1
|
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
|
2
|
Xueying C, Hua Z, Lujing Z, Ruomeng L, Wen D, Anyong C, Ronghua G, Ying G, Shufang Z, Guoliang Y, Lixin L, Shijun W, Shaohui Z, Lijun G. Which patients with coronary artery disease should include oral nitrates on their discharge medication list? Acta Cardiol 2024; 79:136-148. [PMID: 37961760 DOI: 10.1080/00015385.2023.2271763] [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: 06/07/2023] [Accepted: 10/11/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Which patients with coronary artery disease (CAD) should have oral nitrates on their discharge medication list after coronary angiography (CAG)? To assess the relationship between oral nitrates included in the discharge medication list and major adverse cardiovascular events (MACEs) among CAD patients, we designed this retrospective cohort study. METHODS A total of 2979 CAD patients hospitalised in the Department of Cardiology, Affiliated Hospital of Jining Medical University from May 2013 to October 2015 were enrolled, grouped according to whether oral nitrates were included at discharge after CAG, and followed up for MACEs for a mean of 4.42 years after discharge. The primary endpoint was MACEs. Multivariate Cox proportional hazards models were used to analyses potential confounding factors. Stratified analysis was used to observe the relationship between oral nitrates and MACEs by different covariates. RESULTS The median follow-up time was 4.61 years, and 296 (9.94%) patients experienced MACEs. Multivariate Cox proportional hazards model analysis showed no association between oral nitrates on the discharge medication list and the occurrence of MACEs among patients with CAD (p > 0.05) after adjusting for some covariates, such as SYNTAX score (hazard ratio (HR): 1.18, 95% confidence interval (CI): 0.90-1.55, p = 0.2420). Stratified analysis revealed a higher incidence of MACEs among hypertensive patients prescribed oral nitrates at discharge (HR: 1.67, 95% confidence CI: 1.13-2.46, p = 0.0046). However, prescribing nitrates at discharge for patients with low uric acid levels increased the incidence of MACEs, which showed a possible trend towards significance (HR: 1.44, 95% CI: 0.99-2.09, p = 0.0525). CONCLUSION There was no association between oral nitrates included in the discharge medication list and the development of MACEs among patients with CAD after adjusting for some covariates, such as SYNTAX score. Oral nitrates after discharge for CAD patients combined with hypertension increased the occurrence of MACEs. Oral nitrates after discharge for CAD patients combined with low uric acid levels may increase theoccurrence of MACEs, and close monitoring for any adverse events is recommended.
Collapse
Affiliation(s)
- Chen Xueying
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhang Hua
- Colleague of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Zhao Lujing
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Li Ruomeng
- Emergency Care Unit, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Dai Wen
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Chen Anyong
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Gao Ronghua
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Guo Ying
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Zhang Shufang
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Yang Guoliang
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Liu Lixin
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Wang Shijun
- Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhang Shaohui
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Gan Lijun
- Department of Cardiology, Jining Key Laboratory for Diagnosis and Treatment of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| |
Collapse
|
3
|
Asgari A, Jurasz P. Role of Nitric Oxide in Megakaryocyte Function. Int J Mol Sci 2023; 24:ijms24098145. [PMID: 37175857 PMCID: PMC10179655 DOI: 10.3390/ijms24098145] [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: 02/07/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Megakaryocytes are the main members of the hematopoietic system responsible for regulating vascular homeostasis through their progeny platelets, which are generally known for maintaining hemostasis. Megakaryocytes are characterized as large polyploid cells that reside in the bone marrow but may also circulate in the vasculature. They are generated directly or through a multi-lineage commitment step from the most primitive progenitor or Hematopoietic Stem Cells (HSCs) in a process called "megakaryopoiesis". Immature megakaryocytes enter a complicated development process defined as "thrombopoiesis" that ultimately results in the release of extended protrusions called proplatelets into bone marrow sinusoidal or lung microvessels. One of the main mediators that play an important modulatory role in hematopoiesis and hemostasis is nitric oxide (NO), a free radical gas produced by three isoforms of nitric oxide synthase within the mammalian cells. In this review, we summarize the effect of NO and its signaling on megakaryopoiesis and thrombopoiesis under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Amir Asgari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G-2H7, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G-2S2, Canada
- Mazankowski Alberta Heart Institute, Edmonton, AB T6G-2R7, Canada
| |
Collapse
|
4
|
Giles GI, Erickson JR, Bussey CT. Photoactivation of tDodSNO induces localized vasodilation in rats: Metabolically stable S-nitrosothiols can act as targeted nitric oxide donors in vivo. Nitric Oxide 2022; 129:53-62. [DOI: 10.1016/j.niox.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
|
5
|
Qadir OK, Seal CJ, Ashor AW, Tassotti M, Mena P, Del Rio D, Siervo M, Brandt K. Double-blind controlled dietary cross-over intervention with differentially fertilised intact lettuce leaves shows acute reduction in blood pressure in young adults, associated with faster uptake of nitrate than of phenolics. Eur J Nutr 2022; 61:4191-4203. [PMID: 35871120 PMCID: PMC9596532 DOI: 10.1007/s00394-022-02961-5] [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: 07/19/2021] [Accepted: 07/08/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To compare acute effects on blood pressure (BP) of ingestion of visually similar lettuce with controlled high and low content of either nitrate or phenolic compounds. METHODS In a randomised cross-over design, 19 healthy participants (22-31 years) received 50 g of lettuce containing either 530 mg (8.4 mmol) nitrate + 11 mg (0.03 mmol) phenolic compounds (HNLP); or 3 mg nitrate (0.05 mmol) + 77 mg (0.2 mmol) phenolic compounds (LNHP), obtained by differential fertilisation. Ambulatory BP was recorded along with plasma, salivary and urinary nitrate and nitrite and plasma concentrations of cyclic guanosine monophosphate (cGMP), phenolic metabolites, Trolox equivalent antioxidant capacity (TEAC) and ferric reducing antioxidant power (FRAP). RESULTS Compared with LNHP, 3 h post ingestion of HNLP, plasma nitrate increased 0.31 ± (95%CI) 0.12 mM (+ 240%), and salivary nitrate 5.5 ± 1.4 mM (+ 910%); accumulated urinary nitrate excretion increased 188 ± 72 mg (+ 296%) (all P < 0.001). Systolic BP was reduced 4.9 ± 4.2 mmHg (P = 0.031) between 3 and 6 h after ingestion of HNLP compared with LNHP; systolic BP differences were negatively correlated (P = 0.004) with differences in saliva nitrate concentrations. LNHP increased plasma phenolics at 6 h, predominantly 3'-methoxycinnamic acid-4'-glucuronide (ferulic acid-4'-glucuronide), 116%, 204 ± 138 nM more than HNLP (P = 0.001); increased cGMP 14% (P = 0.019); and reduced FRAP 3.1% (P = 0.009). CONCLUSION The acute BP difference within 6 h of consumption matched the plasma/saliva nitrate peak, not the slower changes of plasma phenolics. This is the first double-blind controlled dietary intervention demonstrating differential effects on human physiology by consumption of an intact plant food, where compositional differences were obtained by controlling growing conditions, indicating potential opportunities for health claims relating to precision/vertical farming. CLINICAL TRIAL REGISTRATION The trial was retrospectively registered on ClinicalTrials.gov, with identifier NCT02701959, on March 8, 2016.
Collapse
Affiliation(s)
- Othman K Qadir
- Human Nutrition Research Centre, Population Health Sciences Institute, William Leech Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Food Science and Quality Control Department, College of Agricultural Engineering Sciences, University of Sulaimani, 46001, Sulaymaniah, Kurdistan Region, Iraq
| | - Chris J Seal
- Human Nutrition Research Centre, Population Health Sciences Institute, William Leech Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Ammar W Ashor
- Human Nutrition Research Centre, Population Health Sciences Institute, William Leech Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Department of Internal Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Michele Tassotti
- Human Nutrition Unit, Department of Food and Drugs, University of Parma, Medical School Building C, Via Volturno, 39, 43125, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food and Drugs, University of Parma, Medical School Building C, Via Volturno, 39, 43125, Parma, Italy
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food and Drugs, University of Parma, Medical School Building C, Via Volturno, 39, 43125, Parma, Italy
- School of Advanced Studies on Food and Nutrition, University of Parma, Parma, Italy
| | - Mario Siervo
- Human Nutrition Research Centre, Population Health Sciences Institute, William Leech Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- School of Life Sciences, Queen's Medical Centre, Nottingham University, Nottingham, NG7 2UH, UK
| | - Kirsten Brandt
- Human Nutrition Research Centre, Population Health Sciences Institute, William Leech Building, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
6
|
Quantification of cyclic AMP and cyclic GMP levels in Krebs-Henseleit solution by LC-MS/MS: application in washed platelet aggregation samples. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1211:123472. [DOI: 10.1016/j.jchromb.2022.123472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022]
|
7
|
Tawa P, Zhang L, Metwally E, Hou Y, McCoy MA, Seganish WM, Zhang R, Frank E, Sheth P, Hanisak J, Sondey C, Bauman D, Soriano A. Mechanistic insights on novel small molecule allosteric activators of cGMP-dependent protein kinase PKG1α. J Biol Chem 2022; 298:102284. [PMID: 35868561 PMCID: PMC9425037 DOI: 10.1016/j.jbc.2022.102284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/30/2022] Open
Abstract
cGMP-dependent protein kinase (PKG) represents a compelling drug target for treatment of cardiovascular diseases. PKG1 is the major effector of beneficial cGMP signaling which is involved in smooth muscle relaxation and vascular tone, inhibition of platelet aggregation and signaling that leads to cardioprotection. In this study, a novel piperidine series of activators previously identified from an ultrahigh-throughput screen were validated to directly bind partially activated PKG1α and subsequently enhance its kinase activity in a concentration-dependent manner. Compounds from initial optimization efforts showed an ability to activate PKG1α independent of the endogenous activator, cGMP. We demonstrate these small molecule activators mimic the effect of cGMP on the kinetic parameters of PKG1α by positively modulating the KM of the peptide substrate and negatively modulating the apparent KM for ATP with increase in catalytic efficiency, kcat. In addition, these compounds also allosterically modulate the binding affinity of cGMP for PKG1α by increasing the affinity of cGMP for the high-affinity binding site (CNB-A) and decreasing the affinity of cGMP for the low-affinity binding site (CNB-B). We show the mode of action of these activators involves binding to an allosteric site within the regulatory domain, near the CNB-B binding site. To the best of our knowledge, these are the first reported non-cGMP mimetic small molecules shown to directly activate PKG1α. Insights into the mechanism of action of these compounds will enable future development of cardioprotective compounds that function through novel modes of action for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Paul Tawa
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Lei Zhang
- Biologics AR&D Immunoassay Group, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Essam Metwally
- Computational & Structural Chemistry, Merck & Co., Inc., South San Francisco, CA, USA
| | - Yan Hou
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Mark A McCoy
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Rumin Zhang
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Emily Frank
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Payal Sheth
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - David Bauman
- Discovery Biology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Aileen Soriano
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
8
|
Inhaled nitric oxide improves post-cardiac arrest outcomes via guanylate cyclase-1 in bone marrow-derived cells. Nitric Oxide 2022; 125-126:47-56. [PMID: 35716999 DOI: 10.1016/j.niox.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022]
Abstract
RATIONALE Nitric oxide (NO) exerts its biological effects primarily via activation of guanylate cyclase (GC) and production of cyclic guanosine monophosphate. Inhaled NO improves outcomes after cardiac arrest and cardiopulmonary resuscitation (CPR). However, mechanisms of the protective effects of breathing NO after cardiac arrest are incompletely understood. OBJECTIVE To elucidate the mechanisms of beneficial effects of inhaled NO on outcomes after cardiac arrest. METHODS Adult male C57BL/6J wild-type (WT) mice, GC-1 knockout mice, and chimeric WT mice with WT or GC-1 knockout bone marrow were subjected to 8 min of potassium-induced cardiac arrest to determine the role of GC-1 in bone marrow-derived cells. Mice breathed air or 40 parts per million NO for 23 h starting at 1 h after CPR. RESULTS Breathing NO after CPR prevented hypercoagulability, cerebral microvascular occlusion, an increase in circulating polymorphonuclear neutrophils and neutrophil-to-lymphocyte ratio, and right ventricular dysfunction in WT mice, but not in GC-1 knockout mice, after cardiac arrest. The lack of GC-1 in bone marrow-derived cells diminished the beneficial effects of NO breathing after CPR. CONCLUSIONS GC-dependent signaling in bone marrow-derived cells is essential for the beneficial effects of inhaled NO after cardiac arrest and CPR.
