1
|
Chiodi D, Ishihara Y. The role of the methoxy group in approved drugs. Eur J Med Chem 2024; 273:116364. [PMID: 38781921 DOI: 10.1016/j.ejmech.2024.116364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 05/25/2024]
Abstract
The methoxy substituent is prevalent in natural products and, consequently, is present in many natural product-derived drugs. It has also been installed in modern drug molecules with no remnant of natural product features because medicinal chemists have been taking advantage of the benefits that this small functional group can bestow on ligand-target binding, physicochemical properties, and ADME parameters. Herein, over 230 methoxy-containing small-molecule drugs, as well as several fluoromethoxy-containing drugs, are presented from the vantage point of the methoxy group. Biochemical mechanisms of action, medicinal chemistry SAR studies, and numerous X-ray cocrystal structures are analyzed to identify the precise role of the methoxy group for many of the drugs and drug classes. Although the methoxy substituent can be considered as the hybridization of a hydroxy and a methyl group, the combination of these functionalities often results in unique effects that can amount to more than the sum of the individual parts.
Collapse
Affiliation(s)
- Debora Chiodi
- Department of Chemistry, Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, CA, 92121, USA
| | - Yoshihiro Ishihara
- Department of Chemistry, Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
2
|
Walzik D, Wences Chirino TY, Zimmer P, Joisten N. Molecular insights of exercise therapy in disease prevention and treatment. Signal Transduct Target Ther 2024; 9:138. [PMID: 38806473 PMCID: PMC11133400 DOI: 10.1038/s41392-024-01841-0] [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: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Despite substantial evidence emphasizing the pleiotropic benefits of exercise for the prevention and treatment of various diseases, the underlying biological mechanisms have not been fully elucidated. Several exercise benefits have been attributed to signaling molecules that are released in response to exercise by different tissues such as skeletal muscle, cardiac muscle, adipose, and liver tissue. These signaling molecules, which are collectively termed exerkines, form a heterogenous group of bioactive substances, mediating inter-organ crosstalk as well as structural and functional tissue adaption. Numerous scientific endeavors have focused on identifying and characterizing new biological mediators with such properties. Additionally, some investigations have focused on the molecular targets of exerkines and the cellular signaling cascades that trigger adaption processes. A detailed understanding of the tissue-specific downstream effects of exerkines is crucial to harness the health-related benefits mediated by exercise and improve targeted exercise programs in health and disease. Herein, we review the current in vivo evidence on exerkine-induced signal transduction across multiple target tissues and highlight the preventive and therapeutic value of exerkine signaling in various diseases. By emphasizing different aspects of exerkine research, we provide a comprehensive overview of (i) the molecular underpinnings of exerkine secretion, (ii) the receptor-dependent and receptor-independent signaling cascades mediating tissue adaption, and (iii) the clinical implications of these mechanisms in disease prevention and treatment.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Tiffany Y Wences Chirino
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, 37075, Göttingen, Lower Saxony, Germany.
| |
Collapse
|
3
|
Liu X, Zhou H, Zhang H, Jin H, He Y. Advances in the research of sulfur dioxide and pulmonary hypertension. Front Pharmacol 2023; 14:1282403. [PMID: 37900169 PMCID: PMC10602757 DOI: 10.3389/fphar.2023.1282403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023] Open
Abstract
Pulmonary hypertension (PH) is a fatal disease caused by progressive pulmonary vascular remodeling (PVR). Currently, the mechanisms underlying the occurrence and progression of PVR remain unclear, and effective therapeutic approaches to reverse PVR and PH are lacking. Since the beginning of the 21st century, the endogenous sulfur dioxide (SO2)/aspartate transaminase system has emerged as a novel research focus in the fields of PH and PVR. As a gaseous signaling molecule, SO2 metabolism is tightly regulated in the pulmonary vasculature and is associated with the development of PH as it is involved in the regulation of pathological and physiological activities, such as pulmonary vascular cellular inflammation, proliferation and collagen metabolism, to exert a protective effect against PH. In this review, we present an overview of the studies conducted to date that have provided a theoretical basis for the development of SO2-related drug to inhibit or reverse PVR and effectively treat PH-related diseases.
Collapse
Affiliation(s)
- Xin Liu
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - He Zhou
- Departments of Medicine and Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Hongsheng Zhang
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yan He
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Tan R, Baranauskas MN, Karl ST, Ortiz de Zevallos J, Shei RJ, Paris HL, Wiggins CC, Bailey SJ. Effects of dietary nitrate supplementation on peak power output: Influence of supplementation strategy and population. Nitric Oxide 2023; 138-139:105-119. [PMID: 37438201 DOI: 10.1016/j.niox.2023.06.004] [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: 07/14/2023]
Abstract
Increasing evidence indicates that dietary nitrate supplementation has the potential to increase muscular power output during skeletal muscle contractions. However, there is still a paucity of data characterizing the impact of different nitrate dosing regimens on nitric oxide bioavailability and its potential ergogenic effects across various population groups. This review discusses the potential influence of different dietary nitrate supplementation strategies on nitric oxide bioavailability and muscular peak power output in healthy adults, athletes, older adults and some clinical populations. Effect sizes were calculated for peak power output and absolute and/or relative nitrate doses were considered where applicable. There was no relationship between the effect sizes of peak power output change following nitrate supplementation and when nitrate dosage when considered in absolute or relative terms. Areas for further research are also recommended including a focus on nitrate dosing regimens that optimize nitric oxide bioavailability for enhancing peak power at times of increased muscular work in a variety of healthy and disease populations.
Collapse
Affiliation(s)
- Rachel Tan
- Department of Sports Medicine, Pepperdine University, Malibu, CA, 90263, USA.
| | - Marissa N Baranauskas
- Department of Human Physiology & Nutrition, University of Colorado, Colorado Springs, CO, 80918, USA
| | - Sean T Karl
- Department of Sports Medicine, Pepperdine University, Malibu, CA, 90263, USA
| | | | - Ren-Jay Shei
- Indiana University Alumni Association, Indiana University, Bloomington, IN, 47408, USA
| | - Hunter L Paris
- Department of Sports Medicine, Pepperdine University, Malibu, CA, 90263, USA
| | - Chad C Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| |
Collapse
|
5
|
Tan R, Baranauskas MN, Karl ST, Ortiz de Zevallos J, Shei RJ, Paris HL, Wiggins CC, Bailey SJ. Effects of dietary nitrate supplementation on muscular power output: Influence of supplementation strategy and population. Nitric Oxide 2023:S1089-8603(23)00047-2. [PMID: 37244391 DOI: 10.1016/j.niox.2023.05.003] [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: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
Increasing evidence indicates that dietary nitrate supplementation has the potential to increase muscular power output during skeletal muscle contractions. However, there is still a paucity of data characterizing the impact of different nitrate dosing regimens on nitric oxide bioavailability its potential ergogenic effects across various population groups. This narrative review discusses the potential influence of different dietary nitrate supplementation strategies on nitric oxide bioavailability and muscular power output in healthy adults, athletes, older adults and some clinical populations. Areas for further research are also recommended including a focus individualized nitrate dosing regimens to optimize nitric oxide bioavailability and to promote muscular power enhancements in different populations.
Collapse
Affiliation(s)
- Rachel Tan
- Department of Sports Medicine, Pepperdine University, Malibu, CA, 90263, USA.
| | - Marissa N Baranauskas
- Department of Human Physiology & Nutrition, University of Colorado, Colorado Springs, CO, 80918, USA
| | - Sean T Karl
- Department of Sports Medicine, Pepperdine University, Malibu, CA, 90263, USA
| | | | - Ren-Jay Shei
- Indiana University Alumni Association, Indiana University, Bloomington, IN, 47408, USA
| | - Hunter L Paris
- Department of Sports Medicine, Pepperdine University, Malibu, CA, 90263, USA
| | - Chad C Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| |
Collapse
|
6
|
Piknova B, Woessner MN, de Zevallos JO, Kraus WE, VanBruggen MD, Schechter AN, Allen JD. Human skeletal muscle nitrate and nitrite in individuals with peripheral arterial disease: Effect of inorganic nitrate supplementation and exercise. Physiol Rep 2022; 10:e15531. [PMID: 36461652 PMCID: PMC9718944 DOI: 10.14814/phy2.15531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/28/2022] [Accepted: 11/11/2022] [Indexed: 05/15/2023] Open
Abstract
Skeletal muscle may act as a reservoir for N-oxides following inorganic nitrate supplementation. This idea is most intriguing in individuals with peripheral artery disease (PAD) who are unable to endogenously upregulate nitric oxide. This study analyzed plasma and skeletal muscle nitrate and nitrite concentrations along with exercise performance, prior to and following 12-weeks of exercise training combined with oral inorganic nitrate supplementation (EX+BR) or placebo (EX+PL) in participants with PAD. Non-supplemented, at baseline, there were no differences in plasma and muscle nitrate. For nitrite, muscle concentration was higher than plasma (+0.10 nmol.g-1 ). After 12 -weeks, acute oral nitrate increased both plasma and muscle nitrate (455.04 and 121.14 nmol.g-1 , p < 0.01), which were correlated (r = 0.63, p < 0.01), plasma nitrate increase was greater than in muscle (p < 0.01). Nitrite increased in the plasma (1.01 nmol.g-1 , p < 0.05) but not in the muscle (0.22 nmol.g-1 ) (p < 0.05 between compartments). Peak walk time (PWT) increased in both groups (PL + 257.6 s;BR + 315.0 s). Six-minute walk (6 MW) distance increased only in the (EX+BR) group (BR + 75.4 m). We report no substantial gradient of nitrate (or nitrite) from skeletal muscle to plasma, suggesting a lack of reservoir-like function in participants with PAD. Oral nitrate supplementation produced increases in skeletal muscle nitrate, but not skeletal muscle nitrite. The related changes in nitrate concentration between plasma and muscle suggests a potential for inter-compartmental nitrate "communication". Skeletal muscle did not appear to play a role in within compartment nitrate reduction. Muscle nitrate and nitrite concentrations did not appear to contribute to exercise performance in patients with PAD.
