1
|
Zhu S, Liu Z, Hu B, Feng Y, Pan G. Nitrite Reductases in Biomedicine: From Natural Enzymes to Artificial Mimics. RESEARCH (WASHINGTON, D.C.) 2025; 8:0710. [PMID: 40438154 PMCID: PMC12117333 DOI: 10.34133/research.0710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 06/01/2025]
Abstract
Nitrite reductases (NiRs) are natural enzymes that facilitate the reduction of nitrite. They are essential for the microbial nitrogen cycle and play a vital role in regulating numerous physiological and pathological processes associated with nitric oxide (NO) in living organisms. By the merits of protein engineering, a variety of artificial NiR mimics have been developed. These include traditional artificial proteins, metal-azacycle complexes, and nanozymes such as metal, metal oxide/sulfide nanoparticles, metal-organic frameworks, bioinorganic nanohybrids, and advanced single-atom nanozymes. This development marks an important milestone in broadening the application of enzyme-like catalytic nitrite reduction across various fields, such as biomedicine, biosensing, food science, and environmental science. In this review, we first outline the different types of NiRs, along with their active center structures and catalytic mechanisms, drawing from recent research and discoveries. We then classify the reported NiR mimic materials, discussing their active center structures and enzyme-like catalytic mechanisms. Additionally, we explore the potential future applications and challenges facing NiR mimics in the field of biomedicine.
Collapse
Affiliation(s)
- Sai Zhu
- Institute for Advanced Materials, School of Materials Science and Engineering,
Jiangsu University, Zhenjiang 212013, China
| | - Zhengbiao Liu
- Department of Orthopedics,
Suzhou Industrial Park Xinghu Hospital, Suzhou, Jiangsu 215000, China
| | - Bo Hu
- Jilin Provincial Key Laboratory of Western Jilin’s Clean Energy,
Baicheng Normal University, Baicheng 137000, China
| | - Yonghai Feng
- Institute for Advanced Materials, School of Materials Science and Engineering,
Jiangsu University, Zhenjiang 212013, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering,
Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
2
|
Deepali, Goel N, Khandnor P. DeepOmicsSurv: a deep learning-based model for survival prediction of oral cancer. Discov Oncol 2025; 16:614. [PMID: 40278990 PMCID: PMC12031713 DOI: 10.1007/s12672-025-02346-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE Oral cancer is an important health challenge worldwide and accurate survival time prediction of this disease can guide treatment decisions. This study aims to propose a deep learning-based model, DeepOmicsSurv, to predict survival in oral cancer patients using clinical and multi-omics data. METHODS DeepOmicsSurv builds on the DeepSurv model, incorporating multi-head attention convolutional layers, dropout, pooling, and batch normalization to boost its strength and precision. Various dimensionality reduction techniques, including Principal Component Analysis (PCA), Kernel PCA, Non-Negative Matrix Factorization (NMF), Singular Value Decomposition (SVD), Partial Least Squares (PLS), Multidimensional Scaling (MDS), and Autoencoders, were employed to manage the high-dimensional omics data. The model's performance was evaluated against DeepSurv, DeepHit, Cox Proportional Hazards (CoxPH), Convolutional Neural Networks (CNN), and Recurrent Neural Networks (RNN). Additionally, SHapley Additive Explanations (SHAP) was used to analyze the impact of clinical features on survival predictions. RESULTS DeepOmicsSurv achieved a C-index of 0.966, MSE of 0.0138, RMSE of 0.1174, MAE of 0.0795, and MedAE of 0.0515, outperforming other deep learning models. Among various dimensionality reduction techniques, autoencoder performed the best with DeepOmicsSurv. SHAP analysis showed that Age, AJCC N Stage, alcohol history and patient smoking history are prevalent clinical features for survival time. CONCLUSION In conclusion, DeepOmicsSurv has the potential to predict survival time in oral cancer patients. This model achieved high accuracy with various data types including Clinical, DNAmethylation + clinical, mRNA + clinical, Copy number alteration + clinical, or multi-omics data. Additionally, SHAP analysis reveals clinical factors that influence survival time.
Collapse
Affiliation(s)
- Deepali
- University Institute of Engineering and Technology, Panjab University, Chandigarh, 160014, India
- Department of Computer Science, Guru Nanak College, Budhlada, 151502, India
| | - Neelam Goel
- University Institute of Engineering and Technology, Panjab University, Chandigarh, 160014, India.
| | - Padmavati Khandnor
- Department of Computer Science, Punjab Engineering College (Deemed to be University), Chandigarh, 160012, India
| |
Collapse
|
3
|
Chiletti R, Fincher SH, Horton SB, Peek GJ, Checchia P, Butt W. The Role of Nitric Oxide in the Sweep Gas for Patients Receiving Extracorporeal Membrane Oxygenation or Cardiopulmonary Bypass. Can J Cardiol 2025; 41:621-629. [PMID: 39733940 DOI: 10.1016/j.cjca.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024] Open
Abstract
Nitric oxide (NO) was proclaimed the 1992 "molecule of the year" by Culotta in Science magazine because of its importance in neuroscience, physiology, and immunology. Inhaled NO has been in clinical use for over 35 years to decrease pulmonary hypertension and improve oxygenation. Over the past 20 years, there has been much research into understanding the role of NO on cell surface receptors, mitochondria, and intracellular processes that involve calcium and superoxide radicals. This research has shown that, irrespective of the cause, NO has a major role in the systemic inflammatory response syndrome and ischemia-reperfusion injury.1 More recent clinical research has focussed on NO use in patients undergoing cardiopulmonary bypass and receiving extracorporeal life support, with some centres incorporating NO into sweep gas as part of routine practice. In this article we review NO pathways in humans, the biologic effects of NO, the interplay between NO and red blood cells, and animal and human studies on the effects of exogenously administered NO.
Collapse
Affiliation(s)
- Roberto Chiletti
- Department of Paediatric Intensive Care, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Sophie H Fincher
- Department of Paediatric Intensive Care, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Critical Care, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen B Horton
- Department of Cardiac Surgery, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Melbourne, Victoria, Australia; Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Giles J Peek
- Congenital Heart Centre, University of Florida, Gainesville, Florida, USA
| | - Paul Checchia
- Division of Critical Care Medicine, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - Warwick Butt
- Department of Paediatric Intensive Care, Royal Children's Hospital, Melbourne, Victoria, Australia; Department of Critical Care, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Melbourne, Victoria, Australia; ICU Research Clinical Sciences Theme MCRI, Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Liu H, Yao K, Hu M, Li S, Yang S, Zhao A. On-Chip Electrochemical Sensor Based on 3D Graphene Assembly Decorated Ultrafine RuCu Alloy Nanocatalyst for In Situ Detection of NO in Living Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:417. [PMID: 40137592 PMCID: PMC11946219 DOI: 10.3390/nano15060417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025]
Abstract
In this work, we developed 3D ionic liquid (IL) functionalized graphene assemblies (GAs) decorated by ultrafine RuCu alloy nanoparticles (RuCu-ANPs) via a one-step synthesis process, and integrated it into a microfluidic sensor chip for in situ electrochemical detection of NO released from living cells. Our findings have demonstrated that RuCu-ANPs on 3D IL-GA exhibit high density, uniform distribution, lattice-shaped arrangement of atoms, and extremely ultrafine size, and possess high electrocatalytic activity to NO oxidation on the electrode. Meanwhile, the 3D IL-GA with hierarchical porous structures can facilitate the efficient electron/mass transfer at the electrode/electrolyte interface and the cell culture. Moreover, the graft of IL molecules on GA endows it with high hydrophilicity for facile and well-controllable printing on the electrode. Consequently, the resultant electrochemical microfluidic sensor demonstrated excellent sensing performances including fast response time, high sensitivity, good anti-interference ability, high reproducibility, long-term stability, as well as good biocompatibility, which can be used as an on-chip sensing system for cell culture and real-time in situ electrochemical detection of NO released from living cells with accurate and stable characteristics in physiological conditions.
Collapse
Affiliation(s)
- Haibo Liu
- Technology Inspection Center of ShengLi Oil Filed, China Petrochemical Corporation, Dongying 257000, China;
| | - Kaiyuan Yao
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China;
| | - Min Hu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, China (S.L.); (S.Y.)
| | - Shanting Li
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, China (S.L.); (S.Y.)
| | - Shengxiong Yang
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, China (S.L.); (S.Y.)
| | - Anshun Zhao
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China;
| |
Collapse
|
5
|
Mostahsan Z, Azizi S, Soleimanzadeh A, Shalizar-Jalali A. Protective effects of Mito-TEMPO on ischemia-reperfusion injury in a mouse testicular torsion and detorsion model. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2024; 15:665-672. [PMID: 39816632 PMCID: PMC11729104 DOI: 10.30466/vrf.2024.2024815.4202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/07/2024] [Indexed: 01/18/2025]
Abstract
Testicular ischemia-reperfusion (I/R) injury during testicular torsion is strongly influenced by oxidative stress caused by excessive accumulation of unscavenged reactive oxygen species. This study aimed to investigate the effects of intra-peritoneal administration of Mito-TEMPO (MT) on I/R injury in testicular torsion/detorsion (T/D) in mice. Forty-two male mice were divided into seven groups including 1 control and 6 treatment groups (360° T/D, 720° T/D, 360° T/D + 0.70 mg kg-1 MT, 360° T/D + 1.00 mg kg-1 MT, 720° T/D + 0.70 mg kg-1 MT, and 720° T/D + 1.00 mg kg-1 MT). After inducing 360° and 720° clockwise testicular torsions for 2 hr, sperm parameters, apoptosis-related genes expression, and in vivo fertility index were evaluated. The results showed that 720° T/D can lead to increased abnormal sperm morphology, sperm DNA damage, and Bax expression, while the Bcl-2 expression was reduced compared to the other groups. In addition, it also had negative effects on sperm total and progressive motilities as well as viability and plasma membrane functionality (PMF). The results also showed that administration of MT to T/D mice can result in a reduction in abnormal sperm morphology, DNA damage, and Bax expression. It could also increase sperm total and progressive motilities, viability and PMF, Bcl-2 expression, and in vivo fertility index. Based on our results, it is concluded that MT, when administered after spermatic cord torsion in mice, provides significant protection against acute testicular T/D injury.
Collapse
Affiliation(s)
- Zohreh Mostahsan
- DVSc Candidate, Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran;
| | - Saeed Azizi
- Department of Surgery and Diagnostic Imaging, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran;
| | - Ali Soleimanzadeh
- Department of Theriogenology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran;
| | - Ali Shalizar-Jalali
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| |
Collapse
|
6
|
Massimo G, Dyson N, Olotu F, Khambata RS, Ahluwalia A. Potential Opportunities for Pharmacogenetic-Based Therapeutic Exploitation of Xanthine Dehydrogenase in Cardiovascular Disease. Antioxidants (Basel) 2024; 13:1439. [PMID: 39765766 PMCID: PMC11672463 DOI: 10.3390/antiox13121439] [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: 08/11/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
The majority of naturally occurring mutations of the human gene XDH, are associated with reduced or completely absent xanthine oxidoreductase (XOR) activity, leading to a disease known as classical xanthinuria, which is due to the accumulation and excretion of xanthine in urine. Three types of classical xanthinuria have been identified: type I, characterised by XOR deficiency, type II, caused by XOR and aldehyde oxidase (AO) deficiency, and type III due to XOR, AO, and sulphite oxidase (SO) deficiency. Type I and II are considered rare autosomal recessive disorders, a condition where two copies of the mutated gene must be present to develop the disease or trait. In most cases, xanthinuria type I and II result to be asymptomatic, and only occasionally lead to renal failure due to urolithiasis caused by xanthine deposition. However, in the last 10-15 years, new observations have been made about the link between naturally occurring mutations and pathological phenotypes particularly pertinent to cardiovascular diseases (CVD). These links have been attributed to a genetically driven increase of XOR expression and activity that is responsible for what is thought to be damaging uric acid (UA) and reactive oxygen species (ROS) accumulation, nitric oxide (·NO) depletion and endothelial dysfunction. In this review, we discuss the importance of genetics for interindividual variability of XOR expression and activity while focusing mainly on those variants thought to be relevant for CVD. In addition, we discuss the potential exploitation of the genetically driven increase of XOR activity to deliver more beneficial bioavailable ·NO. Finally, we examine the effect that non-synonymous mutations have on the tertiary structure of the protein and consequently on its capacity to interact with glycosaminoglycans (GAGs) localised on the outer surface of endothelial cells.