Collapse
|
9
|
Letson HL, Dobson GP. The Role of Nitric Oxide in the Efficacy of Adenosine, Lidocaine, and Magnesium Treatment for Experimental Hemorrhagic Shock in Rats. Curr Ther Res Clin Exp 2021; 95:100655. [PMID: 34917219 PMCID: PMC8665347 DOI: 10.1016/j.curtheres.2021.100655] [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: 10/12/2021] [Accepted: 11/12/2021] [Indexed: 11/25/2022] Open
Abstract
Background Nitric oxide (NO) plays multiple roles regulating the central nervous, cardiovascular, and immune systems. Objective Our aim was to investigate the role of NO in the efficacy of hypertonic saline (7.5% sodium chloride [NaCl]) adenosine, lidocaine, and magnesium (ALM) to improve mean arterial pressure (MAP) and heart rate following hemorrhagic shock. Methods One hundred one male Sprague-Dawley rats (mean [SD] weight = 425 [6] g) were randomly assigned to 20 groups (groups of 4–8 rats each). Hemorrhagic shock (MAP < 40 mm Hg) was induced by 20-minute pressure-controlled bleeding (∼40% blood volume), and the animal was left in shock (MAP = 35-40 mm Hg) for 60 minutes. The NO synthase (NOS) inhibitor L-NAME was administered with a 0.3-mL bolus of different combinations of 7.5% NaCl ALM active ingredients and hemodynamic parameters were monitored for 60 minutes. A number of specific NOS and NO inhibitors were tested. Results We found that 7.5% NaCl ALM corrected MAP after hemorrhagic shock. In contrast, the addition of L-NAME to 7.5% NaCl ALM led to a rapid fall in MAP, sustained ventricular arrhythmias, and 100% mortality. Saline controls receiving 7.5% NaCl with NG-nitro-l-arginine methyl ester (L-NAME) showed improved MAP with no deaths. None of the specific NOS and NO inhibitors mimicked L-NAME's effect on ALM. The addition of inducible NOS inhibitor 1400W to 7.5% NaCl ALM failed to resuscitate, whereas the NO scavenger PTIO and the PI3K inhibitor wortmannin reduced MAP recovery during 60-minute resuscitation. Conclusions The ability of 7.5% NaCl ALM to resuscitate appears to be linked to 1 or more NO-producing pathways. Nonspecific NOS inhibition with L-NAME blocked ALM resuscitation and led to cardiovascular collapse. More studies are required to examine NO site-specific contributions to ALM resuscitation. (Curr Ther Res Clin Exp. 2022; 82:XXX–XXX)
Collapse
Affiliation(s)
- Hayley L Letson
- Heart, Trauma, and Sepsis Research Laboratory, College of Medicine & Dentistry, James Cook University, Townsville, Queensland, Australia
| | - Geoffrey P Dobson
- Heart, Trauma, and Sepsis Research Laboratory, College of Medicine & Dentistry, James Cook University, Townsville, Queensland, Australia
| |
Collapse
|
10
|
Tan CS, Tew WY, Jingying C, Yam MF. Vasorelaxant effect of 5,7,4'- Trihydroxyflavanone (Naringenin) via endothelium dependent, potassium and calcium channels in Sprague Dawley rats: Aortic ring model. Chem Biol Interact 2021; 348:109620. [PMID: 34411564 DOI: 10.1016/j.cbi.2021.109620] [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: 06/16/2020] [Revised: 11/24/2020] [Accepted: 08/04/2021] [Indexed: 11/19/2022]
Abstract
Naringenin is a naturally occurring flavanone (flavonoid) known to have bioactive effects on human health. It has been reported to show cardiovascular effects. This study aimed to investigate the possible vasorelaxant effect of naringenin and the mechanism behind it by using a Sprague Dawley rat aortic ring assay model. Naringenin caused significant vasorelaxation of endothelium-intact aortic rings precontracted with phenylephrine (pD2 = 4.27 ± 0.05; Rmax = 121.70 ± 4.04%) or potassium chloride (pD2 = 4.00 ± 0.04; Rmax = 103.40 ± 3.82%). The vasorelaxant effect decreased in the absence of an endothelium (pD2 = 3.34 ± 0.10; Rmax = 62.29 ± 2.73%). The mechanisms of the vasorelaxant effect of naringenin in the presence of antagonists were also investigated. Indomethacin, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, atropine, 4-aminopyridine, Nω-nitro-l-arginine methyl ester, glibenclamide and propranolol significantly reduced the relaxation stimulated by naringenin in the presence of endothelium. Besides that, the effect of naringenin on the voltage-operated calcium channel (VOCC) in the endothelium-intact aortic ring was studied, as was intracellular Ca2+ release from the sarcoplasmic reticulum (SR) in the endothelium-denuded aortic ring. The results showed that naringenin also significantly blocked the entry of Ca2+ via the VOCC, SERCA/SOCC and suppressed the release of Ca2+ from the SR. Thus, the vasorelaxant effect shown by naringenin mostly involve the COX pathway, the endothelium-dependent pathway via NO/sGC/prostaglandin, calcium and potassium channels.
Collapse
Affiliation(s)
- Chu Shan Tan
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Shangjie, Minhou, Fuzhou, 350122, Fujian, China; School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Wan Yin Tew
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Shangjie, Minhou, Fuzhou, 350122, Fujian, China; School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Chen Jingying
- Research Center for Medicinal Plant, Institute of Agricultural Bio-resource, Fujian Academy of Agricultural Sciences, Fuzhou, 350003, Fujian, China.
| | - Mun Fei Yam
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Shangjie, Minhou, Fuzhou, 350122, Fujian, China; School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia.
| |
Collapse
|
11
|
Mukherjee D, Konduri GG. Pediatric Pulmonary Hypertension: Definitions, Mechanisms, Diagnosis, and Treatment. Compr Physiol 2021; 11:2135-2190. [PMID: 34190343 PMCID: PMC8289457 DOI: 10.1002/cphy.c200023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pediatric pulmonary hypertension (PPH) is a multifactorial disease with diverse etiologies and presenting features. Pulmonary hypertension (PH), defined as elevated pulmonary artery pressure, is the presenting feature for several pulmonary vascular diseases. It is often a hidden component of other lung diseases, such as cystic fibrosis and bronchopulmonary dysplasia. Alterations in lung development and genetic conditions are an important contributor to pediatric pulmonary hypertensive disease, which is a distinct entity from adult PH. Many of the causes of pediatric PH have prenatal onset with altered lung development due to maternal and fetal conditions. Since lung growth is altered in several conditions that lead to PPH, therapy for PPH includes both pulmonary vasodilators and strategies to restore lung growth. These strategies include optimal alveolar recruitment, maintaining physiologic blood gas tension, nutritional support, and addressing contributing factors, such as airway disease and gastroesophageal reflux. The outcome for infants and children with PH is highly variable and largely dependent on the underlying cause. The best outcomes are for neonates with persistent pulmonary hypertension (PPHN) and reversible lung diseases, while some genetic conditions such as alveolar capillary dysplasia are lethal. © 2021 American Physiological Society. Compr Physiol 11:2135-2190, 2021.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| | - Girija G. Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| |
Collapse
|
12
|
Thakar S, Gabarin N, Gupta A, Radford M, Warkentin TE, Arnold DM. Anemia-Induced Bleeding in Patients with Platelet Disorders. Transfus Med Rev 2021; 35:22-28. [PMID: 34332828 DOI: 10.1016/j.tmrv.2021.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 11/26/2022]
Abstract
Anemia is not only a consequence of bleeding, but also a modifiable risk factor for bleeding in patients with thrombocytopenia or platelet function defects. In this review we outline the mechanism of anemia-induced bleeding in patients with platelet disorders, which involves a disturbance in normal red blood cell (RBC) rheology and reduced platelet margination to the endothelial surface due to a decrease in RBC mass, leading to impaired primary hemostasis and bleeding. Biologically, anemia reduces the mass of RBCs in the central column of flowing blood through a vessel resulting in fewer platelets coming into contact with the endothelial surface at the periphery of the flowing blood column. Thus, anemia results in impaired primary hemostasis. Von Willebrand factor (vWF) is another component of primary hemostasis and vWF deficiency, especially a deficiency of the highest vWF multimers, can also manifest with bleeding when concomitant anemia occurs. Clinically, patients at greatest risk for anemia-induced bleeding include patients with hematological malignancies in whom anemia and thrombocytopenia occur as a result of the underlying disease or the myelotoxic effects of treatment; patients with renal insufficiency with uremic thrombocytopathy and hypoproliferative anemia; and patients with inherited or acquired bleeding disorders affecting primary hemostasis (eg, Bernard-Soulier syndrome, von Willebrand disease) with chronic blood loss and iron deficiency anemia. Underlying abnormalities of any components of primary hemostasis plus concomitant anemia may result in major bleeding disorders; therefore, correction of remediable abnormalities-most notably, correction of the anemia- would be expected to have important clinical benefit. In this review we discuss how the correction of the anemia may lead to improvement of bleeding outcomes in patients with a primary hemostatic defect, supported by evidence from animal models, clinical trials and clinical experience.
Collapse
Affiliation(s)
- Swarni Thakar
- McMaster Center for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
| | - Nadia Gabarin
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Akash Gupta
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael Radford
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Theodore E Warkentin
- McMaster Center for Transfusion Research, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Donald M Arnold
- McMaster Center for Transfusion Research, McMaster University, Hamilton, Ontario, Canada; Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
13
|
Reyes-Pérez VI, Granados-Soto V, Rangel-Grimaldo M, Déciga-Campos M, Mata R. Pharmacological Analysis of the Anti-inflammatory and Antiallodynic Effects of Zinagrandinolide E from Zinnia grandiflora in Mice. JOURNAL OF NATURAL PRODUCTS 2021; 84:713-723. [PMID: 32870011 DOI: 10.1021/acs.jnatprod.0c00793] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Zinagrandinolide E (1, ZGE) is an elemanolide with antinociceptive action isolated from Zinnia grandiflora (Asteraceae), valued in North México and southwestern United States for pain relief. Herein, we report the anti-inflammatory and antiallodynic action of ZGE (1) in carrageenan-induced inflammation and tactile allodynia in mice and in a neuropathic pain model in hyperglycemic mice. Local peripheral administration of ZGE (1-30 μg/paw) induced dose-dependent acute anti-inflammatory and antiallodynic effects. The anti-inflammatory effect was comparable to diclofenac (30 μg/paw). Intrathecal (i.t.) administration of ZGE (30 μg) in acute experiments did not affect carrageenan-induced inflammation but significantly reduced tactile allodynia in a dose-dependent fashion. In long-term experiments (15 or 6 days), using two different scheme treatments (pretreatment or post-treatment), ZGE (3-30 μg/paw) showed antiallodynic but not anti-inflammatory action. Local peripheral (3-30 μg/paw) or intrathecal (3-30 μg) administration of ZGE partially reversed tactile allodynia in hyperglycemic mice, better or comparable, respectively, with those of pregabalin (30 μg/paw or 30 μg i.t.). The effects were dose-dependent. According to the pharmacological tools employed, the anti-inflammatory and antiallodynic activities of ZGE are multitarget; these involve the opioidergic, serotoninergic, and GABAergic systems, as well as the NO-cGMP-ATP-sensitive K+ channel signaling pathway.
Collapse
Affiliation(s)
| | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City 14330, Mexico
| | | | - Myrna Déciga-Campos
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Rachel Mata
- Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
14
|
de Moraes LHO, Mancini MW, Almeida-Lopes L, Rodrigues GJ. Violet LED induces vasodilation in rat aortic rings by soluble guanylate cyclase-dependent mechanism and increases SOD activity. Lasers Med Sci 2021; 37:537-544. [PMID: 33735421 DOI: 10.1007/s10103-021-03293-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/09/2021] [Indexed: 11/29/2022]
Abstract
We found several studies that have used the aortic rings as an experimental model, mainly for the testing of new drugs or new therapies that try to reverse or prevent endothelial dysfunction or characterize its mechanism of action in a biological system, creating the knowledge necessary to obtain the treatment of those several diseases, where many of these treatments involve photobiomodulation therapies. We also found numerous wavelengths represented by different colors of LASER or LED in which frequently, the mechanism of action in biological systems is unknown. This study has as main objective to investigate the effects of the Violet LED Light (405 nm) by using isolated aortic rings, looking for nitric oxide (NO) release, and evaluating if Violet LED Light can modulate the superoxide dismutase (SOD) activity. We performed a vascular reactivity study in isolated aortic rings from normotensive rats with a single LED application. Besides it, the rings were pre-incubated with soluble guanylate cyclase (sGC) inhibitor or endothelial NO synthase inhibitor and subsequently underwent the application of the Violet LED. The cell viability and nitric oxide release in cell culture of human umbilical codon vein cells (HUVEC) were analyzed. In the vascular reactivity experiment, we observed a peak of vasodilation when applying light to the aortic rings. The soluble guanylate cyclase inhibitor abolished the relaxation induced by the Violet LED Light. However, the NO synthase inhibitor did not modify the Violet LED effect. In an isolated system, we verified that the Violet LED Light can increase SOD activity. Our results suggest that Violet LED Light induces vasodilation by a mechanism dependent on sGC activation, and not by NOS activation, and part of this effect could be due to the increase of SOD activity.