Collapse
Affiliation(s)
- Barbora Piknova
- Molecular Medicine Branch, NIDDKNational Institutes of HealthBethesdaMarylandUSA
| | - Mary N. Woessner
- Institute for Health and Sport (IHES)Victoria UniversityMelbourneAustralia
| | - Joaquin Ortiz de Zevallos
- Department of Kinesiology, School of Health and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - William E. Kraus
- Duke Molecular Physiology Institute, Duke University School of MedicineDurhamNorth CarolinaUSA
| | - Mitch D. VanBruggen
- Duke Molecular Physiology Institute, Duke University School of MedicineDurhamNorth CarolinaUSA
| | - Alan N. Schechter
- Molecular Medicine Branch, NIDDKNational Institutes of HealthBethesdaMarylandUSA
| | - Jason D. Allen
- Department of Kinesiology, School of Health and Human DevelopmentUniversity of VirginiaCharlottesvilleVirginiaUSA
- Division of Cardiovascular Medicine, School of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
7
|
Bryan NS. Nitric oxide deficiency is a primary driver of hypertension. Biochem Pharmacol 2022; 206:115325. [DOI: 10.1016/j.bcp.2022.115325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/08/2022]
|
8
|
Chen X, Chen L, Lin G, Wang Z, Kodali MC, Li M, Chen H, Lebovitz SG, Ortyl TC, Li L, Ismael S, Singh P, Malik KU, Ishrat T, Zhou FM, Zheng W, Liao FF. White matter damage as a consequence of vascular dysfunction in a spontaneous mouse model of chronic mild chronic hypoperfusion with eNOS deficiency. Mol Psychiatry 2022; 27:4754-4769. [PMID: 35948662 PMCID: PMC9734049 DOI: 10.1038/s41380-022-01701-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Vascular cognitive impairment and dementia (VCID) is the second most common form of dementia after Alzheimer's disease (AD). Currently, the mechanistic insights into the evolution and progression of VCID remain elusive. White matter change represents an invariant feature. Compelling clinical neuroimaging and pathological evidence suggest a link between white matter changes and neurodegeneration. Our prior study detected hypoperfused lesions in mice with partial deficiency of endothelial nitric oxide (eNOS) at very young age, precisely matching to those hypoperfused areas identified in preclinical AD patients. White matter tracts are particularly susceptible to the vascular damage induced by chronic hypoperfusion. Using immunohistochemistry, we detected severe demyelination in the middle-aged eNOS-deficient mice. The demyelinated areas were confined to cortical and subcortical areas including the corpus callosum and hippocampus. The intensity of demyelination correlated with behavioral deficits of gait and associative recognition memory performances. By Evans blue angiography, we detected blood-brain barrier (BBB) leakage as another early pathological change affecting frontal and parietal cortex in eNOS-deficient mice. Sodium nitrate fortified drinking water provided to young and middle-aged eNOS-deficient mice completely prevented non-perfusion, BBB leakage, and white matter pathology, indicating that impaired endothelium-derived NO signaling may have caused these pathological events. Furthermore, genome-wide transcriptomic analysis revealed altered gene clusters most related to mitochondrial respiratory pathways selectively in the white matter of young eNOS-deficient mice. Using eNOS-deficient mice, we identified BBB breakdown and hypoperfusion as the two earliest pathological events, resulting from insufficient vascular NO signaling. We speculate that the compromised BBB and mild chronic hypoperfusion trigger vascular damage, along with oxidative stress and astrogliosis, accounting for the white matter pathological changes in the eNOS-deficient mouse model. We conclude that eNOS-deficient mice represent an ideal spontaneous evolving model for studying the earliest events leading to white matter changes, which will be instrumental to future therapeutic testing of drug candidates and for targeting novel/specific vascular mechanisms contributing to VCID and AD.
Collapse
Affiliation(s)
- Xingyong Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, PR China
| | - Ling Chen
- Department of Cell Biology and Genetics, The school of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350001, PR China
| | - Geng Lin
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
- Teaching Center of Basic Medical Experiment, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Zhengjun Wang
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Mahesh C Kodali
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Mingqi Li
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Huimin Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Sarah G Lebovitz
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Tyler C Ortyl
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Lexiao Li
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Saifudeen Ismael
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Purnima Singh
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Kafait U Malik
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Fu-Ming Zhou
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Wei Zheng
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA.
- Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Francesca-Fang Liao
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA.
| |
Collapse
|
9
|
Skeletal muscle as a reservoir for nitrate and nitrite: The role of xanthine oxidase reductase (XOR). Nitric Oxide 2022; 129:102-109. [DOI: 10.1016/j.niox.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 09/16/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
|
10
|
Whitehead AK, Fried ND, Li Z, Neelamegam K, Pearson CS, LaPenna KB, Sharp TE, Lefer DJ, Lazartigues E, Gardner JD, Yue X. Alpha7 nicotinic acetylcholine receptor mediates chronic nicotine inhalation-induced cardiopulmonary dysfunction. Clin Sci (Lond) 2022; 136:973-987. [PMID: 35678315 PMCID: PMC10199464 DOI: 10.1042/cs20220083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/27/2022] [Accepted: 06/09/2022] [Indexed: 12/17/2022]
Abstract
Cigarette smoking remains the leading modifiable risk factor for cardiopulmonary diseases; however, the effects of nicotine alone on cardiopulmonary function remain largely unknown. Previously, we have shown that chronic nicotine vapor inhalation in mice leads to the development of pulmonary hypertension (PH) with right ventricular (RV) remodeling. The present study aims to further examine the cardiopulmonary effects of nicotine and the role of the α7 nicotinic acetylcholine receptor (α7-nAChR), which is widely expressed in the cardiovascular system. Wild-type (WT) and α7-nAChR knockout (α7-nAChR-/-) mice were exposed to room air (control) or nicotine vapor daily for 12 weeks. Consistent with our previous study, echocardiography and RV catheterization reveal that male WT mice developed increased RV systolic pressure with RV hypertrophy and dilatation following 12-week nicotine vapor exposure; in contrast, these changes were not observed in male α7-nAChR-/- mice. In addition, chronic nicotine inhalation failed to induce PH and RV remodeling in female mice regardless of genotype. The effects of nicotine on the vasculature were further examined in male mice. Our results show that chronic nicotine inhalation led to impaired acetylcholine-mediated vasodilatory response in both thoracic aortas and pulmonary arteries, and these effects were accompanied by altered endothelial nitric oxide synthase phosphorylation (enhanced inhibitory phosphorylation at threonine 495) and reduced plasma nitrite levels in WT but not α7-nAChR-/- mice. Finally, RNA sequencing revealed up-regulation of multiple inflammatory pathways in thoracic aortas from WT but not α7-nAChR-/- mice. We conclude that the α7-nAChR mediates chronic nicotine inhalation-induced PH, RV remodeling and vascular dysfunction.
Collapse
Affiliation(s)
- Anna K. Whitehead
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Nicholas D. Fried
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Zhen Li
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Kandasamy Neelamegam
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Charlotte S. Pearson
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Kyle B. LaPenna
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Thomas E. Sharp
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Department of Medicine Section of Cardiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - David J. Lefer
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
- Southeast Louisiana Veterans Health Care Systems, New Orleans, LA 70119, U.S.A
| | - Jason D. Gardner
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| | - Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, U.S.A
| |
Collapse
|
11
|
Bryan NS, Burleigh MC, Easton C. The oral microbiome, nitric oxide and exercise performance. Nitric Oxide 2022; 125-126:23-30. [PMID: 35636654 DOI: 10.1016/j.niox.2022.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 05/12/2022] [Accepted: 05/22/2022] [Indexed: 12/15/2022]
Abstract
The human microbiome comprises ∼1013-1014 microbial cells which form a symbiotic relationship with the host and play a critical role in the regulation of human metabolism. In the oral cavity, several species of bacteria are capable of reducing nitrate to nitrite; a key precursor of the signaling molecule nitric oxide. Nitric oxide has myriad physiological functions, which include the maintenance of cardiovascular homeostasis and the regulation of acute and chronic responses to exercise. This article provides a brief narrative review of the research that has explored how diversity and plasticity of the oral microbiome influences nitric oxide bioavailability and related physiological outcomes. There is unequivocal evidence that dysbiosis (e.g. through disease) or disruption (e.g. by use of antiseptic mouthwash or antibiotics) of the oral microbiota will suppress nitric oxide production via the nitrate-nitrite-nitric oxide pathway and negatively impact blood pressure. Conversely, there is preliminary evidence to suggest that proliferation of nitrate-reducing bacteria via the diet or targeted probiotics can augment nitric oxide production and improve markers of oral health. Despite this, it is yet to be established whether purposefully altering the oral microbiome can have a meaningful impact on exercise performance. Future research should determine whether alterations to the composition and metabolic activity of bacteria in the mouth influence the acute responses to exercise and the physiological adaptations to exercise training.
Collapse
Affiliation(s)
- Nathan S Bryan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Mia C Burleigh
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Blantyre, UK
| | - Chris Easton
- Institute for Clinical Exercise and Health Science, University of the West of Scotland, Blantyre, UK.
| |
Collapse
|
12
|
Goh CE, Bohn B, Marotz C, Molinsky R, Roy S, Paster BJ, Chen C, Rosenbaum M, Yuzefpolskaya M, Colombo PC, Desvarieux M, Papapanou PN, Jacobs DR, Knight R, Demmer RT. Nitrite Generating and Depleting Capacity of the Oral Microbiome and Cardiometabolic Risk: Results from ORIGINS. J Am Heart Assoc 2022; 11:e023038. [PMID: 35574962 PMCID: PMC9238569 DOI: 10.1161/jaha.121.023038] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background
The enterosalivary nitrate–nitrite–nitric oxide (NO
3
–NO
2
–NO) pathway generates NO following oral microbiota‐mediated production of salivary nitrite, potentially linking the oral microbiota to reduced cardiometabolic risk. Nitrite depletion by oral bacteria may also be important for determining the net nitrite available systemically. We examine if higher abundance of oral microbial genes favoring increased oral nitrite generation and decreased nitrite depletion is associated with a better cardiometabolic profile cross‐sectionally.
Methods and Results
This study includes 764 adults (mean [SD] age 32 [9] years, 71% women) enrolled in ORIGINS (Oral Infections, Glucose Intolerance, and Insulin Resistance Study). Microbial DNA from subgingival dental plaques underwent 16S rRNA gene sequencing; PICRUSt2 was used to estimate functional gene profiles. To represent the different components and pathways of nitrogen metabolism in bacteria, predicted gene abundances were operationalized to create summary scores by (1) bacterial nitrogen metabolic pathway or (2) biochemical product (NO
2
, NO, or ammonia [NH
3
]) formed by the action of the bacterial reductases encoded. Finally, nitrite generation‐to‐depletion ratios of gene abundances were created from the above summary scores. A composite cardiometabolic
Z
score was created from cardiometabolic risk variables, with higher scores associated with worse cardiometabolic health. We performed multivariable linear regression analysis with cardiometabolic
Z
score as the outcome and the gene abundance summary scores and ratios as predictor variables, adjusting for sex, age, race, and ethnicity in the simple adjusted model. A 1 SD higher NO versus NH
3
summary ratio was inversely associated with a −0.10 (false discovery rate
q
=0.003) lower composite cardiometabolic
Z
score in simple adjusted models. Higher NH
3
summary score (suggestive of nitrite depletion) was associated with higher cardiometabolic risk, with a 0.06 (false discovery rate
q
=0.04) higher composite cardiometabolic
Z
score.
Conclusions
Increased net capacity for nitrite generation versus depletion by oral bacteria, assessed through a metagenome estimation approach, is associated with lower levels of cardiometabolic risk.
