship between the nitrogen utilization and conidiation in fungi.
Collapse
|
9
|
Naskar T, Jana M, Majumdar A. Binuclear manganese(II)-thiolate complexes: Synthesis, characterization and nitrite induced structural changes. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
10
|
Solanki K, Rajpoot S, Bezsonov EE, Orekhov AN, Saluja R, Wary A, Axen C, Wary K, Baig MS. The expanding roles of neuronal nitric oxide synthase (NOS1). PeerJ 2022; 10:e13651. [PMID: 35821897 PMCID: PMC9271274 DOI: 10.7717/peerj.13651] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/07/2022] [Indexed: 01/17/2023] Open
Abstract
The nitric oxide synthases (NOS; EC 1.14.13.39) use L-arginine as a substrate to produce nitric oxide (NO) as a by-product in the tissue microenvironment. NOS1 represents the predominant NO-producing enzyme highly enriched in the brain and known to mediate multiple functions, ranging from learning and memory development to maintaining synaptic plasticity and neuronal development, Alzheimer's disease (AD), psychiatric disorders and behavioral deficits. However, accumulating evidence indicate both canonical and non-canonical roles of NOS1-derived NO in several other tissues and chronic diseases. A better understanding of NOS1-derived NO signaling, and identification and characterization of NO-metabolites in non-neuronal tissues could become useful in diagnosis and prognosis of diseases associated with NOS1 expression. Continued investigation on the roles of NOS1, therefore, will synthesize new knowledge and aid in the discovery of small molecules which could be used to titrate the activities of NOS1-derived NO signaling and NO-metabolites. Here, we address the significance of NOS1 and its byproduct NO in modifying pathophysiological events, which could be beneficial in understanding both the disease mechanisms and therapeutics.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Sajjan Rajpoot
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny E. Bezsonov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alexander N. Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Rohit Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Anita Wary
- Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Cassondra Axen
- Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Kishore Wary
- Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Mirza S. Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| |
Collapse
|
11
|
Elevated Nitrite/Nitrate Ratio as a Potential Biomarker for the Differential Diagnosis of Pleural Effusions. Antioxidants (Basel) 2022; 11:antiox11071327. [PMID: 35883818 PMCID: PMC9312090 DOI: 10.3390/antiox11071327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 12/10/2022] Open
Abstract
Pleural effusions (PEs) are common in clinical practice and can be due to many different underlying diseases such as cancer, congestive heart failure, or pneumonia. An accurate differential diagnostic categorization is essential, as the treatment and prognosis of PEs largely depend on its cause. In this study, we tested the hypothesis that nitrite and nitrate concentrations in PEs are associated with the inflammation and infection conditions. We therefore measured the nitrite and nitrate levels in 143 PE samples using a sensitive liquid chromatography-tandem mass spectrometry method and investigated their diagnostic potential in differentiating PEs. The results showed that nitrite concentrations and nitrite/nitrate ratios were higher in exudates than in transudates (NO2-: 2.12 vs. 1.49 μM; NO2-/NO3-: 23.3 vs. 14.0). Both the nitrite concentrations and the nitrite/nitrate ratios were positively correlated with the three Light's criteria. Moreover, the receiver operating characteristic curve analysis revealed that the nitrite/nitrate ratio with an area under the curve of 0.71 could be a potential diagnostic biomarker in separating infectious PEs (IPEs) from other types of PEs. Taken together, the nitrite/nitrate ratio not only reflected the statuses of inflammation, but also the nitrate reduction by pathogenic bacteria infection in the pleural cavity. The nitrite/nitrate ratio could be a better biomarker in the differential diagnosis of PEs than the nitrite concentration alone.
Collapse
|
12
|
Liu T, Schroeder H, Power GG, Blood AB. A physiologically relevant role for NO stored in vascular smooth muscle cells: A novel theory of vascular NO signaling. Redox Biol 2022; 53:102327. [PMID: 35605454 PMCID: PMC9126848 DOI: 10.1016/j.redox.2022.102327] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/16/2022] [Accepted: 04/29/2022] [Indexed: 01/16/2023] Open
Abstract
S-nitrosothiols (SNO), dinitrosyl iron complexes (DNIC), and nitroglycerine (NTG) dilate vessels via activation of soluble guanylyl cyclase (sGC) in vascular smooth muscle cells. Although these compounds are often considered to be nitric oxide (NO) donors, attempts to ascribe their vasodilatory activity to NO-donating properties have failed. Even more puzzling, many of these compounds have vasodilatory potency comparable to or even greater than that of NO itself, despite low membrane permeability. This raises the question: How do these NO adducts activate cytosolic sGC when their NO moiety is still outside the cell? In this review, we classify these compounds as ‘nitrodilators’, defined by their potent NO-mimetic vasoactivities despite not releasing requisite amounts of free NO. We propose that nitrodilators activate sGC via a preformed nitrodilator-activated NO store (NANOS) found within the vascular smooth muscle cell. We reinterpret vascular NO handling in the framework of this NANOS paradigm, and describe the knowledge gaps and perspectives of this novel model.
Collapse
|
13
|
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
|
14
|
Abbasi-Habashi S, Jickling GC, Winship IR. Immune Modulation as a Key Mechanism for the Protective Effects of Remote Ischemic Conditioning After Stroke. Front Neurol 2021; 12:746486. [PMID: 34956045 PMCID: PMC8695500 DOI: 10.3389/fneur.2021.746486] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Remote ischemic conditioning (RIC), which involves a series of short cycles of ischemia in an organ remote to the brain (typically the limbs), has been shown to protect the ischemic penumbra after stroke and reduce ischemia/reperfusion (IR) injury. Although the exact mechanism by which this protective signal is transferred from the remote site to the brain remains unclear, preclinical studies suggest that the mechanisms of RIC involve a combination of circulating humoral factors and neuronal signals. An improved understanding of these mechanisms will facilitate translation to more effective treatment strategies in clinical settings. In this review, we will discuss potential protective mechanisms in the brain and cerebral vasculature associated with RIC. We will discuss a putative role of the immune system and circulating mediators of inflammation in these protective processes, including the expression of pro-and anti-inflammatory genes in peripheral immune cells that may influence the outcome. We will also review the potential role of extracellular vesicles (EVs), biological vectors capable of delivering cell-specific cargo such as proteins and miRNAs to cells, in modulating the protective effects of RIC in the brain and vasculature.
Collapse
Affiliation(s)
- Sima Abbasi-Habashi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Division of Neurology, Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Mondal P, Tolbert GB, Wijeratne GB. Bio-inspired nitrogen oxide (NO x) interconversion reactivities of synthetic heme Compound-I and Compound-II intermediates. J Inorg Biochem 2021; 226:111633. [PMID: 34749065 DOI: 10.1016/j.jinorgbio.2021.111633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 10/20/2022]
Abstract
Dioxygen activating heme enzymes have long predicted to be powerhouses for nitrogen oxide interconversion, especially for nitric oxide (NO) oxidation which has far-reaching biological and/or environmental impacts. Lending credence, reactivity of NO with high-valent heme‑oxygen intermediates of globin proteins has recently been implicated in the regulation of a variety of pivotal physiological events such as modulating catalytic activities of various heme enzymes, enhancing antioxidant activity to inhibit oxidative damage, controlling inflammatory and infectious properties within the local heme environments, and NO scavenging. To reveal insights into such crucial biological processes, we have investigated low temperature NO reactivities of two classes of synthetic high-valent heme intermediates, Compound-II and Compound-I. In that, Compound-II rapidly reacts with NO yielding the six-coordinate (NO bound) heme ferric nitrite complex, which upon warming to room temperature converts into the five-coordinate heme ferric nitrite species. These ferric nitrite complexes mediate efficient substrate oxidation reactions liberating NO; i.e., shuttling NO2- back to NO. In contrast, Compound-I and NO proceed through an oxygen-atom transfer process generating the strong nitrating agent NO2, along with the corresponding ferric nitrosyl species that converts to the naked heme ferric parent complex upon warmup. All reaction components have been fully characterized by UV-vis, 2H NMR and EPR spectroscopic methods, mass spectrometry, elemental analyses, and semi-quantitative determination of NO2- anions. The clean, efficient, potentially catalytic NOx interconversions driven by high-valent heme species presented herein illustrate the strong prospects of a heme enzyme/O2/NOx dependent unexplored territory that is central to human physiology, pathology, and therapeutics.