Collapse
Affiliation(s)
| | | | - Luciana Almeida-Lopes
- Nucleus of Research and Teaching of Phototherapy in Health Sciences, NuPEn, São Carlos, SP, Brazil
| | | |
Collapse
|
15
|
Wajih N, Alipour E, Rigal F, Zhu J, Perlegas A, Caudell DL, Kim-Shapiro D. Effects of nitrite and far-red light on coagulation. Nitric Oxide 2021; 107:11-18. [PMID: 33271226 PMCID: PMC7855911 DOI: 10.1016/j.niox.2020.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/18/2020] [Accepted: 11/26/2020] [Indexed: 01/14/2023]
Abstract
Nitric oxide, NO, has been explored as a therapeutic agent to treat thrombosis. In particular, NO has potential in treating mechanical device-associated thrombosis due to its ability to reduce platelet activation and due to the central role of platelet activation and adhesion in device thrombosis. Nitrite is a unique NO donor that reduces platelet activation in that it's activity requires the presence of red blood cells whereas NO activity of other NO donors is blunted by red blood cells. Interestingly, we have previously shown that red blood cell mediated inhibition of platelet activation by adenosine diphosophate (ADP) is dramatically enhanced by illumination with far-red light that is likely due to photolysis of red cell surface bound NO congeners. We now report the effects of nitrite, far-red light, and their combination on several measure of blood coagulation using a variety of agonists. We employed turbidity assays in platelet rich plasma, platelet activation using flow cytometry analysis of a fluorescently labeled antibody to the activated platelet fibrinogen binding site, multiplate impedance-based platelet aggregometry, and assessment of platelet adhesion to collagen coated flow-through microslides. In all cases, the combination of far-red light and nitrite treatment decreased measures of coagulation, but in some cases mono-treatment with nitrite or light alone had no effect. Perhaps most relevant to device thrombosis, we observed that platelet adhesions was inhibited by the combination of nitrite and light treatment while nitrite alone and far-red light alone trended to decrease adhesion, but the results were mixed. These results support the potential of combined far-red light and nitrite treatment for preventing thrombosis in extra-corporeal or shallow-tissue depth devices where the far-red light can penetrate. Such a combined treatment could be advantageous due to the localized treatment afforded by far-red light illumination with minimal systemic effects. Given the role of thrombosis in COVID 19, application to treatment of patients infected with SARS Cov-2 might also be considered.
Collapse
Affiliation(s)
- Nadeem Wajih
- Department of Physics, Wake Forest University, Winston-Salem, NC, 27109, United States; Translational Science Center, Wake Forest University, Winston-Salem, NC, 27109, United States.
| | - Elmira Alipour
- Department of Physics, Wake Forest University, Winston-Salem, NC, 27109, United States.
| | - Fernando Rigal
- Department of Physics, Wake Forest University, Winston-Salem, NC, 27109, United States.
| | - Jiqing Zhu
- Department of Physics, Wake Forest University, Winston-Salem, NC, 27109, United States.
| | - Andreas Perlegas
- Department of Physics, Wake Forest University, Winston-Salem, NC, 27109, United States
| | - David L Caudell
- Department of Pathology,-Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, United States.
| | - Daniel Kim-Shapiro
- Department of Physics, Wake Forest University, Winston-Salem, NC, 27109, United States; Translational Science Center, Wake Forest University, Winston-Salem, NC, 27109, United States.
| |
Collapse
|
16
|
Abstract
Cyclic guanosine 3',5'-monophosphate (cGMP) is the key second messenger molecule in nitric oxide signaling. Its rapid generation and fate, but also its role in mediating acute cellular functions has been extensively studied. In the past years, genetic studies suggested an important role for cGMP in affecting the risk of chronic cardiovascular diseases, for example, coronary artery disease and myocardial infarction. Here, we review the role of cGMP in atherosclerosis and other cardiovascular diseases and discuss recent genetic findings and identified mechanisms. Finally, we highlight open questions and promising research topics.
Collapse
|
17
|
Hall KT, Kessler T, Buring JE, Passow D, Sesso HD, Zee RYL, Ridker PM, Chasman DI, Schunkert H. Genetic variation at the coronary artery disease risk locus GUCY1A3 modifies cardiovascular disease prevention effects of aspirin. Eur Heart J 2020; 40:3385-3392. [PMID: 31228190 DOI: 10.1093/eurheartj/ehz384] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 02/26/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
AIMS Efficacy of aspirin in primary prevention of cardiovascular disease (CVD) may be influenced by a common allele in guanylate cyclase GUCY1A3, which has been shown to modify platelet function and increase CVD risk. METHODS AND RESULTS We investigated whether homozygotes of the GUCY1A3 rs7692387 risk (G) allele benefited from aspirin in two long-term, randomized placebo-controlled trials of aspirin in primary CVD prevention: the Women's Genome Health Study (WGHS, N = 23 294) and a myocardial infarction (MI, N = 550) and stroke (N = 382) case-control set from the Physician's Health Study (PHS, N = 22 071). Bleeding risk was evaluated in the WGHS. In the placebo group of the WGHS, the GUCY1A3 risk (G) allele was confirmed to increase CVD risk [hazard ratio 1.38; 95% confidence interval (CI) 1.08-1.78; P = 0.01]. Random-effects meta-analysis of the WGHS and PHS revealed that aspirin reduced CVD events among risk allele homozygotes [G/G: odds ratio (OR) 0.79; 95% CI 0.65-0.97; P = 0.03] but increased CVD events among non-risk allele carriers (e.g. G/A: OR 1.39; 95% CI 1.03-1.87; P = 0.03) thus implying an interaction between genotype stratum and aspirin intake (Pinteraction = 0.01). Bleeding associated with aspirin increased in all genotype groups, with higher risks in heterozygotes. CONCLUSION In two randomized placebo-controlled trials in the setting of primary prevention, aspirin reduced the incidence of CVD events in individuals homozygous for the GUCY1A3 risk (G) allele, whereas heterozygote individuals had more events when taking aspirin.
Collapse
Affiliation(s)
- Kathryn T Hall
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02146, USA
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Lazarettstrasse 36, 80636 Munich, Germany
| | - Julie E Buring
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02146, USA
| | - Dani Passow
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02146, USA
| | - Howard D Sesso
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02146, USA
| | - Robert Y L Zee
- Department of Pediatric Dentistry, Tufts University School of Dental Medicine, Boston, MA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02146, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02146, USA
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Deutsches Zentrum für Herz- und Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Lazarettstrasse 36, 80636 Munich, Germany
| |
Collapse
|
18
|
Kessler T, Schunkert H, von Hundelshausen P. Novel Approaches to Fine-Tune Therapeutic Targeting of Platelets in Atherosclerosis: A Critical Appraisal. Thromb Haemost 2020; 120:1492-1504. [PMID: 32772352 DOI: 10.1055/s-0040-1714352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pathogenesis of atherosclerotic vascular disease is driven by a multitude of risk factors intertwining metabolic and inflammatory pathways. Increasing knowledge about platelet biology sheds light on how platelets take part in these processes from early to later stages of plaque development. Recent insights from experimental studies and mouse models substantiate platelets as initiators and amplifiers in atherogenic leukocyte recruitment. These studies are complemented by results from genetics studies shedding light on novel molecular mechanisms which provide an interesting prospect as novel targets. For instance, experimental studies provide further details how platelet-decorated von Willebrand factor tethered to activated endothelial cells plays a role in atherogenic monocyte recruitment. Novel aspects of platelets as atherogenic inductors of neutrophil extracellular traps and particularities in signaling pathways such as cyclic guanosine monophosphate and the inhibitory adaptor molecule SHB23/LNK associating platelets with atherogenesis are shared. In summary, it was our intention to balance insights from recent experimental data that support a plausible role for platelets in atherogenesis against a paucity of clinical evidence needed to validate this concept in humans.
Collapse
Affiliation(s)
- Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany
| | - Philipp von Hundelshausen
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., Partner Site Munich Heart Alliance, Munich, Germany.,Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, Klinikum der Universität, Ludwig-Maximilians-Universität, Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
19
|
Dabiré H, Dramé F, Cita N, Ghaleh B. The hypertensive effect of sorafenib is abolished by sildenafil. CARDIO-ONCOLOGY 2020; 6:7. [PMID: 32685197 PMCID: PMC7358208 DOI: 10.1186/s40959-020-00064-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/03/2020] [Indexed: 12/30/2022]
Abstract
Background Contrasting to the well documented tyrosine kinase inhibitor (TKI)-induced hypertension, little is known on their intrinsic vasomotor effects. We investigated the vasomotor effects of sorafenib, a widely used multikinase inhibitor in the treatment of hepatocellular and renal cell carcinoma and tested the hypothesis that sildenafil, a phosphodiesterase-5 (PDE-5) inhibitor, could represent a pharmacological strategy for the treatment of TKI-induced hypertension. Methods Concentration-response curves of sorafenib were constructed in endothelium-intact or denuded precontracted rat aorta, in the presence or absence of several inhibitors. Acute intravenous effects of sorafenib on arterial blood pressure were also investigated in anaesthetized rats. Finally, rats were chronically treated with sorafenib during 4 weeks in the presence and absence of sildenafil. Results In endothelium intact aortic ring, sorafenib induced a potent concentration-dependent relaxation of precontracted rat aorta. Removal of the endothelium shifted the concentration-response curve of sorafenib to the right and significantly reduced its maximal effects, demonstrating that sorafenib-induced vasorelaxation is endothelium-dependent and endothelium-independent. Inhibition of the different pathways implicated in the endothelium-dependent and independent vasorelaxation revealed that the endothelium-dependent effects of sorafenib result mainly from the activation of prostaglandin and the nitric oxide (NO) pathways. The endothelium-independent vasodilatory effects of sorafenib may result mainly from the activation of Na/K-ATPase and soluble guanylate cyclase. These vasodilatory effects observed in vitro were confirmed by the decrease in arterial blood pressure observed during acute administrations of sorafenib in anesthetized rats. Finally, and most importantly, we report here for the first time that chronic administration of sorafenib in rats induced an increase in SBP that was abolished by sildenafil. Conclusion The multikinase inhibitor sorafenib induced in vitro vasorelaxation of large conductance artery, primary by activating soluble guanylate cyclase. Its chronic administration led to arterial blood hypertension that was counteracted by a PDE-5 inhibitor, sildenafil. Our results suggest that targeting the cGMP pathway including NO signalling might be an interesting pharmacological strategy for the treatment of TKI-induced hypertension.
Collapse
Affiliation(s)
- Hubert Dabiré
- U955 - IMRB, Inserm, UPEC, École Nationale Vétérinaire d'Alfort, Créteil, France
| | - Fatou Dramé
- U955 - IMRB, Inserm, UPEC, École Nationale Vétérinaire d'Alfort, Créteil, France
| | - Nelly Cita
- U955 - IMRB, Inserm, UPEC, École Nationale Vétérinaire d'Alfort, Créteil, France
| | - Bijan Ghaleh
- U955 - IMRB, Inserm, UPEC, École Nationale Vétérinaire d'Alfort, Créteil, France.,INSERM U955 Équipe 03, Faculté de Médecine, 8 rue du Général Sarrail, 94000 Créteil, France
| |
Collapse
|
20
|
Armstrong PC, Ferreira PM, Chan MV, Lundberg Slingsby MH, Crescente M, Shih CC, Kirkby NS, Hobbs AJ, Warner TD. Combination of cyclic nucleotide modulators with P2Y 12 receptor antagonists as anti-platelet therapy. J Thromb Haemost 2020; 18:1705-1713. [PMID: 32278335 DOI: 10.1111/jth.14826] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 03/02/2020] [Accepted: 03/25/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Endothelium-derived prostacyclin and nitric oxide elevate platelet cyclic nucleotide levels and maintain quiescence. We previously demonstrated that a synergistic relationship exists between cyclic nucleotides and P2Y12 receptor inhibition. A number of clinically approved drug classes can modulate cyclic nucleotide tone in platelets including activators of NO-sensitive guanylyl cyclase (GC) and phosphodiesterase (PDE) inhibitors. However, the doses required to inhibit platelets produce numerous side effects including headache. OBJECTIVE We investigated using GC-activators in combination with P2Y12 receptor antagonists as a way to selectively amplify the anti-thrombotic effect of both drugs. METHODS In vitro light transmission aggregation and platelet adhesion under flow were performed on washed platelets and platelet rich plasma. Aggregation in whole blood and a ferric chloride-induced arterial thrombosis model were also performed. RESULTS The GC-activator BAY-70 potentiated the action of the P2Y12 receptor inhibitor prasugrel active metabolite in aggregation and adhesion studies and was associated with raised intra-platelet cyclic nucleotide levels. Furthermore, mice administered sub-maximal doses of the GC activator cinaciguat together with the PDE inhibitor dipyridamole and prasugrel, showed significant inhibition of ex vivo platelet aggregation and significantly reduced in vivo arterial thrombosis in response to injury without alteration in basal carotid artery blood flow. CONCLUSIONS Using in vitro, ex vivo, and in vivo functional studies, we show that low dose GC activators synergize with P2Y12 inhibition to produce powerful anti-platelet effects without altering blood flow. Therefore, modulation of intra-platelet cyclic nucleotide levels alongside P2Y12 inhibition can provide a strong, focused anti-thrombotic regimen while minimizing vasodilator side effects.