Collapse
Affiliation(s)
- Charlene E. Goh
- Faculty of DentistryNational University of SingaporeSingapore
| | - Bruno Bohn
- Division of Epidemiology and Community HealthSchool of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Clarisse Marotz
- Department of PediatricsUniversity of California San DiegoLa JollaCA
| | - Rebecca Molinsky
- Division of Epidemiology and Community HealthSchool of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Sumith Roy
- Department of EpidemiologyMailman School of Public HealthColumbia UniversityNew YorkNY
| | - Bruce J. Paster
- The Forsyth InstituteCambridgeMA
- Department of Oral Medicine, Infection, and ImmunityHarvard School of Dental MedicineBostonMA
| | - Ching‐Yuan Chen
- Division of PeriodonticsSection of Oral, Diagnostic and Rehabilitation SciencesCollege of Dental MedicineColumbia UniversityNew YorkNY
| | - Michael Rosenbaum
- Division of Molecular GeneticsDepartments of Pediatrics and MedicineColumbia UniversityNew YorkNY
| | - Melana Yuzefpolskaya
- Division of CardiologyDepartment of MedicineNew York Presbyterian HospitalColumbia UniversityNew YorkNY
| | - Paolo C. Colombo
- Division of CardiologyDepartment of MedicineNew York Presbyterian HospitalColumbia UniversityNew YorkNY
| | - Moïse Desvarieux
- Department of EpidemiologyMailman School of Public HealthColumbia UniversityNew YorkNY
- INSERM UMR 1153Centre de Recherche Epidemiologie et Statistique Paris Sorbonne Cité (CRESS)METHODS CoreParisFrance
| | - Panos N. Papapanou
- Division of PeriodonticsSection of Oral, Diagnostic and Rehabilitation SciencesCollege of Dental MedicineColumbia UniversityNew YorkNY
| | - David R. Jacobs
- Division of Epidemiology and Community HealthSchool of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Rob Knight
- Department of Computer Science & EngineeringJacobs School of EngineeringUniversity of California San DiegoLa JollaCA
- Department of BioengineeringUniversity of California San DiegoLa JollaCA
- Center for Microbiome InnovationUniversity of California San DiegoLa JollaCA
| | - Ryan T. Demmer
- Division of Epidemiology and Community HealthSchool of Public HealthUniversity of MinnesotaMinneapolisMN
- Department of EpidemiologyMailman School of Public HealthColumbia UniversityNew YorkNY
| |
Collapse
|
13
|
Kim F, Maynard C, Dezfulian C, Sayre M, Kudenchuk P, Rea T, Sampson D, Olsufka M, May S, Nichol G. Effect of Out-of-Hospital Sodium Nitrite on Survival to Hospital Admission After Cardiac Arrest: A Randomized Clinical Trial. JAMA 2021; 325:138-145. [PMID: 33433575 PMCID: PMC7804921 DOI: 10.1001/jama.2020.24326] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IMPORTANCE Therapeutic delivery of sodium nitrite during resuscitation improved survival in animal models of cardiac arrest, but efficacy has not been evaluated in clinical trials in humans. OBJECTIVE To determine whether parenteral administration of sodium nitrite given by paramedics during resuscitation for out-of-hospital cardiac arrest improved survival to hospital admission. DESIGN, SETTING, AND PARTICIPANTS Double-blind, placebo-controlled, phase 2 randomized clinical trial including 1502 adults in King County, Washington, with out-of-hospital cardiac arrest from ventricular fibrillation or nonventricular fibrillation. Patients underwent resuscitation by paramedics and were enrolled between February 8, 2018, and August 19, 2019; follow-up and data abstraction were completed by December 31, 2019. INTERVENTIONS Eligible patients with out-of-hospital cardiac arrest were randomized (1:1:1) to receive 45 mg of sodium nitrite (n = 500), 60 mg of sodium nitrite (n = 498), or placebo (n = 499), which was given via bolus injection by the paramedics as soon as possible during active resuscitation. MAIN OUTCOMES AND MEASURES The primary outcome was survival to hospital admission and was evaluated with 1-sided hypothesis testing. The secondary outcomes included out-of-hospital variables (rate of return of spontaneous circulation, rate of rearrest, and use of norepinephrine to support blood pressure) and in-hospital variables (survival to hospital discharge; neurological outcomes at hospital discharge; cumulative survival to 24 hours, 48 hours, and 72 hours; and number of days in the intensive care unit). RESULTS Among 1502 patients with out-of-hospital cardiac arrest who were randomized (mean age, 64 years [SD, 17 years]; 34% were women), 99% completed the trial. Overall, 205 patients (41%) in the 45 mg of sodium nitrite group and 212 patients (43%) in the 60 mg of sodium nitrite group compared with 218 patients (44%) in the placebo group survived to hospital admission; the mean difference for the 45-mg dose vs placebo was -2.9% (1-sided 95% CI, -8.0% to ∞; P = .82) and the mean difference for the 60-mg dose vs placebo was -1.3% (1-sided 95% CI, -6.5% to ∞; P = .66). None of the 7 prespecified secondary outcomes were significantly different, including survival to hospital discharge for 66 patients (13.2%) in the 45 mg of sodium nitrite group and 72 patients (14.5%) in the 60 mg of sodium nitrite group compared with 74 patients (14.9%) in the placebo group; the mean difference for the 45-mg dose vs placebo was -1.7% (2-sided 95% CI, -6.0% to 2.6%; P = .44) and the mean difference for the 60-mg dose vs placebo was -0.4% (2-sided 95% CI, -4.9% to 4.0%; P = .85). CONCLUSIONS AND RELEVANCE Among patients with out-of-hospital cardiac arrest, administration of sodium nitrite, compared with placebo, did not significantly improve survival to hospital admission. These findings do not support the use of sodium nitrite during resuscitation from out-of-hospital cardiac arrest. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03452917.
Collapse
Affiliation(s)
- Francis Kim
- Department of Medicine, School of Medicine, University of Washington, Seattle
| | - Charles Maynard
- Department of Health Services, School of Public Health, University of Washington, Seattle
| | - Cameron Dezfulian
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael Sayre
- Department of Emergency Medicine, School of Medicine, University of Washington, Seattle
| | - Peter Kudenchuk
- Department of Medicine, School of Medicine, University of Washington, Seattle
| | - Thomas Rea
- Department of Medicine, School of Medicine, University of Washington, Seattle
| | - Deborah Sampson
- Department of Medicine, School of Medicine, University of Washington, Seattle
| | - Michele Olsufka
- Department of Medicine, School of Medicine, University of Washington, Seattle
| | - Susanne May
- Department of Biostatistics, School of Public Health, University of Washington, Seattle
| | - Graham Nichol
- Department of Medicine, School of Medicine, University of Washington, Seattle
- Department of Emergency Medicine, School of Medicine, University of Washington, Seattle
| |
Collapse
|
14
|
Kapil V, Khambata RS, Jones DA, Rathod K, Primus C, Massimo G, Fukuto JM, Ahluwalia A. The Noncanonical Pathway for In Vivo Nitric Oxide Generation: The Nitrate-Nitrite-Nitric Oxide Pathway. Pharmacol Rev 2020; 72:692-766. [PMID: 32576603 DOI: 10.1124/pr.120.019240] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
In contrast to nitric oxide, which has well established and important roles in the regulation of blood flow and thrombosis, neurotransmission, the normal functioning of the genitourinary system, and the inflammation response and host defense, its oxidized metabolites nitrite and nitrate have, until recently, been considered to be relatively inactive. However, this view has been radically revised over the past decade and more. Much evidence has now accumulated demonstrating that nitrite serves as a storage form of nitric oxide, releasing nitric oxide preferentially under acidic and/or hypoxic conditions but also occurring under physiologic conditions: a phenomenon that is catalyzed by a number of distinct mammalian nitrite reductases. Importantly, preclinical studies demonstrate that reduction of nitrite to nitric oxide results in a number of beneficial effects, including vasodilatation of blood vessels and lowering of blood pressure, as well as cytoprotective effects that limit the extent of damage caused by an ischemia/reperfusion insult, with this latter issue having been translated more recently to the clinical setting. In addition, research has demonstrated that the other main metabolite of the oxidation of nitric oxide (i.e., nitrate) can also be sequentially reduced through processing in vivo to nitrite and then nitrite to nitric oxide to exert a range of beneficial effects-most notably lowering of blood pressure, a phenomenon that has also been confirmed recently to be an effective method for blood pressure lowering in patients with hypertension. This review will provide a detailed description of the pathways involved in the bioactivation of both nitrate and nitrite in vivo, their functional effects in preclinical models, and their mechanisms of action, as well as a discussion of translational exploration of this pathway in diverse disease states characterized by deficiencies in bioavailable nitric oxide. SIGNIFICANCE STATEMENT: The past 15 years has seen a major revision in our understanding of the pathways for nitric oxide synthesis in the body with the discovery of the noncanonical pathway for nitric oxide generation known as the nitrate-nitrite-nitric oxide pathway. This review describes the molecular components of this pathway, its role in physiology, potential therapeutics of targeting this pathway, and their impact in experimental models, as well as the clinical translation (past and future) and potential side effects.
Collapse
Affiliation(s)
- V Kapil
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - R S Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - D A Jones
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - K Rathod
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - C Primus
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - G Massimo
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - J M Fukuto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - A Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| |
Collapse
|
15
|
Bahadoran Z, Carlström M, Mirmiran P, Ghasemi A. Nitric oxide: To be or not to be an endocrine hormone? Acta Physiol (Oxf) 2020; 229:e13443. [PMID: 31944587 DOI: 10.1111/apha.13443] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 01/05/2020] [Accepted: 01/10/2020] [Indexed: 01/02/2023]
Abstract
Nitric oxide (NO), a highly reactive gasotransmitter, is critical for a number of cellular processes and has multiple biological functions. Due to its limited lifetime and diffusion distance, NO has been mainly believed to act in autocrine/paracrine fashion. The increasingly recognized effects of pharmacologically delivered and endogenous NO at a distant site have changed the conventional wisdom and introduced NO as an endocrine signalling molecule. The notion is greatly supported by the detection of a number of NO adducts and their circulatory cycles, which in turn contribute to the transport and delivery of NO bioactivity, remote from the sites of its synthesis. The existence of endocrine sites of synthesis, negative feedback regulation of biosynthesis, integrated storage and transport systems, having an exclusive receptor, that is, soluble guanylyl cyclase (sGC), and organized circadian rhythmicity make NO something beyond a simple autocrine/paracrine signalling molecule that could qualify for being an endocrine signalling molecule. Here, we discuss hormonal features of NO from the classical endocrine point of view and review available knowledge supporting NO as a true endocrine hormone. This new insight can provide a new framework within which to reinterpret NO biology and its clinical applications.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center Research Institute for Endocrine Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mattias Carlström
- Department of Physiology and Pharmacology Karolinska Institutet Stockholm Sweden
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics Faculty of Nutrition Sciences and Food Technology National Nutrition and Food Technology Research Institute Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center Research Institute for Endocrine Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
16
|
Bryan NS, Lefer DJ. Update on Gaseous Signaling Molecules Nitric Oxide and Hydrogen Sulfide: Strategies to Capture their Functional Activity for Human Therapeutics. Mol Pharmacol 2019; 96:109-114. [PMID: 31061006 PMCID: PMC6592147 DOI: 10.1124/mol.118.113910] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 05/02/2019] [Indexed: 11/22/2022] Open
Abstract
Discovery of the production of gaseous molecules, such as nitric oxide and hydrogen sulfide, within the human body began a new concept in cellular signaling. Over the past 30 years, these molecules have been investigated and found to have extremely important beneficial effects in numerous chronic diseases. Gaseous signaling molecules that diffuse in three dimensions apparently contradict the selectivity and specificity afforded by normal ligand receptor binding and activation. This new concept has also created hurdles in the development of safe and efficacious drug therapy based on these molecules. Mechanisms involving formation of more stable intermediates and second messengers allow for new strategies for safe and effective delivery of these molecules for human disease. The purpose of this review is to highlight the biologic effects of nitric oxide and hydrogen sulfide, their seemingly indistinguishable effects, and how these molecules can be safely harnessed for drug development and precursors or substrates administered for human consumption through applied physiology.