Collapse
Affiliation(s)
| | | | | | | | - Amrita Ahluwalia
- Barts & The London Faculty of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK; (G.M.); (N.D.); (F.O.); (R.S.K.)
| |
Collapse
|
7
|
Rathod KS, Mathur A, Shabbir A, Khambata RS, Lau C, Beirne AM, Chhetri I, Ono M, Belgaid DR, Massimo G, Ramasamy A, Tufaro V, Jain AK, Poulter N, Falaschetti E, Jones DA, Garcia-Garcia HM, Bourantas C, Learoyd A, Warren HR, Ahluwalia A. The NITRATE-OCT study-inorganic nitrate reduces in-stent restenosis in patients with stable coronary artery disease: a double-blind, randomised controlled trial. EClinicalMedicine 2024; 77:102885. [PMID: 39469537 PMCID: PMC11513660 DOI: 10.1016/j.eclinm.2024.102885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024] Open
Abstract
Background Coronary angioplasty and stent insertion is a first line treatment for patients with coronary artery disease, however it is complicated in the long-term by in-stent restenosis (ISR) in a proportion of patients with an associated morbidity. Despite this, currently there are no effective treatments available for the prevention of ISR. Repeat percutaneous revascularisation carries increased risks of major adverse cardiovascular events and a higher incidence of stent failure. In this study we report the efficacy of dietary inorganic nitrate in the prevention of ISR in a prospective, double-blind, randomised controlled trial. Methods NITRATE-OCT is a double-blind, randomised, single-centre, placebo-controlled phase II trial. 300 patients who were planned to undergo percutaneous coronary intervention (PCI) and drug eluting stent (DES) implantation for stable angina were randomised on a 1:1 basis to receive a daily dose of either dietary inorganic nitrate or placebo for 6 months. Block randomisation was used and patients stratified according to diabetes status. The patients then underwent quantitative coronary angiography (QCA) at baseline and at 6 months and optical coherence tomography at 6 months to quantify ISR. The primary endpoint was the QCA quantified decrease of in-stent/in-segment diameter from the baseline measure at 6 months i.e., in-stent and in-segment late-lumen loss (LLL). The study is registered with ClinicalTrials.gov, number NCT02529189. Findings From November 1st 2015 and March 31st 2020, NITRATE-OCT enrolled 300 patients with angina, with 150 each randomised to receive 70 mL of nitrate-containing beetroot juice or placebo (nitrate-deplete) juice for 6 months. Procedural characteristics were similar between the groups. The primary endpoint was available in 208 patients: 107 and 101 in the nitrate and placebo groups, respectively. There was a statistically significant effect of inorganic nitrate on both primary endpoints: in-stent LLL decreased by 0.16 mm (95% CI:0.06-0.25; P = 0.001) with mean = 0.09 ± 0.38 mm in the inorganic nitrate group versus 0.24 ± 0.33 mm in the placebo group; (P = 0.0052); and in-segment LLL decreased by 0.24 mm (95% CI:0.12-0.36; P < 0.001) with mean = 0.02 ± 0.52 mm in the inorganic nitrate group and 0.26 ± 0.37 mm in the placebo group (P = 0.0002). Inorganic nitrate treatment was associated with a rise in the plasma nitrate concentration of ∼6.1-fold and plasma nitrite (NO2 -) of ∼2.0-fold at 6 months. These rises were associated with sustained decreases in systolic blood pressure (SBP) at 6 months compared to baseline with a change SBP of -12.06 ± 15.88 mmHg compared to the placebo group of 2.52 ± 14.60 mmHg (P < 0.0001). Interpretation In patients who underwent PCI for stable coronary artery disease, a once-a-day oral inorganic nitrate treatment was associated with a significant decrease in both in-stent and in-segment LLL. Funding This trial and KSR was funded by the National Institute for Health and Care Research (NIHR) (DRF-2014-07-008) and NIHR ACL, HW and this study were supported by The NIHR Barts Biomedical Research Centre, IC was funded by The North and East London Clinical Research Network, CL, GM were funded by The Barts Charity Cardiovascular Programme MRG00913 and MO was funded by The British Heart Foundation Project Grant PG/19/4/33995.
Collapse
Affiliation(s)
- Krishnaraj S. Rathod
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Anthony Mathur
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Asad Shabbir
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rayomand S. Khambata
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Clement Lau
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Anne-Marie Beirne
- Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Ismita Chhetri
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Mutsumi Ono
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Gianmichele Massimo
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Vincenzo Tufaro
- Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Ajay K. Jain
- Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | - Neil Poulter
- Imperial College Trials Unit, London, United Kingdom
| | | | - Daniel A. Jones
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St. Bartholomew's Hospital, London, United Kingdom
| | | | | | - Anna Learoyd
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Helen R. Warren
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Amrita Ahluwalia
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
8
|
Saberi S, Najafipour H, Rajizadeh MA, Etminan A, Jafari E, Iranpour M. NaHS protects brain, heart, and lungs as remote organs from renal ischemia/reperfusion-induced oxidative stress in male and female rats. BMC Nephrol 2024; 25:373. [PMID: 39438873 PMCID: PMC11515705 DOI: 10.1186/s12882-024-03824-3] [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: 06/17/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Acute Kidney Injury (AKI) is frequently observed in hospitalized patients in intensive care units, often caused by renal ischemia-reperfusion injury (IRI). IRI disrupts the function of various 'remote organs' such as the lungs, pancreas, intestine, liver, heart, and brain through inflammation, oxidative stress, apoptosis, leukocyte infiltration, and increased urea and creatinine levels. Gender differences in renal IRI-induced injury are noted. H2S, an endogenous gaseous modulator, shows potential in vasodilation, bronchodilation, and hypotension and can regulate apoptosis, inflammation, angiogenesis, metabolism, and oxidative stress. This study aims to investigate the protective effects of NaHS on brain, heart, and lung injuries following renal IR and to assess the oxidative system status as a potential mechanism in male and female rats.Forty-eight Wistar rats were randomly divided into eight groups (n = 6): Control/Saline, Sham/Saline, IR/Saline, and IR/NaHS in both sexes. Forty-five minutes of bilateral renal ischemia followed by 24-hour reperfusion was induced in the IR groups. NaHS (100µM/Kg, IP) was administered 10 min before clamp release in treated groups. BUN, SCr, BUN/SCr, albuminuria, histopathology, and oxidative stress biomarkers of the brain, heart, and lung were assessed as remote organs. IR increased serum markers of renal function, albuminuria, malondialdehyde levels, and tissue injury scores while reducing nitrite levels and superoxide dismutase and glutathione peroxidase activities. NaHS treatment reversed the adverse effects of IR in remote organs in both sexes, although it showed limited improvement in renal function. Our findings demonstrate that NaHS has a beneficial effect on remote organ injury following renal IR by mitigating oxidative stress, with noted tissue-specific and gender-specific differences in response. These findings suggest NaHS as a potential therapeutic agent for mitigating multi-organ injury after renal IR, with effects varying by tissue and gender.
Collapse
Affiliation(s)
- Shadan Saberi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Hamid Najafipour
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Rajizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Endocrinology and Metabolism Research Center, Institute of basic and clinical physiology sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abbas Etminan
- Physiology Research Center, Departments of Nephrology, Urology and Renal Transplantation, Kerman University of Medical Sciences, Kerman, Iran.
| | - Elham Jafari
- Department of Pathology, Pathology and Stem Cell Research Center, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Iranpour
- Department of Pathology, Pathology and Stem Cell Research Center, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
9
|
Carlström M, Weitzberg E, Lundberg JO. Nitric Oxide Signaling and Regulation in the Cardiovascular System: Recent Advances. Pharmacol Rev 2024; 76:1038-1062. [PMID: 38866562 DOI: 10.1124/pharmrev.124.001060] [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: 02/02/2024] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
Nitric oxide (NO) from endothelial NO synthase importantly contributes to vascular homeostasis. Reduced NO production or increased scavenging during disease conditions with oxidative stress contribute to endothelial dysfunction and NO deficiency. In addition to the classical enzymatic NO synthases (NOS) system, NO can also be generated via the nitrate-nitrite-NO pathway. Dietary and pharmacological approaches aimed at increasing NO bioactivity, especially in the cardiovascular system, have been the focus of much research since the discovery of this small gaseous signaling molecule. Despite wide appreciation of the biological role of NOS/NO signaling, questions still remain about the chemical nature of NOS-derived bioactivity. Recent studies show that NO-like bioactivity can be efficiently transduced by mobile NO-ferroheme species, which can transfer between proteins, partition into a hydrophobic phase, and directly activate the soluble guanylyl cyclase-cGMP-protein kinase G pathway without intermediacy of free NO. Moreover, interaction between red blood cells and the endothelium in the regulation of vascular NO homeostasis have gained much attention, especially in conditions with cardiometabolic disease. In this review we discuss both classical and nonclassical pathways for NO generation in the cardiovascular system and how these can be modulated for therapeutic purposes. SIGNIFICANCE STATEMENT: After four decades of intensive research, questions persist about the transduction and control of nitric oxide (NO) synthase bioactivity. Here we discuss NO signaling in cardiovascular health and disease, highlighting new findings, such as the important role of red blood cells in cardiovascular NO homeostasis. Nonclassical signaling modes, like the nitrate-nitrite-NO pathway, and therapeutic opportunities related to the NO system are discussed. Existing and potential pharmacological treatments/strategies, as well as dietary components influencing NO generation and signaling are covered.
Collapse
Affiliation(s)
- Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| |
Collapse
|
10
|
Martinc B, Grabnar I, Milosheska D, Lorber B, Vovk T. A Cross-Sectional Study Comparing Oxidative Stress in Patients with Epilepsy Treated with Old and New Generation Antiseizure Medications. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1299. [PMID: 39202580 PMCID: PMC11356379 DOI: 10.3390/medicina60081299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: Oxidative stress resulting from a disturbance of the endogenous redox system is suspected in numerous diseases of the central nervous system, including epilepsy. In addition, antiseizure medications (ASMs), especially those of the old generation, may further increase oxidative stress. To evaluate the effects of ASM generation on oxidative stress, we conducted a cross-sectional study in patients with epilepsy treated with old, new, and polytherapy. Materials and Methods: The antioxidant activity of superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase, as well as the concentrations of malondialdehyde, protein carbonyl, nitrate, nitrite, and glutathione in reduced and oxidized forms, were measured in 49 patients with epilepsy and 14 healthy controls. In addition, the plasma concentrations of ASMs and metabolites of carbamazepine and valproic acid were measured in the patients. Results: Patients with epilepsy showed increased activities of superoxide dismutase and catalase (p < 0.001), concentrations of glutathione disulfide and markers of nitric oxide metabolism (p < 0.001), and decreased activities of glutathione peroxidase, glutathione reductase, glutathione, and nitrite concentrations (p ≤ 0.005) compared to healthy controls. A comparison of ASM generations revealed increased levels of superoxide dismutase and catalase (p ≤ 0.007) and decreased levels of glutathione peroxidase and glutathione reductase (p ≤ 0.01) in patients treated with old ASMs compared to those treated with new generation ASMs. In addition, an increase in protein carbonyl and nitric oxide metabolites (p ≤ 0.002) was observed in patients treated with old generation ASMs compared to those treated with new generation ASMs. Most oxidative stress parameters in patients receiving polytherapy with ASMs were intermediate between the results of patients treated with the old and new generations of ASMs. Conclusions: An increase in oxidative stress markers and modulation of antioxidant enzyme activities was observed in patients with epilepsy compared to controls. The results of our study showed significantly higher oxidative stress in patients treated with old ASMs compared to those treated with new generation ASMs.