Collapse
Affiliation(s)
- Paul C Armstrong
- Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Plinio M Ferreira
- Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Melissa V Chan
- Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Martina H Lundberg Slingsby
- Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Marilena Crescente
- Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Chih-Chin Shih
- Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Nicholas S Kirkby
- Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Timothy D Warner
- Blizard Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
21
|
Affiliation(s)
- Colin Baigent
- MRC Population Health Research Unit, Nuffield Department of Population Health, Oxford, UK
| | - Michael V Holmes
- MRC Population Health Research Unit, Nuffield Department of Population Health, Oxford, UK
| |
Collapse
|
22
|
Ahmad T, Khan T, Alamgeer, Shah AJ. Juglone as antihypertensive agent acts through multiple vascular mechanisms. Clin Exp Hypertens 2020; 42:335-344. [PMID: 31523996 DOI: 10.1080/10641963.2019.1665674] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background: Juglone, a natural phenolic compound obtained from the walnut tree, is known for its wide range of biological activities. However, it has yet to be tested for its effects on hypertension and vascular tone. This investigation was aimed to explore the antihypertensive effect and the nature of vascular reactivity of juglone in rat models.Methods: Juglone was tested in in vivo and in vitro experiments in rats. The responses were analyzed and recorded through a PowerLab data acquisition system.Results: Intravenous injection of juglone significantly decreased the mean arterial blood pressure (MAP) in normotensive and hypertensive rats (Max. fall, 43.50 ± 2.96 vs 49.66 ± 3.28 mmHg). In rats pretreated with Nω-Nitro l-arginine methyl ester (L-NAME), the effect of juglone on MAP was reduced as compared to the control. However, in rats pretreated with atropine the fall in MAP by juglone was not altered. Juglone induced relaxation in the phenylephrine, K+ (80 mM), and angiotensin II pretreated isolated rat aortic rings. This vasorelaxant effect was reduced with L-NAME pretreatment. Atropine pretreatment did not modify the vasorelaxant effect of juglone. Pre-incubation with juglone attenuated the intracellular Ca2+ release by suppressing phenylephrine peak formation and also shifted CaCl2 concentration-response curves (CRCs) to the right. Of note, combined treatment with 4-aminopyridine and barium chloride also reduced juglone-mediated vasorelaxation suggesting a role of K+-channels as well.Conclusion: In conclusion, juglone exerts its antihypertensive effect through vasorelaxation, which is mediated by nitric oxide, inhibition of intracellular calcium release and opening of K+-channels.
Collapse
Affiliation(s)
- Taseer Ahmad
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad, Pakistan.,Laboratory of Cardiovascular Research and Integrative Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Taous Khan
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad, Pakistan
| | - Alamgeer
- Laboratory of Cardiovascular Research and Integrative Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Abdul Jabbar Shah
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad, Pakistan
| |
Collapse
|
23
|
Kessler T, Schunkert H. Genomic Strategies Toward Identification of Novel Therapeutic Targets. Handb Exp Pharmacol 2020; 270:429-462. [PMID: 32399778 DOI: 10.1007/164_2020_360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Coronary artery disease, myocardial infarction, and secondary damages of the myocardium in the form of ischemic heart disease remain major causes of death in Western countries. Beyond traditional risk factors such as smoking, hypertension, dyslipidemia, or diabetes, a positive family history is known to increase risk. The genetic factors underlying this observation remained unknown for decades until genetic studies were able to identify multiple genomic loci contributing to the heritability of the trait. Knowledge of the affected genes and the resulting molecular and cellular mechanisms leads to improved understanding of the pathophysiology leading to coronary atherosclerosis. Major goals are also to improve prevention and therapy of coronary artery disease and its sequelae via improved risk prediction tools and pharmacological targets. In this chapter, we recapitulate recent major findings. We focus on established novel targets and discuss possible further targets which are currently explored in translational studies.
Collapse
Affiliation(s)
- Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany. .,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany.
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
24
|
Premont RT, Reynolds JD, Zhang R, Stamler JS. Role of Nitric Oxide Carried by Hemoglobin in Cardiovascular Physiology: Developments on a Three-Gas Respiratory Cycle. Circ Res 2019; 126:129-158. [PMID: 31590598 DOI: 10.1161/circresaha.119.315626] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A continuous supply of oxygen is essential for the survival of multicellular organisms. The understanding of how this supply is regulated in the microvasculature has evolved from viewing erythrocytes (red blood cells [RBCs]) as passive carriers of oxygen to recognizing the complex interplay between Hb (hemoglobin) and oxygen, carbon dioxide, and nitric oxide-the three-gas respiratory cycle-that insures adequate oxygen and nutrient delivery to meet local metabolic demand. In this context, it is blood flow and not blood oxygen content that is the main driver of tissue oxygenation by RBCs. Herein, we review the lines of experimentation that led to this understanding of RBC function; from the foundational understanding of allosteric regulation of oxygen binding in Hb in the stereochemical model of Perutz, to blood flow autoregulation (hypoxic vasodilation governing oxygen delivery) observed by Guyton, to current understanding that centers on S-nitrosylation of Hb (ie, S-nitrosohemoglobin; SNO-Hb) as a purveyor of oxygen-dependent vasodilatory activity. Notably, hypoxic vasodilation is recapitulated by native S-nitrosothiol (SNO)-replete RBCs and by SNO-Hb itself, whereby SNO is released from Hb and RBCs during deoxygenation, in proportion to the degree of Hb deoxygenation, to regulate vessels directly. In addition, we discuss how dysregulation of this system through genetic mutation in Hb or through disease is a common factor in oxygenation pathologies resulting from microcirculatory impairment, including sickle cell disease, ischemic heart disease, and heart failure. We then conclude by identifying potential therapeutic interventions to correct deficits in RBC-mediated vasodilation to improve oxygen delivery-steps toward effective microvasculature-targeted therapies. To the extent that diseases of the heart, lungs, and blood are associated with impaired tissue oxygenation, the development of new therapies based on the three-gas respiratory system have the potential to improve the well-being of millions of patients.
Collapse
Affiliation(s)
- Richard T Premont
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| | - James D Reynolds
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Anesthesiology and Perioperative Medicine (J.D.R.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| | - Rongli Zhang
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Medicine, Cardiovascular Research Institute (R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH
| | - Jonathan S Stamler
- From the Institute for Transformative Molecular Medicine (R.T.P., J.D.R., R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Department of Medicine, Cardiovascular Research Institute (R.Z., J.S.S.), Case Western Reserve University School of Medicine, OH.,Harrington Discovery Institute (R.T.P., J.D.R., J.S.S.), University Hospitals Cleveland Medical Center, OH
| |
Collapse
|
25
|
Quiñonez-Bastidas GN, Pineda-Farias JB, Flores-Murrieta FJ, Rodríguez-Silverio J, Reyes-García JG, Godínez-Chaparro B, Granados-Soto V, Rocha-González HI. Antinociceptive effect of (-)-epicatechin in inflammatory and neuropathic pain in rats. Behav Pharmacol 2019; 29:270-279. [PMID: 28590304 DOI: 10.1097/fbp.0000000000000320] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The aim of this study was to investigate the antinociceptive potential of (-)-epicatechin and the possible mechanisms of action involved in its antinociceptive effect. The carrageenan and formalin tests were used as inflammatory pain models. A plethysmometer was used to measure inflammation and L5/L6 spinal nerve ligation as a neuropathic pain model. Oral (-)-epicatechin reduced carrageenan-induced inflammation and nociception by about 59 and 73%, respectively, and reduced formalin- induced and nerve injury-induced nociception by about 86 and 43%, respectively. (-)-Epicatechin-induced antinociception in the formalin test was prevented by the intraperitoneal administration of antagonists: methiothepin (5-HT1/5 receptor), WAY-100635 (5-HT1A receptor), SB-224289 (5-HT1B receptor), BRL-15572 (5-HT1D receptor), SB-699551 (5-HT5A receptor), naloxone (opioid receptor), CTAP (μ opioid receptor), nor-binaltorphimine (κ opioid receptor), and 7-benzylidenenaltrexone (δ1 opioid receptor). The effect of (-)-epicatechin was also prevented by the intraperitoneal administration of L-NAME [nitric oxide (NO) synthase inhibitor], 7-nitroindazole (neuronal NO synthase inhibitor), ODQ (guanylyl cyclase inhibitor), glibenclamide (ATP-sensitive K channel blocker), 4-aminopyridine (voltage-dependent K channel blocker), and iberiotoxin (large-conductance Ca-activated K channel blocker), but not by amiloride (acid sensing ion channel blocker). The data suggest that (-)-epicatechin exerts its antinociceptive effects by activation of the NO-cyclic GMP-K channels pathway, 5-HT1A/1B/1D/5A serotonergic receptors, and μ/κ/δ opioid receptors.
Collapse
Affiliation(s)
- Geovanna N Quiñonez-Bastidas
- Section of Postgraduate Studies and Research, Escuela Superior de Medicina, Instituto Politecnico Nacional.,Department of Biological Systems, Division of Biological Sciences and Health, UAM-Xochimilco
| | | | - Francisco J Flores-Murrieta
- Section of Postgraduate Studies and Research, Escuela Superior de Medicina, Instituto Politecnico Nacional.,Pharmacology Research Unit, INER, Ismael Cosio Villegas, Secretaria de Salud, Mexico City, Mexico
| | - Juan Rodríguez-Silverio
- Section of Postgraduate Studies and Research, Escuela Superior de Medicina, Instituto Politecnico Nacional
| | - Juan G Reyes-García
- Section of Postgraduate Studies and Research, Escuela Superior de Medicina, Instituto Politecnico Nacional
| | | | | | - Héctor I Rocha-González
- Section of Postgraduate Studies and Research, Escuela Superior de Medicina, Instituto Politecnico Nacional
| |
Collapse
|
26
|
Elgert C, Rühle A, Sandner P, Behrends S. A novel soluble guanylyl cyclase activator, BR 11257, acts as a non-stabilising partial agonist of sGC. Biochem Pharmacol 2019; 163:142-153. [DOI: 10.1016/j.bcp.2019.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/06/2019] [Indexed: 01/05/2023]
|
27
|
Qiu H, Qi P, Liu J, Yang Y, Tan X, Xiao Y, Maitz MF, Huang N, Yang Z. Biomimetic engineering endothelium-like coating on cardiovascular stent through heparin and nitric oxide-generating compound synergistic modification strategy. Biomaterials 2019; 207:10-22. [PMID: 30947118 DOI: 10.1016/j.biomaterials.2019.03.033] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/15/2019] [Accepted: 03/22/2019] [Indexed: 01/23/2023]
Abstract
Co-immobilization of two or more molecules with different and complementary functions to prevent thrombosis, suppress smooth muscle cell (SMC) proliferation, and support endothelial cell (EC) growth is generally considered to be promising for the re-endothelialization on cardiovascular stents. However, integration of molecules with distinct therapeutic effects does not necessarily result in synergistic physiological functions due to the lack of interactions among them, limiting their practical efficacy. Herein, we apply heparin and nitric oxide (NO), two key molecules of the physiological functions of endothelium, to develop an endothelium-mimetic coating. Such coating is achieved by sequential conjugation of heparin and the NO-generating compound selenocystamine (SeCA) on an amine-bearing film of plasma polymerized allylamine. The resulting surface combines the anti-coagulant (anti-FXa) function provided by the heparin and the anti-platelet activity of the catalytically produced NO. It also endows the stents with the ability to simultaneously up-regulate α-smooth muscle actin (α-SMA) expression and to increase cyclic guanylate monophosphate (cGMP) synthesis of SMC, thereby significantly promoting their contractile phenotype and suppressing their proliferation. Importantly, this endothelium-biomimetic coating creates a favorable microenvironment for EC over SMC. These features impressively improve the antithrombogenicity, re-endothelialization and anti-restenosis of vascular stents in vivo.