Collapse
Affiliation(s)
- Nathan S Bryan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (N.S.B.); and Louisiana State University School of Medicine, New Orleans, Louisiana (D.J.L.)
| | - David J Lefer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas (N.S.B.); and Louisiana State University School of Medicine, New Orleans, Louisiana (D.J.L.)
| |
Collapse
|
17
|
Affiliation(s)
- Nathan S. Bryan
- Department of Molecular and Human GeneticsBaylor College of Medicine One Baylor Plaza Alkek Building for Biomedical Research R-850 Houston TX 77030
| |
Collapse
|
18
|
Tribble GD, Angelov N, Weltman R, Wang BY, Eswaran SV, Gay IC, Parthasarathy K, Dao DHV, Richardson KN, Ismail NM, Sharina IG, Hyde ER, Ajami NJ, Petrosino JF, Bryan NS. Frequency of Tongue Cleaning Impacts the Human Tongue Microbiome Composition and Enterosalivary Circulation of Nitrate. Front Cell Infect Microbiol 2019; 9:39. [PMID: 30881924 PMCID: PMC6406172 DOI: 10.3389/fcimb.2019.00039] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/07/2019] [Indexed: 01/25/2023] Open
Abstract
The oral microbiome has the potential to provide an important symbiotic function in human blood pressure physiology by contributing to the generation of nitric oxide (NO), an essential cardiovascular signaling molecule. NO is produced by the human body via conversion of arginine to NO by endogenous nitric oxide synthase (eNOS) but eNOS activity varies by subject. Oral microbial communities are proposed to supplement host NO production by reducing dietary nitrate to nitrite via bacterial nitrate reductases. Unreduced dietary nitrate is delivered to the oral cavity in saliva, a physiological process termed the enterosalivary circulation of nitrate. Previous studies demonstrated that disruption of enterosalivary circulation via use of oral antiseptics resulted in increases in systolic blood pressure. These previous studies did not include detailed information on the oral health of enrolled subjects. Using 16S rRNA gene sequencing and analysis, we determined whether introduction of chlorhexidine antiseptic mouthwash for 1 week was associated with changes in tongue bacterial communities and resting systolic blood pressure in healthy normotensive individuals with documented oral hygiene behaviors and free of oral disease. Tongue cleaning frequency was a predictor of chlorhexidine-induced changes in systolic blood pressure and tongue microbiome composition. Twice-daily chlorhexidine usage was associated with a significant increase in systolic blood pressure after 1 week of use and recovery from use resulted in an enrichment in nitrate-reducing bacteria on the tongue. Individuals with relatively high levels of bacterial nitrite reductases had lower resting systolic blood pressure. These results further support the concept of a symbiotic oral microbiome contributing to human health via the enterosalivary nitrate-nitrite-NO pathway. These data suggest that management of the tongue microbiome by regular cleaning together with adequate dietary intake of nitrate provide an opportunity for the improvement of resting systolic blood pressure.
Collapse
Affiliation(s)
- Gena D. Tribble
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Nikola Angelov
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Robin Weltman
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Bing-Yan Wang
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Sridhar V. Eswaran
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Isabel C. Gay
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Kavitha Parthasarathy
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Doan-Hieu V. Dao
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Katherine N. Richardson
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Nadia M. Ismail
- Department of Periodontics, School of Dentistry, The University of Texas Health Science Center Houston, Houston, TX, United States
| | - Iraida G. Sharina
- Division of Cardiology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center Houston, Houston, TX, United States
| | | | - Nadim J. Ajami
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX, United States
| | - Joseph F. Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of MedicineHouston, TX, United States
| | - Nathan S. Bryan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
19
|
Rushing AM, Donnarumma E, Polhemus DJ, Au KR, Victoria SE, Schumacher JD, Li Z, Jenkins JS, Lefer DJ, Goodchild TT. Effects of a novel hydrogen sulfide prodrug in a porcine model of acute limb ischemia. J Vasc Surg 2019; 69:1924-1935. [PMID: 30777693 DOI: 10.1016/j.jvs.2018.08.172] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 08/07/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Previous studies have shown that hydrogen sulfide (H2S) exerts potent proangiogenic properties under in vitro conditions and in rodent models. We sought to determine whether a novel H2S prodrug promotes peripheral revascularization in a swine model of acute limb ischemia (ALI). METHODS ALI was induced in 17 female miniswine via intravascular occlusion of the external iliac. At day 7 after ALI induction, miniswine (n = 17) were randomized to received placebo or the H2S prodrug, SG-1002 (800 mg per os twice a day), for 35 days. At day 35 SG-1002 increased circulating levels of H2S (5.0 ± 1.2 μmol/L vs 1.8 ± 0.50 μmol/L; P < .05), sulfane sulfur (10.6 ± 2.3 μmol/L vs 2.6 ± 0.8 μmol/L; P < .05), and nitrite (0.5 ± 0.05 μmol/L vs 0.3 ± 0.03 μmol/L; P < .005) compared with placebo. SG-1002 therapy increased angiographic scoring in ischemic limb vessel number (27.6 ± 1.6 vs 22.2 ± 1.8; P < .05) compared with placebo. Treatment with SG-1002 preserved existing capillaries in ischemic limbs (128.3 ± 18.7 capillaries/mm2 vs 79.0 ± 9.8 capillaries/mm2; P < .05) compared with placebo. Interestingly, treatment with SG-1002 also improved coronary vasorelaxation responses to bradykinin and substance P in miniswine with ALI. CONCLUSIONS Our results suggest that daily administration of the H2S prodrug, SG-1002, leads to an increase in circulating H2S and nitric oxide signaling and preserves vessel number and density in ischemic limbs. Furthermore, SG-1002 therapy improved endothelial-dependent coronary artery vasorelaxation in the setting of ALI. Our data demonstrate that SG-1002 preserves the vascular architecture in ischemic limbs and exerts vascular protective effects in the coronary vasculature in a model of peripheral vascular disease.
Collapse
Affiliation(s)
- Amanda M Rushing
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - Erminia Donnarumma
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - David J Polhemus
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La; Department of Pharmacology, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - Kevin R Au
- Department of Vascular Surgery, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - Samuel E Victoria
- Department of Vascular Surgery, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - Jeffrey D Schumacher
- Department of Animal Care, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - Zhen Li
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La; Department of Pharmacology, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - J Stephen Jenkins
- Heart and Vascular Institute, Ochsner Medical Center, New Orleans, La
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La; Department of Pharmacology, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La
| | - Traci T Goodchild
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La; Department of Pharmacology, Louisiana State University Health Sciences Center-New Orleans, New Orleans, La.
| |
Collapse
|
20
|
Tolaymat Y, Doré S, Griffin HW, Shih S, Edwards ME, Weiss MD. Inhaled Gases for Neuroprotection of Neonates: A Review. Front Pediatr 2019; 7:558. [PMID: 32047729 PMCID: PMC6996209 DOI: 10.3389/fped.2019.00558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/20/2019] [Indexed: 11/30/2022] Open
Abstract
Importance: Hypoxic-ischemic encephalopathy (HIE) is a significant cause of morbidity and mortality in neonates. The incidence of HIE is 1-8 per 1,000 live births in developed countries. Whole-body hypothermia reduces the risk of disability or death, but 7 infants needed to be treated to prevent death or major neurodevelopmental disability. Inhalational gases may be promising synergistic agents due to their rapid onset and easy titratability. Objective: To review current data on different inhaled gases with neuroprotective properties that may serve as adjunct therapies to hypothermia. Evidence review: Literature review was performed using the PubMed database, google scholar, and ClinicalTrials.Gov. Results focused on articles published from January 1, 2005, through December 31, 2017. Articles published earlier than 2005 were included when appropriate for historical perspective. Our review emphasized preclinical and clinical studies relevant to the use of inhaled agents for neuroprotection. Findings: Based on the relevance to our topic, 111 articles were selected pertaining to the incidence of HIE, pathophysiology of HIE, therapeutic hypothermia, and emerging therapies for hypoxic-ischemic encephalopathy in preclinical and clinical settings. Supplemental tables summarizes highly relevant 49 publications that were included in this review. The selected publications emphasize the emergence of promising inhaled gases that may improve neurologic survival and alleviate neurodevelopmental disability when combined with therapeutic hypothermia in the future. Conclusions: Many inhaled agents have neuroprotective properties and could serve as an adjunct therapy to whole-body hypothermia. Inhaled agents are ideal due to their easy administration, titrability, and rapid onset and offset.