Collapse
Affiliation(s)
- Pritam Mondal
- Department of Chemistry and O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35205, United States
| | - Garrett B Tolbert
- Department of Chemistry and O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35205, United States
| | - Gayan B Wijeratne
- Department of Chemistry and O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35205, United States.
| |
Collapse
|
16
|
Miller L, Hébert CD, Grimes SD, Toomey JS, Oh JY, Rose JJ, Patel RP. Safety and toxicology assessment of sodium nitrite administered by intramuscular injection. Toxicol Appl Pharmacol 2021; 429:115702. [PMID: 34464673 PMCID: PMC8459319 DOI: 10.1016/j.taap.2021.115702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 10/20/2022]
Abstract
Intramuscular (IM) injection of nitrite (1-10 mg/kg) confers survival benefit and protects against lung injury after exposure to chlorine gas in preclinical models. Herein, we evaluated safety/toxicity parameters after single, and repeated (once daily for 7 days) IM injection of nitrite in male and female Sprague Dawley rats and Beagle dogs. The repeat dose studies were performed in compliance with the Federal Drug Administration's (FDA) Good Laboratory Practices Code of Federal Regulations (21 CFR Part 58). Parameters evaluated consisted of survival, clinical observations, body weights, clinical pathology, plasma drug levels, methemoglobin and macroscopic and microscopic pathology. In rats and dogs, single doses of ≥100 mg/kg and 60 mg/kg resulted in death and moribundity, while repeated administration of ≤30 or ≤ 10 mg/kg/day, respectively, was well tolerated. Therefore, the maximum tolerated dose following repeated administration in rats and dogs were determined to be 30 mg/kg/day and 10 mg/kg/day, respectively. Effects at doses below the maximum tolerated dose (MTD) were limited to emesis (in dogs only) and methemoglobinemia (in both species) with clinical signs (e.g. blue discoloration of lips) being dose-dependent, transient and reversible. These signs were not considered adverse, therefore the No Observed Adverse Effect Level (NOAEL) for both rats and dogs was 10 mg/kg/day in males (highest dose tested for dogs), and 3 mg/kg/day in females. Toxicokinetic assessment of plasma nitrite showed no difference between male and females, with Cmax occurring between 5 mins and 0.5 h (rats) or 0.25 h (dogs). In summary, IM nitrite was well tolerated in rats and dogs at doses previously shown to confer protection against chlorine gas toxicity.
Collapse
Affiliation(s)
- Lutfiya Miller
- Intertek Health Sciences, Inc., Pharmaceuticals & Healthcare, Mississauga, ON, Canada
| | | | | | - James S Toomey
- Southern Research, Birmingham, AL, United States of America
| | - Joo-Yeun Oh
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason J Rose
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rakesh P Patel
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
17
|
Pekas EJ, Wooden TK, Yadav SK, Park SY. Body mass-normalized moderate dose of dietary nitrate intake improves endothelial function and walking capacity in patients with peripheral artery disease. Am J Physiol Regul Integr Comp Physiol 2021; 321:R162-R173. [PMID: 34161745 DOI: 10.1152/ajpregu.00121.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peripheral artery disease (PAD) is characterized by the accumulation of atherosclerotic plaques in the lower extremity conduit arteries, which impairs blood flow and walking capacity. Dietary nitrate has been used to reduce blood pressure (BP) and improve walking capacity in PAD. However, a standardized dose for PAD has not been determined. Therefore, we sought to determine the effects of a body mass-normalized moderate dose of nitrate (0.11 mmol nitrate/kg) as beetroot juice on serum nitrate/nitrite, vascular function, walking capacity, and tissue oxygen utilization capacity in patients with PAD. A total of 11 patients with PAD received either nitrate supplement or placebo in a randomized crossover design. Total serum nitrate/nitrite, resting BP, brachial and popliteal artery endothelial function (flow-mediated dilation, FMD), arterial stiffness (pulse-wave velocity, PWV), augmentation index (AIx), maximal walking distance and time, claudication onset time, and skeletal muscle oxygen utilization were measured pre- and postnitrate and placebo intake. There were significant group × time interactions (P < 0.05) for serum nitrate/nitrite, FMD, BP, walking distance and time, and skeletal muscle oxygen utilization. The nitrate group showed significantly increased serum nitrate/nitrite (Δ1.32 μM), increased brachial and popliteal FMD (Δ1.3% and Δ1.7%, respectively), reduced peripheral and central systolic BP (Δ-4.7 mmHg and Δ-8.2 mmHg, respectively), increased maximal walking distance (Δ92.7 m) and time (Δ56.3 s), and reduced deoxygenated hemoglobin during walking. There were no changes in PWV, AIx, or claudication (P > 0.05). These results indicate that a body-mass normalized moderate dose of nitrate may be effective and safe for reducing BP, improving endothelial function, and improving walking capacity in patients with PAD.
Collapse
Affiliation(s)
- Elizabeth J Pekas
- School of Health & Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska
| | - TeSean K Wooden
- School of Health & Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska
| | - Santosh K Yadav
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Song-Young Park
- School of Health & Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska
| |
Collapse
|
18
|
Nitrite and tempol combination promotes synergic effects and alleviates right ventricular wall stress during acute pulmonary thromboembolism. Nitric Oxide 2021; 115:23-29. [PMID: 34133975 DOI: 10.1016/j.niox.2021.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/29/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION The mechanical obstruction and pulmonary vasoconstriction are major determinants of the sudden right ventricular (RV) afterload increases observed during acute pulmonary thromboembolism (APT). Vasodilators and antioxidants agents have been shown to mitigate pulmonary hypertension. We examined whether sodium nitrite and the antioxidant tempol combination could be advantageous in an APT sheep model. METHODS APT was induced in anesthetized sheep by autologous blood clots (250 mg/kg) into the right atrium. Thirty minutes after APT induction, the animals received a continuous infusion of tempol (1.0 mg/kg/min), increasing sodium nitrite infusion (5, 15, and 50 μmol/kg), or a simultaneous combination of both drugs. Saline was used as a control treatment. Hemodynamic measurements were carried out every 15 min. Also, whole blood nitrite and serum 8-isoprostanes levels were measured. RESULTS APT induced sustained pulmonary hypertension, increased dp/dtmax, and rate pressure product (RPP). Nitrite or tempol treatments attenuated these increases (P < 0.05). When both drugs were combined, we found a robust reduction in the RV RPP compared with the treatments alone (P < 0.05). The sole nitrite infusion increased blood nitrite concentrations by 35 ± 6 μM (P < 0.05), whereas the nitrite and tempol combination produced higher blood nitrite concentrations by approximately 54 ± 7 μM. Tempol or nitrite infusions, both alone or combined, blunted the increases in 8-isoprostane concentrations observed after APT. CONCLUSIONS Nitrite and tempol combination protects against APT-induced RV wall stress. The association of both drugs may offer an advantage to treat RV failure during severe APT.