Collapse
Affiliation(s)
- Hua Qiu
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Pengkai Qi
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Jingxia Liu
- Physical Education Department, Southwest Jiaotong University, Chengdu, 610031, China
| | - Ying Yang
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 4059, Australia
| | - Xing Tan
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yu Xiao
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Manfred F Maitz
- Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden, 01069, Germany
| | - Nan Huang
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Zhilu Yang
- Key Lab of Advanced Technology of Materials of Education Ministry, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
28
|
Tan CS, Loh YC, Ch'ng YS, Ng CH, Yeap ZQ, Ahmad M, Asmawi MZ, Yam MF. Vasorelaxant and chemical fingerprint studies of Citrus reticulatae pericarpium extracts. JOURNAL OF ETHNOPHARMACOLOGY 2019; 232:135-144. [PMID: 30543913 DOI: 10.1016/j.jep.2018.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/16/2018] [Accepted: 12/08/2018] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Citrus reticulatae Pericarpium (Chen pi) was widely used as an important ingredient in the prescription of TCM to treat phlegm fluid retention type hypertension. Since Chen pi is involved in treatment as antihypertensive TCM formula, we have reasonable expectation in believing that it might possess vasorelaxant activity. AIM OF THE STUDY This study is designed to investigate the vasorelaxant effect of Chen pi and to study its pharmacology effects. MATERIALS AND METHODS The vasorelaxant effect of water extract of Chen pi (CRW) were evaluated on thoracic aortic rings isolated from Sprague Dawley rats. The fingerprint of Chen pi and the extracts were developed with quantification of hesperidin content by HPTLC. RESULTS CRW exhibited the strongest vasorelaxant activity. CRW caused the relaxation of the phenylephrine pre-contracted aortic rings in the presence and absence of endothelium as well as in potassium chloride pre-contracted endothelium-intact aortic ring. The incubation of propranolol (β-adrenergic receptor blocker), atropine (muscarinic receptor blocker), Nω-nitro-L-arginine methyl ester (NO synthase inhibitor), ODQ (sGC inhibitor), indomethacin (COX inhibitor), 4-aminopyridine (KV blocker), barium chloride (Kir blocker), and glibenclamide (KATP blocker) significantly reduced the vasorelaxant effects of CRW. CRW was also found to be active in reducing Ca2+ releases from the sarcoplasmic reticulum and suppressing the voltage-operated calcium channels. CONCLUSION The vasorelaxant effect of CRW on rat aorta involves NO/sGC, calcium and potassium channels, muscarinic and β-adrenergic receptors.
Collapse
Affiliation(s)
- Chu Shan Tan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Yean Chun Loh
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Yung Sing Ch'ng
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Chiew Hoong Ng
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Zhao Qin Yeap
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Mariam Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Mun Fei Yam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia; College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| |
Collapse
|
29
|
|
30
|
Tropea T, Wareing M, Greenwood SL, Feelisch M, Sibley CP, Cottrell EC. Nitrite mediated vasorelaxation in human chorionic plate vessels is enhanced by hypoxia and dependent on the NO-sGC-cGMP pathway. Nitric Oxide 2018; 80:82-88. [PMID: 30179715 PMCID: PMC6199414 DOI: 10.1016/j.niox.2018.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/05/2018] [Accepted: 08/30/2018] [Indexed: 11/25/2022]
Abstract
Adequate perfusion of the placental vasculature is essential to meet the metabolic demands of fetal growth and development. Lacking neural control, local tissue metabolites, circulating and physical factors contribute significantly to blood flow regulation. Nitric oxide (NO) is a key regulator of fetoplacental vascular tone. Nitrite, previously considered an inert end-product of NO oxidation, has been shown to provide an important source of NO. Reduction of nitrite to NO may be particularly relevant in tissue when the oxygen-dependent NO synthase (NOS) activity is compromised, e.g. in hypoxia. The contribution of this pathway in the placenta is currently unknown. We hypothesised that nitrite vasodilates human placental blood vessels, with enhanced efficacy under hypoxia. Placentas were collected from uncomplicated pregnancies and the vasorelaxant effect of nitrite (10-6-5x10-3 M) was assessed using wire myography on isolated pre-constricted chorionic plate arteries (CPAs) and veins (CPVs) under normoxic (pO2 ∼5%) and hypoxic (pO2 ∼1%) conditions. The dependency on the NO-sGC-cGMP pathway and known nitrite reductase (NiR) activities was also investigated. Nitrite caused concentration-dependent vasorelaxation in both arteries and veins, and this effect was enhanced by hypoxia, significantly in CPVs (P < 0.01) and with a trend in CPAs (P = 0.054). Pre-incubation with NO scavengers (cPTIO and oxyhemoglobin) attenuated (P < 0.01 and P < 0.0001, respectively), and the sGC inhibitor ODQ completely abolished nitrite-mediated vasorelaxation, confirming the involvement of NO and sGC. Inhibition of potential NiR enzymes xanthine oxidoreductase, mitochondrial aldehyde dehydrogenase and mitochondrial bc1 complex did not attenuate vasorelaxation. This data suggests that nitrite may provide an important reservoir of NO bioactivity within the placenta to enhance blood flow when fetoplacental oxygenation is impaired, as occurring in pregnancy diseases such as pre-eclampsia and fetal growth restriction.
Collapse
Affiliation(s)
- Teresa Tropea
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom.
| | - Mark Wareing
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| | - Susan L Greenwood
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, Southampton General Hospital and Institute for Life Sciences, University of Southampton, United Kingdom
| | - Colin P Sibley
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| | - Elizabeth C Cottrell
- Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Maternal & Fetal Health Research Centre, University of Manchester, United Kingdom
| |
Collapse
|
31
|
Radziwon-Balicka A, Lesyk G, Back V, Fong T, Loredo-Calderon EL, Dong B, El-Sikhry H, El-Sherbeni AA, El-Kadi A, Ogg S, Siraki A, Seubert JM, Santos-Martinez MJ, Radomski MW, Velazquez-Martinez CA, Winship IR, Jurasz P. Differential eNOS-signalling by platelet subpopulations regulates adhesion and aggregation. Cardiovasc Res 2018; 113:1719-1731. [PMID: 29016749 DOI: 10.1093/cvr/cvx179] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/01/2017] [Indexed: 12/22/2022] Open
Abstract
Aims In addition to maintaining haemostasis, circulating blood platelets are the cellular culprits that form occlusive thrombi in arteries and veins. Compared to blood leucocytes, which exist as functionally distinct subtypes, platelets are considered to be relatively simple cell fragments that form vascular system plugs without a differentially regulated cellular response. Hence, investigation into platelet subpopulations with distinct functional roles in haemostasis/thrombosis has been limited. In our present study, we investigated whether functionally distinct platelet subpopulations exist based on their ability to generate and respond to nitric oxide (NO), an endogenous platelet inhibitor. Methods and results Utilizing highly sensitive and selective flow cytometry protocols, we demonstrate that human platelet subpopulations exist based on the presence and absence of endothelial nitric oxide synthase (eNOS). Platelets lacking eNOS (approximately 20% of total platelets) fail to produce NO and have a down-regulated soluble guanylate cyclase-protein kinase G (sGC-PKG)-signalling pathway. In flow chamber and aggregation experiments eNOS-negative platelets primarily initiate adhesion to collagen, more readily activate integrin αIIbβ3 and secrete matrix metalloproteinase-2, and form larger aggregates than their eNOS-positive counterparts. Conversely, platelets having an intact eNOS-sGC-PKG-signalling pathway (approximately 80% of total platelets) form the bulk of an aggregate via increased thromboxane synthesis and ultimately limit its size via NO generation. Conclusion These findings reveal previously unrecognized characteristics and complexity of platelets and their regulation of adhesion/aggregation. The identification of platelet subpopulations also has potentially important consequences to human health and disease as impaired platelet NO-signalling has been identified in patients with coronary artery disease.
Collapse
Affiliation(s)
- Aneta Radziwon-Balicka
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Gabriela Lesyk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Valentina Back
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Teresa Fong
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Erica L Loredo-Calderon
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Bin Dong
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G-2R3, Canada
| | - Haitham El-Sikhry
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Ahmed A El-Sherbeni
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Ayman El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Stephen Ogg
- Department of Medical Microbiology and Immunology, University of Alberta Edmonton, AB T6G-2E1, Canada
| | - Arno Siraki
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada.,Department of Pharmacology, University of Alberta Edmonton, AB T6G-2H7, Canada.,Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G-2S2, Canada.,Mazankowski Heart Institute, Edmonton, AB T6G-2R7
| | | | - Marek W Radomski
- College of Medicine, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | | | - Ian R Winship
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G-2R3, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada.,Department of Pharmacology, University of Alberta Edmonton, AB T6G-2H7, Canada.,Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G-2S2, Canada.,Mazankowski Heart Institute, Edmonton, AB T6G-2R7
| |
Collapse
|
32
|
Friebe A, Voußen B, Groneberg D. NO-GC in cells 'off the beaten track'. Nitric Oxide 2018; 77:12-18. [DOI: 10.1016/j.niox.2018.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/14/2018] [Accepted: 02/23/2018] [Indexed: 02/08/2023]
|
33
|
Zhang X, Yu S, Deng G, He Y, Li Q, Yu L, Yu Y. Effects of nitric oxide donor S-nitrosoglutathione on apoptosis of apheresis platelets. Hematology 2018; 23:574-580. [PMID: 29890936 DOI: 10.1080/10245332.2018.1483547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Xiongxin Zhang
- The Department of Anesthesiology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Shifang Yu
- The Department of Transfusion Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Gang Deng
- The Ningbo Central Blood Station, Ningbo, People’s Republic of China
| | - Yunlei He
- The Ningbo Central Blood Station, Ningbo, People’s Republic of China
| | - Qiang Li
- The Department of Laboratory Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Lu Yu
- The Ningbo Central Blood Station, Ningbo, People’s Republic of China
| | - Yong Yu
- The Department of Transfusion Medicine, Ningbo No. 2 Hospital, Ningbo, People’s Republic of China
| |
Collapse
|
34
|
Liu T, Zhang M, Terry MH, Schroeder H, Wilson SM, Power GG, Li Q, Tipple TE, Borchardt D, Blood AB. Hemodynamic Effects of Glutathione-Liganded Binuclear Dinitrosyl Iron Complex: Evidence for Nitroxyl Generation and Modulation by Plasma Albumin. Mol Pharmacol 2018; 93:427-437. [PMID: 29476040 PMCID: PMC5878675 DOI: 10.1124/mol.117.110957] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/21/2018] [Indexed: 12/25/2022] Open
Abstract
Glutathione-liganded binuclear dinitrosyl iron complex (glut-BDNIC) has been proposed to be a donor of nitric oxide (NO). This study was undertaken to investigate the mechanisms of vasoactivity, systemic hemodynamic effects, and pharmacokinetics of glut-BDNIC. To test the hypothesis that glut-BDNICs vasodilate by releasing NO in its reduced [nitroxyl (HNO)] state, a bioassay method of isolated, preconstricted ovine mesenteric arterial rings was used in the presence of selective scavengers of HNO or NO free radical (NO•); the vasodilatory effects of glut-BDNIC were found to have characteristics similar to those of an HNO donor and markedly different than an NO• donor. In addition, products of the reaction of glut-BDNIC with CPTIO [2-(4-carboxyphenyl)-4,4,5-tetramethyl imidazoline-1-oxyl-3-oxide] were found to have electron paramagnetic characteristics similar to those of an HNO donor compared with an NO• donor. In contrast to S-nitroso-glutathione, which was vasodilative both in vitro and in vivo, the potency of glut-BDNIC-mediated vasodilation was markedly diminished in both rats and sheep. Wire myography showed that plasma albumin contributed to this loss of hypotensive effects, an effect abolished by modification of the cysteine-thiol residue of albumin. High doses of glut-BDNIC caused long-lasting hypotension in rats that can be at least partially attributed to its long circulating half-life of ∼44 minutes. This study suggests that glut-BDNIC is an HNO donor, and that its vasoactive effects are modulated by binding to the cysteine residue of plasma proteins, such as albumin.
Collapse
Affiliation(s)
- Taiming Liu
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Meijuan Zhang
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Michael H Terry
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Hobe Schroeder
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Sean M Wilson
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Gordon G Power
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Qian Li
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Trent E Tipple
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Dan Borchardt
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| | - Arlin B Blood
- Division of Neonatology, Department of Pediatrics (T.L., M.Z., A.B.B.), Department of Respiratory Care (M.H.T.), and Center for Perinatal Biology (H.S., S.M.W., G.G.P., A.B.B.), Loma Linda University School of Medicine, Loma Linda, California; Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, Alabama (Q.L., T.E.T.); and Department of Chemistry, University of California, Riverside, California (D.B.)
| |
Collapse
|
35
|
Wobst J, Schunkert H, Kessler T. Genetic alterations in the NO-cGMP pathway and cardiovascular risk. Nitric Oxide 2018; 76:105-112. [PMID: 29601927 DOI: 10.1016/j.niox.2018.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/18/2018] [Accepted: 03/26/2018] [Indexed: 12/18/2022]
Abstract
In the past ten years, several chromosomal loci have been identified by genome-wide association studies to influence the risk of coronary artery disease (CAD) and its risk factors. The GUCY1A3 gene encoding the α1 subunit of the soluble guanylyl cyclase (sGC) resides at one of these loci and has been strongly associated with blood pressure and CAD risk. More recently, further genes in the pathway encoding the endothelial nitric oxide synthase, the phosphodiesterases 3A and 5A, and the inositol 1,4,5-trisphosphate receptor I-associated protein (IRAG), i.e., NOS3, PDE3A, PDE5A, and MRVI1, respectively, were likewise identified as CAD risk genes. In this review, we highlight the genetic findings linking variants in NO-cGMP signaling and cardiovascular disease, discuss the potential underlying mechanisms which might propagate the development of atherosclerosis, and speculate about therapeutic implications.