Collapse
Affiliation(s)
- Youness Tolaymat
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Sylvain Doré
- Departments of Neurology, Psychiatry, Pharmaceuticals and Neuroscience, University of Florida, Gainesville, FL, United States
| | - Hudson W Griffin
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Susana Shih
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Mary E Edwards
- Health Science Center Libraries, University of Florida, Gainesville, FL, United States
| | - Michael D Weiss
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
21
|
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
|
22
|
Kim F, Dezfulian C, Empey PE, Morrell M, Olsufka M, Scruggs S, Kudenchuk P, May S, Maynard C, Sayre MR, Nichol G. Usefulness of Intravenous Sodium Nitrite During Resuscitation for the Treatment of Out-of-Hospital Cardiac Arrest. Am J Cardiol 2018; 122:554-559. [PMID: 30205886 DOI: 10.1016/j.amjcard.2018.04.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 04/24/2018] [Accepted: 04/30/2018] [Indexed: 01/02/2023]
Abstract
It is hypothesized that intravenous (IV) sodium nitrite given during resuscitation of out-of-hospital cardiac arrest (OHCA) will improve survival. We performed a phase 1 open-label study of IV sodium nitrite given during resuscitation of 120 patents with OHCA from ventricular fibrillation or nonventricular fibrillation initial rhythms by Seattle Fire Department paramedics. A total of 59 patients received 25 mg (low) and 61 patients received 60 mg (high) of sodium nitrite during resuscitation from OHCA. Treatment effects were compared between high- and low-dose nitrite groups, and all patients in a concurrent local Emergency Medical Services registry of OHCA. Whole blood nitrite levels were measured in 97 patients. The rate of return of spontaneous circulation (48% vs 49%), rearrest in the field (15% vs 25%), use of norepinephrine (12% vs 12%), first systolic blood pressure (124 ± 32 vs 125 ± 38 mm Hg), survival to discharge (23.7% vs 16.4%), and neurologically favorable survival (18.6% vs 11.5%) were not significantly different in the low and high nitrite groups. There were no significant differences in these outcomes among patients who received IV nitrite compared with concurrent registry controls. We estimate that 60 mg achieves whole blood nitrite levels of 22 to 38 μM 10 minutes after administration, whereas 25 mg achieves a level of 9 to 16 μM 10 minutes after delivery. In conclusion, administration of IV nitrite is feasible and appears to be safe in patients with OHCA, permitting subsequent evaluation of the effectiveness of IV nitrite for the treatment of OHCA.
Collapse
Affiliation(s)
- Francis Kim
- Department of Medicine, Harborview Medical Center, University of Washington, Seattle, Washington.
| | - Cameron Dezfulian
- Department of Critical Care Medicine, Safar Center for Resuscitation Research and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Philip E Empey
- Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew Morrell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michele Olsufka
- Department of Medicine, Harborview Medical Center, University of Washington, Seattle, Washington
| | - Sue Scruggs
- Department of Medicine, Harborview Medical Center, University of Washington, Seattle, Washington
| | - Peter Kudenchuk
- Department of Medicine, Harborview Medical Center, University of Washington, Seattle, Washington
| | - Susanne May
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Charles Maynard
- Department of Health Services, University of Washington, Seattle, Washington
| | - Michael R Sayre
- Department of Emergency Medicine, Harborview Medical Center, University of Washington, Seattle, Washington
| | - Graham Nichol
- Department of Medicine, Harborview Medical Center, University of Washington, Seattle, Washington; Department of Emergency Medicine, Harborview Medical Center, University of Washington, Seattle, Washington
| |
Collapse
|
23
|
Imbrogno S, Filice M, Cerra MC, Gattuso A. NO, CO and H 2 S: What about gasotransmitters in fish and amphibian heart? Acta Physiol (Oxf) 2018; 223:e13035. [PMID: 29338122 DOI: 10.1111/apha.13035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 12/25/2022]
Abstract
The gasotransmitters nitric oxide (NO), carbon monoxide (CO), and hydrogen sulphide (H2 S), long considered only toxicant, are produced in vivo during the catabolism of common biological molecules and are crucial for a large variety of physiological processes. Mounting evidence is emerging that in poikilotherm vertebrates, as in mammals, they modulate the basal performance of the heart and the response to stress challenges. In this review, we will focus on teleost fish and amphibians to highlight the evolutionary importance in vertebrates of the cardiac control elicited by NO, CO and H2 S, and the conservation of the intracellular cascades they activate. Although many gaps are still present due to discontinuous information, we will use examples obtained by studies from our and other laboratories to illustrate the complexity of the mechanisms that, by involving gasotransmitters, allow beat-to-beat, short-, medium- and long-term cardiac homoeostasis. By presenting the latest data, we will also provide a framework in which the peculiar morpho-functional arrangement of the teleost and amphibian heart can be considered as a reference tool to decipher cardiac regulatory networks which are difficult to explore using more conventional vertebrates, such as mammals.
Collapse
Affiliation(s)
- S. Imbrogno
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - M. Filice
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - M. C. Cerra
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| | - A. Gattuso
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende; Italy
| |
Collapse
|
24
|
Wang D, Gao Q, Wang T, Zhao G, Qian F, Huang J, Wang H, Zhang X, Wang Y. Green tea infusion protects against alcoholic liver injury by attenuating inflammation and regulating the PI3K/Akt/eNOS pathway in C57BL/6 mice. Food Funct 2018; 8:3165-3177. [PMID: 28782772 DOI: 10.1039/c7fo00791d] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Alcohol intake is a major risk factor for the pathogenesis of alcoholic liver diseases. Accumulating evidence suggests that green tea protects against alcoholic liver injury; however, the underlying mechanisms remain unclear. The present study investigated the role of endothelial nitric oxide synthase (eNOS) in the protective effects of green tea against alcohol-induced liver injury and inflammation. Ethanol was intragastrically administered to male C57BL/6 mice once a day, and the mice were allowed free access to green tea infusion or water for two weeks. We assessed the plasma levels of alanine aminotransferase and aspartate aminotransferase, hepatic contents of thiobarbituric acid reactive substances, malondialdehyde and triglyceride and hepatic mRNA expression of pro-inflammatory cytokines (interleukin-1β, tumor necrosis factor-α, and interleukin-6). Our results showed that compared with water alone, green tea infusion markedly reduced liver damage, hepatic oxidative stress, hepatic lipid accumulation and inflammatory response. Green tea infusion also significantly reduced hepatic nuclear factor-κB expression and its downstream inflammatory mediators (inducible nitric oxide synthase and cyclooxygenase-2) mRNA levels in ethanol-treated mice. Additionally, green tea infusion significantly activated hepatic phosphorylated phosphatidylinositol 3-kinase (PI3K) and phosphorylated protein kinase B (Akt), which are associated with the upregulation of phosphorylated eNOS expression and the increase of plasma nitric oxide levels in ethanol-treated mice. Furthermore, the protective effects of green tea infusion were considerably inhibited by the eNOS inhibitor NG-nitro-l-arginine methyl ester in ethanol-treated mice. In conclusion, our study demonstrated that the protective effects of green tea infusion on alcohol-induced liver injury and inflammation involve the modulation of the PI3K/AKT/eNOS pathway.
Collapse
Affiliation(s)
- Dongxu Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science & Technology, Anhui Agricultural University, Hefei, Anhui, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Dezfulian C, Taft M, Corey C, Hill G, Krehel N, Rittenberger JC, Guyette FX, Shiva S. Biochemical signaling by remote ischemic conditioning of the arm versus thigh: Is one raise of the cuff enough? Redox Biol 2017; 12:491-498. [PMID: 28334684 PMCID: PMC5362138 DOI: 10.1016/j.redox.2017.03.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 03/08/2017] [Indexed: 01/23/2023] Open
Abstract
Remote Ischemic Conditioning (RIC), induced by brief cycles of ischemia and reperfusion, protects vital organs from a prolonged ischemic insult. While several biochemical mediators have been implicated in RIC's mechanism of action, it remains unclear whether the localization or “dose” of RIC affects the extent of protective signaling. In this randomized crossover study of healthy individuals, we tested whether the number of cycles of RIC and its localization (arm versus thigh) determines biochemical signaling and cytoprotection. Subjects received either arm or thigh RIC and then were crossed over to receive RIC in the other extremity. Blood flow, tissue perfusion, concentrations of the circulating protective mediator nitrite, and platelet mitochondrial function were measured after each RIC cycle. We found that plasma nitrite concentration peaked after the first RIC cycle and remained elevated throughout RIC. This plasma nitrite conferred cytoprotection in an in vitro myocyte model of hypoxia/reoxygenation. Notably, though plasma nitrite returned to baseline at 24 h, RIC conditioned plasma still mediated protection. Additionally, no difference in endpoints between RIC in thigh versus arm was found. These data demonstrate that localization and “dose” of RIC does not affect cytoprotection and further elucidate the mechanisms by which nitrite contributes to RIC-dependent protection.
Collapse
Affiliation(s)
- Cameron Dezfulian
- Safar Center for Resuscitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Maia Taft
- Safar Center for Resuscitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Catherine Corey
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Gabrielle Hill
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Nicholas Krehel
- Safar Center for Resuscitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Jon C Rittenberger
- Applied Physiology Laboratory, Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Frank X Guyette
- Applied Physiology Laboratory, Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Applied Physiology Laboratory, Department of Emergency Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
26
|
Jones DA, Rathod KS, Ahluwalia A. Update on Nitrite Reduction in Ischemic Disease: Mechanisms and Clinical Translation. NITRIC OXIDE 2017:195-211. [DOI: 10.1016/b978-0-12-804273-1.00015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
27
|
|
28
|
Yuan S, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Remodeling. Microcirculation 2016; 23:134-45. [PMID: 26381654 DOI: 10.1111/micc.12248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 09/13/2015] [Indexed: 12/22/2022]
Abstract
Blockage or restriction of blood flow through conduit arteries results in tissue ischemia downstream of the disturbed area. Local tissues can adapt to this challenge by stimulating vascular remodeling through angiogenesis and arteriogenesis thereby restoring blood perfusion and removal of wastes. Multiple molecular mechanisms of vascular remodeling during ischemia have been identified and extensively studied. However, therapeutic benefits from these findings and insights are limited due to the complexity of various signaling networks and a lack of understanding central metabolic regulators governing these responses. The gasotransmitters NO and H2 S have emerged as master regulators that influence multiple molecular targets necessary for ischemic vascular remodeling. In this review, we discuss how NO and H2 S are individually regulated under ischemia, what their roles are in angiogenesis and arteriogenesis, and how their interaction controls ischemic vascular remodeling.
Collapse
Affiliation(s)
- Shuai Yuan
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| | - Christopher G Kevil
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
29
|
Li M, Tejada T, Lambert JP, Nicholson CK, Yahiro E, Ambai VT, Ali SF, Bradley EW, Graham RM, Dell’Italia LJ, Calvert JW, Naqvi N. Angiotensin type 2-receptor (AT2R) activation induces hypotension in apolipoprotein E-deficient mice by activating peroxisome proliferator-activated receptor-γ. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2016; 6:118-128. [PMID: 27679746 PMCID: PMC5030391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/19/2016] [Indexed: 06/06/2023]
Abstract
Angiotensin II (Ang II) modulates blood pressure and atherosclerosis development through its vascular type-1 (AT1R) and type-2 (AT2R) receptors, which have opposing effects. AT2R activation produces hypotension, and is anti-atherogenic. Targeted overexpression of AT2Rs in vascular smooth muscle cells (VSMCs) indicates that these effects are due to increased nitric oxide (NO) generation. However, the role of endogenous VSMC AT2Rs in these events is unknown. Effect of 7-day low-dose Ang II-infusion (12 µg/kg/hr) on blood pressure was tested in 9-week-old apoE((-/-)) mice fed a low or high cholesterol diet (LCD or HCD, respectively). Cardiac output was measured by echocardiography. Immunohistochemistry was performed to localize and quantify AT2Rs and p-Ser(1177)-endothelial nitric oxide synthase (eNOS) levels in the aortic arch. PD123319 and GW-9662 were used to selectively block the AT2R and peroxisome proliferator-activated receptor-γ (PPAR-γ), respectively. Ang II infusion decreased blood pressure by 12 mmHg (P < 0.001) in LCD/apoE((-/-)) mice without altering cardiac output; a response blocked by PD123319. Although, AT2R stimulation neither activated eNOS (p-Ser(1177)-eNOS) nor changed plasma NO metabolites, it caused an ~6-fold increase in VSMC PPAR-γ levels (P < 0.001) and the AT2R-mediated hypotension was abolished by GW-9662. AT2R-mediated hypotension was also inhibited by HCD, which selectively decreased VSMC AT2R expression by ~6-fold (P < 0.01). These findings suggest a novel pathway for the Ang II/AT2R-mediated hypotensive response that involves PPAR-γ, and is down regulated by a HCD.