Collapse
Affiliation(s)
- Boštjan Martinc
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.M.); (I.G.)
| | - Iztok Grabnar
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.M.); (I.G.)
| | - Daniela Milosheska
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.M.); (I.G.)
| | - Bogdan Lorber
- Department of Neurology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia;
| | - Tomaž Vovk
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.M.); (I.G.)
| |
Collapse
|
11
|
Korsmo HW, Ekperikpe US, Daehn IS. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:712. [PMID: 38929151 PMCID: PMC11200862 DOI: 10.3390/antiox13060712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Xanthine Oxidoreductase (XOR) is a ubiquitous, essential enzyme responsible for the terminal steps of purine catabolism, ultimately producing uric acid that is eliminated by the kidneys. XOR is also a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various physiological pathways, as well as contribute to the development and the progression of chronic conditions including kidney diseases, which are increasing in prevalence worldwide. XOR activity can promote oxidative distress, endothelial dysfunction, and inflammation through the biological effects of reactive oxygen species; nitric oxide and uric acid are the major products of XOR activity. However, the complex relationship of these reactions in disease settings has long been debated, and the environmental influences and genetics remain largely unknown. In this review, we give an overview of the biochemistry, biology, environmental, and current clinical impact of XOR in the kidney. Finally, we highlight recent genetic studies linking XOR and risk for kidney disease, igniting enthusiasm for future biomarker development and novel therapeutic approaches targeting XOR.
Collapse
Affiliation(s)
| | | | - Ilse S. Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1243, New York, NY 10029, USA
| |
Collapse
|
12
|
Delgado Spicuzza JM, Gosalia J, Zhong L, Bondonno C, Petersen KS, De Souza MJ, Alipour E, Kim-Shapiro DB, Somani YB, Proctor DN. Seven-day dietary nitrate supplementation clinically significantly improves basal macrovascular function in postmenopausal women: a randomized, placebo-controlled, double-blind, crossover clinical trial. Front Nutr 2024; 11:1359671. [PMID: 38915856 PMCID: PMC11194363 DOI: 10.3389/fnut.2024.1359671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/09/2024] [Indexed: 06/26/2024] Open
Abstract
Introduction Cardiovascular disease (CVD) is the leading cause of death in women, with increased risk following menopause. Dietary intake of beetroot juice and other plant-based nitrate-rich foods is a promising non-pharmacological strategy for increasing systemic nitric oxide and improving endothelial function in elderly populations. The purpose of this randomized, placebo-controlled, double-blind, crossover clinical trial was to determine the effects of short-term dietary nitrate (NO3 -) supplementation, in the form of beetroot juice, on resting macrovascular endothelial function and endothelial resistance to whole-arm ischemia-reperfusion (IR) injury in postmenopausal women at two distinct stages of menopause. Methods Early-postmenopausal [1-6 years following their final menstrual period (FMP), n = 12] and late-postmenopausal (6+ years FMP, n = 12) women consumed nitrate-rich (400 mg NO3 -/70 mL) and nitrate-depleted beetroot juice (approximately 40 mg NO3 -/70 mL, placebo) daily for 7 days. Brachial artery flow-mediated dilation (FMD) was measured pre-supplementation (Day 0), and approximately 24 h after the last beetroot juice (BR) dose (Day 8, post-7-day BR). Consequently, FMD was measured immediately post-IR injury and 15 min later (recovery). Results Results of the linear mixed-effects model revealed a significantly greater increase in resting FMD with 7 days of BRnitrate compared to BRplacebo (mean difference of 2.21, 95% CI [0.082, 4.34], p = 0.042); however, neither treatment blunted the decline in post-IR injury FMD in either postmenopausal group. Our results suggest that 7-day BRnitrate-mediated endothelial protection is lost within the 24-h period following the final dose of BRnitrate. Conclusion Our findings demonstrate that nitrate-mediated postmenopausal endothelial protection is dependent on the timing of supplementation in relation to IR injury and chronobiological variations in dietary nitrate metabolism. Clinical trial registration https://classic.clinicaltrials.gov/ct2/show/NCT03644472.
Collapse
Affiliation(s)
- Jocelyn M. Delgado Spicuzza
- Integrative Vascular Physiology Lab, Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Jigar Gosalia
- Integrative Vascular Physiology Lab, Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| | - Liezhou Zhong
- Nutrition and Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Catherine Bondonno
- Nutrition and Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Kristina S. Petersen
- Cardiometabolic Nutrition Research Lab, Department of Nutritional Sciences, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| | - Mary Jane De Souza
- Integrative Vascular Physiology Lab, Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
- Women’s Health and Exercise Lab, Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| | - Elmira Alipour
- Department of Physics, Wake Forest University, Winston-Salem, NC, United States
| | | | - Yasina B. Somani
- Faculty of Biological Sciences, Department of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - David N. Proctor
- Integrative Vascular Physiology Lab, Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
- Integrative Vascular Physiology Lab, Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
13
|
Silva-Cunha M, Lacchini R, Tanus-Santos JE. Facilitating Nitrite-Derived S-Nitrosothiol Formation in the Upper Gastrointestinal Tract in the Therapy of Cardiovascular Diseases. Antioxidants (Basel) 2024; 13:691. [PMID: 38929130 PMCID: PMC11200996 DOI: 10.3390/antiox13060691] [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: 04/23/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are often associated with impaired nitric oxide (NO) bioavailability, a critical pathophysiological alteration in CVDs and an important target for therapeutic interventions. Recent studies have revealed the potential of inorganic nitrite and nitrate as sources of NO, offering promising alternatives for managing various cardiovascular conditions. It is now becoming clear that taking advantage of enzymatic pathways involved in nitrite reduction to NO is very relevant in new therapeutics. However, recent studies have shown that nitrite may be bioactivated in the acidic gastric environment, where nitrite generates NO and a variety of S-nitrosating compounds that result in increased circulating S-nitrosothiol concentrations and S-nitrosation of tissue pharmacological targets. Moreover, transnitrosation reactions may further nitrosate other targets, resulting in improved cardiovascular function in patients with CVDs. In this review, we comprehensively address the mechanisms and relevant effects of nitrate and nitrite-stimulated gastric S-nitrosothiol formation that may promote S-nitrosation of pharmacological targets in various CVDs. Recently identified interfering factors that may inhibit these mechanisms and prevent the beneficial responses to nitrate and nitrite therapy were also taken into consideration.
Collapse
Affiliation(s)
- Mila Silva-Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil;
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto 14040-902, Brazil;
| | - Jose E. Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil;
| |
Collapse
|
14
|
Webb AJ. "Every beet you take": lowering systolic blood pressure and improving vascular function/exercise capacity via the dietary nitrate-nitrite-NO pathway in patients with COPD. Eur Respir J 2024; 63:2302238. [PMID: 38302179 DOI: 10.1183/13993003.02238-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 02/03/2024]
Affiliation(s)
- Andrew J Webb
- Department of Clinical Pharmacology, King's College London British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, London, UK
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
15
|
Majerczak J, Drzymala‐Celichowska H, Grandys M, Kij A, Kus K, Celichowski J, Krysciak K, Molik WA, Szkutnik Z, Zoladz JA. Exercise Training Decreases Nitrite Concentration in the Heart and Locomotory Muscles of Rats Without Changing the Muscle Nitrate Content. J Am Heart Assoc 2024; 13:e031085. [PMID: 38214271 PMCID: PMC10926815 DOI: 10.1161/jaha.123.031085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Skeletal muscles are postulated to be a potent regulator of systemic nitric oxide homeostasis. In this study, we aimed to evaluate the impact of physical training on the heart and skeletal muscle nitric oxide bioavailability (judged on the basis of intramuscular nitrite and nitrate) in rats. METHODS AND RESULTS Rats were trained on a treadmill for 8 weeks, performing mainly endurance running sessions with some sprinting runs. Muscle nitrite (NO2-) and nitrate (NO3-) concentrations were measured using a high-performance liquid chromatography-based method, while amino acids, pyruvate, lactate, and reduced and oxidized glutathione were determined using a liquid chromatography coupled with tandem mass spectrometry technique. The content of muscle nitrite reductases (electron transport chain proteins, myoglobin, and xanthine oxidase) was assessed by western immunoblotting. We found that 8 weeks of endurance training decreased basal NO2- in the locomotory muscles and in the heart, without changes in the basal NO3-. In the slow-twitch oxidative soleus muscle, the decrease in NO2- was already present after the first week of training, and the content of nitrite reductases remained unchanged throughout the entire period of training, except for the electron transport chain protein content, which increased no sooner than after 8 weeks of training. CONCLUSIONS Muscle NO2- level, opposed to NO3-, decreases in the time course of training. This effect is rapid and already visible in the slow-oxidative soleus after the first week of training. The underlying mechanisms of training-induced muscle NO2- decrease may involve an increase in the oxidative stress, as well as metabolite changes related to an increased muscle anaerobic glycolytic activity contributing to (1) direct chemical reduction of NO2- or (2) activation of muscle nitrite reductases.
Collapse
Affiliation(s)
- Joanna Majerczak
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health SciencesJagiellonian University Medical CollegeKrakowPoland
| | - Hanna Drzymala‐Celichowska
- Department of Neurobiology, Faculty of Health SciencesPoznan University of Physical EducationPoznanPoland
- Department of Physiology and Biochemistry, Faculty of Health SciencesPoznan University of Physical EducationPoznanPoland
| | - Marcin Grandys
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health SciencesJagiellonian University Medical CollegeKrakowPoland
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET)Jagiellonian UniversityKrakowPoland
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET)Jagiellonian UniversityKrakowPoland
| | - Jan Celichowski
- Department of Neurobiology, Faculty of Health SciencesPoznan University of Physical EducationPoznanPoland
| | - Katarzyna Krysciak
- Department of Neurobiology, Faculty of Health SciencesPoznan University of Physical EducationPoznanPoland
| | - Weronika A. Molik
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health SciencesJagiellonian University Medical CollegeKrakowPoland
- University of FloridaGainesvilleFLUSA
| | | | - Jerzy A. Zoladz
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health SciencesJagiellonian University Medical CollegeKrakowPoland
| |
Collapse
|
16
|
Yassaghi Y, Jeddi S, Kashfi K, Ghasemi A. Myocardial infarct size is reduced by nitrite and nitrate administration: a systematic review and meta-analysis of animal studies. EXCLI JOURNAL 2024; 23:18-33. [PMID: 38357094 PMCID: PMC10864704 DOI: 10.17179/excli2023-6740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/18/2023] [Indexed: 02/16/2024]
Abstract
Ischemic heart disease (IHD) is the leading cause of mortality worldwide and can be complicated by myocardial infarction (MI), leading to cardiac failure. Inorganic nitrite and nitrate, which release nitric oxide (NO), can protect the heart against myocardial injury. This animal systematic review and meta-analysis aims to assess whether the administration of nitrite/nitrate decreases myocardial infarct size. We systematically searched PubMed, Scopus, and Web of Science databases until October 2023; 15 eligible animal studies (35 study arms for in-vivo and 10 for in-vitro studies) published between 1989 and 2023 were included. In-vivo studies were conducted on rats, mice, cats, and dogs, and in-vitro studies on rats and mice with an overall exposure of 0.03 to 12713 mg/kg to nitrate/nitrite administrated before, after, or during ischemia mainly by intravenous single bolus or by oral over 270 days. All in-vitro studies used nitrite/nitrate before ischemia, with the concentration ranging between 0.34 to 201 μM. MI was induced by occlusion of the left anterior diagonal or left circumflex arteries in in-vitro studies and by isoproterenol in in-vivo studies. Infarct size was measured by direct staining of the sliced heart sections. In in-vivo studies, nitrite (overall effect size (ES)=-17.0 %, 95 % confidence interval (CI)=-21.3, -12.8, P<0.001) and nitrate (overall ES= -9.6 %, 95 % CI=-15.7, -3.4, P=0.002) reduced myocardial infarct size. In in-vitro studies, nitrite (overall ES=-15.8 %, 95 % CI=-25.5, -6.2, P=0.001) reduced the infarct size. Sensitivity analysis showed that the overall effect of nitrite on myocardial infarct size was unaffected by doses or health conditions in in-vivo and in-vitro studies. In conclusion, our meta-analysis showed that nitrite/nitrate administration can effectively reduce myocardial infarct size. However, these results should be approached with caution because of the limitations of animal studies and the existing high heterogeneity.