Collapse
Affiliation(s)
- Jana Wobst
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
36
|
Liu T, Zhang M, Terry MH, Schroeder H, Wilson SM, Power GG, Li Q, Tipple TE, Borchardt D, Blood AB. Nitrite potentiates the vasodilatory signaling of S-nitrosothiols. Nitric Oxide 2018; 75:60-69. [PMID: 29428841 DOI: 10.1016/j.niox.2018.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 12/27/2022]
Abstract
Nitrite and S-nitrosothiols (SNOs) are both byproducts of nitric oxide (NO) metabolism and are proposed to cause vasodilation via activation of soluble guanylate cyclase (sGC). We have previously reported that while SNOs are potent vasodilators at physiological concentrations, nitrite itself only produces vasodilation at supraphysiological concentrations. Here, we tested the hypothesis that sub-vasoactive concentrations of nitrite potentiate the vasodilatory effects of SNOs. Multiple exposures of isolated sheep arteries to S-nitroso-glutathione (GSNO) resulted in a tachyphylactic decreased vasodilatory response to GSNO but not to NO, suggesting attenuation of signaling steps upstream from sGC. Exposure of arteries to 1 μM nitrite potentiated the vasodilatory effects of GSNO in naive arteries and abrogated the tachyphylactic response to GSNO in pre-exposed arteries, suggesting that nitrite facilitates GSNO-mediated activation of sGC. In intact anesthetized sheep and rats, inhibition of NO synthases to decrease plasma nitrite levels attenuated vasodilatory responses to exogenous infusions of GSNO, an effect that was reversed by exogenous infusion of nitrite at sub-vasodilating levels. This study suggests nitrite potentiates SNO-mediated vasodilation via a mechanism that lies upstream from activation of sGC.
Collapse
Affiliation(s)
- Taiming Liu
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Meijuan Zhang
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Michael H Terry
- Department of Respiratory Care, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Hobe Schroeder
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Sean M Wilson
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Gordon G Power
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Qian Li
- Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Trent E Tipple
- Neonatal Redox Biology Laboratory, Division of Neonatology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Dan Borchardt
- Department of Chemistry, University of California, Riverside, CA 92521, United States
| | - Arlin B Blood
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States; Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States.
| |
Collapse
|
37
|
Zhang Q, Tian P, Zhai M, Lei X, Yang Z, Liu Y, Liu M, Huang H, Zhang X, Yang X, Zhao Y, Meng Z. Formaldehyde regulates vascular tensions through nitric oxide-cGMP signaling pathway and ion channels. CHEMOSPHERE 2018; 193:60-73. [PMID: 29126066 DOI: 10.1016/j.chemosphere.2017.11.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/26/2017] [Accepted: 11/03/2017] [Indexed: 06/07/2023]
Abstract
Formaldehyde (FA) has been linked to the detrimental cardiovascular effects. Here, we explored the effects and mechanisms of FA on rat aortas both in vivo and in vitro. The results presented that FA evidently lowered the blood pressures of rats. The expression levels of BKCa subunits α and β1 and iNOS of the aortas were up-regulated by FA in vivo. However, FA markedly reduced the levels of Cav1.2 and Cav1.3, which are the subunits of L-Ca2+ channel. Furthermore, the contents of NO, cGMP and iNOS in the aortas were augmented by FA. To further confirm these findings, the mechanisms accredited to these effects were examined in vitro. The data showed that FA contracted the isolated aortic rings at low concentrations (<300 μM), while it relaxed the rings at high concentrations (>500 μM). The FA-induced vasoconstriction at low concentrations was blocked partly by an inhibitor of ACE. The relaxation caused by FA at high concentrations was attenuated by the inhibitors of NO-cGMP pathway, L-Ca2+ channel and BKCa channel, respectively. Similarly, the expression of iNOS was strongly enhanced by FA in vitro. The effects of FA on the aortic rings with endothelium were significantly greater than those on the rings without endothelium. Our results indicate that the vasoconstriction of FA at low concentrations might be partially pertinent to endothelin, and the FA-caused vasorelaxation at high concentrations is possibly associated with the NO-cGMP pathway, L-Ca2+ channel and BKCa channel. This study will improve our understanding of the pathogenic mechanisms for FA-related cardiovascular diseases.
Collapse
Affiliation(s)
- Quanxi Zhang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China.
| | - Peiru Tian
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Miaomiao Zhai
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Xiaodong Lei
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Zhenhua Yang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Yan Liu
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Mengting Liu
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Hao Huang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Xiri Zhang
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - Xu Yang
- Laboratory of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Huazhong Normal University, Wuhan 430079, China.
| | - Yun Zhao
- Laboratory of Environmental Biomedicine, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Sciences, Huazhong Normal University, Wuhan 430079, China
| | - Ziqiang Meng
- Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
38
|
Overview of the Microenvironment of Vasculature in Vascular Tone Regulation. Int J Mol Sci 2018; 19:ijms19010120. [PMID: 29301280 PMCID: PMC5796069 DOI: 10.3390/ijms19010120] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/11/2017] [Accepted: 12/16/2017] [Indexed: 12/16/2022] Open
Abstract
Hypertension is asymptomatic and a well-known “silent killer”, which can cause various concomitant diseases in human population after years of adherence. Although there are varieties of synthetic antihypertensive drugs available in current market, their relatively low efficacies and major application in only single drug therapy, as well as the undesired chronic adverse effects associated, has drawn the attention of worldwide scientists. According to the trend of antihypertensive drug evolution, the antihypertensive drugs used as primary treatment often change from time-to-time with the purpose of achieving the targeted blood pressure range. One of the major concerns that need to be accounted for here is that the signaling mechanism pathways involved in the vasculature during the vascular tone regulation should be clearly understood during the pharmacological research of antihypertensive drugs, either in vitro or in vivo. There are plenty of articles that discussed the signaling mechanism pathways mediated in vascular tone in isolated fragments instead of a whole comprehensive image. Therefore, the present review aims to summarize previous published vasculature-related studies and provide an overall depiction of each pathway including endothelium-derived relaxing factors, G-protein-coupled, enzyme-linked, and channel-linked receptors that occurred in the microenvironment of vasculature with a full schematic diagram on the ways their signals interact. Furthermore, the crucial vasodilative receptors that should be included in the mechanisms of actions study on vasodilatory effects of test compounds were suggested in the present review as well.
Collapse
|
39
|
Kim-Shapiro DB, Gladwin MT. Nitric oxide pathology and therapeutics in sickle cell disease. Clin Hemorheol Microcirc 2018; 68:223-237. [PMID: 29614634 PMCID: PMC5911689 DOI: 10.3233/ch-189009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sickle cell disease is caused by a mutant form of hemoglobin that polymerizes under hypoxic conditions which leads to red blood cell (RBC) distortion, calcium-influx mediated RBC dehydration, increased RBC adhesivity, reduced RBC deformability, increased RBC fragility, and hemolysis. These impairments in RBC structure and function result in multifaceted downstream pathology including inflammation, endothelial cell activation, platelet and leukocyte activation and adhesion, and thrombosis, all of which contribute vascular occlusion and substantial morbidity and mortality. Hemoglobin released upon RBC hemolysis scavenges nitric oxide (NO) and generates reactive oxygen species (ROS) and thereby decreases bioavailability of this important signaling molecule. As the endothelium-derived relaxing factor, NO acts as a vasodilator and also decreases platelet, leukocyte, and endothelial cell activation. Thus, low NO bioavailability contributes to pathology in sickle cell disease and its restoration could serve as an effective treatment. Despite its promise, clinical trials based on restoring NO bioavailability have so far been mainly disappointing. However, particular "NO donating" agents such as nitrite, which unlike some other NO donors can improve sickle RBC properties, may yet prove effective.
Collapse
Affiliation(s)
- Daniel B. Kim-Shapiro
- Department of Physics and the Translational Science Center, Wake Forest University, Winston-Salem NC 27109
| | - Mark T. Gladwin
- Heart, Lung, Blood and Vascular Medicine Institute and the Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
40
|
Rathod KS, Jones DA, Van-Eijl TJA, Tsang H, Warren H, Hamshere SM, Kapil V, Jain AK, Deaner A, Poulter N, Caulfield MJ, Mathur A, Ahluwalia A. Randomised, double-blind, placebo-controlled study investigating the effects of inorganic nitrate on vascular function, platelet reactivity and restenosis in stable angina: protocol of the NITRATE-OCT study. BMJ Open 2016; 6:e012728. [PMID: 27998900 PMCID: PMC5223652 DOI: 10.1136/bmjopen-2016-012728] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION The mainstay treatment for reducing the symptoms of angina and long-term risk of heart attacks in patients with heart disease is stent implantation in the diseased coronary artery. While this procedure has revolutionised treatment, the incidence of secondary events remains a concern. These repeat events are thought to be due, in part, to continued enhanced platelet reactivity, endothelial dysfunction and ultimately restenosis of the stented artery. In this study, we will investigate whether a once a day inorganic nitrate administration might favourably modulate platelet reactivity and endothelial function leading to a decrease in restenosis. METHODS AND DESIGN NITRATE-OCT is a double-blind, randomised, single-centre, placebo-controlled phase II trial that will enrol 246 patients with stable angina due to have elective percutaneous coronary intervention procedure with stent implantation. Patients will be randomised to receive 6 months of a once a day dose of either nitrate-rich beetroot juice or nitrate-deplete beetroot juice (placebo) starting up to 1 week before their procedure. The primary outcome is reduction of in-stent late loss assessed by quantitative coronary angiography and optical coherence tomography at 6 months. The study is powered to detect a 0.22±0.55 mm reduction in late loss in the treatment group compared with the placebo group. Secondary end points include change from baseline assessment of endothelial function measured using flow-mediated dilation at 6 months, target vessel revascularisation (TVR), restenosis rate (diameter>50%) and in-segment late loss at 6 months, markers of inflammation and platelet reactivity and major adverse cardiac events (ie, myocardial infarction, death, cerebrovascular accident, TVR) at 12 and 24 months. ETHICS AND DISSEMINATION The study was approved by the Local Ethics Committee (15/LO/0555). Trial results will be published according to the CONSORT statement and will be presented at conferences and reported in peer-reviewed journals. TRIAL REGISTRATION NUMBERS NCT02529189 and ISRCTN17373946, Pre-results.