Collapse
|
19
|
Hu Q, Shi J, Zhang J, Wang Y, Guo Y, Zhang Z. Progress and Prospects of Regulatory Functions Mediated by Nitric Oxide on Immunity and Immunotherapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Qian Hu
- Tongji School of Pharmacy Huazhong University of Science and Technology Wuhan Hubei 430030 China
| | - Jingyu Shi
- Liyuan Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei 430077 China
| | - Jiao Zhang
- Tongji School of Pharmacy Huazhong University of Science and Technology Wuhan Hubei 430030 China
| | - Yi Wang
- Tongji School of Pharmacy Huazhong University of Science and Technology Wuhan Hubei 430030 China
| | - Yuanyuan Guo
- Liyuan Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei 430077 China
| | - Zhiping Zhang
- Tongji School of Pharmacy, National Engineering Research Centre for Nanomedicine, Hubei Engineering Research Centre for Novel Drug Delivery System Huazhong University of Science and Technology Wuhan Hubei 430030 China
| |
Collapse
|
20
|
Oral nitrite treatment increases S-nitrosylation of vascular protein kinase C and attenuates the responses to angiotensin II. Redox Biol 2020; 38:101769. [PMID: 33126056 PMCID: PMC7596338 DOI: 10.1016/j.redox.2020.101769] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023] Open
Abstract
Nitrate and nitrite supplement deficient endogenous nitric oxide (NO) formation. While these anions may generate NO, recent studies have shown that circulating nitrite levels do not necessarily correlate with the antihypertensive effect of oral nitrite administration and that formation of nitrosylated species (RXNO) in the stomach is critically involved in this effect. This study examined the possibility that RXNO formed in the stomach after oral nitrite administration promotes target protein nitrosylation in the vasculature, inhibits vasoconstriction and the hypertensive responses to angiotensin II. Our results show that oral nitrite treatment enhances circulating RXNO concentrations (measured by ozone-based chemiluminescence methods), increases aortic protein kinase C (PKC) nitrosylation (measured by resin-assisted capture SNO-RAC method), and reduces both angiotensin II-induced vasoconstriction (isolated aortic ring preparation) and hypertensive (in vivo invasive blood pressure measurements) effects implicating PKC nitrosylation as a key mechanism for the responses to oral nitrite. Treatment of rats with the nitrosylating compound S-nitrosoglutathione (GSNO) resulted in the same effects described for oral nitrite. Moreover, partial depletion of thiols with buthionine sulfoximine prevented PKC nitrosylation and the blood pressure effects of oral nitrite. Further confirming a role for PKC nitrosylation, preincubation of aortas with GSNO attenuated the responses to both angiotensin II and to a direct PKC activator, and this effect was attenuated by ascorbate (reverses GSNO-induced nitrosylation). GSNO-induced nitrosylation also inhibited the increases in Ca2+ mobilization in angiotensin II-stimulated HEK293T cells expressing angiotensin type 1 receptor. Together, these results are consistent with the idea that PKC nitrosylation in the vasculature may underlie oral nitrite treatment-induced reduction in the vascular and hypertensive responses to angiotensin II. Oral nitrite treatment exerts antihypertensive effects. The mechanisms explaining such effects are not entirely known. Oral nitrite treatment increases circulating concentrations of nitrosylating species. Vascular PKC nitrosylation attenuates the vascular responses to angiotensin II.
Collapse
|
21
|
The solution chemistry of nitric oxide and other reactive nitrogen species. Nitric Oxide 2020; 103:31-46. [DOI: 10.1016/j.niox.2020.07.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022]
|
22
|
Oral Administration of Sodium Nitrate to Metabolic Syndrome Patients Attenuates Mild Inflammatory and Oxidative Responses to Acute Exercise. Antioxidants (Basel) 2020; 9:antiox9070596. [PMID: 32646062 PMCID: PMC7402183 DOI: 10.3390/antiox9070596] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 01/24/2023] Open
Abstract
The beneficial effects of exercise for the treatment and prevention of metabolic syndrome pathologies have been related to its anti-inflammatory and antioxidant effects. Dietary nitrate supplementation is an emerging treatment strategy to alleviate the symptoms of metabolic syndrome affections and to improve vascular function. In this double-blind crossover trial, metabolic syndrome patients performed two exercise tests for 30 min at 60–70% maximal heart rate after the intake of a placebo or a nitrate-enriched beverage. Acute exercise increased the plasma concentration of TNFα, intercellular adhesion molecule ICAM1, PGE1, PGE2 and the newly detected 16-hydroxypalmitic acid (16-HPAL) in metabolic syndrome patients. The cytokine and oxylipin production by peripheral blood mononuclear cells (PBMCs) and neutrophils could be responsible for the plasma concentrations of TNFα and IL6, but not for the plasma concentration of oxylipins nor its post-exercise increase. The intake of sodium nitrate 30 min before exercise increased the concentration of nitrate and nitrite in the oral cavity and plasma and reduced the oxygen cost of exercise. Additionally, nitrate intake prevented the enhancing effects of acute exercise on the plasma concentration of TNFα, ICAM1, PGE1, PGE2 and 16-HPAL, while reducing the capabilities of PBMCs and neutrophils to produce oxylipins.
Collapse
|
23
|
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]
|
24
|
Abstract
Significance: Cytoglobin (Cygb) was discovered as a new addition to the globin superfamily and subsequently identified to have potent nitric oxide (NO) dioxygenase function. Cygb plays a critical role in the oxygen-dependent regulation of NO levels and vascular tone. Recent Advances: In recent years, the mechanism of the Cygb-mediated NO dioxygenation has been studied in isolated protein, smooth muscle cell, isolated blood vessel, and in vivo animal model systems. Studies in Cygb-/- mice have demonstrated that Cygb plays a critical role in regulating blood pressure and vascular tone. This review summarizes advances in the knowledge of NO dioxygenation/metabolism regulated by Cygb. Advances in measurement of NO diffusion dynamics across blood vessels and kinetic modeling of Cygb-mediated NO dioxygenation are summarized. The oxygen-dependent regulation of NO degradation by Cygb is also reviewed along with how Cygb paradoxically generates NO from nitrite under anaerobic conditions. The important role of Cygb in the regulation of vascular function and disease is reviewed. Critical Issues: Cygb is a more potent NO dioxygenase (NOD) than previously known globins with structural differences in heme coordination and environment, conferring it with a higher rate of reduction and more rapid process of NO dioxygenation with unique oxygen dependence. Various cellular reducing systems regenerate the catalytic oxyferrous Cygb species, supporting a high rate of NO dioxygenation. Future Directions: There remains a critical need to further characterize the factors and processes that modulate Cygb-mediated NOD function, and to develop pharmacological or other approaches to modulate Cygb function and expression.
Collapse
Affiliation(s)
- Jay L Zweier
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Govindasamy Ilangovan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
25
|
Gantner BN, LaFond KM, Bonini MG. Nitric oxide in cellular adaptation and disease. Redox Biol 2020; 34:101550. [PMID: 32438317 PMCID: PMC7235643 DOI: 10.1016/j.redox.2020.101550] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/15/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide synthases are the major sources of nitric oxide, a critical signaling molecule involved in a wide range of cellular and physiological processes. These enzymes comprise a family of genes that are highly conserved across all eukaryotes. The three family members found in mammals are important for inter- and intra-cellular signaling in tissues that include the nervous system, the vasculature, the gut, skeletal muscle, and the immune system, among others. We summarize major advances in the understanding of biochemical and tissue-specific roles of nitric oxide synthases, with a focus on how these mechanisms enable tissue adaptation and health or dysfunction and disease. We highlight the unique mechanisms and processes of neuronal nitric oxide synthase, or NOS1. This was the first of these enzymes discovered in mammals, and yet much remains to be understood about this highly conserved and complex gene. We provide examples of two areas that will likely be of increasing importance in nitric oxide biology. These include the mechanisms by which these critical enzymes promote adaptation or disease by 1) coordinating communication by diverse cell types within a tissue and 2) directing cellular differentiation/activation decisions processes.