Collapse
Affiliation(s)
- Younes Yassaghi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Mollace R, Scarano F, Bava I, Carresi C, Maiuolo J, Tavernese A, Gliozzi M, Musolino V, Muscoli S, Palma E, Muscoli C, Salvemini D, Federici M, Macrì R, Mollace V. Modulation of the nitric oxide/cGMP pathway in cardiac contraction and relaxation: Potential role in heart failure treatment. Pharmacol Res 2023; 196:106931. [PMID: 37722519 DOI: 10.1016/j.phrs.2023.106931] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Evidence exists that heart failure (HF) has an overall impact of 1-2 % in the global population being often associated with comorbidities that contribute to increased disease prevalence, hospitalization, and mortality. Recent advances in pharmacological approaches have significantly improved clinical outcomes for patients with vascular injury and HF. Nevertheless, there remains an unmet need to clarify the crucial role of nitric oxide/cyclic guanosine 3',5'-monophosphate (NO/cGMP) signalling in cardiac contraction and relaxation, to better identify the key mechanisms involved in the pathophysiology of myocardial dysfunction both with reduced (HFrEF) as well as preserved ejection fraction (HFpEF). Indeed, NO signalling plays a crucial role in cardiovascular homeostasis and its dysregulation induces a significant increase in oxidative and nitrosative stress, producing anatomical and physiological cardiac alterations that can lead to heart failure. The present review aims to examine the molecular mechanisms involved in the bioavailability of NO and its modulation of downstream pathways. In particular, we focus on the main therapeutic targets and emphasize the recent evidence of preclinical and clinical studies, describing the different emerging therapeutic strategies developed to counteract NO impaired signalling and cardiovascular disease (CVD) development.
Collapse
Affiliation(s)
- Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Irene Bava
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Jessica Maiuolo
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Saverio Muscoli
- Division of Cardiology, Foundation PTV Polyclinic Tor Vergata, Rome 00133, Italy
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Italy
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy.
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro 88100, Italy; Renato Dulbecco Institute, Lamezia Terme, Catanzaro 88046, Italy.
| |
Collapse
|
18
|
Martins LZ, da Silva MLS, Rodrigues SD, Gomes SEB, Molezini L, Rizzi E, Montenegro MF, Dias-Junior CA. Sodium Nitrite Attenuates Reduced Activity of Vascular Matrix Metalloproteinase-2 and Vascular Hyper-Reactivity and Increased Systolic Blood Pressure Induced by the Placental Ischemia Model of Preeclampsia in Anesthetized Rats. Int J Mol Sci 2023; 24:12818. [PMID: 37628999 PMCID: PMC10454117 DOI: 10.3390/ijms241612818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Preeclampsia is a maternal hypertension disorder associated with vascular dysfunction and fetal and placental growth restrictions. Placental ischemia is suggested as the primary trigger of preeclampsia-associated impairments of both endothelium-derived nitric oxide (NO) and the vascular activity of extracellular matrix metalloproteinase-2 (MMP-2). Reduced uteroplacental perfusion pressure (RUPP) is a placental ischemia model of preeclampsia. Reduction of sodium nitrite to NO may occur during ischemic conditions. However, sodium nitrite effects in the RUPP model of preeclampsia have not yet been investigated. Pregnant rats were divided into four groups: normotensive pregnant rats (Norm-Preg), pregnant rats treated with sodium nitrite (Preg + Nitrite), preeclamptic rats (RUPP), and preeclamptic rats treated with sodium nitrite (RUPP + Nitrite). Maternal blood pressure and fetal and placental parameters were recorded. Vascular function, circulating NO metabolites, and the gelatinolytic activity of vascular MMP-2 were also examined. Sodium nitrite attenuates increased blood pressure, prevents fetal and placental weight loss, counteracts vascular hyper-reactivity, and partially restores NO metabolites and MMP-2 activity. In conclusion, sodium nitrite reduction to NO may occur during RUPP-induced placental ischemia, thereby attenuating increased blood pressure, fetal and placental growth restriction, and vascular hyper-reactivity associated with preeclampsia and possibly restoring NO and MMP-2 activity, which underlie the blood pressure-lowering effects.
Collapse
Affiliation(s)
- Laisla Zanetoni Martins
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, Brazil; (L.Z.M.); (M.L.S.d.S.); (S.D.R.); (S.E.B.G.)
| | - Maria Luiza Santos da Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, Brazil; (L.Z.M.); (M.L.S.d.S.); (S.D.R.); (S.E.B.G.)
| | - Serginara David Rodrigues
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, Brazil; (L.Z.M.); (M.L.S.d.S.); (S.D.R.); (S.E.B.G.)
| | - Sáskia Estela Biasotti Gomes
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, Brazil; (L.Z.M.); (M.L.S.d.S.); (S.D.R.); (S.E.B.G.)
| | - Laura Molezini
- Unit of Biotechnology, University of Ribeirao Preto (UNAERP), Ribeirao Preto 14096-900, Brazil; (L.M.); (E.R.)
| | - Elen Rizzi
- Unit of Biotechnology, University of Ribeirao Preto (UNAERP), Ribeirao Preto 14096-900, Brazil; (L.M.); (E.R.)
| | - Marcelo Freitas Montenegro
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 10691 Stockholm, Sweden;
| | - Carlos Alan Dias-Junior
- Department of Biophysics and Pharmacology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, Brazil; (L.Z.M.); (M.L.S.d.S.); (S.D.R.); (S.E.B.G.)
| |
Collapse
|
19
|
Maia LB. Bringing Nitric Oxide to the Molybdenum World-A Personal Perspective. Molecules 2023; 28:5819. [PMID: 37570788 PMCID: PMC10420851 DOI: 10.3390/molecules28155819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Molybdenum-containing enzymes of the xanthine oxidase (XO) family are well known to catalyse oxygen atom transfer reactions, with the great majority of the characterised enzymes catalysing the insertion of an oxygen atom into the substrate. Although some family members are known to catalyse the "reverse" reaction, the capability to abstract an oxygen atom from the substrate molecule is not generally recognised for these enzymes. Hence, it was with surprise and scepticism that the "molybdenum community" noticed the reports on the mammalian XO capability to catalyse the oxygen atom abstraction of nitrite to form nitric oxide (NO). The lack of precedent for a molybdenum- (or tungsten) containing nitrite reductase on the nitrogen biogeochemical cycle contributed also to the scepticism. It took several kinetic, spectroscopic and mechanistic studies on enzymes of the XO family and also of sulfite oxidase and DMSO reductase families to finally have wide recognition of the molybdoenzymes' ability to form NO from nitrite. Herein, integrated in a collection of "personal views" edited by Professor Ralf Mendel, is an overview of my personal journey on the XO and aldehyde oxidase-catalysed nitrite reduction to NO. The main research findings and the path followed to establish XO and AO as competent nitrite reductases are reviewed. The evidence suggesting that these enzymes are probable players of the mammalian NO metabolism is also discussed.
Collapse
Affiliation(s)
- Luisa B Maia
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology (FCT NOVA), 2829-516 Caparica, Portugal
| |
Collapse
|
20
|
Mace EH, Kimlinger MJ, Billings FT, Lopez MG. Targeting Soluble Guanylyl Cyclase during Ischemia and Reperfusion. Cells 2023; 12:1903. [PMID: 37508567 PMCID: PMC10378692 DOI: 10.3390/cells12141903] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Ischemia and reperfusion (IR) damage organs and contribute to many disease states. Few effective treatments exist that attenuate IR injury. The augmentation of nitric oxide (NO) signaling remains a promising therapeutic target for IR injury. NO binds to soluble guanylyl cyclase (sGC) to regulate vasodilation, maintain endothelial barrier integrity, and modulate inflammation through the production of cyclic-GMP in vascular smooth muscle. Pharmacologic sGC stimulators and activators have recently been developed. In preclinical studies, sGC stimulators, which augment the reduced form of sGC, and activators, which activate the oxidized non-NO binding form of sGC, increase vasodilation and decrease cardiac, cerebral, renal, pulmonary, and hepatic injury following IR. These effects may be a result of the improved regulation of perfusion and decreased oxidative injury during IR. sGC stimulators are now used clinically to treat some chronic conditions such as heart failure and pulmonary hypertension. Clinical trials of sGC activators have been terminated secondary to adverse side effects including hypotension. Additional clinical studies to investigate the effects of sGC stimulation and activation during acute conditions, such as IR, are warranted.
Collapse
Affiliation(s)
- Eric H Mace
- Department of Surgery, Vanderbilt University Medical Center, Medical Center North, Suite CCC-4312, 1161 21st Avenue South, Nashville, TN 37232-2730, USA
| | - Melissa J Kimlinger
- Vanderbilt University School of Medicine, 428 Eskind Family Biomedical Library and Learning Center, Nashville, TN 37240-0002, USA
| | - Frederic T Billings
- Department of Anesthesiology, Division of Critical Care Medicine, Vanderbilt University Medical Center, Medical Arts Building, Suite 422, 1211 21st Avenue South, Nashville, TN 37212-1750, USA
| | - Marcos G Lopez
- Department of Anesthesiology, Division of Critical Care Medicine, Vanderbilt University Medical Center, Medical Arts Building, Suite 422, 1211 21st Avenue South, Nashville, TN 37212-1750, USA
| |
Collapse
|
21
|
Liu Y, Croft KD, Mori TA, Gaspari TA, Kemp-Harper BK, Ward NC. Long-term dietary nitrate supplementation slows the progression of established atherosclerosis in ApoE -/- mice fed a high fat diet. Eur J Nutr 2023; 62:1845-1857. [PMID: 36853380 PMCID: PMC10195750 DOI: 10.1007/s00394-023-03127-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/20/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is associated with a reduction in the bioavailability and/or bioactivity of endogenous nitric oxide (NO). Dietary nitrate has been proposed as an alternate source when endogenous NO production is reduced. Our previous study demonstrated a protective effect of dietary nitrate on the development of atherosclerosis in the apoE-/- mouse model. However most patients do not present clinically until well after the disease is established. The aims of this study were to determine whether chronic dietary nitrate supplementation can prevent or reverse the progression of atherosclerosis after disease is already established, as well as to explore the underlying mechanism of these cardiovascular protective effects. METHODS 60 apoE-/- mice were given a high fat diet (HFD) for 12 weeks to allow for the development of atherosclerosis. The mice were then randomized to (i) control group (HFD + 1 mmol/kg/day NaCl), (ii) moderate-dose group (HFD +1 mmol/kg/day NaNO3), or (iii) high-dose group (HFD + 10 mmol/kg/day NaNO3) (20/group) for a further 12 weeks. A group of apoE-/- mice (n = 20) consumed a normal laboratory chow diet for 24 weeks and were included as a reference group. RESULTS Long-term supplementation with high dose nitrate resulted in ~ 50% reduction in plaque lesion area. Collagen expression and smooth muscle accumulation were increased, and lipid deposition and macrophage accumulation were reduced within atherosclerotic plaques of mice supplemented with high dose nitrate. These changes were associated with an increase in nitrite reductase as well as activation of the endogenous eNOS-NO pathway. CONCLUSION Long-term high dose nitrate significantly attenuated the progression of established atherosclerosis in the apoE-/- mice fed a HFD. This appears to be mediated in part through a XOR-dependent reduction of nitrate to NO, as well as enhanced eNOS activation via increased Akt and eNOS phosphorylation.