Collapse
Affiliation(s)
- Krishnaraj S Rathod
- Barts NIHR Cardiovascular Biomedical Research Unit, Centre of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University, London, UK
- Department of Cardiology, Barts Heart Centre,2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Daniel A Jones
- Barts NIHR Cardiovascular Biomedical Research Unit, Centre of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University, London, UK
- Department of Cardiology, Barts Heart Centre,2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - T J A Van-Eijl
- Department of Cardiology, Barts Heart Centre,2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Hilda Tsang
- Imperial Clinical Trials Unit, Imperial College, London, UK
| | - Helen Warren
- Barts NIHR Cardiovascular Biomedical Research Unit, Centre of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University, London, UK
| | - Stephen M Hamshere
- Department of Cardiology, Barts Heart Centre,2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Vikas Kapil
- Barts NIHR Cardiovascular Biomedical Research Unit, Centre of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University, London, UK
| | - Ajay K Jain
- Department of Cardiology, Barts Heart Centre,2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- King George Hospital, Barking and Havering NHS Trust, London, UK
| | - Andrew Deaner
- Department of Cardiology, Barts Heart Centre,2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- King George Hospital, Barking and Havering NHS Trust, London, UK
| | - Neil Poulter
- Imperial Clinical Trials Unit, Imperial College, London, UK
| | - Mark J Caulfield
- Barts NIHR Cardiovascular Biomedical Research Unit, Centre of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University, London, UK
| | - Anthony Mathur
- Barts NIHR Cardiovascular Biomedical Research Unit, Centre of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University, London, UK
- Department of Cardiology, Barts Heart Centre,2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Amrita Ahluwalia
- Barts NIHR Cardiovascular Biomedical Research Unit, Centre of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University, London, UK
| |
Collapse
|
41
|
Jayakumar T, Lin KC, Lu WJ, Lin CY, Pitchairaj G, Li JY, Sheu JR. Nobiletin, a citrus flavonoid, activates vasodilator-stimulated phosphoprotein in human platelets through non-cyclic nucleotide-related mechanisms. Int J Mol Med 2016; 39:174-182. [PMID: 27959381 PMCID: PMC5179174 DOI: 10.3892/ijmm.2016.2822] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 11/28/2016] [Indexed: 11/19/2022] Open
Abstract
Nobiletin, a bioactive polymethoxylated flavone, has been described to possess a diversity of biological effects through its antioxidant and anti-inflammatory properties. Vasodilator-stimulated phosphoprotein (VASP) is a common substrate for cyclic AMP and cyclic GMP-regulated protein kinases [i.e., cyclic AMP-dependent protein kinase (PKA; also known as protein kinase A) and cyclic GMP-dependent protein kinase (PKG; also known as protein kinase G)] and it has been shown to be directly phosphorylated by protein kinase C (PKC). In the present study, we demonstrate that VASP is phosphorylated by nobiletin in human platelets via a non-cyclic nucleotide-related mechanism. This was confirmed by the use of inhibitors of adenylate cyclase (SQ22536) and guanylate cyclase [1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ)], since they prevented VASP phosphorylation induced by nobiletin. Furthormore, this event was also not affected by specific inhibitors of PKA (H-89), PKG (KT5823) and PKC (Ro318220), representing cyclic nucleotide-dependent pathways upon nobiletin-induced VASP phosphorylation. Similarly, inhibitors of p38 mitogen-activated protein kinase (MAPK; SB203580), extracellular signal-regulated kinase 2 (ERK2; PD98059), c-Jun N-terminal kinase 1 (JNK1; SP600125), Akt (LY294002) and nuclear factor-κB (NF-κB; Bay11-7082) did not affect nobiletin-induced VASP phosphorylation. Moreover, electron spin resonance, dichlorofluorescein fluorescence and western blotting techniques revealed that nobiletin did not affect hydroxyl radicals (OH•), intracellular reactive oxygen species (ROS) and on protein carbonylation, respectively. Furthermore, the nobiletin-induced VASP phosphorylation was surprisingly reversed by the intracellular antioxidant, N-acetylcysteine (NAC), but not by the inhibitor of NADPH oxidase, diphenyleneiodonium chloride (DPI). It was surprising to observe the differential effects of nobiletin and NAC on VASP phosphorylation in human platelets, since they both have been reported to have antioxidant properties. The likely explanation for this discrepancy is that NAC may bind to allosteric sites on the receptor different from those that nobiletin binds to in human platelets. Taken together, our findings suggest that nobiletin induces VASP phosphorylation in human platelets through non-cyclic nucleotide-related mechanisms. Nevertheless, the exact mechanisms responsible for these effects need to be further confirmed in future studies.
Collapse
Affiliation(s)
- Thanasekaran Jayakumar
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Kao-Chang Lin
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Wan-Jung Lu
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Chia-Ying Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Geraldine Pitchairaj
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Jiun-Yi Li
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Joen-Rong Sheu
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| |
Collapse
|
42
|
Lambert MP. Platelets in liver and renal disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:251-255. [PMID: 27913488 PMCID: PMC6142504 DOI: 10.1182/asheducation-2016.1.251] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
This review will discuss how 2 common and morbid conditions, renal disease and liver disease, alter platelet number and function. It will review the impact of thrombocytopenia on bleeding complications in patients with these disorders and whether the low platelet count actually correlates with bleeding risk. Emerging data also suggest that platelets are much more than bystanders in both renal and liver disease, but instead play an active role in the pathobiology of these disorders. This review will briefly cover the emerging information on novel roles of platelets in the biology of renal and liver disease.
Collapse
Affiliation(s)
- Michele P Lambert
- Divisions of Hematology, Departments of Pediatrics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
43
|
Thalidomide attenuates the development and expression of antinociceptive tolerance to μ-opioid agonist morphine through l-arginine-iNOS and nitric oxide pathway. Biomed Pharmacother 2016; 85:493-502. [PMID: 27899254 DOI: 10.1016/j.biopha.2016.11.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/28/2016] [Accepted: 11/14/2016] [Indexed: 12/30/2022] Open
Abstract
Morphine is a μ-opioid analgesic drug which is used in the treatment and management of chronic pain. However, due to development of antinociceptive tolerance its clinical use is limited. Thalidomide is an old glutamic acid derivative which recently reemerged because of its potential to counteract a number of disorders including neurodegenerative disorders. The potential underlying mechanisms and effects of thalidomide on morphine-induced antinociceptive tolerance is still elusive. Hence, the present study was designed to explore the effect of thalidomide on the development and expression of morphine antinociceptive tolerance targeting l-arginine-nitric oxide (NO) pathway in mice and T98G human glioblastoma cell line. When thalidomide was administered in a dose of 17.5mg/kg before each dose of morphine chronically for 5days it prevented the development of antinociceptive tolerance. Also, a single dose of thalidomide 20mg/kg attenuated the expression phase of antinociceptive tolerance. The protective effect of thalidomide was augmented in development phase when co-administration with NOS inhibitors like L-NAME (non- selective NOS inhibitor; 2mg/kg) or aminoguanidine (selective inducible NOS inhibitor; 50mg/kg). Also, the reversal effect of thalidomide in expression phase was potentiated when concomitantly administrated with L-NAME (5mg/kg) or aminoguanidine (100mg/kg). Co-administration of ODQ (a guanylyl cyclase inhibitor) 10mg/kg in developmental phase or 20mg/kg in expression phase also progressively increased the pain threshold. In addition, thalidomide (20μM) also significantly inhibited the overexpression of iNOS gene induced by morphine (2.5μM) in T98G cell line. Hence, our findings suggest that thalidomide has protective effect both in the development and expression phases of morphine antinociceptive tolerance. It is also evident that this effect of thalidomide is induced by the inhibition of NOS enzyme predominantly iNOS.
Collapse
|
44
|
Bai Y, Zhang Q, Yang Z, Meng Z, Zhao Q. The vasorelaxant mechanisms of methanol on isolated rat aortic rings: Involvement of ion channels and signal transduction pathways. Hum Exp Toxicol 2016; 36:1031-1038. [PMID: 27852936 DOI: 10.1177/0960327116678301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is reported that methanol is generally used as an industrial solvent, antifreeze, windshield washer fluid, cooking fuel and perfume. Methanol ingestion can lead to severe metabolic disturbances, blindness, or even death. So far, few studies about its negative effects on cardiovascular system have been reported. The purpose of this study was to determine the vasoactive effect of methanol and roles of ion channels and signal transduction pathways on isolated rat aorta. The results suggested that the mechanism of methanol-induced vasorelaxation at low concentrations (<500 mM) was mediated by ATP-sensitive K+ (KATP) and L-type Ca2+ channels, but the mechanism at high concentrations (>600 mM) was related to KATP, voltage-dependent K+, big-conductance Ca2+-activated K+, L-type Ca2+ channels as well as prostacyclin, protein kinase C, β-adrenoceptors pathways. In addition, methanol induced a dose-dependent inhibition of vasoconstrictions caused by calcium chloride, potassium chloride, or norepinephrine. Further work is needed to investigate the relative contribution of each channel and pathway in methanol-induced vasoactive effect.
Collapse
Affiliation(s)
- Y Bai
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Q Zhang
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Z Yang
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Z Meng
- 1 Institute of Environmental Medicine and Toxicology, Institute of Environmental Science, Shanxi University, Taiyuan, People's Republic of China
| | - Q Zhao
- 2 State Key Laboratory of Environmental Chemistry and Eco-toxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
45
|
de Los Monteros-Zuñiga AE, Izquierdo T, Quiñonez-Bastidas GN, Rocha-González HI, Godínez-Chaparro B. Anti-allodynic effect of mangiferin in neuropathic rats: Involvement of nitric oxide-cyclic GMP-ATP sensitive K + channels pathway and serotoninergic system. Pharmacol Biochem Behav 2016; 150-151:190-197. [PMID: 27984097 DOI: 10.1016/j.pbb.2016.10.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/14/2016] [Accepted: 10/19/2016] [Indexed: 11/17/2022]
Abstract
The neurobiology of neuropathic pain is caused by injury in the central or peripheral nervous system. Recent evidence points out that mangiferin shows anti-nociceptive effect in inflammatory pain. However, its role in inflammatory and neuropathic pain and the possible mechanisms of action are not yet established. The purpose of this study was to determine the possible anti-allodynic effect of mangiferin in rats with spinal nerve ligation (SNL). Furthermore, we sought to investigate the possible mechanisms of action that contribute to these effects. Mechanical allodynia to stimulation with the von Frey filaments was measured by the up and down method. Intrathecal administration of mangiferin prevented, in a dose-dependent fashion, SNL-induced mechanical allodynia. Mangiferin-induced anti-allodynia was prevented by the intrathecal administration of L-NAME (100μg/rat, non-selective nitric oxide synthase inhibitor), ODQ (10μg/rat, inhibitor of guanylate-cyclase) and glibenclamide (50μg/rat, channel blocker of ATP-sensitive K+ channels). Moreover, methiothepin (30μg/rat, non-selective 5-HT receptor antagonist), WAY-100635 (6μg/rat, selective 5-HT1A receptor antagonist), SB-224289 (5μg/rat, selective 5-HT1B receptor antagonist), BRL-15572 (4μg/rat, selective 5-HT1D receptor antagonist) and SB-659551 (6μg/rat, selective 5-HT5A receptor antagonist), but not naloxone (50μg/rat, non-selective opioid receptor antagonist), were able to prevent mangiferin-induced anti-allodynic effect. These data suggest that the anti-allodynic effect induced by mangiferin is mediated at least in part by the serotoninergic system, involving the activation of 5-HT1A/1B/1D/5A receptors, as well as the nitric oxide-cyclic GMP-ATP-sensitive K+ channels pathway, but not by the opioidergic system, in the SNL model of neuropathic pain in rats.
Collapse
Affiliation(s)
- Antonio Espinosa de Los Monteros-Zuñiga
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960 Mexico, D.F., Mexico
| | - Teresa Izquierdo
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960 Mexico, D.F., Mexico
| | - Geovanna Nallely Quiñonez-Bastidas
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960 Mexico, D.F., Mexico
| | - Héctor Isaac Rocha-González
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Miguel Hidalgo, 11340 Mexico, D.F., Mexico
| | - Beatriz Godínez-Chaparro
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960 Mexico, D.F., Mexico.