Collapse
Affiliation(s)
- Ming Li
- Division of Cardiology, Department of Medicine, Emory UniversityAtlanta, Georgia, USA
- Cardiac Regeneration Research Institute, Wenzhou Medical UniversityWenzhou 325035, China
| | - Thor Tejada
- Division of Cardiology, Department of Medicine, Emory UniversityAtlanta, Georgia, USA
| | - Jonathan P Lambert
- Division of Cardiothoracic Surgery, Department of Surgery, Carlyle Fraser Heart Center, Emory UniversityAtlanta, Georgia, USA
| | - Chad K Nicholson
- Division of Cardiothoracic Surgery, Department of Surgery, Carlyle Fraser Heart Center, Emory UniversityAtlanta, Georgia, USA
| | - Eiji Yahiro
- Division of Cardiology, Department of Medicine, Emory UniversityAtlanta, Georgia, USA
| | - Vats T Ambai
- Division of Cardiology, Department of Medicine, Emory UniversityAtlanta, Georgia, USA
| | - Syeda F Ali
- Division of Cardiology, Department of Medicine, Emory UniversityAtlanta, Georgia, USA
| | - Eddie W Bradley
- Department of Medicine, University of Alabama at BirminghamBirmingham, Alabama, USA
| | - Robert M Graham
- Victor Chang Cardiac Research InstituteDarlinghurst, NSW 2010, Australia
| | - Louis J Dell’Italia
- Department of Medicine, University of Alabama at BirminghamBirmingham, Alabama, USA
- VA Medical CenterBirmingham, Alabama, USA
| | - John W Calvert
- Division of Cardiothoracic Surgery, Department of Surgery, Carlyle Fraser Heart Center, Emory UniversityAtlanta, Georgia, USA
| | - Nawazish Naqvi
- Division of Cardiology, Department of Medicine, Emory UniversityAtlanta, Georgia, USA
| |
Collapse
|
30
|
Donnarumma E, Bhushan S, Bradley JM, Otsuka H, Donnelly EL, Lefer DJ, Islam KN. Nitrite Therapy Ameliorates Myocardial Dysfunction via H2S and Nuclear Factor-Erythroid 2-Related Factor 2 (Nrf2)-Dependent Signaling in Chronic Heart Failure. J Am Heart Assoc 2016; 5:JAHA.116.003551. [PMID: 27473036 PMCID: PMC5015282 DOI: 10.1161/jaha.116.003551] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bioavailability of nitric oxide (NO) and hydrogen sulfide (H2S) is reduced in heart failure (HF). Recent studies suggest cross-talk between NO and H2S signaling. We previously reported that sodium nitrite (NaNO2) ameliorates myocardial ischemia-reperfusion injury and HF. Nuclear factor-erythroid-2-related factor 2 (Nrf2) regulates the antioxidant proteins expression and is upregulated by H2S. We examined the NaNO2 effects on endogenous H2S bioavailability and Nrf2 activation in mice subjected to ischemia-induced chronic heart failure (CHF). METHODS AND RESULTS Mice underwent 60 minutes of left coronary artery occlusion and 4 weeks of reperfusion. NaNO2 (165 μg/kgic) or vehicle was administered at reperfusion and then in drinking water (100 mg/L) for 4 weeks. Left ventricular (LV), ejection fraction (EF), LV end diastolic (LVEDD) and systolic dimensions (LVESD) were determined at baseline and at 4 weeks of reperfusion. Myocardial tissue was analyzed for oxidative stress and respective gene/protein-related assays. We found that NaNO2 therapy preserved LVEF, LVEDD and LVSD at 4 weeks during ischemia-induced HF. Myocardial malondialdehyde and protein carbonyl content were significantly reduced in NaNO2-treated mice as compared to vehicle, suggesting a reduction in oxidative stress. NaNO2 therapy markedly increased expression of Cu,Zn-superoxide dismutase, catalase, and glutathione peroxidase during 4 weeks of reperfusion. Furthermore, NaNO2 upregulated the activity of Nrf2, as well as H2S-producing enzymes, and ultimately increased H2S bioavailability in ischemia-induced CHF in mice as compared with vehicle. CONCLUSIONS Our results demonstrate that NaNO2 therapy significantly improves LV function via increasing H2S bioavailability, Nrf2 activation, and antioxidant defenses.
Collapse
Affiliation(s)
- Erminia Donnarumma
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Shashi Bhushan
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Jessica M Bradley
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Hiroyuki Otsuka
- Department of Surgery, Kurume University School of Medicine Kurume, Japan
| | - Erinn L Donnelly
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - David J Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Kazi N Islam
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA
| |
Collapse
|
31
|
Dimassi S, Chahed K, Boumiza S, Canault M, Tabka Z, Laurant P, Riva C. Role of eNOS- and NOX-containing microparticles in endothelial dysfunction in patients with obesity. Obesity (Silver Spring) 2016; 24:1305-12. [PMID: 27130266 DOI: 10.1002/oby.21508] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/19/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To explore the pathophysiological profile of patients who have obesity and to investigate the potential role of circulating microparticles (MPs) in endothelial dysfunction in patients who have obesity. METHODS The inflammatory and oxidative status and the cutaneous microvascular blood flow were characterized in 69 patients with android obesity and 46 subjects with normal weight (controls) by using laser Doppler flowmetry. Circulating MP levels were measured by flow cytometry, and endothelial nitric oxide synthase (eNOS) and NADPH oxidase (NOX) expression in MPs was investigated by Western blotting. MP effect on vascular reactivity was assessed in rat aorta rings. RESULTS Patients with obesity showed endothelial dysfunction, hyperglycemia, inflammation, and oxidative stress. In controls, low MP levels were positively correlated with normal microvascular function. Western blot analysis revealed reduced eNOS and increased NOX4D expression in MPs from subjects with obesity compared with controls. However, this was not correlated with endothelial dysfunction parameters and did not impair ex vivo endothelium-dependent vasodilation. CONCLUSIONS These results suggest that MPs do not contribute directly to endothelial dysfunction associated with obesity. Conversely, eNOS- and NOX-containing MPs could be involved in the compensatory mechanism of vascular endothelial cells to counteract the pathologic mechanisms underlying endothelial dysfunction.
Collapse
Affiliation(s)
- Saloua Dimassi
- Avignon University, LAPEC EA4278, Avignon, France
- Faculty of Medicine, Sousse University, UR12ES06, Physiologie de l'Exercice et Physiopathologie: de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Sousse, Tunisia
| | - Karim Chahed
- Faculty of Medicine, Sousse University, UR12ES06, Physiologie de l'Exercice et Physiopathologie: de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Sousse, Tunisia
| | - Soumaya Boumiza
- Faculty of Medicine, Sousse University, UR12ES06, Physiologie de l'Exercice et Physiopathologie: de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Sousse, Tunisia
| | - Matthias Canault
- INRA, UMR 1260, Marseille, France
- INSERM, UMR 1062, Nutrition, Obésité et Risque Thrombotique, Marseille, France
- Aix Marseille University, Faculté de Médecine, Marseille, France
| | - Zouhair Tabka
- Faculty of Medicine, Sousse University, UR12ES06, Physiologie de l'Exercice et Physiopathologie: de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Sousse, Tunisia
| | | | | |
Collapse
|
32
|
Sethumadhavan S, Whitsett J, Bennett B, Ionova IA, Pieper GM, Vasquez-Vivar J. Increasing tetrahydrobiopterin in cardiomyocytes adversely affects cardiac redox state and mitochondrial function independently of changes in NO production. Free Radic Biol Med 2016; 93:1-11. [PMID: 26826575 PMCID: PMC5498285 DOI: 10.1016/j.freeradbiomed.2016.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 01/25/2016] [Indexed: 02/07/2023]
Abstract
Tetrahydrobiopterin (BH4) represents a potential strategy for the treatment of cardiac remodeling, fibrosis and/or diastolic dysfunction. The effects of oral treatment with BH4 (Sapropterin™ or Kuvan™) are however dose-limiting with high dose negating functional improvements. Cardiomyocyte-specific overexpression of GTP cyclohydrolase I (mGCH) increases BH4 several-fold in the heart. Using this model, we aimed to establish the cardiomyocyte-specific responses to high levels of BH4. Quantification of BH4 and BH2 in mGCH transgenic hearts showed age-based variations in BH4:BH2 ratios. Hearts of mice (<6 months) have lower BH4:BH2 ratios than hearts of older mice while both GTPCH activity and tissue ascorbate levels were higher in hearts of young than older mice. No evident changes in nitric oxide (NO) production assessed by nitrite and endogenous iron-nitrosyl complexes were detected in any of the age groups. Increased BH4 production in cardiomyocytes resulted in a significant loss of mitochondrial function. Diminished oxygen consumption and reserve capacity was verified in mitochondria isolated from hearts of 12-month old compared to 3-month old mice, even though at 12 months an improved BH4:BH2 ratio is established. Accumulation of 4-hydroxynonenal (4-HNE) and decreased glutathione levels were found in the mGCH hearts and isolated mitochondria. Taken together, our results indicate that the ratio of BH4:BH2 does not predict changes in neither NO levels nor cellular redox state in the heart. The BH4 oxidation essentially limits the capacity of cardiomyocytes to reduce oxidant stress. Cardiomyocyte with chronically high levels of BH4 show a significant decline in redox state and mitochondrial function.