Collapse
Affiliation(s)
- Benjamin N Gantner
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, USA.
| | - Katy M LaFond
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, USA
| | - Marcelo G Bonini
- Department of Medicine, Division of Endocrinology, Medical College of Wisconsin, USA; Feinberg School of Medicine, Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, USA
| |
Collapse
|
26
|
Glutamine and citrulline concentrations reflect nitric oxide synthesis in the human nervous system. NEUROLOGÍA (ENGLISH EDITION) 2020. [DOI: 10.1016/j.nrleng.2017.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
27
|
Su H, Liu X, Du J, Deng X, Fan Y. The role of hemoglobin in nitric oxide transport in vascular system. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2020. [DOI: 10.1016/j.medntd.2020.100034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
28
|
Zhao Y, Lim J, Xu J, Yu J, Zheng W. Nitric oxide as a developmental and metabolic signal in filamentous fungi. Mol Microbiol 2020; 113:872-882. [DOI: 10.1111/mmi.14465] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Yanxia Zhao
- Key Laboratory for Biotechnology of Medicinal Plants Jiangsu Normal University Xuzhou China
| | - Jieyin Lim
- Departments of Bacteriology and Genetics Food Research Institute University of Wisconsin‐Madison Madison Wisconsin USA
| | - Jianyang Xu
- Department of Traditional Chinese Medicine General Hospital of Shenzhen University Shenzhen China
| | - Jae‐Hyuk Yu
- Departments of Bacteriology and Genetics Food Research Institute University of Wisconsin‐Madison Madison Wisconsin USA
- Department of Systems Biotechnology Konkuk University Seoul Republic of Korea
| | - Weifa Zheng
- Key Laboratory for Biotechnology of Medicinal Plants Jiangsu Normal University Xuzhou China
| |
Collapse
|
29
|
Pazarín-Villaseñor L, Alejandro Gutiérrez-Prieto J, Soto-Vargas J, Parra-Michel R, García-Sanchez A, Guillermina Miranda-Diaz A. Increase of Oxidants and Antioxidant Consumption in Patients with Type 2 Diabetes Mellitus in Peritoneal Dialysis. Antioxidants (Basel) 2019. [DOI: 10.5772/intechopen.82880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
30
|
Neto-Neves EM, Pinheiro LC, Nogueira RC, Portella RL, Batista RI, Tanus-Santos JE. Sodium nitrite improves hypertension-induced myocardial dysfunction by mechanisms involving cardiac S-nitrosylation. J Mol Cell Cardiol 2019; 134:40-50. [DOI: 10.1016/j.yjmcc.2019.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
|
31
|
Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther 2019; 201:137-180. [PMID: 31128989 DOI: 10.1016/j.pharmthera.2019.05.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/27/2019] [Indexed: 11/29/2022]
Abstract
Aldehyde oxidase (AO) is a cytosolic enzyme that belongs to the family of structurally related molybdoflavoproteins like xanthine oxidase (XO). The enzyme is characterized by broad substrate specificity and marked species differences. It catalyzes the oxidation of aromatic and aliphatic aldehydes and various heteroaromatic rings as well as reduction of several functional groups. The references to AO and its role in metabolism date back to the 1950s, but the importance of this enzyme in the metabolism of drugs has emerged in the past fifteen years. Several reviews on the role of AO in drug metabolism have been published in the past decade indicative of the growing interest in the enzyme and its influence in drug metabolism. Here, we present a comprehensive monograph of AO as a drug metabolizing enzyme with emphasis on marketed drugs as well as other xenobiotics, as substrates and inhibitors. Although the number of drugs that are primarily metabolized by AO are few, the impact of AO on drug development has been extensive. We also discuss the effect of AO on the systemic exposure and clearance these clinical candidates. The review provides a comprehensive analysis of drug discovery compounds involving AO with the focus on developmental candidates that were reported in the past five years with regards to pharmacokinetics and toxicity. While there is only one known report of AO-mediated clinically relevant drug-drug interaction (DDI), a detailed description of inhibitors and inducers of AO known to date has been presented here and the potential risks associated with DDI. The increasing recognition of the importance of AO has led to significant progress in predicting the site of AO-mediated metabolism using computational methods. Additionally, marked species difference in expression of AO makes it is difficult to predict human clearance with high confidence. The progress made towards developing in vivo, in vitro and in silico approaches for predicting AO metabolism and estimating human clearance of compounds that are metabolized by AO have also been discussed.
Collapse
Affiliation(s)
- Deepak Dalvie
- Drug Metabolism and Pharmacokinetics, Celgene Corporation, 10300, Campus Point Drive, San Diego, CA 92121, USA.
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, UK
| |
Collapse
|
32
|
Cortés-Puch I, Sun J, Schechter AN, Solomon SB, Park JW, Feng J, Gilliard C, Natanson C, Piknova B. Inhaled nebulized nitrite and nitrate therapy in a canine model of hypoxia-induced pulmonary hypertension. Nitric Oxide 2019; 91:1-14. [PMID: 31299340 DOI: 10.1016/j.niox.2019.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/17/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Dysfunction in the nitric oxide (NO) signaling pathway can lead to the development of pulmonary hypertension (PH) in mammals. Discovery of an alternative pathway to NO generation involving reduction from nitrate to nitrite and to NO has motivated the evaluation of nitrite as an alternative to inhaled NO for PH. In contrast, inhaled nitrate has not been evaluated to date, and potential benefits include a prolonged half-life and decreased risk of methemoglobinemia. In a canine model of acute hypoxia-induced PH we evaluated the effects of inhaled nitrate to reduce pulmonary arterial pressure (PAP). In a randomized controlled trial, inhaled nitrate was compared to inhaled nitrite and inhaled saline. Exhaled NO, PAP and systemic blood pressures were continuously monitored. Inhaled nitrite significantly decreased PAP and increased exhaled NO. In contrast, inhaled nitrate and inhaled saline did not decrease PAP or increase exhaled NO. Unexpectedly, we found that inhaled nitrite resulted in prolonged (>5 h) exhaled NO release, increase in nitrate venous/arterial levels and a late surge in venous nitrite levels. These findings do not support a therapeutic role for inhaled nitrate in PH but may have therapeutic implications for inhaled nitrite in various disease states.
Collapse
Affiliation(s)
- Irene Cortés-Puch
- National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, MD, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California Davis, Sacramento, CA, USA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Alan N Schechter
- National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, MD, USA
| | - Steven B Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Ji Won Park
- National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, MD, USA
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Cameron Gilliard
- National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, MD, USA; Penn State Health Milton S. Hershey Medical Center, Department of Anesthesia and Perioperative Medicine, Hershey, PA, USA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Barbora Piknova
- National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Oliveira-Paula GH, Pinheiro LC, Tanus-Santos JE. Mechanisms impairing blood pressure responses to nitrite and nitrate. Nitric Oxide 2019; 85:35-43. [PMID: 30716418 DOI: 10.1016/j.niox.2019.01.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/13/2018] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
Hypertension is a multifactorial disease associated with impaired nitric oxide (NO) production and bioavailability. In this respect, restoring NO activity by using nitrite and nitrate has been considered a potential therapeutic strategy to treat hypertension. This possibility is justified by the understanding that both nitrite and nitrate may be recycled back to NO and also promote the generation of other bioactive species. This process involves a complex biological circuit known as the enterosalivary cycle of nitrate, where this anion is actively taken up by the salivary glands and converted to nitrite by nitrate-reducing bacteria in the oral cavity. Nitrite is then ingested and reduced to NO and other nitroso species under the acid conditions of the stomach, whereas reminiscent nitrite that escapes gastric reduction is absorbed systemically and can be converted into NO by nitrite-reductases in tissues. While there is no doubt that nitrite and nitrate exert antihypertensive effects, several agents can impair the blood pressure responses to these anions by disrupting the enterosalivary cycle of nitrate. These agents include dietary and smoking-derived thiocyanate, antiseptic mouthwash, proton pump inhibitors, ascorbate at high concentrations, and xanthine oxidoreductase inhibitors. In this article, we provide an overview of the physiological aspects of nitrite and nitrate bioactivation and the therapeutic potential of these anions in hypertension. We also discuss mechanisms by which agents counteracting the antihypertensive responses to nitrite and nitrate mediate their effects. These critical aspects should be taken into consideration when suggesting nitrate or nitrite-based therapies to patients.