| |
Collapse
|
46
|
Tourki B, Matéo P, Morand J, Elayeb M, Godin-Ribuot D, Marrakchi N, Belaidi E, Messadi E. Lebetin 2, a Snake Venom-Derived Natriuretic Peptide, Attenuates Acute Myocardial Ischemic Injury through the Modulation of Mitochondrial Permeability Transition Pore at the Time of Reperfusion. PLoS One 2016; 11:e0162632. [PMID: 27618302 PMCID: PMC5019389 DOI: 10.1371/journal.pone.0162632] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/25/2016] [Indexed: 12/28/2022] Open
Abstract
Cardiac ischemia is one of the leading causes of death worldwide. It is now well established that natriuretic peptides can attenuate the development of irreversible ischemic injury during myocardial infarction. Lebetin 2 (L2) is a new discovered peptide isolated from Macrovipera lebetina venom with structural similarity to B-type natriuretic peptide (BNP). Our objectives were to define the acute cardioprotective actions of L2 in isolated Langendorff-perfused rat hearts after regional or global ischemia-reperfusion (IR). We studied infarct size, left ventricular contractile recovery, survival protein kinases and mitochondrial permeability transition pore (mPTP) opening in injured myocardium. L2 dosage was determined by preliminary experiments at its ability to induce cyclic guanosine monophosphate (cGMP) release without changing hemodynamic effects in normoxic hearts. L2 was found to be as effective as BNP in reducing infarct size after the induction of either regional or global IR. Both peptides equally improved contractile recovery after regional IR, but only L2 increased coronary flow and reduced severe contractile dysfunction after global ischemia. Cardioprotection afforded by L2 was abolished after isatin or 5-hydroxydecanote pretreatment suggesting the involvement of natriuretic peptide receptors and mitochondrial KATP (mitoKATP) channels in the L2-induced effects. L2 also increased survival protein expression in the reperfused myocardium as evidenced by phosphorylation of signaling pathways PKCε/ERK/GSK3β and PI3K/Akt/eNOS. IR induced mitochondrial pore opening, but this effect was markedly prevented by L2 treatment. These data show that L2 has strong cardioprotective effect in acute ischemia through stimulation of natriuretic peptide receptors. These beneficial effects are mediated, at least in part, by mitoKATP channel opening and downstream activated survival kinases, thus delaying mPTP opening and improving IR-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Bochra Tourki
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
- Université Carthage Tunis, Bizerte, Tunisia
| | - Philippe Matéo
- Laboratoire de Signalisation et Physiopathologie Cardiovasculaire, UMR-S 1180, Faculté de Pharmacie, Université Paris Sud, Paris, France
| | - Jessica Morand
- Laboratoire d’Hypoxie et Physiopathologie Cardiaque, Inserm U1042, Faculté de Pharmacie, Université Grenoble Alpes, Grenoble, France
| | - Mohamed Elayeb
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Diane Godin-Ribuot
- Laboratoire d’Hypoxie et Physiopathologie Cardiaque, Inserm U1042, Faculté de Pharmacie, Université Grenoble Alpes, Grenoble, France
| | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Elise Belaidi
- Laboratoire d’Hypoxie et Physiopathologie Cardiaque, Inserm U1042, Faculté de Pharmacie, Université Grenoble Alpes, Grenoble, France
| | - Erij Messadi
- Laboratoire des Venins et Biomolécules Thérapeutiques (LR11IPT08) et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires (P2C), Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
- * E-mail:
| |
Collapse
|
47
|
Hassanipour M, Amini-Khoei H, Shafaroodi H, Shirzadian A, Rahimi N, Imran-Khan M, Rezayat SM, Dehpour A. Atorvastatin attenuates the antinociceptive tolerance of morphine via nitric oxide dependent pathway in male mice. Brain Res Bull 2016; 125:173-80. [PMID: 27381980 DOI: 10.1016/j.brainresbull.2016.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/24/2016] [Accepted: 07/01/2016] [Indexed: 01/01/2023]
Abstract
The development of morphine-induced antinociceptive tolerance limits its therapeutic efficacy in pain management. Atorvastatin, or competitive inhibitor of 3-hydroxy-methyl-glutaryl coenzyme A (HMG-CoA) reductase, is mainstay agent in hypercholesterolemia treatment. Beyond the cholesterol-lowering activity, exploration of neuroprotective properties of this statin indicates its potential benefit in central nervous disorders. The aim of the present study was to assess the effects of atorvastatin in development and expression of morphine-induced analgesic tolerance in male mice and probable involvement of nitric oxide. Chronic and acute treatment with atorvastatin 10 and 20mg/kg, respectively, could alleviate morphine tolerance in development and expression phases. Chronic co-administration of nitric oxide synthase (NOS) inhibitors including L-NAME (non selective NOS inhibitor; 2mg/kg), aminoguanidine (selective inducible NOS inhibitor; 50mg/kg) and 7-NI (selective neuronal NOS inhibitor; 15mg/kg) with atorvastatin blocked the protective effect of atorvastatin in tolerance reversal. Moreover, reversing the atorvastatin effect was also observed in acute simultaneous treatment of L-NAME (5mg/kg) and aminoguanidine (100mg/kg) with atorvastatin. Co-treatment of guanylyl cyclase inhibitor, ODQ (chronic dose: 10mg/kg and acute dose: 20mg/kg) was associated with prevention of atorvastatin anti-tolerance properties. Our results revealed that the atorvastatin modulating role in morphine antinociceptive tolerance is mediated at least in part via nitric oxide in animal pain models of hot plate and tail flick.
Collapse
Affiliation(s)
- Mahsa Hassanipour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Hossein Amini-Khoei
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of pharmacology, School of medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hamed Shafaroodi
- Department of Pharmacology and Toxicology, School of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran.
| | - Armin Shirzadian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Nastaran Rahimi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Muhammad Imran-Khan
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed-Mahdi Rezayat
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology and Toxicology, School of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran.
| | - Ahmadreza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Overview of Antagonists Used for Determining the Mechanisms of Action Employed by Potential Vasodilators with Their Suggested Signaling Pathways. Molecules 2016; 21:495. [PMID: 27092479 PMCID: PMC6274436 DOI: 10.3390/molecules21040495] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 01/04/2023] Open
Abstract
This paper is a review on the types of antagonists and the signaling mechanism pathways that have been used to determine the mechanisms of action employed for vasodilation by test compounds. Thus, we exhaustively reviewed and analyzed reports related to this topic published in PubMed between the years of 2010 till 2015. The aim of this paperis to suggest the most appropriate type of antagonists that correspond to receptors that would be involved during the mechanistic studies, as well as the latest signaling pathways trends that are being studied in order to determine the route(s) that atest compound employs for inducing vasodilation. The methods to perform the mechanism studies were included. Fundamentally, the affinity, specificity and selectivity of the antagonists to their receptors or enzymes were clearly elaborated as well as the solubility and reversibility. All the signaling pathways on the mechanisms of action involved in the vascular tone regulation have been well described in previous review articles. However, the most appropriate antagonists that should be utilized have never been suggested and elaborated before, hence the reason for this review.
Collapse
|
49
|
Chan MV, Knowles RBM, Lundberg MH, Tucker AT, Mohamed NA, Kirkby NS, Armstrong PCJ, Mitchell JA, Warner TD. P2Y12 receptor blockade synergizes strongly with nitric oxide and prostacyclin to inhibit platelet activation. Br J Clin Pharmacol 2016; 81:621-33. [PMID: 26561399 PMCID: PMC4799935 DOI: 10.1111/bcp.12826] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/16/2015] [Accepted: 11/05/2015] [Indexed: 12/22/2022] Open
Abstract
AIMS In vivo platelet function is a product of intrinsic platelet reactivity, modifiable by dual antiplatelet therapy (DAPT), and the extrinsic inhibitory endothelial mediators, nitric oxide (NO) and prostacyclin (PGI2 ), that are powerfully potentiated by P2Y12 receptor blockade. This implies that for individual patients endothelial mediator production is an important determinant of DAPT effectiveness. Here, we have investigated this idea using platelets taken from healthy volunteers treated with anti-platelet drugs. METHODS Three groups of male volunteers (n = 8) received either prasugrel (10 mg), aspirin (75 mg) or DAPT (prasugrel + aspirin) once daily for 7 days. Platelet reactivity in the presence of diethylammonium (Z)-1-(N,N-diethylamino)diazen-1-ium-1,2-diolate (DEA/NONOate) and PGI2 was studied before and following treatment. RESULTS Ex vivo, PGI2 and/or DEA/NONOate had little inhibitory effect on TRAP-6-induced platelet reactivity in control conditions. However, in the presence of DAPT, combination of DEA/NONOate + PGI2 reduced platelet aggregation (74 ± 3% to 19 ± 6%, P < 0.05). In vitro studies showed even partial (25%) P2Y12 receptor blockade produced a significant (67 ± 2% to 39 ± 10%, P < 0.05) inhibition when DEA/NONOate + PGI2 was present. CONCLUSIONS We have demonstrated that PGI2 and NO synergize with P2Y12 receptor antagonists to produce powerful platelet inhibition. Furthermore, even with submaximal P2Y12 blockade the presence of PGI2 and NO greatly enhances platelet inhibition. Our findings highlight the importance of endothelial mediator in vivo modulation of P2Y12 inhibition and introduces the concept of refining ex vivo platelet function testing by incorporating an assessment of endothelial function to predict thrombotic outcomes better and adjust therapy to prevent adverse outcomes in individual patients.
Collapse
Affiliation(s)
- Melissa V. Chan
- The William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Rebecca B. M. Knowles
- The William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Martina H. Lundberg
- The William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Arthur T. Tucker
- The William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Nura A. Mohamed
- Qatar Foundation Research and Development DivisionDohaQatar
- National Heart & Lung InstituteImperial College LondonLondonUK
| | - Nicholas S. Kirkby
- The William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
- National Heart & Lung InstituteImperial College LondonLondonUK
| | - Paul C. J. Armstrong
- The William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | | | - Timothy D. Warner
- The William Harvey Research Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
50
|
Choi SH, Ruggiero D, Sorice R, Song C, Nutile T, Vernon Smith A, Concas MP, Traglia M, Barbieri C, Ndiaye NC, Stathopoulou MG, Lagou V, Maestrale GB, Sala C, Debette S, Kovacs P, Lind L, Lamont J, Fitzgerald P, Tönjes A, Gudnason V, Toniolo D, Pirastu M, Bellenguez C, Vasan RS, Ingelsson E, Leutenegger AL, Johnson AD, DeStefano AL, Visvikis-Siest S, Seshadri S, Ciullo M. Six Novel Loci Associated with Circulating VEGF Levels Identified by a Meta-analysis of Genome-Wide Association Studies. PLoS Genet 2016; 12:e1005874. [PMID: 26910538 PMCID: PMC4766012 DOI: 10.1371/journal.pgen.1005874] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/26/2016] [Indexed: 12/31/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic and neurotrophic factor, secreted by endothelial cells, known to impact various physiological and disease processes from cancer to cardiovascular disease and to be pharmacologically modifiable. We sought to identify novel loci associated with circulating VEGF levels through a genome-wide association meta-analysis combining data from European-ancestry individuals and using a dense variant map from 1000 genomes imputation panel. Six discovery cohorts including 13,312 samples were analyzed, followed by in-silico and de-novo replication studies including an additional 2,800 individuals. A total of 10 genome-wide significant variants were identified at 7 loci. Four were novel loci (5q14.3, 10q21.3, 16q24.2 and 18q22.3) and the leading variants at these loci were rs114694170 (MEF2C, P = 6.79x10-13), rs74506613 (JMJD1C, P = 1.17x10-19), rs4782371 (ZFPM1, P = 1.59x10-9) and rs2639990 (ZADH2, P = 1.72x10-8), respectively. We also identified two new independent variants (rs34528081, VEGFA, P = 1.52x10-18; rs7043199, VLDLR-AS1, P = 5.12x10-14) at the 3 previously identified loci and strengthened the evidence for the four previously identified SNPs (rs6921438, LOC100132354, P = 7.39x10-1467; rs1740073, C6orf223, P = 2.34x10-17; rs6993770, ZFPM2, P = 2.44x10-60; rs2375981, KCNV2, P = 1.48x10-100). These variants collectively explained up to 52% of the VEGF phenotypic variance. We explored biological links between genes in the associated loci using Ingenuity Pathway Analysis that emphasized their roles in embryonic development and function. Gene set enrichment analysis identified the ERK5 pathway as enriched in genes containing VEGF associated variants. eQTL analysis showed, in three of the identified regions, variants acting as both cis and trans eQTLs for multiple genes. Most of these genes, as well as some of those in the associated loci, were involved in platelet biogenesis and functionality, suggesting the importance of this process in regulation of VEGF levels. This work also provided new insights into the involvement of genes implicated in various angiogenesis related pathologies in determining circulating VEGF levels. The understanding of the molecular mechanisms by which the identified genes affect circulating VEGF levels could be important in the development of novel VEGF-related therapies for such diseases. Vascular Endothelial Growth Factor (VEGF) is a protein with a fundamental role in development of vascular system. The protein, produced by many types of cells, is released in the blood. High levels of VEGF have been observed in different pathological conditions especially in cancer, cardiovascular, and inflammatory diseases. Therefore, identifying the genetic factors influencing VEGF levels is important for predicting and treating such pathologies. The number of genetic variants associated with VEGF levels has been limited. To identify new loci, we have performed a Genome Wide Association Study meta-analysis on a sample of more than 16,000 individuals from 10 cohorts, using a high-density genetic map. This analysis revealed 10 variants associated with VEGF circulating levels, 6 of these being novel associations. The 10 variants cumulatively explain more than 50% of the variability of VEGF serum levels. Our analyses have identified genes known to be involved in angiogenesis related diseases and genes implicated in platelet metabolism, suggesting the importance of links between this process and VEGF regulation. Overall, these data have improved our understanding of the genetic variation underlying circulating VEGF levels. This in turn could guide our response to the challenge posed by various VEGF-related pathologies.
Collapse
Affiliation(s)
- Seung Hoan Choi
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Rossella Sorice
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Ci Song
- Population Sciences Branch, National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Teresa Nutile
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Maria Pina Concas
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Michela Traglia
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Caterina Barbieri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Ndeye Coumba Ndiaye
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Maria G. Stathopoulou
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Stephanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Neurology, Bordeaux University Hospital, Bordeaux, France
- INSERM U897, Bordeaux, France
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases, Leipzig, Germany
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - John Lamont
- Randox Laboratories, Crumlin, United Kingdom
| | | | - Anke Tönjes
- University of Leipzig, Department of Medicine, Leipzig, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Celine Bellenguez
- Institut Pasteur de Lille, Lille, France
- INSEM U744, Lille, France
- Université Lille-Nord de France, Lille, France
| | - Ramachandran S. Vasan
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, United States of America
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anne-Louise Leutenegger
- INSERM U946, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, IUH, UMR-S 946, Paris, France
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Anita L. DeStefano
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Sophie Visvikis-Siest
- UMR INSERM U1122, IGE-PCV “Interactions Gène-Environnement en Physiopathologie Cardio-Vasculaire”, Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- National Heart, Lung and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- * E-mail: (SS); (MC)
| | - Marina Ciullo
- Institute of Genetics and Biophysics, National Research Council of Italy, Naples, Italy
- * E-mail: (SS); (MC)
| |
Collapse
|