Collapse
Affiliation(s)
- Yang Liu
- School of Biomedical Sciences, University of Western Australia, Perth, WA Australia
| | - Kevin D. Croft
- School of Biomedical Sciences, University of Western Australia, Perth, WA Australia
| | - Trevor A. Mori
- Medical School, University of Western Australia, Perth, WA Australia
| | - Tracey A. Gaspari
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC Australia
| | - Barbara K. Kemp-Harper
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC Australia
| | - Natalie C. Ward
- Medical School, University of Western Australia, Perth, WA Australia
- Dobney Hypertension Centre, Medical School, University of Western Australia, G.P.O Box X2213, Perth, WA 6847 Australia
| |
Collapse
|
22
|
Dou C, Han X, Xie H, Liao H, Xiao X, Huang Z, Luo G, Zhang X, Yao W. Protective role of nitric oxide donors on endothelium in ischemia-reperfusion injury: a meta-analysis of randomized controlled trials. BMC Anesthesiol 2023; 23:189. [PMID: 37259069 DOI: 10.1186/s12871-023-02117-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 04/29/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Decreased bioavailability of nitric oxide (NO) under hypoxic conditions can lead to endothelial dysfunction. NO supplementation may protect endothelial function in ischemia-reperfusion (IR) injury. Therefore, a meta-analysis of randomized controlled trials (RCTs) was performed to verify the protective effect of NO donors on endothelium in IR injury. METHODS Medline, Embase, Cochrane Library, and Web of Science databases were searched from inception to April 1, 2023. The specific inclusion criteria were as follows: (1) RCTs; (2) trials comparing NO donors with placebo control groups; and (3) trials reporting the effects of these interventions on vascular endothelial functional outcomes in IR injury. Random-effects models were used to assess pooled effect sizes, which were expressed as standardized mean differences (SMD). RESULTS Seven studies satisfied the inclusion criteria and consisted of a total of 149 participants. NO donors were protective of endothelial function in IR injury (SMD: - 1.60; 95% confidence interval [CI]: - 2.33, - 0.88, P < 0.0001; heterogeneity [I2 = 66%, P = 0.001]). Results of the subgroup analysis showed the following: absence of protective effect of NO donor use following ischemia on endothelial function in IR injury - 1.78 (95% CI: - 2.50, - 1.07) and loss of protective effect on endothelial function after prolonged NO donor use - 0.89 (95% CI: - 2.06, 0.28). CONCLUSION The short-period use of NO donors before the onset of ischemia can protect endothelial function in IR injury.
Collapse
Grants
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
- 81974081,81601724,2021A1515012318, 2019A1515011852,202201010765, 202102010190, National Natural Science Foundation of China ,Guangdong Basic and Applied Basic Research Foundation,Science and Technology Program of Guangzhou, China
Collapse
Affiliation(s)
- Chaoxun Dou
- Department of Anesthesiology, The third Affiliated hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xue Han
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Hanbin Xie
- Department of Anesthesiology, The third Affiliated hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Haofeng Liao
- Department of Anesthesiology, The third Affiliated hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xue Xiao
- Department of Anesthesiology, The third Affiliated hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Ziyan Huang
- Department of Anesthesiology, The third Affiliated hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Gangjian Luo
- Department of Anesthesiology, The third Affiliated hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xinmin Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Weifeng Yao
- Department of Anesthesiology, The third Affiliated hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
23
|
Maiuolo J, Oppedisano F, Carresi C, Gliozzi M, Musolino V, Macrì R, Scarano F, Coppoletta A, Cardamone A, Bosco F, Mollace R, Muscoli C, Palma E, Mollace V. The Generation of Nitric Oxide from Aldehyde Dehydrogenase-2: The Role of Dietary Nitrates and Their Implication in Cardiovascular Disease Management. Int J Mol Sci 2022; 23:ijms232415454. [PMID: 36555095 PMCID: PMC9779284 DOI: 10.3390/ijms232415454] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Reduced bioavailability of the nitric oxide (NO) signaling molecule has been associated with the onset of cardiovascular disease. One of the better-known and effective therapies for cardiovascular disorders is the use of organic nitrates, such as glyceryl trinitrate (GTN), which increases the concentration of NO. Unfortunately, chronic use of this therapy can induce a phenomenon known as "nitrate tolerance", which is defined as the loss of hemodynamic effects and a reduction in therapeutic effects. As such, a higher dosage of GTN is required in order to achieve the same vasodilatory and antiplatelet effects. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is a cardioprotective enzyme that catalyzes the bio-activation of GTN to NO. Nitrate tolerance is accompanied by an increase in oxidative stress, endothelial dysfunction, and sympathetic activation, as well as a loss of the catalytic activity of ALDH2 itself. On the basis of current knowledge, nitrate intake in the diet would guarantee a concentration of NO such as to avoid (or at least reduce) treatment with GTN and the consequent onset of nitrate tolerance in the course of cardiovascular diseases, so as not to make necessary the increase in GTN concentrations and the possible inhibition/alteration of ALDH2, which aggravates the problem of a positive feedback mechanism. Therefore, the purpose of this review is to summarize data relating to the introduction into the diet of some natural products that could assist pharmacological therapy in order to provide the NO necessary to reduce the intake of GTN and the phenomenon of nitrate tolerance and to ensure the correct catalytic activity of ALDH2.
Collapse
Affiliation(s)
- Jessica Maiuolo
- Pharmaceutical Biology Laboratory, in Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Correspondence: (J.M.); (F.O.)
| | - Francesca Oppedisano
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Correspondence: (J.M.); (F.O.)
| | - Cristina Carresi
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, in Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Roberta Macrì
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Scarano
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Annarita Coppoletta
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Antonio Cardamone
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Bosco
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Rocco Mollace
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Ernesto Palma
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Renato Dulbecco Institute, Lamezia Terme, 88046 Catanzaro, Italy
| |
Collapse
|
24
|
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]
|
25
|
Gao J, Cheng Z, Jiang S, Wills M, Wehbe A, Rajah GB, Geng X, Ding Y. Arterial Glyceryl Trinitrate in Acute Ischemic Stroke after Thrombectomy for Neuroprotection (AGAIN): Rationale, design and protocol for a prospective randomized controlled trial. BMC Geriatr 2022; 22:804. [PMID: 36253714 PMCID: PMC9575243 DOI: 10.1186/s12877-022-03506-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although endovascular recanalization therapy demonstrates robust clinical efficacy in acute ischemic stroke (AIS), not all victims of these cerebrovascular accidents can benefit from it and achieve a favorable prognosis after successful reperfusion. Therefore, alternative neuroprotective strategies are urgently needed for AIS patients after vessel recanalization. Nitric oxide (NO) levels are low after AIS and NO donor drugs may be neuroprotective against cerebral ischemia-reperfusion injury. Glyceryl trinitrate (GTN), often used in the clinic as a NO donor, may provide a novel neuroprotective strategy. This rationale, design, and protocol for a prospective pilot study plans to explore the preliminary safety, feasibility, and neuroprotective benefits of Arterial Glyceryl Trinitrate in Acute Ischemic Stroke after Thrombectomy for Neuroprotection (AGAIN). METHODS AGAIN, a prospective RCT, is proposed for AIS patients after mechanical thrombectomy. Subjects will be randomly assigned in a 1:1 fashion (n = 40) to either the control group or the intervention group. Participants assigned to the intervention group will be administered 800 μg GTN in the catheter immediately after recanalization, whereas those in the control group will be administered the same volume of normal saline. All participants from either group will be given concurrent treatment with standard of care therapies in accordance with the current guidelines for stroke management. The primary outcome is safety [symptomatic intracranial hemorrhage (ICH), hypotension, neurological deterioration, ICH, fatal ICH, as well as headache, tachycardia, emesis, and seizures], whereas secondary outcomes included changes in poststroke functional outcomes, infarction volumes, and blood nitrate index detection. DISCUSSIONS This study is a prospective randomized controlled trial to test the safety and efficacy of intra-arterial GTN in AIS patients after endovascular therapy. The results from this study will give insight for future GTN studies and new neuroprotective strategies for future AIS treatment strategies. TRIAL REGISTRATION NUMBER ChiCTR2100045254. Registered on March 21, 2021.
Collapse
Affiliation(s)
- Jie Gao
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Road, Tongzhou District, Beijing, 101149, China
| | - Zhe Cheng
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Road, Tongzhou District, Beijing, 101149, China
| | - Shangqian Jiang
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Road, Tongzhou District, Beijing, 101149, China
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, 101100, China
| | - Melissa Wills
- Department of Neurosurgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
| | - Alexandra Wehbe
- Department of Neurosurgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, 02138, USA
| | - Gary B Rajah
- Department of Neurosurgery, Munson Healthcare, Traverse City, MI, USA
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, Department of Neurosurgery, University at Buffalo, Gates Vascular Institute at Kaleida Health, Buffalo, NY, USA
| | - Xiaokun Geng
- Department of Neurology and Stroke Center, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Road, Tongzhou District, Beijing, 101149, China.
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, 101100, China.
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA.
| |
Collapse
|
26
|
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]
|
27
|
Quantitative aspects of nitric oxide production in the heart. Mol Biol Rep 2022; 49:11113-11122. [DOI: 10.1007/s11033-022-07889-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022]
|
28
|
On the Role of Dietary Nitrate in the Maintenance of Systemic and Oral Health. Dent J (Basel) 2022; 10:dj10050084. [PMID: 35621537 PMCID: PMC9139378 DOI: 10.3390/dj10050084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 01/27/2023] Open
Abstract
The assessment of the significance of nitrates ingested with food has undergone a fundamental change in recent years after many controversial discussions. While for a long time, a diet as low in nitrates as possible was advocated on the basis of epidemiological data suggesting a cancer-promoting effect of nitrate-rich diets, more recent findings show that dietary nitrate, after its conversion to nitrite by nitrate-reducing bacteria of the oral microbiota, is an indispensable alternative source for the formation of nitric oxide (NO), which comprises a key element in the physiology of a variety of central body functions such as blood pressure control, defense against invading bacteria and maintenance of a eubiotic microbiota in the gut and oral cavity. This compact narrative review aims to present the evidence supported by clinical and in vitro studies on the ambivalent nature of dietary nitrates for general and oral health and to explain how the targeted adjuvant use of nitrate-rich diets could open new opportunities for a more cause-related control of caries and periodontal disease.
Collapse
|
29
|
Jeddi S, Gheibi S, Afzali H, Carlström M, Kashfi K, Ghasemi A. Hydrogen sulfide potentiates the protective effects of nitrite against myocardial ischemia-reperfusion injury in type 2 diabetic rats. Nitric Oxide 2022; 124:15-23. [PMID: 35504499 DOI: 10.1016/j.niox.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/06/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
|
30
|
Bajic Z, Sobot T, Skrbic R, Stojiljkovic MP, Ponorac N, Matavulj A, Djuric DM. Homocysteine, Vitamins B6 and Folic Acid in Experimental Models of Myocardial Infarction and Heart Failure—How Strong Is That Link? Biomolecules 2022; 12:biom12040536. [PMID: 35454125 PMCID: PMC9027107 DOI: 10.3390/biom12040536] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death and the main cause of disability. In the last decade, homocysteine has been found to be a risk factor or a marker for cardiovascular diseases, including myocardial infarction (MI) and heart failure (HF). There are indications that vitamin B6 plays a significant role in the process of transsulfuration in homocysteine metabolism, specifically, in a part of the reaction in which homocysteine transfers a sulfhydryl group to serine to form α-ketobutyrate and cysteine. Therefore, an elevated homocysteine concentration (hyperhomocysteinemia) could be a consequence of vitamin B6 and/or folate deficiency. Hyperhomocysteinemia in turn could damage the endothelium and the blood vessel wall and induce worsening of atherosclerotic process, having a negative impact on the mechanisms underlying MI and HF, such as oxidative stress, inflammation, and altered function of gasotransmitters. Given the importance of the vitamin B6 in homocysteine metabolism, in this paper, we review its role in reducing oxidative stress and inflammation, influencing the functions of gasotransmitters, and improving vasodilatation and coronary flow in animal models of MI and HF.
Collapse
Affiliation(s)
- Zorislava Bajic
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Tanja Sobot
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Ranko Skrbic
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (R.S.); (M.P.S.)
| | - Milos P. Stojiljkovic
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (R.S.); (M.P.S.)
| | - Nenad Ponorac
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Amela Matavulj
- Department of Physiology, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina; (Z.B.); (T.S.); (N.P.); (A.M.)
| | - Dragan M. Djuric
- Faculty of Medicine, Institute of Medical Physiology “Richard Burian”, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
31
|
Stamm P, Kalinovic S, Oelze M, Steven S, Czarnowski A, Kvandova M, Bayer F, Reinhardt C, Münzel T, Daiber A. Mechanistic Insights into Inorganic Nitrite-Mediated Vasodilation of Isolated Aortic Rings under Oxidative/Hypertensive Conditions and S-Nitros(yl)ation of Proteins in Germ-Free Mice. Biomedicines 2022; 10:biomedicines10030730. [PMID: 35327532 PMCID: PMC8945819 DOI: 10.3390/biomedicines10030730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 02/07/2023] Open
Abstract
The prevalence and clinical importance of arterial hypertension are still growing. Inorganic nitrite (NO2-) represents an attractive dietary antihypertensive agent, but its metabolism and mode of action, which we aimed to investigate with the present study, are not completely understood. Isolated aortic rings from rats were treated ex vivo with oxidants, and rats were infused in vivo with angiotensin-II. Vascular responses to acetylcholine (ACh) and nitrite were assessed by isometric tension recording. The loss of vasodilatory potency in response to oxidants was much more pronounced for ACh as compared to nitrite ex vivo (but not in vivo with angiotensin-II). This effect may be caused by the redox regulation of conversion to xanthine oxidase (XO). Conventionally raised and germ-free mice were treated with nitrite by gavage, which did not improve ACh-mediated vasodilation, but did increase the plasma levels of S-nitros(yl)ated proteins in the conventionally-raised, but not in the germ-free mice. In conclusion, inorganic nitrite represents a dietary drug option to treat arterial hypertension in addition to already established pharmacological treatment. Short-term oxidative stress did not impair the vasodilatory properties of nitrite, which may be beneficial in cardiovascular disease patients. The gastrointestinal microbiome appears to play a key role in nitrite metabolism and bioactivation.