Collapse
Affiliation(s)
- Gustavo H Oliveira-Paula
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
34
|
Lim YJ, Foo TC, Yeung AWS, Tu X, Ma Y, Hawkins CL, Witting PK, Jameson GNL, Terentis AC, Thomas SR. Human Indoleamine 2,3-Dioxygenase 1 Is an Efficient Mammalian Nitrite Reductase. Biochemistry 2019; 58:974-986. [PMID: 30585477 DOI: 10.1021/acs.biochem.8b01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The heme enzyme indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the first reaction of l-tryptophan oxidation along the kynurenine pathway. IDO1 is a central immunoregulatory enzyme with important implications for inflammation, infectious disease, autoimmune disorders, and cancer. Here we demonstrate that IDO1 is a mammalian nitrite reductase capable of chemically reducing nitrite to nitric oxide (NO) under hypoxia. Ultraviolet-visible absorption and resonance Raman spectroscopy showed that incubation of dithionite-reduced, ferrous-IDO1 protein (FeII-IDO1) with nitrite under anaerobic conditions resulted in the time-dependent formation of an FeII-nitrosyl IDO1 species, which was inhibited by substrate l-tryptophan, dependent on the concentration of nitrite or IDO1, and independent of the concentration of the reductant, dithionite. The bimolecular rate constant for IDO1 nitrite reductase activity was determined as 5.4 M-1 s-1 (pH 7.4, 23 °C), which was comparable to that measured for myoglobin (3.6 M-1 s-1; pH 7.4, 23 °C), an efficient and biologically important mammalian heme-based nitrite reductase. IDO1 nitrite reductase activity was pH-dependent but differed with myoglobin in that it showed a reduced proton dependency at pH >7. Electron paramagnetic resonance studies measuring NO production showed that the conventional IDO1 dioxygenase reducing cofactors, ascorbate and methylene blue, enhanced IDO1's nitrite reductase activity and the time- and IDO1 concentration-dependent release of NO in a manner inhibited by l-tryptophan or the IDO inhibitor 1-methyl-l-tryptophan. These data identify IDO1 as an efficient mammalian nitrite reductase that is capable of generating NO under anaerobic conditions. IDO1's nitrite reductase activity may have important implications for the enzyme's biological actions when expressed within hypoxic tissues.
Collapse
Affiliation(s)
| | - Timothy C Foo
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | | | | | | - Clare L Hawkins
- Department of Biomedical Sciences , University of Copenhagen , Copenhagen N DK-2200 , Denmark
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Guy N L Jameson
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , VIC 3010 , Australia
| | - Andrew C Terentis
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | |
Collapse
|
35
|
del Carmen Baez M, Tarán M, Moya M, de la Paz Scribano Parada M. Oxidative Stress in Metabolic Syndrome: Experimental Model of Biomarkers. MODULATION OF OXIDATIVE STRESS IN HEART DISEASE 2019:313-338. [DOI: 10.1007/978-981-13-8946-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 PMCID: PMC6442925 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/30/2018] [Accepted: 05/06/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
37
|
Jankov RP, Daniel KL, Iny S, Kantores C, Ivanovska J, Ben Fadel N, Jain A. Sodium nitrite augments lung S-nitrosylation and reverses chronic hypoxic pulmonary hypertension in juvenile rats. Am J Physiol Lung Cell Mol Physiol 2018; 315:L742-L751. [DOI: 10.1152/ajplung.00184.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Deficient nitric oxide (NO) signaling plays a critical role in the pathogenesis of chronic neonatal pulmonary hypertension (PHT). Physiological NO signaling is regulated by S-nitrosothiols (SNOs), which act both as a reservoir for NO and as a reversible modulator of protein function. We have previously reported that therapy with inhaled NO (iNO) increased peroxynitrite-mediated nitration in the juvenile rat lung, although having minimal reversing effects on vascular remodeling. We hypothesized that sodium nitrite (NaNO2) would be superior to iNO in enhancing lung SNOs, thereby contributing to reversal of chronic hypoxic PHT. Rat pups were exposed to air or hypoxia (13% O2) from postnatal days 1 to 21. Dose-response prevention studies were conducted from days 1–21 to determine the optimal dose of NaNO2. Animals then received rescue therapy with daily subcutaneous NaNO2 (20 mg/kg), vehicle, or were continuously exposed to iNO (20 ppm) from days 14–21. Chronic PHT secondary to hypoxia was both prevented and reversed by treatment with NaNO2. Rescue NaNO2 increased lung NO and SNO contents to a greater extent than iNO, without causing nitration. Seven lung SNO proteins upregulated by treatment with NaNO2 were identified by multiplex tandem mass tag spectrometry, one of which was leukotriene A4 hydrolase (LTA4H). Rescue therapy with a LTA4H inhibitor, SC57461A (10 mg·kg−1·day−1 sc), partially reversed chronic hypoxic PHT. We conclude that NaNO2 was superior to iNO in increasing tissue NO and SNO generation and reversing chronic PHT, in part via upregulated SNO-LTA4H.
Collapse
Affiliation(s)
- Robert P. Jankov
- Molecular Biomedicine Program, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Kathrine L. Daniel
- Molecular Biomedicine Program, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Shira Iny
- Molecular Biomedicine Program, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Crystal Kantores
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Julijana Ivanovska
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Nadya Ben Fadel
- Faculty of Medicine, Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Amish Jain
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Troutman AD, Gallardo EJ, Brown MB, Coggan AR. Measurement of nitrate and nitrite in biopsy-sized muscle samples using HPLC. J Appl Physiol (1985) 2018; 125:1475-1481. [PMID: 30113272 DOI: 10.1152/japplphysiol.00625.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Studies of rats have indicated that skeletal muscle plays a central role in whole-body nitrate ( NO3- )/nitrite ( NO2- )/nitric oxide (NO) metabolism. Extending these results to humans, however, is challenging due to the small size of needle biopsy samples. We therefore developed a method to precisely and accurately quantify NO3- and NO2- in biopsy-sized muscle samples. NO3- and NO2- were extracted from rat soleus samples using methanol combined with mechanical homogenization + ultrasound, bead beating, pulverization at liquid N2 temperature or pulverization + 0.5% Triton X-100. After centrifugation to remove proteins, NO3- and NO2- were measured using HPLC. Mechanical homogenization + ultrasound resulted in the lowest NO3- content (62 ± 20 pmol/mg), with high variability [coefficient of variation (CV) >50%] across samples from the same muscle. The NO2- / NO3- ratio (0.019 ± 0.006) was also elevated, suggestive of NO3- reduction during tissue processing. Bead beating or pulverization yielded lower NO2- and slightly higher NO3- levels, but reproducibility was still poor. Pulverization + 0.5% Triton X-100 provided the highest NO3- content (124 ± 12 pmol/mg) and lowest NO2- / NO3- ratio (0.008 ± 0.001), with the least variability between duplicate samples (CV ~15%). These values are consistent with literature data from larger rat muscle samples analyzed using chemiluminescence. Samples were stable for at least 5 wk at -80°C, provided residual xanthine oxidoreductase activity was blocked using 0.1 mmol/l oxypurinol. We have developed a method capable of measuring NO3- and NO2- in <1 mg of muscle. This method should prove highly useful in investigating the role of skeletal muscle in NO3- / NO2- /NO metabolism in human health and disease. NEW & NOTEWORTHY Measurement of nitrate and especially nitrite in small, i.e., biopsy-sized, muscle samples is analytically challenging. We have developed a precise, accurate, and convenient method for doing so using an affordable commercial HPLC system.