Collapse
Affiliation(s)
- Savitha Sethumadhavan
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jennifer Whitsett
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Brian Bennett
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; Department of Physics, Marquette University, Milwaukee, 1250 W Wisconsin Ave, Milwaukee, WI 53233, USA
| | - Irina A Ionova
- Department of Surgery Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Galen M Pieper
- Department of Surgery Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jeannette Vasquez-Vivar
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
33
|
Hess DC, Khan MB, Hoda N, Morgan JC. Remote ischemic conditioning: a treatment for vascular cognitive impairment. Brain Circ 2015; 1:133-139. [PMID: 30221201 DOI: 10.4103/2394-8108.172885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
There is a strong link between hypoperfusion and white matter (WM) damage in patients with leukoaraiosis and vascular cognitive impairment (VCI). Other than management of vascular risk factors, there is no treatment for WM damage and VCI that delays progression of the disease process to dementia. Observational studies suggest that exercise may prevent or slow down the progression of Alzheimer's disease (AD) and VCI. However, getting patients to exercise is challenging especially with advancing age and disability. Remote ischemic conditioning, an "exercise equivalent", allows exercise to be given with a "device" in the home for long periods of time. Since RIC increases CBF in pre-clinical studies and in humans, RIC may be an ideal therapy to treat VCI and WM disease and perhaps even sporadic AD. By using MRI imaging of WM progression, a sample size in the range of about 100 subjects per group could determine if RIC has activity in WM disease and VCI.
Collapse
Affiliation(s)
- David C Hess
- Department of Neurology Medical College of Georgia, Georgia Regent's University, Augusta, GA USA
| | - Mohammad B Khan
- Department of Neurology Medical College of Georgia, Georgia Regent's University, Augusta, GA USA
| | - Nasrul Hoda
- Department of Medical Laboratory, Imaging, and Radiologic Sciences, College of Allied Health Sciences, Georgia Regent's University, Augusta, GA USA
| | - John C Morgan
- Department of Neurology Medical College of Georgia, Georgia Regent's University, Augusta, GA USA
| |
Collapse
|
34
|
de Lima Portella R, Lynn Bickta J, Shiva S. Nitrite Confers Preconditioning and Cytoprotection After Ischemia/Reperfusion Injury Through the Modulation of Mitochondrial Function. Antioxid Redox Signal 2015; 23:307-27. [PMID: 26094636 DOI: 10.1089/ars.2015.6260] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Nitrite is now recognized as an intrinsic signaling molecule that mediates a number of biological processes. One of the most reproducible effects of nitrite is its ability to mediate cytoprotection after ischemia/reperfusion (I/R). This robust phenomenon has been reproduced by a number of investigators in varying animal models focusing on different target organs. Furthermore, nitrite's cytoprotective versatility is highlighted by its ability to mediate delayed preconditioning and remote conditioning in addition to acute protection. RECENT ADVANCES In the last 10 years, significant progress has been made in elucidating the mechanisms underlying nitrite-mediated ischemic tolerance. CRITICAL ISSUES The mitochondrion, which is essential to both the progression of I/R injury and the protection afforded by preconditioning, has emerged as a major subcellular target for nitrite. This review will outline the role of the mitochondrion in I/R injury and preconditioning, review the accumulated preclinical studies demonstrating nitrite-mediated cytoprotection, and finally focus on the known interactions of nitrite with mitochondria and their role in the mechanism of nitrite-mediated ischemic tolerance. FUTURE DIRECTIONS These studies set the stage for current clinical trials testing the efficacy of nitrite to prevent warm and cold I/R injury.
Collapse
Affiliation(s)
- Rafael de Lima Portella
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Janelle Lynn Bickta
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 Department of Bioengineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- 1 Vascular Medicine Institute, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Abstract
It is becoming increasingly clear that many diseases are characterized or associated with perturbations in nitric oxide (NO) production/signaling. Therapeutics or strategies designed to restore normal NO homeostasis will likely have broad application and utility. This highly complex and multistep pathway for NO production and subsequent target activation provides many steps in the endogenous pathway that may be useful targets for drug development for cardiovascular disease, antimicrobial, cancer, wound healing, etc. This article will summarize known strategies that are currently available or in development for enhancing NO production or availability in the human body. Each strategy will be discussed including exogenous sources of NO, use of precursors to promote NO production and downstream pathways affected by NO production with advantages and disadvantages highlighted for each. Development of NO-based therapeutics is and will continue to be a major focus of biotech, academia as well as pharmaceutical companies. Application of safe and effective strategies will certainly transform health and disease.
Collapse
|
36
|
Uryash A, Bassuk J, Kurlansky P, Altamirano F, Lopez JR, Adams JA. Antioxidant Properties of Whole Body Periodic Acceleration (pGz). PLoS One 2015; 10:e0131392. [PMID: 26133377 PMCID: PMC4489838 DOI: 10.1371/journal.pone.0131392] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/02/2015] [Indexed: 11/30/2022] Open
Abstract
The recognition that oxidative stress is a major component of several chronic diseases has engendered numerous trials of antioxidant therapies with minimal or no direct benefits. Nanomolar quantities of nitric oxide released into the circulation by pharmacologic stimulation of eNOS have antioxidant properties but physiologic stimulation as through increased pulsatile shear stress of the endothelium has not been assessed. The present study utilized a non-invasive technology, periodic acceleration (pGz) that increases pulsatile shear stress such that upregulation of cardiac eNOS occurs, We assessed its efficacy in normal mice and mouse models with high levels of oxidative stress, e.g. Diabetes type 1 and mdx (Duchene Muscular Dystrophy). pGz increased protein expression and upregulated eNOS in hearts. Application of pGz was associated with significantly increased expression of endogenous antioxidants (Glutathioneperoxidase-1(GPX-1), Catalase (CAT), Superoxide, Superoxide Dismutase 1(SOD1). This led to an increase of total cardiac antioxidant capacity along with an increase in the antioxidant response element transcription factor Nrf2 translocation to the nucleus. pGz decreased reactive oxygen species in both mice models of oxidative stress. Thus, pGz is a novel non-pharmacologic method to harness endogenous antioxidant capacity.
Collapse
Affiliation(s)
- Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, Florida, United States of America
| | - Jorge Bassuk
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, Florida, United States of America
| | - Paul Kurlansky
- Department of Surgery, Columbia University, New York, New York, United States of America
| | - Francisco Altamirano
- Department of Molecular Biosciences, University of California Davis, Davis, California, United States of America
| | - Jose R. Lopez
- Department of Molecular Biosciences, University of California Davis, Davis, California, United States of America
| | - Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, Florida, United States of America
- * E-mail:
| |
Collapse
|
37
|
Ramezani Tehrani F, Behboudi-Gandevani S, Ghasemi A, Azizi F. Association between serum concentrations of nitric oxide and transition to menopause. Acta Obstet Gynecol Scand 2015; 94:708-714. [DOI: 10.1111/aogs.12655] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 04/08/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center; Research Institute for Endocrine Sciences; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Samira Behboudi-Gandevani
- Reproductive Endocrinology Research Center; Research Institute for Endocrine Sciences; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center; Research Institute for Endocrine Sciences; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Fereidoun Azizi
- Endocrine Research Center; Research Institute for Endocrine Sciences; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
38
|
Bradley JM, Islam KN, Polhemus DJ, Donnarumma E, Brewster LP, Tao YX, Goodchild TT, Lefer DJ. Sustained release nitrite therapy results in myocardial protection in a porcine model of metabolic syndrome with peripheral vascular disease. Am J Physiol Heart Circ Physiol 2015; 309:H305-17. [PMID: 25957218 DOI: 10.1152/ajpheart.00163.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/06/2015] [Indexed: 11/22/2022]
Abstract
Metabolic syndrome (MetS) reduces endothelial nitric oxide (NO) bioavailability and exacerbates vascular dysfunction in patients with preexisting vascular diseases. Nitrite, a storage form of NO, can mediate vascular function during pathological conditions when endogenous NO is reduced. The aims of the present study were to characterize the effects of severe MetS and obesity on dyslipidemia, myocardial oxidative stress, and endothelial NO synthase (eNOS) regulation in the obese Ossabaw swine (OS) model and to examine the effects of a novel, sustained-release formulation of sodium nitrite (SR-nitrite) on coronary vascular reactivity and myocardial redox status in obese OS subjected to critical limb ischemia (CLI). After 6 mo of an atherogenic diet, obese OS displayed a MetS phenotype. Obese OS had decreased eNOS functionality and NO bioavailability. In addition, obese OS exhibited increased oxidative stress and a significant reduction in antioxidant enzymes. The efficacy of SR-nitrite therapy was examined in obese OS subjected to CLI. After 3 wk of treatment, SR-nitrite (80 mg · kg(-1) · day(-1) bid po) increased myocardial nitrite levels and eNOS function. Treatment with SR-nitrite reduced myocardial oxidative stress while increasing myocardial antioxidant capacity. Ex vivo assessment of vascular reactivity of left anterior descending coronary artery segments demonstrated marked improvement in vasoreactivity to sodium nitroprusside but not to substance P and bradykinin in SR-nitrite-treated animals compared with placebo-treated animals. In conclusion, in a clinically relevant, large-animal model of MetS and CLI, treatment with SR-nitrite enhanced myocardial NO bioavailability, attenuated oxidative stress, and improved ex vivo coronary artery vasorelaxation.
Collapse
Affiliation(s)
- Jessica M Bradley
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Kazi N Islam
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - David J Polhemus
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Erminia Donnarumma
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Luke P Brewster
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia; Surgery and Research Services, Atlanta Veterans Affairs Medical Center, Decatur, Georgia; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia; and
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, Alabama
| | - Traci T Goodchild
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - David J Lefer
- Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisiana;
| |
Collapse
|
39
|
Bryan NS. The potential use of salivary nitrite as a marker of NO status in humans. Nitric Oxide 2015; 45:4-6. [DOI: 10.1016/j.niox.2014.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 12/16/2014] [Accepted: 12/17/2014] [Indexed: 01/22/2023]
|
40
|
Farah C, Reboul C. NO Better Way to Protect the Heart during Ischemia-Reperfusion: To be in the Right Place at the Right Time. Front Pediatr 2015; 3:6. [PMID: 25705614 PMCID: PMC4319379 DOI: 10.3389/fped.2015.00006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/26/2015] [Indexed: 11/24/2022] Open
Affiliation(s)
- Charlotte Farah
- EA4278, LaPEC, Université d'Avignon , Avignon , France ; UMR-CNRS 9214, INSERM U1046, Université de Montpellier , Montpellier , France
| | - Cyril Reboul
- EA4278, LaPEC, Université d'Avignon , Avignon , France
| |
Collapse
|
41
|
Rassaf T, Ferdinandy P, Schulz R. Nitrite in organ protection. Br J Pharmacol 2014; 171:1-11. [PMID: 23826831 DOI: 10.1111/bph.12291] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 06/09/2013] [Accepted: 06/21/2013] [Indexed: 12/15/2022] Open
Abstract
In the last decade, the nitrate-nitrite-nitric oxide pathway has emerged to therapeutical importance. Modulation of endogenous nitrate and nitrite levels with the subsequent S-nitros(yl)ation of the downstream signalling cascade open the way for novel cytoprotective strategies. In the following, we summarize the actual literature and give a short overview on the potential of nitrite in organ protection.