Collapse
Affiliation(s)
- Paul Stamm
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany;
| | - Sanela Kalinovic
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
| | - Matthias Oelze
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
| | - Sebastian Steven
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, 55131 Mainz, Germany;
| | - Alexander Czarnowski
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
| | - Miroslava Kvandova
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
| | - Franziska Bayer
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, 55131 Mainz, Germany;
| | - Christoph Reinhardt
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany;
- Center for Thrombosis and Hemostasis Mainz, University Medical Center Mainz, 55131 Mainz, Germany;
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany;
- Correspondence: (T.M.); (A.D.); Tel.: +49-6131-17-6280 (A.D.)
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center Mainz, 55131 Mainz, Germany; (P.S.); (S.K.); (M.O.); (S.S.); (A.C.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany;
- Correspondence: (T.M.); (A.D.); Tel.: +49-6131-17-6280 (A.D.)
| |
Collapse
|
32
|
Nitrite Concentration in the Striated Muscles Is Reversely Related to Myoglobin and Mitochondrial Proteins Content in Rats. Int J Mol Sci 2022; 23:ijms23052686. [PMID: 35269826 PMCID: PMC8910716 DOI: 10.3390/ijms23052686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscles are an important reservoir of nitric oxide (NO•) stored in the form of nitrite [NO2−] and nitrate [NO3−] (NOx). Nitrite, which can be reduced to NO• under hypoxic and acidotic conditions, is considered a physiologically relevant, direct source of bioactive NO•. The aim of the present study was to determine the basal levels of NOx in striated muscles (including rat heart and locomotory muscles) with varied contents of tissue nitrite reductases, such as myoglobin and mitochondrial electron transport chain proteins (ETC-proteins). Muscle NOx was determined using a high-performance liquid chromatography-based method. Muscle proteins were evaluated using western-immunoblotting. We found that oxidative muscles with a higher content of ETC-proteins and myoglobin (such as the heart and slow-twitch locomotory muscles) have lower [NO2−] compared to fast-twitch muscles with a lower content of those proteins. The muscle type had no observed effect on the [NO3−]. Our results demonstrated that fast-twitch muscles possess greater potential to generate NO• via nitrite reduction than slow-twitch muscles and the heart. This property might be of special importance for fast skeletal muscles during strenuous exercise and/or hypoxia since it might support muscle blood flow via additional NO• provision (acidic/hypoxic vasodilation) and delay muscle fatigue.
Collapse
|
33
|
Heme oxygenase-1, carbon monoxide, and malaria – The interplay of chemistry and biology. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
34
|
Lehnert N, Kim E, Dong HT, Harland JB, Hunt AP, Manickas EC, Oakley KM, Pham J, Reed GC, Alfaro VS. The Biologically Relevant Coordination Chemistry of Iron and Nitric Oxide: Electronic Structure and Reactivity. Chem Rev 2021; 121:14682-14905. [PMID: 34902255 DOI: 10.1021/acs.chemrev.1c00253] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Nitric oxide (NO) is an important signaling molecule that is involved in a wide range of physiological and pathological events in biology. Metal coordination chemistry, especially with iron, is at the heart of many biological transformations involving NO. A series of heme proteins, nitric oxide synthases (NOS), soluble guanylate cyclase (sGC), and nitrophorins, are responsible for the biosynthesis, sensing, and transport of NO. Alternatively, NO can be generated from nitrite by heme- and copper-containing nitrite reductases (NIRs). The NO-bearing small molecules such as nitrosothiols and dinitrosyl iron complexes (DNICs) can serve as an alternative vehicle for NO storage and transport. Once NO is formed, the rich reaction chemistry of NO leads to a wide variety of biological activities including reduction of NO by heme or non-heme iron-containing NO reductases and protein post-translational modifications by DNICs. Much of our understanding of the reactivity of metal sites in biology with NO and the mechanisms of these transformations has come from the elucidation of the geometric and electronic structures and chemical reactivity of synthetic model systems, in synergy with biochemical and biophysical studies on the relevant proteins themselves. This review focuses on recent advancements from studies on proteins and model complexes that not only have improved our understanding of the biological roles of NO but also have provided foundations for biomedical research and for bio-inspired catalyst design in energy science.
Collapse
Affiliation(s)
- Nicolai Lehnert
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Eunsuk Kim
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Hai T Dong
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Jill B Harland
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Andrew P Hunt
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Elizabeth C Manickas
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Kady M Oakley
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - John Pham
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Garrett C Reed
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Victor Sosa Alfaro
- Department of Chemistry and Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| |
Collapse
|
35
|
Carvalho LRRA, Guimarães DD, Flôr AFL, Leite EG, Ruiz CR, de Andrade JT, Monteiro MMO, Balarini CM, Lucena RBD, Sandrim VC, Lundberg JO, Weitzberg E, Carlström M, Braga VDA. Effects of chronic dietary nitrate supplementation on longevity, vascular function and cancer incidence in rats. Redox Biol 2021; 48:102209. [PMID: 34915448 PMCID: PMC8683768 DOI: 10.1016/j.redox.2021.102209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022] Open
Abstract
RATIONALE Dietary nitrate and nitrite have a notoriously bad reputation because of their proposed association with disease, in particular cancer. However, more recent lines of research have challenged this dogma suggesting that intake of these anions also possess beneficial effects after in vivo conversion to the vital signaling molecule nitric oxide. Such effects include improvement in cardiovascular, renal and metabolic function, which is partly mediated via reduction of oxidative stress. A recent study even indicates that low dose of dietary nitrite extends life span in fruit flies. METHODS In this study, 200 middle-aged Wistar rats of both sexes were supplemented with nitrate or placebo in the drinking water throughout their remaining life and we studied longevity, biochemical markers of disease, vascular reactivity along with careful determination of the cause of death. RESULTS Dietary nitrate did not affect life span or the age-dependent changes in markers of oxidative stress, kidney and liver function, or lipid profile. Ex vivo examination of vascular function, however, showed improvements in endothelial function in rats treated with nitrate. Neoplasms were not more common in the nitrate group. CONCLUSION We conclude that chronic treatment with dietary nitrate does not affect life span in rats nor does it increase the incidence of cancer. In contrast, vascular function was improved by nitrate, possibly suggesting an increase in health span.
Collapse
Affiliation(s)
| | - Drielle D Guimarães
- Department of Physiology and Pharmacology - Karolinska Institutet, Stockholm, Sweden
| | | | - Ericka G Leite
- Department of Biotechnology - Federal University of Paraiba, João Pessoa, Brazil
| | - Clara R Ruiz
- Department of Biotechnology - Federal University of Paraiba, João Pessoa, Brazil
| | - Juliana T de Andrade
- Department of Biotechnology - Federal University of Paraiba, João Pessoa, Brazil
| | - Matheus M O Monteiro
- Department of Biotechnology - Federal University of Paraiba, João Pessoa, Brazil
| | - Camille M Balarini
- Department of Biotechnology - Federal University of Paraiba, João Pessoa, Brazil
| | | | | | - Jon O Lundberg
- Department of Physiology and Pharmacology - Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology - Karolinska Institutet, Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology - Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
36
|
Rocha BS. The Nitrate-Nitrite-Nitric Oxide Pathway on Healthy Ageing: A Review of Pre-clinical and Clinical Data on the Impact of Dietary Nitrate in the Elderly. FRONTIERS IN AGING 2021; 2:778467. [PMID: 35821990 PMCID: PMC9261383 DOI: 10.3389/fragi.2021.778467] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/20/2021] [Indexed: 01/01/2023]
Abstract
We are living longer. Are we living healthier? As we age, cellular and molecular damage reshape our physiological responses towards environmental and endogenous stimuli. The free radical theory of ageing has been proposed long before ageing has been considered a “scientific discipline” and, since then, has been discussed and upgraded as a major contributor to aberrant ageing. Assuming that ageing results merely from the accumulation of oxidative modifications of biomolecules is not only a simplistic and reductive view of such a complex and dynamic process, but also free radicals and related oxidants are now considered pivotal signalling molecules. The fine modulation of critical signalling pathways by redox compounds demands a novel approach to tackle the role of free radicals in ageing. Nitric oxide (⋅NO) is a paradigmatic example given its biological functions in cardiovascular, neurologic and immune systems. In addition to the canonical ⋅NO synthesis by a family of enzymes, nitrate from green leafy vegetables, is reduced to nitrite in the oral cavity which is further reduced to ⋅NO in the stomach. Boosting this nitrate-nitrite-NO pathway has been shown to improve gastrointestinal, cardiovascular, metabolic and cognitive performance both in humans and in animal models of disease. In the elderly, nitrate-derived ⋅NO has been shown improve several physiological functions that typically decline during ageing. In this paper, the role of nitrate and derived nitrogen oxides will be discussed while reviewing pre-clinical and clinical data on the cardiovascular, neuronal, musculoskeletal and metabolic effects of nitrate during healthy ageing.
Collapse
|
37
|
Galinato MGI, Trail AM, Steinbeck OR, Si Z, Rodland AM, Gowen J. Influence of heme propionates on the nitrite reductase activity of myoglobin. J Inorg Biochem 2021; 226:111630. [PMID: 34688205 DOI: 10.1016/j.jinorgbio.2021.111630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 11/20/2022]
Abstract
The heme propionates in myoglobin (Mb) form a H-bonding network among several residues within its second-sphere coordination, providing a key structural role towards Mb's functional properties. Our work aims to understand the role of the heme propionates on the nitrite reductase (NiR) activity (e.g. reduction of NO2- to NO) of this globin by studying an artificial dimethylester heme-substituted horse heart Mb (DME-Mb). The minor structural change brought about by esterification of the heme propionates causes the NiR rate to increase by more than over two-fold (5.6 ± 0.1 M-1 s-1) relative to wildtype (wt) Mb (2.3 ± 0.1 M-1 s-1). The lower pKa observed in DME-Mb may enhance the tendency of His64 towards protonation, therefore increasing the NiR rate. In addition, the nitrite binding constant (Knitrite) for DME-MbIII is greater than wt MbIII (350 M-1 versus 120 M-1). The disparity in the NiR activity correlates with the differences in electrostatic behavior, which influences the system's reactivity towards the approaching NO2- ion, and thus the formation of the FeII-NO2- intermediate.