Collapse
Affiliation(s)
- Ashley D Troutman
- Department of Kinesiology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana.,Department of Physical Therapy, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Edgar J Gallardo
- Department of Kinesiology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Mary Beth Brown
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana
| | - Andrew R Coggan
- Department of Kinesiology, Indiana University-Purdue University Indianapolis , Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana-University Purdue University Indianapolis , Indianapolis, Indiana
| |
Collapse
|
39
|
Ma L, Hu L, Feng X, Wang S. Nitrate and Nitrite in Health and Disease. Aging Dis 2018; 9:938-945. [PMID: 30271668 PMCID: PMC6147587 DOI: 10.14336/ad.2017.1207] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 12/07/2017] [Indexed: 12/23/2022] Open
Abstract
The source of dietary nitrate (NO3) is mainly green, leafy vegetables, partially absorbed into blood through intestinal mucosa. The recycled nitrate is reabsorbed and concentrated by the salivary glands and then secreted into saliva. In 2012, sialin was first discovered as the mammalian membrane nitrate transporter in salivary glands and plays a key role in circulation of inorganic nitrate, providing a scientific basis for further investigation into the circulation and functions of nitrate. Dietary nitrate can be converted to nitrite (NO2) by oral commensal bacteria under the tongue or in the stomach, following which nitrite is converted to nitric oxide (NO) through non-enzymatic synthesis. Previously, nitrate and nitrite were thought to be carcinogenic due to the potential formation of nitrogen compounds, whereas the beneficial functions of NO3--NO2--NO pathway were ignored. Under conditions of hypoxia and ischemia, the production of endogenous NO from L-arginine is inhibited, while the activity of exogenous NO3--NO2--NO is enhanced. Recently, a greater amount of evidence has shown that nitrate and nitrite serve as a reservoir and perform positive biological NO-like functions. Therefore, exogenous dietary nitrate plays an important role in various physiological activities as an effective supplement of nitrite and NO in human body. Here we generally review the source, circulation and bio-functions of dietary nitrate.
Collapse
Affiliation(s)
- Linsha Ma
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100069, China
| | - Liang Hu
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100069, China
| | - Xiaoyu Feng
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100069, China
| | - Songlin Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University, Beijing 100069, China
| |
Collapse
|
40
|
Li F, Meyer RL, Carpenter SH, VanGelder LE, Nichols AW, Machan CW, Neidig ML, Matson EM. Nitric oxide activation facilitated by cooperative multimetallic electron transfer within an iron-functionalized polyoxovanadate-alkoxide cluster. Chem Sci 2018; 9:6379-6389. [PMID: 30310566 PMCID: PMC6115649 DOI: 10.1039/c8sc00987b] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/30/2018] [Indexed: 01/06/2023] Open
Abstract
Cooperative multimetallic electron transfer to accommodate substrate binding.
A series of NO-bound, iron-functionalized polyoxovanadate–alkoxide (FePOV–alkoxide) clusters have been synthesized, providing insight into the role of multimetallic constructs in the coordination and activation of a substrate. Upon exposure of the heterometallic cluster to NO, the vanadium-oxide metalloligand is oxidized by a single electron, shuttling the reducing equivalent to the {FeNO} subunit to form a {FeNO}7 species. Four NO-bound clusters with electronic distributions ranging from [VV3VIV2]{FeNO}7 to [VIV5]{FeNO}7 have been synthesized, and characterized via1H NMR, infrared, and electronic absorption spectroscopies. The ability of the FePOV–alkoxide cluster to store reducing equivalents in the metalloligand for substrate coordination and activation highlights the ultility of the metal-oxide scaffold as a redox reservoir.
Collapse
Affiliation(s)
- F Li
- Department of Chemistry , University of Rochester , Rochester , New York 14627 , USA .
| | - R L Meyer
- Department of Chemistry , University of Rochester , Rochester , New York 14627 , USA .
| | - S H Carpenter
- Department of Chemistry , University of Rochester , Rochester , New York 14627 , USA .
| | - L E VanGelder
- Department of Chemistry , University of Rochester , Rochester , New York 14627 , USA .
| | - A W Nichols
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904-4319 , USA
| | - C W Machan
- Department of Chemistry , University of Virginia , Charlottesville , Virginia 22904-4319 , USA
| | - M L Neidig
- Department of Chemistry , University of Rochester , Rochester , New York 14627 , USA .
| | - E M Matson
- Department of Chemistry , University of Rochester , Rochester , New York 14627 , USA .
| |
Collapse
|
41
|
Bender D, Schwarz G. Nitrite-dependent nitric oxide synthesis by molybdenum enzymes. FEBS Lett 2018; 592:2126-2139. [DOI: 10.1002/1873-3468.13089] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Daniel Bender
- Department of Chemistry; Institute for Biochemistry; University of Cologne; Germany
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Germany
| | - Guenter Schwarz
- Department of Chemistry; Institute for Biochemistry; University of Cologne; Germany
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD); University of Cologne; Germany
| |
Collapse
|
42
|
Does dietary nitrate say NO to cardiovascular ageing? Current evidence and implications for research. Proc Nutr Soc 2018; 77:112-123. [DOI: 10.1017/s0029665118000058] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CVD are characterised by a multi-factorial pathogenesis. Key pathogenetic steps in the development of CVD are the occurrence of endothelial dysfunction and formation of atherosclerotic lesions. Reduced nitric oxide (NO) bioavailability is a primary event in the initiation of the atherosclerotic cascade. NO is a free radical with multiple physiological functions including the regulation of vascular resistance, coagulation, immunity and oxidative metabolism. The synthesis of NO proceeds via two distinct pathways identified as enzymatic and non-enzymatic. The former involves the conversion of arginine into NO by the NO synthases, whilst the latter comprises a two-step reducing process converting inorganic nitrate $({\rm NO}_3^ - )$ into nitrite and subsequently NO.Inorganic ${\rm NO}_3^ - $ is present in water and food, particularly beetroot and green leafy vegetables. Several investigations have therefore used the non-enzymatic NO pathway as a target for nutritional supplementation (${\rm NO}_3^ - $ salts) or dietary interventions (high-${\rm NO}_3^ - $ foods) to increase NO bioavailability and impact on cardiovascular outcomes. Some studies have reported positive effects of dietary ${\rm NO}_3^ - $ on systolic blood pressure and endothelial function in patients with hypertension and chronic heart failure. Nevertheless, results have been inconsistent and the size of the effect appears to be declining in older individuals. Additionally, there is a paucity of studies for disorders such as diabetes, CHD and chronic kidney failure. Thus, whilst dietary ${\rm NO}_3^ - $ supplementation could represent an effective and viable strategy for the primary and secondary prevention of age-related cardiovascular and metabolic diseases, more large-scale, robust studies are awaited to confirm or refute this notion.
Collapse
|
43
|
Hassanpour SH, Dehghani MA, Karami SZ. Study of respiratory chain dysfunction in heart disease. J Cardiovasc Thorac Res 2018; 10:1-13. [PMID: 29707171 PMCID: PMC5913686 DOI: 10.15171/jcvtr.2018.01] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/25/2017] [Indexed: 02/06/2023] Open
Abstract
The relentlessly beating heart has the greatest oxygen consumption of any organ in the body at rest reflecting its huge metabolic turnover and energetic demands. The vast majority of its energy is produced and cycled in form of ATP which stems mainly from oxidative phosphorylation occurring at the respiratory chain in the mitochondria. A part from energy production, the respiratory chain is also the main source of reactive oxygen species and plays a pivotal role in the regulation of oxidative stress. Dysfunction of the respiratory chain is therefore found in most common heart conditions. The pathophysiology of mitochondrial respiratory chain dysfunction in hereditary cardiac mitochondrial disease, the aging heart, in LV hypertrophy and heart failure, and in ischaemia-reperfusion injury is reviewed. We introduce the practicing clinician to the complex physiology of the respiratory chain, highlight its impact on common cardiac disorders and review translational pharmacological and non-pharmacological treatment strategies.