Collapse
Affiliation(s)
- Tienush Rassaf
- Department of Medicine, Division of Cardiology, Pulmonary and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|
42
|
Evaluation of endothelial function by peripheral arterial tonometry and relation with the nitric oxide pathway. Nitric Oxide 2014; 42:1-8. [PMID: 25064180 DOI: 10.1016/j.niox.2014.07.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/30/2014] [Accepted: 07/10/2014] [Indexed: 10/25/2022]
Abstract
Endothelial dysfunction is an important component in the development of cardiovascular diseases. Endothelial function may be evaluated by peripheral arterial tonometry (PAT) which measures the vasodilator function in the microvasculature of the fingertip during reactive hyperaemia. The reactive hyperaemia index (RHI) is decreased in the presence of cardiovascular risk factors and thus far several studies have shown that PAT-RHI may provide reliable prediction of outcome. The technique is operator independent and easy to perform. Abnormalities measured by PAT follow the same trend as those measured by flow-mediated dilation in the brachial artery, but the two methods are not interchangeable. We have reviewed the recent literature in an effort to evaluate peripheral arterial tonometry as a method to assess the function of the endothelium and additionally suggest directions for future research. Special attention will be directed to the nitric oxide dependency of the reactive hyperaemia index obtained by peripheral arterial tonometry.
Collapse
|
43
|
Affiliation(s)
- Paola Corti
- From the Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Department of Medicine (P.C., M.T.G.) and Department of Pulmonary, Allergy and Critical Care Medicine (M.T.G.), University of Pittsburgh, PA
| | | |
Collapse
|
44
|
Omentin Prevents Myocardial Ischemic Injury Through AMP-Activated Protein Kinase- and Akt-Dependent Mechanisms. J Am Coll Cardiol 2014; 63:2722-33. [DOI: 10.1016/j.jacc.2014.03.032] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 11/17/2022]
|
45
|
Hydrogen sulfide cytoprotective signaling is endothelial nitric oxide synthase-nitric oxide dependent. Proc Natl Acad Sci U S A 2014; 111:3182-7. [PMID: 24516168 DOI: 10.1073/pnas.1321871111] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Previous studies have demonstrated that hydrogen sulfide (H2S) protects against multiple cardiovascular disease states in a similar manner as nitric oxide (NO). H2S therapy also has been shown to augment NO bioavailability and signaling. The purpose of this study was to investigate the impact of H2S deficiency on endothelial NO synthase (eNOS) function, NO production, and ischemia/reperfusion (I/R) injury. We found that mice lacking the H2S-producing enzyme cystathionine γ-lyase (CSE) exhibit elevated oxidative stress, dysfunctional eNOS, diminished NO levels, and exacerbated myocardial and hepatic I/R injury. In CSE KO mice, acute H2S therapy restored eNOS function and NO bioavailability and attenuated I/R injury. In addition, we found that H2S therapy fails to protect against I/R in eNOS phosphomutant mice (S1179A). Our results suggest that H2S-mediated cytoprotective signaling in the setting of I/R injury is dependent in large part on eNOS activation and NO generation.
Collapse
|
46
|
Jiang H, Torregrossa AC, Potts A, Pierini D, Aranke M, Garg HK, Bryan NS. Dietary nitrite improves insulin signaling through GLUT4 translocation. Free Radic Biol Med 2014; 67:51-7. [PMID: 24157451 DOI: 10.1016/j.freeradbiomed.2013.10.809] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 01/31/2023]
Abstract
Diabetes mellitus type 2 is a syndrome of disordered metabolism with inappropriate hyperglycemia owing to a reduction in the biological effectiveness of insulin. Type 2 diabetes is associated with an impaired nitric oxide (NO) pathway that probably serves as the key link between metabolic disorders and cardiovascular disease. Insulin-mediated translocation of GLUT4 involves the PI3K/Akt kinase signal cascade that results in activation of endothelial NO synthase (eNOS). eNOS is dysfunctional during diabetes. We hypothesize that loss of eNOS-derived NO terminates the signaling cascade and therefore cannot activate GLUT4 translocation and that dietary nitrite may repair this pathway. In this study, we administered 50mg/L sodium nitrite to db/db diabetic mice for 4 weeks. After 4 weeks treatment, the db/db mice experienced less weight gain, improved fasting glucose levels, and reduced insulin levels. Cell culture experiments using CHO-HIRc-myc-GLUT4eGFP cell lines stably expressing insulin receptor and myc-GLUT4eGFP protein, as well as L6 skeletal muscle cells stably expressing rat GLUT4 with a Myc epitope (L6-GLUT4myc), showed that NO, nitrite, and GSNO stimulate GLUT4 translocation independent of insulin, which is inhibited by NEM. Collectively our data suggest that nitrite improves insulin signaling through restoration of NO-dependent nitrosation of GLUT4 signaling translocation. These data suggest that NO-mediated nitrosation of GLUT4 by nitrite or other nitrosating agents is necessary and sufficient for GLUT4 translocation in target tissue. Description of this pathway may justify a high-nitrate/nitrite diet along with the glycemic index to provide a safe and nutritional regimen for the management and treatment of diabetes.
Collapse
Affiliation(s)
- Hong Jiang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Ashley C Torregrossa
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Amy Potts
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Dan Pierini
- California State University at Fullerton, Fullerton, CA 92831, USA
| | - Mayank Aranke
- The University of Texas at Austin, Austin, TX 78712, USA
| | - Harsha K Garg
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA
| | - Nathan S Bryan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, Health Science Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, The University of Texas at Houston, Houston, TX 77030, USA.
| |
Collapse
|
47
|
Cox AG, Saunders DC, Kelsey PB, Conway AA, Tesmenitsky Y, Marchini JF, Brown KK, Stamler JS, Colagiovanni DB, Rosenthal GJ, Croce KJ, North TE, Goessling W. S-nitrosothiol signaling regulates liver development and improves outcome following toxic liver injury. Cell Rep 2014; 6:56-69. [PMID: 24388745 DOI: 10.1016/j.celrep.2013.12.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 10/26/2013] [Accepted: 12/03/2013] [Indexed: 12/21/2022] Open
Abstract
Toxic liver injury is a leading cause of liver failure and death because of the organ's inability to regenerate amidst massive cell death, and few therapeutic options exist. The mechanisms coordinating damage protection and repair are poorly understood. Here, we show that S-nitrosothiols regulate liver growth during development and after injury in vivo; in zebrafish, nitric-oxide (NO) enhanced liver formation independently of cGMP-mediated vasoactive effects. After acetaminophen (APAP) exposure, inhibition of the enzymatic regulator S-nitrosoglutathione reductase (GSNOR) minimized toxic liver damage, increased cell proliferation, and improved survival through sustained activation of the cytoprotective Nrf2 pathway. Preclinical studies of APAP injury in GSNOR-deficient mice confirmed conservation of hepatoprotective properties of S-nitrosothiol signaling across vertebrates; a GSNOR-specific inhibitor improved liver histology and acted with the approved therapy N-acetylcysteine to expand the therapeutic time window and improve outcome. These studies demonstrate that GSNOR inhibitors will be beneficial therapeutic candidates for treating liver injury.
Collapse
Affiliation(s)
- Andrew G Cox
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Diane C Saunders
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter B Kelsey
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Allie A Conway
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yevgenia Tesmenitsky
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Julio F Marchini
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kristin K Brown
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine and Department of Medicine, Harrington Discovery Institute, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | | | | | - Kevin J Croce
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E North
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | - Wolfram Goessling
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Gastroenterology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Gastrointestinal Cancer Center, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
48
|
Bir SC, Pattillo CB, Pardue S, Kolluru GK, Shen X, Giordano T, Kevil CG. Nitrite anion therapy protects against chronic ischemic tissue injury in db/db diabetic mice in a NO/VEGF-dependent manner. Diabetes 2014; 63:270-81. [PMID: 24009258 PMCID: PMC4179307 DOI: 10.2337/db13-0890] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nitrite anion has been demonstrated to be a prodrug of nitric oxide (NO) with positive effects on tissue ischemia/reperfusion injury, cytoprotection, and vasodilation. However, effects of nitrite anion therapy for ischemic tissue vascular remodeling during diabetes remain unknown. We examined whether sodium nitrite therapy altered ischemic revascularization in BKS-Lepr(db/db) mice subjected to permanent unilateral femoral artery ligation. Sodium nitrite therapy completely restored ischemic hind limb blood flow compared with nitrate or PBS therapy. Importantly, delayed nitrite therapy 5 days after ischemia restored ischemic limb blood flow in aged diabetic mice. Restoration of blood flow was associated with increases in ischemic tissue angiogenesis activity and cell proliferation. Moreover, nitrite but not nitrate therapy significantly prevented ischemia-mediated tissue necrosis in aged mice. Nitrite therapy significantly increased ischemic tissue vascular endothelial growth factor (VEGF) protein expression that was essential for nitrite-mediated reperfusion of ischemic hind limbs. Nitrite significantly increased ischemic tissue NO bioavailability along with concomitant reduction of superoxide formation. Lastly, nitrite treatment also significantly stimulated hypoxic endothelial cell proliferation and migration in the presence of high glucose in an NO/VEGF-dependent manner. These results demonstrate that nitrite therapy effectively stimulates ischemic tissue vascular remodeling in the setting of metabolic dysfunction that may be clinically useful.
Collapse
Affiliation(s)
- Shyamal C. Bir
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Christopher B. Pattillo
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Sibile Pardue
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Gopi K. Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
| | | | - Christopher G. Kevil
- Department of Pathology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, LA
- TheraVasc Inc., Cleveland, OH
- Corresponding author: Christopher G. Kevil,
| |
Collapse
|
49
|
Calvert JW, Lefer DJ. Role of β-adrenergic receptors and nitric oxide signaling in exercise-mediated cardioprotection. Physiology (Bethesda) 2013; 28:216-24. [PMID: 23817796 DOI: 10.1152/physiol.00011.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Exercise promotes cardioprotection in both humans and animals not only by reducing risk factors associated with cardiovascular disease but by reducing myocardial infarction and improving survival following ischemia. This article will define the role that nitric oxide and β-adrenergic receptors play in mediating the cardioprotective effects of exercise in the setting of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- John W Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, Georgia, USA.
| | | |
Collapse
|
50
|
Corti P, Tejero J, Gladwin MT. Evidence mounts that red cells and deoxyhemoglobin can reduce nitrite to bioactive NO to mediate intravascular endocrine NO signaling: commentary on "Anti-platelet effects of dietary nitrate in healthy volunteers: involvement of cGMP and influence of sex". Free Radic Biol Med 2013; 65:1518-1520. [PMID: 24100230 DOI: 10.1016/j.freeradbiomed.2013.09.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/25/2013] [Indexed: 10/26/2022]
Affiliation(s)
- Paola Corti
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jesús Tejero
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mark T Gladwin
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|