Collapse
Affiliation(s)
- Mary Grace I Galinato
- School of Science-Chemistry, Penn State Behrend, Erie, PA 16563, United States of America.
| | - Aaron M Trail
- School of Science-Chemistry, Penn State Behrend, Erie, PA 16563, United States of America
| | - Olivia R Steinbeck
- School of Science-Chemistry, Penn State Behrend, Erie, PA 16563, United States of America
| | - Zhuoyan Si
- School of Science-Chemistry, Penn State Behrend, Erie, PA 16563, United States of America
| | - Anthony M Rodland
- School of Science-Chemistry, Penn State Behrend, Erie, PA 16563, United States of America
| | - Jaclyn Gowen
- School of Science-Chemistry, Penn State Behrend, Erie, PA 16563, United States of America
| |
Collapse
|
38
|
Ganjiani V, Ahmadi N, Divar MR, Sharifiyazdi H, Meimandi-Parizi A. Protective effects of crocin on testicular torsion/detorsion in rats. Theriogenology 2021; 173:241-248. [PMID: 34399388 DOI: 10.1016/j.theriogenology.2021.07.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 07/27/2021] [Accepted: 07/31/2021] [Indexed: 11/19/2022]
Abstract
Oxidative stress, caused by extreme accumulation of un-scavenged reactive oxygen species, plays an integral role in the Ischemia-Reperfusion (I/R) injury to the testicles following testicular torsion. The current research aimed to examine the protective effects of crocin as a natural antioxidant on testicular I/R injury in rats. Animals were divided randomly into five groups (seven each): (1) sham group, (2) torsion/detorsion (T/D) group, (3) intact group with 100 mg/kg crocin, (4) and (5) T/D groups followed by treatment with two different doses of crocin (50 and 100 mg/kg (IP)). I/R injury was induced by 720° clockwise torsion of the left testicles for 2 h. After 24 h of reperfusion, blood samples and epididymal sperms were collected to measure biochemical (GPx, SOD, and MDA), hormonal (testosterone), and sperm parameters (total sperm recovery, motility, viability, and morphology). Moreover, affected testicles were subjected to histopathology examination. I/R injury caused a significant reduction in sperm characteristics (except for morphology) (P < 0.05), which could not be significantly improved by crocin administration at either dose (P > 0.05). Johnsen's testicular score, mean seminiferous tubular diameter, and germinal epithelial cell thickness were significantly decreased in the T/D group compared to the intact and sham groups. However, crocin could significantly improve the histopathological parameters in both treatment groups compared to the T/D group (P < 0.05). T/D reduced SOD and GPx activity and testosterone level significantly (except for GPx) compared to the sham group (P < 0.05). However, crocin administration could significantly reverse them. Also, crocin reduced the amount of MDA significantly in the high-dose treatment group in comparison to T/D group (P < 0.05). The results of the current study revealed that crocin could be a promising agent to protect against I/R injury following surgical correction of the testicular torsion.
Collapse
Affiliation(s)
- Vahid Ganjiani
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Nasrollah Ahmadi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Mohammad Reza Divar
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Hassan Sharifiyazdi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | | |
Collapse
|
39
|
Lee DY, Lee SY, Jo C, Yoon Y, Jeong JY, Hur SJ. Effect on health from consumption of meat and meat products. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:955-976. [PMID: 34796340 PMCID: PMC8564306 DOI: 10.5187/jast.2021.e101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/26/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022]
Abstract
The aim of this study was to investigate the effects of dietary sodium nitrite and meat on human health. Sodium nitrite in processed meat is known to be one of the main precursors of carcinogens, such as N-nitroso compounds. However, we previously found that processed meat is not the primary source of sodium nitrite; nitrate or the conversion of nitrate in vegetables are contribute to generate more than 70% Sodium nitrite or nitrate containing compounds in body. Although the heavy consumption of meat is likely to cause various diseases, meat intake is not the only cause of colorectal cancer. Our review indicates that sodium nitrite derived from foods and endogenous nitric oxide may exhibit positive effects on human health, such as preventing cardiovascular disease or improving reproductive function. Therefore, further epidemiological studies considering various factors, such as cigarette consumption, alcohol consumption, stress index, salt intake, and genetic factors, are required to reliably elucidate the effects of dietary sodium nitrite and meat on the incidence of diseases, such as colorectal cancer.
Collapse
Affiliation(s)
- Da Young Lee
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Seung Yun Lee
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology,
Research Institute of Agriculture and Life Science, Seoul National
University, Seoul 08826, Korea
| | - Yohan Yoon
- Department of Food and Nutrition,
Sookmyung Women’s University, Seoul 04310, Korea
| | - Jong Youn Jeong
- School of Food Biotechnology and
Nutrition, Kyungsung University, Busan 48434, Korea
| | - Sun Jin Hur
- Department of Animal Science and
Technology, Chung-Ang University, Anseong 17546, Korea
| |
Collapse
|
40
|
Filice M, Cerra MC, Imbrogno S. The goldfish Carassius auratus: an emerging animal model for comparative cardiac research. J Comp Physiol B 2021; 192:27-48. [PMID: 34455483 PMCID: PMC8816371 DOI: 10.1007/s00360-021-01402-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
The use of unconventional model organisms is significantly increasing in different fields of research, widely contributing to advance life sciences understanding. Among fishes, the cyprinid Carassius auratus (goldfish) is largely used for studies on comparative and evolutionary endocrinology, neurobiology, adaptive and conservation physiology, as well as for translational research aimed to explore mechanisms that may be useful in an applicative biomedical context. More recently, the research possibilities offered by the goldfish are further expanded to cardiac studies. A growing literature is available to illustrate the complex networks involved in the modulation of the goldfish cardiac performance, also in relation to the influence of environmental signals. However, an overview on the existing current knowledge is not yet available. By discussing the mechanisms that in C. auratus finely regulate the cardiac function under basal conditions and under environmental challenges, this review highlights the remarkable flexibility of the goldfish heart in relation not only to the basic morpho-functional design and complex neuro-humoral traits, but also to its extraordinary biochemical-metabolic plasticity and its adaptive potential. The purpose of this review is also to emphasize the power of the heart of C. auratus as an experimental tool useful to investigate mechanisms that could be difficult to explore using more conventional animal models and complex cardiac designs.
Collapse
Affiliation(s)
- Mariacristina Filice
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Arcavacata di Rende, CS, Italy.
| | - Maria Carmela Cerra
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Arcavacata di Rende, CS, Italy
| | - Sandra Imbrogno
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036, Arcavacata di Rende, CS, Italy
| |
Collapse
|
41
|
Cai L, Rajah G, Duan H, Gao J, Cheng Z, Xin R, Jiang S, Palmer P, Geng X, Ding Y. Rapid Intravenous Glyceryl Trinitrate in Ischemic Damage (RIGID) After Stroke: Rationale, Design and Protocol for a Prospective Randomized Controlled Trial. Front Neurol 2021; 12:693330. [PMID: 34421796 PMCID: PMC8371530 DOI: 10.3389/fneur.2021.693330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Despite intravenous thrombolysis and endovascular therapy for acute ischemic stroke (AIS), many survivors still have varying degrees of disability. Glyceryl trinitrate (GTN), a nitric oxide (NO) donor, has been previously reported to induce neuroprotection after AIS. The use of GTN to reduce brain damage after stroke remains yet to be elucidated. This study was designed to explore the safety, feasibility, and preliminary efficacy of intravenous administration of GTN after AIS. Methods: A prospective randomized controlled trial is proposed with AIS patients. Participants will be randomly allocated to GTN group and control group with a 1:1 ratio (n = 40). Both groups will be treated with standard therapies according to the current stroke guidelines. Participants allocated to the GTN group will receive intravenous administration of GTN (5 mg GTN in 50 ml saline at a rate of 0.4 mg/h that is continued for 12.5 h/day for 2 days) within 24 h of symptom onset. Participants allocated to the control group will receive intravenous administration at equal capacity of 0.9% normal saline (NS) (total 50 ml/day at 4 ml/h that is continued for 12.5 h/day for 2 days). The primary outcome is safety [systolic blood pressure (SBP) <110 mmHg, headache], while the secondary outcomes include changes in functional outcome and infarction volume. Discussion: Rapid Intravenous Glyceryl Trinitrate in Ischemic Damage (RIGID) is a prospective randomized controlled trial that aims to ascertain the safety, feasibility, and preliminary efficacy of intravenous GTN as a neuroprotection strategy after AIS. These results will provide parameters for future studies as well as provide insights into treatment effects. Any possible neuroprotective qualities of GTN in AIS will also be elucidated. Trial Registration:www.chictr.org.cn, identifier: ChiCTR2100046271.
Collapse
Affiliation(s)
- Lipeng Cai
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Gary Rajah
- Department of Neurosurgery, Munson Medical Center, Traverse City, MI, United States
| | - Honglian Duan
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Jie Gao
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhe Cheng
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Ruiqiang Xin
- Department of Medical Imaging, Luhe Hospital, Capital Medical University, Beijing, China
| | - Shangqian Jiang
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Peter Palmer
- Department of Neurology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Xiaokun Geng
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
42
|
Eriksson KE, Eidhagen F, Liska J, Franco-Cereceda A, Lundberg JO, Weitzberg E. Effects of inorganic nitrate on ischaemia-reperfusion injury after coronary artery bypass surgery. Br J Anaesth 2021; 127:547-555. [PMID: 34399982 PMCID: PMC8524391 DOI: 10.1016/j.bja.2021.06.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/30/2022] Open
Abstract
Background Nitric oxide (NO) is an important signalling molecule in the cardiovascular system with protective properties in ischaemia–reperfusion injury. Inorganic nitrate, an oxidation product of endogenous NO production and a constituent in our diet, can be recycled back to bioactive NO. We investigated if preoperative administration of inorganic nitrate could reduce troponin T release and other plasma markers of injury to the heart, liver, kidney, and brain in patients undergoing cardiac surgery. Methods This single-centre, randomised, double-blind, placebo-controlled trial included 82 patients undergoing coronary artery bypass surgery with cardiopulmonary bypass. Oral sodium nitrate (700 mg×2) or placebo (NaCl) were administered before surgery. Biomarkers of ischaemia–reperfusion injury and plasma nitrate and nitrite were collected before and up to 72 h after surgery. Troponin T release was our predefined primary endpoint and biomarkers of renal, liver, and brain injury were secondary endpoints. Results Plasma concentrations of nitrate and nitrite were elevated in nitrate-treated patients compared with placebo. The 72-h release of troponin T did not differ between groups. Other plasma biomarkers of organ injury were also similar between groups. Blood loss was not a predefined outcome parameter, but perioperative bleeding was 18% less in nitrate-treated patients compared with controls. Conclusion Preoperative administration of inorganic nitrate did not influence troponin T release or other plasma biomarkers of organ injury in cardiac surgery. Clinical trial registration NCT01348971.
Collapse
Affiliation(s)
- Karin E Eriksson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.
| | - Fredrik Eidhagen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Stockholm Center for Spine Surgery (RKC), Stockholm, Sweden
| | - Jan Liska
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Cardiothoracic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Franco-Cereceda
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Cardiothoracic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
43
|
Molecular mechanism of Chuanxiong Rhizoma in treating coronary artery diseases. CHINESE HERBAL MEDICINES 2021; 13:396-402. [PMID: 36118926 PMCID: PMC9476474 DOI: 10.1016/j.chmed.2021.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Objective Most of the studies on the herb Chuanxiong Rhizoma (CR) have focused on the l-arginine-nitric oxide (NO) pathway, but the nitrate-nitrite-NO (NO3−–NO2−–NO) pathway was rarely investigated. Therefore, the aim of this study was to evaluate the effects and mechanisms of action of CR in coronary artery disease (CAD). Methods The NO3−, NO2− and NO levels were examined in the NO3−–NO2−–NO pathway. High-performance ion chromatography was used to quantify NO3− and NO2− levels. Then, NO was quantified using a multifunctional enzyme marker with a fluorescent probe. The tension of aortic rings was measured using a multi myograph system. Results High content of NO3− and low content of NO2− was found in CR, and which could potently convert NO3− to NO2− in the presence of endogenous reductase enzyme. Incubating human coronary artery endothelial cells (HCAECs) with CR-containing serum showed that CR significantly decreased the NO3− content and increased the levels of NO2− and NO in the cells under hypoxic conditions. In addition, CR significantly relaxed isolated aortic rings when the l-arginine –NO pathway was blocked. The optimal concentration of CR for relaxation was 200 mg/mL. Conclusion CR supplements large amounts of NO in cells and vessels to achieve relaxation via the NO3−–NO2−–NO pathway, thereby making up for the deficiency caused by the lack of NO after the l-arginine-NO pathway is suppressed. This study also supports the potential use of a traditional Chinese herb for future drug development.