Collapse
Affiliation(s)
| | - Mohammad Amin Dehghani
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
44
|
Dias L, Bekhti N, Kuznetsov ML, Ferreira JAB, Bacariza MC, da Silva JAL. Nitrite Reduction in Aqueous Solution Mediated by Amavadin Homologues: N2O Formation and Water Oxidation. Chemistry 2018; 24:2474-2482. [DOI: 10.1002/chem.201705385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Lúcia Dias
- Centro de Química Estrutural; Instituto Superior Técnico; Universidade de Lisboa; Av. Rovisco Pais, 1 1049-001 Lisbon Portugal
| | - Nihel Bekhti
- Centro de Química Estrutural; Instituto Superior Técnico; Universidade de Lisboa; Av. Rovisco Pais, 1 1049-001 Lisbon Portugal
| | - Maxim L. Kuznetsov
- Centro de Química Estrutural; Instituto Superior Técnico; Universidade de Lisboa; Av. Rovisco Pais, 1 1049-001 Lisbon Portugal
| | - José A. B. Ferreira
- Centro de Química Estrutural; Instituto Superior Técnico; Universidade de Lisboa; Av. Rovisco Pais, 1 1049-001 Lisbon Portugal
| | - Maria C. Bacariza
- Centro de Química Estrutural; Instituto Superior Técnico; Universidade de Lisboa; Av. Rovisco Pais, 1 1049-001 Lisbon Portugal
| | - José Armando L. da Silva
- Centro de Química Estrutural; Instituto Superior Técnico; Universidade de Lisboa; Av. Rovisco Pais, 1 1049-001 Lisbon Portugal
| |
Collapse
|
45
|
Thomas DD, Corey C, Hickok J, Wang Y, Shiva S. Differential mitochondrial dinitrosyliron complex formation by nitrite and nitric oxide. Redox Biol 2017; 15:277-283. [PMID: 29304478 PMCID: PMC5975210 DOI: 10.1016/j.redox.2017.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/14/2017] [Accepted: 12/17/2017] [Indexed: 01/09/2023] Open
Abstract
Nitrite represents an endocrine reserve of bioavailable nitric oxide (NO) that mediates a number of physiological responses including conferral of cytoprotection after ischemia/reperfusion (I/R). It has long been known that nitrite can react with non-heme iron to form dinitrosyliron complexes (DNIC). However, it remains unclear how quickly nitrite-dependent DNIC form in vivo, whether formation kinetics differ from that of NO-dependent DNIC, and whether DNIC play a role in the cytoprotective effects of nitrite. Here we demonstrate that chronic but not acute nitrite supplementation increases DNIC concentration in the liver and kidney of mice. Although DNIC have been purported to have antioxidant properties, we show that the accumulation of DNIC in vivo is not associated with nitrite-dependent cytoprotection after hepatic I/R. Further, our data in an isolated mitochondrial model of anoxia/reoxygenation show that while NO and nitrite demonstrate similar S-nitrosothiol formation kinetics, DNIC formation is significantly greater with NO and associated with mitochondrial dysfunction as well as inhibition of aconitase activity. These data are the first to directly compare mitochondrial DNIC formation by NO and nitrite. This study suggests that nitrite-dependent DNIC formation is a physiological consequence of dietary nitrite. The data presented herein implicate mitochondrial DNIC formation as a potential mechanism underlying the differential cytoprotective effects of nitrite and NO after I/R, and suggest that DNIC formation is potentially responsible for the cytotoxic effects observed at high NO concentrations. Dietary nitrite results in DNIC formation in many tissues, most notably the liver. Nitrite-dependent DNIC accumulate within the mitochondrion. NO generates greater DNIC formation in the mitochondrion than nitrite. At high concentrations of NO DNIC formation is associated with mitochondrial injury.
Collapse
Affiliation(s)
- Douglas D Thomas
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 South Wood St., Chicago IL 60612, USA.
| | - Catherine Corey
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, BST1240E, 200 Lothrop St, Pittsburgh, PA 15261, USA
| | - Jason Hickok
- Department of Medicinal Chemistry & Pharmacognosy, University of Illinois at Chicago, 833 South Wood St., Chicago IL 60612, USA
| | - Yinna Wang
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, BST1240E, 200 Lothrop St, Pittsburgh, PA 15261, USA
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, BST1240E, 200 Lothrop St, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Center for Metabolism & Mitochondrial Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
46
|
Glutamine and citrulline concentrations reflect nitric oxide synthesis in the human nervous system. Neurologia 2017; 35:96-104. [PMID: 28867511 DOI: 10.1016/j.nrl.2017.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/02/2017] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Although citrulline is produced by nitric oxide (NO) synthase upon activation of the NMDA glutamate receptor, nitrite and nitrate (NOx) concentration is considered the best marker of NO synthesis, as citrulline is also metabolised by other enzymes. This study analyses the correlation between human cerebrospinal fluid NOx and citrulline concentrations in order to determine the extent to which citrulline reflects NO synthesis and glutamatergic neurotransmission. METHODS Participants were patients with acute neurological diseases undergoing lumbar puncture (n=240). NOx and amino acid concentrations were determined by HPLC. RESULTS NOx concentrations did not vary significantly where infection (p=0,110) or inflammation (p=0,349) were present. Multiple regression analysis showed that NOx concentration was correlated with glutamine (r=-0,319, p<0,001) and citrulline concentrations (r=0,293, p=0,005) but not with the citrulline/arginine ratio (r=-0,160, p=0,173). ANCOVA confirmed that NOx concentration was correlated with citrulline concentration (F=7,6, p=0,007) but not with the citrulline/arginine ratio (F=2,2, p=0,136), or presence of infection (F=1,8, p=0,173) or inflammation (F=1,4, p=0,227). No association was found between NOx and arginine or glutamate concentrations. CONCLUSION The results suggest that CSF citrulline concentration reflects NOx synthesis to some extent, despite the contribution of other metabolic pathways. In addition, this study shows that glutamine is an important modulator of NO synthase activity, and that arginine and glutamate are not correlated with NOx.
Collapse
|
47
|
Liu Y, Buerk DG, Barbee KA, Jaron D. Nitric oxide release by deoxymyoglobin nitrite reduction during cardiac ischemia: A mathematical model. Microvasc Res 2017; 112:79-86. [PMID: 28363495 DOI: 10.1016/j.mvr.2017.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/16/2017] [Accepted: 03/19/2017] [Indexed: 12/19/2022]
Abstract
Interactions between cardiac myoglobin (Mb), nitrite, and nitric oxide (NO) are vital in regulating O2 storage, transport, and NO homeostasis. Production of NO through the reduction of endogenous myocardial nitrite by deoxygenated myoglobin has been shown to significantly reduce myocardial infarction damage and ischemic injury. We developed a mathematical model for a cardiac arteriole and surrounding myocardium to examine the hypothesis that myoglobin switches functions from being a strong NO scavenger to an NO producer via the deoxymyoglobin nitrite reductase pathway. Our results predict that under ischemic conditions of flow, blood oxygen level, and tissue pH, deoxyMb nitrite reduction significantly elevates tissue and smooth muscle cell NO. The size of the effect is consistent at different flow rates, increases with decreasing blood oxygen and tissue pH and, in extreme pathophysiological conditions, NO can even be elevated above the normoxic levels. Our simulations suggest that cardiac deoxyMb nitrite reduction is a plausible mechanism for preserving or enhancing NO levels using endogenous nitrite despite the rate-limiting O2 levels for endothelial NO production. This NO could then be responsible for mitigating deleterious effects under ischemic conditions.