Collapse
|
44
|
Protective Effects of Topical Application of Nitrite on Testicular Ischemia-Reperfusion Injury in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5514537. [PMID: 34257803 PMCID: PMC8257346 DOI: 10.1155/2021/5514537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/10/2021] [Accepted: 06/02/2021] [Indexed: 11/17/2022]
Abstract
Testicular torsion is a urologic emergency induced by torsion of the spermatic cord, interrupting blood circulation to the testis. Therapeutic options for testicular torsion, except surgical restoration of testis, are rarely applied in clinical practice. This study, therefore, investigated whether topical application of nitrite (NO2−) is beneficial in tissue damage due to testicular ischemia-reperfusion (I/R) injury in rats. Pubertal Sprague-Dawley rats were assigned to seven groups: group A, sham-operated control group; group B, I/R with no treatment; groups C, D, and E, I/R followed by treatment with three different doses of nitrite; group F, I/R followed by administration of nitrite and a NO scavenger, C-PTIO (2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide potassium salt); and group G, I/R followed by administration of nitrate (NO3−). Unilateral testicular ischemia was maintained for 5 h, followed by reperfusion for 24 h. Nitrite and nitrate were topically administered before reperfusion. Compared to group A, germ cell apoptosis, oxidative stress, antioxidant enzymatic function, and lipid peroxidation were significantly increased, along with abnormal morphology and impaired spermatogenesis in group B (P < 0.05). In contrast, testicular damage was generally attenuated in the nitrite treatment groups due to a reduction in superoxide and peroxynitrite levels and the inhibition of caspase-3-dependent apoptosis (P < 0.05 vs. group B). These therapeutic effects of nitrite-derived NO were suppressed after injection of C-PTIO, which showed in group F. Taken together, our results demonstrate that topical application of nitrite may be one of the therapeutic strategies for testicular ischemia-reperfusion injury.
Collapse
|
45
|
Li S, Jin H, Sun G, Zhang C, Wang J, Xu H, Zhang D, Wang S. Dietary Inorganic Nitrate Protects Hepatic Ischemia-Reperfusion Injury Through NRF2-Mediated Antioxidative Stress. Front Pharmacol 2021; 12:634115. [PMID: 34163351 PMCID: PMC8215696 DOI: 10.3389/fphar.2021.634115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Objectives: Hepatic ischemia-reperfusion injury (HIRI) is of common occurrence during liver surgery and liver transplantation and may cause hepatic impairment, resulting in acute liver dysfunction. Nitrate plays an important physiological regulatory role in the human body. Whether dietary nitrate could prevent HIRI is, however, unknown. Methods: A HIRI mouse model was established in that the blood supply to the median lobe and left lateral lobe was blocked for 60 min through the portal vein and related structures using an atraumatic clip. Sodium nitrate (4 mM) was administrated in advance through drinking water to compare the influence of sodium nitrate and normal water on HIRI. Results: Liver necrosis and injury aggravated after HIRI. The group treated with sodium nitrate showed the lowest activities of plasma aminotransferase and lactate dehydrogenase and improved outcomes in histological investigation and TUNEL assay. Mechanistically, sodium nitrate intake increased plasma and liver nitric oxide levels, upregulated nuclear factor erythroid 2-related factor 2 (NRF2)-related molecules to reduce malondialdehyde level, and increased the activities of antioxidant enzymes to modulate hepatic oxidative stress. Conclusions: Dietary inorganic nitrate could prevent HIRI, possibly by activating the NRF2 pathway and modulating oxidative stress. Our study provides a novel therapeutic compound that could potentially prevent HIRI during liver transplantation or hepatic surgery.
Collapse
Affiliation(s)
- Shaorong Li
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hua Jin
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Guangyong Sun
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Chunmei Zhang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jinsong Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medicine, Beijing, China
| | - Hufeng Xu
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Dong Zhang
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Songlin Wang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health, Capital Medical University School of Stomatology, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medicine, Beijing, China
| |
Collapse
|
46
|
Buelna-Chontal M, García-Niño WR, Silva-Palacios A, Enríquez-Cortina C, Zazueta C. Implications of Oxidative and Nitrosative Post-Translational Modifications in Therapeutic Strategies against Reperfusion Damage. Antioxidants (Basel) 2021; 10:749. [PMID: 34066806 PMCID: PMC8151040 DOI: 10.3390/antiox10050749] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022] Open
Abstract
Post-translational modifications based on redox reactions "switch on-off" the biological activity of different downstream targets, modifying a myriad of processes and providing an efficient mechanism for signaling regulation in physiological and pathological conditions. Such modifications depend on the generation of redox components, such as reactive oxygen species and nitric oxide. Therefore, as the oxidative or nitrosative milieu prevailing in the reperfused heart is determinant for protective signaling, in this review we defined the impact of redox-based post-translational modifications resulting from either oxidative/nitrosative signaling or oxidative/nitrosative stress that occurs during reperfusion damage. The role that cardioprotective conditioning strategies have had to establish that such changes occur at different subcellular levels, particularly in mitochondria, is also presented. Another section is devoted to the possible mechanism of signal delivering of modified proteins. Finally, we discuss the possible efficacy of redox-based therapeutic strategies against reperfusion damage.
Collapse
Affiliation(s)
| | | | | | | | - Cecilia Zazueta
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (M.B.-C.); (W.R.G.-N.); (A.S.-P.); (C.E.-C.)
| |
Collapse
|
47
|
Xu X, Sun M, Luo X, Zhang Z, Su L, Cui L, Zhu Z, Lu X, Wang R, Han F, Qian X, Yang Y. One-electron reduction triggered nitric oxide release for ischemia-reperfusion protection. Free Radic Biol Med 2021; 164:13-19. [PMID: 33418107 DOI: 10.1016/j.freeradbiomed.2020.12.443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/25/2022]
Abstract
Nitric oxide donors (NODs) are indispensable in biological research and disease treatment. NODs had been utilized to treat cardiovascular diseases in clinic and many others are under trial. Thiols are typically required for these donors to release NO. Yet, their mechanism is complex and often lead to resistance. Herein, we reported that N-nitrosated electron-deficient dyes are capable of NO release with one-electron reduction. A fluorophore is generated simultaneously, whose fluorescence is harnessed to monitor the profile of NO release. Through electrochemical and spectral studies, NOD f3 was found to exhibit good biocompatibility and high reduction efficiency and its potentials in cell-protection in oxygen and glucose deprivation (OGD) models were showcased with endothelial cells. This work aims at offering a new approach to design reduction-triggered NOD, which have therapeutic potentials in ischemia-reperfusion.
Collapse
Affiliation(s)
- Xiu Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Meiling Sun
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xiao Luo
- School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China
| | - Ziqian Zhang
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Lin Su
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lingfei Cui
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhihui Zhu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xicun Lu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Rui Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Xuhong Qian
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Youjun Yang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
48
|
Griffiths K, Lee JJ, Frenneaux MP, Feelisch M, Madhani M. Nitrite and myocardial ischaemia reperfusion injury. Where are we now? Pharmacol Ther 2021; 223:107819. [PMID: 33600852 DOI: 10.1016/j.pharmthera.2021.107819] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite major advances in technology and treatment, with coronary heart disease (CHD) being a key contributor. Following an acute myocardial infarction (AMI), it is imperative that blood flow is rapidly restored to the ischaemic myocardium. However, this restoration is associated with an increased risk of additional complications and further cardiomyocyte death, termed myocardial ischaemia reperfusion injury (IRI). Endogenously produced nitric oxide (NO) plays an important role in protecting the myocardium from IRI. It is well established that NO mediates many of its downstream functions through the 'canonical' NO-sGC-cGMP pathway, which is vital for cardiovascular homeostasis; however, this pathway can become impaired in the face of inadequate delivery of necessary substrates, in particular L-arginine, oxygen and reducing equivalents. Recently, it has been shown that during conditions of ischaemia an alternative pathway for NO generation exists, which has become known as the 'nitrate-nitrite-NO pathway'. This pathway has been reported to improve endothelial dysfunction, protect against myocardial IRI and attenuate infarct size in various experimental models. Furthermore, emerging evidence suggests that nitrite itself provides multi-faceted protection, in an NO-independent fashion, against a myriad of pathophysiologies attributed to IRI. In this review, we explore the existing pre-clinical and clinical evidence for the role of nitrate and nitrite in cardioprotection and discuss the lessons learnt from the clinical trials for nitrite as a perconditioning agent. We also discuss the potential future for nitrite as a pre-conditioning intervention in man.
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jordan J Lee
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael P Frenneaux
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
49
|
Zhou K, Xu Y, Wang Q, Dong L. Overexpression of miR-431 attenuates hypoxia/reoxygenation-induced myocardial damage via autophagy-related 3. Acta Biochim Biophys Sin (Shanghai) 2021; 53:140-148. [PMID: 33382073 DOI: 10.1093/abbs/gmaa154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Indexed: 11/12/2022] Open
Abstract
Myocardial injury is still a serious condition damaging the public health. Clinically, myocardial injury often leads to cardiac dysfunction and, in severe cases, death. Reperfusion of the ischemic myocardial tissues can minimize acute myocardial infarction (AMI)-induced damage. MicroRNAs are commonly recognized in diverse diseases and are often involved in the development of myocardial ischemia/reperfusion injury. However, the role of miR-431 remains unclear in myocardial injury. In this study, we investigated the underlying mechanisms of miR-431 in the cell apoptosis and autophagy of human cardiomyocytes in hypoxia/reoxygenation (H/R). H/R treatment reduced cell viability, promoted cell apoptotic rate, and down-regulated the expression of miR-431 in human cardiomyocytes. The down-regulation of miR-431 by its inhibitor reduced cell viability and induced cell apoptosis in the human cardiomyocytes. Moreover, miR-431 down-regulated the expression of autophagy-related 3 (ATG3) via targeting the 3'-untranslated region of ATG3. Up-regulated expression of ATG3 by pcDNA3.1-ATG3 reversed the protective role of the overexpression of miR-431 on cell viability and cell apoptosis in H/R-treated human cardiomyocytes. More importantly, H/R treatments promoted autophagy in the human cardiomyocytes, and this effect was greatly alleviated via miR-431-mimic transfection. Our results suggested that miR-431 overexpression attenuated the H/R-induced myocardial damage at least partly through regulating the expression of ATG3.
Collapse
Affiliation(s)
- Kang Zhou
- Department of Cardiac Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yan Xu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qiong Wang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lini Dong
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
50
|
Eissa MM, Ahmed MM, Abd Eldaim MA, Mousa AA, Elkirdasy AF, Mohamed MA, Orabi SH. Chlorella vulgaris ameliorates sodium nitrite-induced hepatotoxicity in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:9731-9741. [PMID: 33151487 DOI: 10.1007/s11356-020-11474-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/28/2020] [Indexed: 06/11/2023]
Abstract
The current was conducted to evaluate the ameliorating effect of Chlorella vulgaris (CV) extract against sodium nitrite-induced hepatotoxicity in rats. Forty-five rats were allocated randomly into 5 groups (n = 9). Group I (GI), control group: orally gavaged with normal saline daily. Group II (GII): orally gavaged with CV extract (70 mg/kg BW) for 3 months. Group III (GIII): orally gavaged with sodium nitrite (80 mg/kg BW) for 3 months. Group IV (GIV): received sodium nitrite as GIII and CV extract as GII simultaneously for 3 months. Group V (GV): received CV extract as GII and then, sodium nitrite as in GIII from the end of first month until the end of the experiment. Sodium nitrite significantly increased the activities of serum alanine aminotransferase, aspartate aminotransferase, and serum concentrations of tumor interleukin 1-β and necrosis factor α. In addition, it increased concentrations of malondialdehyde and nitric oxide and expression level of caspase-3 in the hepatic tissue. However, it decreased activities of hepatic glutathione peroxidase, catalase, and superoxide dismutase and induced degenerative and necrotic changes in hepatic tissues. In contrast, CV extract administration modulated sodium nitrite-induced inflammation, oxidative stress, and alteration in hepatic tissue function and architecture. This study indicated that CV extract modulated sodium nitrite-induced hepatic toxicity through decreasing oxidative stress and inflammation and enhancing antioxidant enzyme activities in hepatic tissue of rats.
Collapse
Affiliation(s)
- Mai M Eissa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| | - Mohamed M Ahmed
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt.
| | - Mabrouk A Abd Eldaim
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Menoufia University, Sheben Elkom, 32511, Egypt
| | - Ahmed A Mousa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| | - Ahmed F Elkirdasy
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| | - Mostafa A Mohamed
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Sheben Elkom, Egypt
| | - Sahar H Orabi
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Egypt
| |
Collapse
|