Collapse
Affiliation(s)
- Yien Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Philadelphia, PA 19104, USA.
| |
Collapse
|
48
|
Pinheiro LC, Ferreira GC, Amaral JH, Portella RL, Tella SDOC, Passos MA, Tanus-Santos JE. Oral nitrite circumvents antiseptic mouthwash-induced disruption of enterosalivary circuit of nitrate and promotes nitrosation and blood pressure lowering effect. Free Radic Biol Med 2016; 101:226-235. [PMID: 27769921 DOI: 10.1016/j.freeradbiomed.2016.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/15/2016] [Accepted: 10/17/2016] [Indexed: 02/04/2023]
Abstract
The nitric oxide (NO•) metabolites nitrite and nitrate exert antihypertensive effects by mechanisms that involve gastric formation of S-nitrosothiols. However, while the use of antiseptic mouthwash (AM) is known to attenuate the responses to nitrate by disrupting its enterosalivary cycle, there is little information about whether AM attenuates the effects of orally administered nitrite. We hypothesized that the antihypertensive effects of orally administered nitrite would not be prevented by AM because, in contrast to oral nitrate, oral nitrite could promote S-nitrosothiols formation in the stomach without intereference by AM. Chronic effects of oral nitrite or nitrate were studied in two-kidney, one-clip (2K1C) hypertensive rats (and normotensive controls) treated with AM (or vehicle) once/day. We found that orally administered nitrite exerts antihypertensive effects that were not affected by AM. This finding contrasts with lack of antihypertensive responses to oral nitrate in 2K1C hypertensive rats treated with AM. Nitrite and nitrate treatments increased plasma nitrites, nitrates, and S-nitrosothiols concentrations. However, while treatment with AM attenuated the increases in plasma nitrite concentrations after both nitrite and nitrate treatments, AM attenuated the increases in S-nitrosothiols in nitrate-treated rats, but not in nitrite-treated rats. Moreover, AM attenuated vascular S-nitrosylation (detected by the SNO-RAC method) after nitrate, but not after nitrite treatment. Significant correlations were found between the hypotensive responses and S-nitrosothiols, and vascular S-nitrosylation levels. These results show for the first time that oral nitrite exerts antihypertensive effects notwithstanding the fact that antiseptic mouthwash disrupts the enterosalivary circulation of nitrate. Our results support a major role for S-nitrosothiols formation resulting in vascular S-nitrosylation as a key mechanism for the antihypertensive effects of both oral nitrite and nitrate.
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
| | - Jefferson H Amaral
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil
| | - Rafael L Portella
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil
| | - Sandra de O C Tella
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil
| | - Madla A Passos
- 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
|
49
|
Chirinos JA, Zamani P. The Nitrate-Nitrite-NO Pathway and Its Implications for Heart Failure and Preserved Ejection Fraction. Curr Heart Fail Rep 2016; 13:47-59. [PMID: 26792295 DOI: 10.1007/s11897-016-0277-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The pathogenesis of exercise intolerance in patients with heart failure and preserved ejection fraction (HFpEF) is likely multifactorial. In addition to cardiac abnormalities (diastolic dysfunction, abnormal contractile reserve, chronotropic incompetence), several peripheral abnormalities are likely to be involved. These include abnormal pulsatile hemodynamics, abnormal arterial vasodilatory responses to exercise, and abnormal peripheral O2 delivery, extraction, and utilization. The nitrate-nitrite-NO pathway is emerging as a potential target to modify key physiologic abnormalities, including late systolic left ventricular (LV) load from arterial wave reflections (which has deleterious short- and long-term consequences for the LV), arterial vasodilatory reserve, muscle O2 delivery, and skeletal muscle mitochondrial function. In a recently completed randomized trial, the administration of a single dose of exogenous inorganic nitrate has been shown to exert various salutary arterial hemodynamic effects, ultimately leading to enhanced aerobic capacity in patients with HFpEF. These effects have the potential for both immediate improvements in exercise tolerance and for long-term "disease-modifying" effects. In this review, we provide an overview of key mechanistic contributors to exercise intolerance in HFpEF, and of the potential therapeutic role of drugs that target the nitrate-nitrite-NO pathway.
Collapse
Affiliation(s)
- Julio A Chirinos
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Hospital of the University of Pennsylvania, Philadelphia, PA, USA. .,Ghent University, Ghent, Belgium.
| | - Payman Zamani
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Kupper C, Rees JA, Dechert S, DeBeer S, Meyer F. Complete Series of {FeNO}(8), {FeNO}(7), and {FeNO}(6) Complexes Stabilized by a Tetracarbene Macrocycle. J Am Chem Soc 2016; 138:7888-98. [PMID: 27191681 DOI: 10.1021/jacs.6b00584] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Use of a macrocyclic tetracarbene ligand, which is topologically reminiscent of tetrapyrrole macrocycles though electronically distinct, has allowed for the isolation, X-ray crystallographic characterization and comprehensive spectroscopic investigation of a complete set of {FeNO}(x) complexes (x = 6, 7, 8). Electrochemical reduction, or chemical reduction with CoCp2, of the {FeNO}(7) complex 1 leads to the organometallic {FeNO}(8) species 2. Its crystallographic structure determination is the first for a nonheme iron nitroxyl {FeNO}(8) and has allowed to identify structural trends among the series of {FeNO}(x) complexes. Combined experimental data including (57)Fe Mössbauer, IR, UV-vis-NIR, NMR and Kβ X-ray emission spectroscopies in concert with DFT calculations suggest a largely metal centered reduction of 1 to form the low spin (S = 0) {FeNO}(8) species 2. The very strong σ-donor character of the tetracarbene ligand imparts unusual properties and spectroscopic signatures such as low (57)Fe Mössbauer isomer shifts and linear Fe-N-O units with high IR stretching frequencies for the NO ligand. The observed metal-centered reduction leads to distinct reactivity patterns of the {FeNO}(8) species. In contrast to literature reported {FeNO}(8) complexes, 2 does not undergo NO protonation under strictly anaerobic conditions. Only in the presence of both dioxygen and protons is rapid and clean oxidation to the {FeNO}(7) complex 1 observed. While 1 is stable toward dioxygen, its reaction with dioxygen under NO atmosphere forms the {FeNO}(6)(ONO) complex 3 that features an unusual O-nitrito ligand trans to the NO. 3 is a rare example of a nonheme octahedral {FeNO}(6) complex. Its electrochemical or chemical reduction triggers dissociation of the O-nitrito ligand and sequential formation of the {FeNO}(7) and {FeNO}(8) compounds 1 and 2. A consistent electronic structure picture has been derived for these unique organometallic variants of the key bioinorganic {FeNO}(x) functional units.
Collapse
Affiliation(s)
- Claudia Kupper
- Institut für Anorganische Chemie, Georg-August-Universität , Tammannstrasse 4, D-37077 Göttingen, Germany
| | - Julian A Rees
- Max Planck Institute for Chemical Energy Conversion , Stiftstrasse 34-36, D-45470 Mülheim an der Ruhr, Germany.,Department of Chemistry, University of Washington , Box 351700, Seattle, Washington 98195-1700, United States
| | - Sebastian Dechert
- Institut für Anorganische Chemie, Georg-August-Universität , Tammannstrasse 4, D-37077 Göttingen, Germany
| | - Serena DeBeer
- Max Planck Institute for Chemical Energy Conversion , Stiftstrasse 34-36, D-45470 Mülheim an der Ruhr, Germany.,Department of Chemistry and Biochemistry, Cornell University , Ithaca, New York 14853, United States
| | - Franc Meyer
- Institut für Anorganische Chemie, Georg-August-Universität , Tammannstrasse 4, D-37077 Göttingen, Germany
| |
Collapse
|