1
|
Guo L, Yang Q, Wei R, Zhang W, Yin N, Chen Y, Xu C, Li C, Carney RP, Li Y, Feng M. Enhanced pericyte-endothelial interactions through NO-boosted extracellular vesicles drive revascularization in a mouse model of ischemic injury. Nat Commun 2023; 14:7334. [PMID: 37957174 PMCID: PMC10643472 DOI: 10.1038/s41467-023-43153-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Despite improvements in medical and surgical therapies, a significant portion of patients with critical limb ischemia (CLI) are considered as "no option" for revascularization. In this work, a nitric oxide (NO)-boosted and activated nanovesicle regeneration kit (n-BANK) is constructed by decorating stem cell-derived nanoscale extracellular vesicles with NO nanocages. Our results demonstrate that n-BANKs could store NO in endothelial cells for subsequent release upon pericyte recruitment for CLI revascularization. Notably, n-BANKs enable endothelial cells to trigger eNOS activation and form tube-like structures. Subsequently, eNOS-derived NO robustly recruits pericytes to invest nascent endothelial cell tubes, giving rise to mature blood vessels. Consequently, n-BANKs confer complete revascularization in female mice following CLI, and thereby achieve limb preservation and restore the motor function. In light of n-BANK evoking pericyte-endothelial interactions to create functional vascular networks, it features promising therapeutic potential in revascularization to reduce CLI-related amputations, which potentially impact regeneration medicine.
Collapse
Affiliation(s)
- Ling Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China.
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, P. R. China.
| | - Qiang Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Runxiu Wei
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Wenjun Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Na Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Yuling Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China
| | - Chao Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, P. R. China
| | - Changrui Li
- Guangzhou Zhixin High School, Zhixin South Road, Guangzhou, 510080, P.R. China
| | - Randy P Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA, 95616, USA.
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, 95616, USA.
| | - Min Feng
- School of Pharmaceutical Sciences, Sun Yat-sen University, University Town, Guangzhou, 510006, P.R. China.
| |
Collapse
|
2
|
Sudhahar V, Okur MN, O'Bryan JP, Minshall RD, Fulton D, Ushio-Fukai M, Fukai T. Caveolin-1 stabilizes ATP7A, a copper transporter for extracellular SOD, in vascular tissue to maintain endothelial function. Am J Physiol Cell Physiol 2020; 319:C933-C944. [PMID: 32936699 PMCID: PMC7789967 DOI: 10.1152/ajpcell.00151.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/31/2022]
Abstract
Caveolin-1 (Cav-1) is a scaffolding protein and a major component of caveolae/lipid rafts. Previous reports have shown that endothelial dysfunction in Cav-1-deficient (Cav-1-/-) mice is mediated by elevated oxidative stress through endothelial nitric oxide synthase (eNOS) uncoupling and increased NADPH oxidase. Oxidant stress is the net balance of oxidant generation and scavenging, and the role of Cav-1 as a regulator of antioxidant enzymes in vascular tissue is poorly understood. Extracellular SOD (SOD3) is a copper (Cu)-containing enzyme that is secreted from vascular smooth muscle cells/fibroblasts and subsequently binds to the endothelial cells surface, where it scavenges extracellular [Formula: see text] and preserves endothelial function. SOD3 activity is dependent on Cu, supplied by the Cu transporter ATP7A, but whether Cav-1 regulates the ATP7A-SOD3 axis and its role in oxidative stress-mediated vascular dysfunction has not been studied. Here we show that the activity of SOD3, but not SOD1, was significantly decreased in Cav-1-/- vessels, which was rescued by re-expression of Cav-1 or Cu supplementation. Loss of Cav-1 reduced ATP7A protein, but not mRNA, and this was mediated by ubiquitination of ATP7A and proteasomal degradation. ATP7A bound to Cav-1 and was colocalized with SOD3 in caveolae/lipid rafts or perinucleus in vascular tissues or cells. Impaired endothelium-dependent vasorelaxation in Cav-1-/- mice was rescued by gene transfer of SOD3 or by ATP7A-overexpressing transgenic mice. These data reveal an unexpected role of Cav-1 in stabilizing ATP7A protein expression by preventing its ubiquitination and proteasomal degradation, thereby increasing SOD3 activity, which in turn protects against vascular oxidative stress-mediated endothelial dysfunction.
Collapse
Affiliation(s)
- Varadarajan Sudhahar
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Mustafa Nazir Okur
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - John P O'Bryan
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Richard D Minshall
- Departments of Anesthesiology and Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
3
|
Revealing the Common Mechanisms of Scutellarin in Angina Pectoris and Ischemic Stroke Treatment via a Network Pharmacology Approach. Chin J Integr Med 2020; 27:62-69. [PMID: 32447519 DOI: 10.1007/s11655-020-2716-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the shared mechanisms of scutellarin in angina pectoris (AP) and ischemic stroke (IS) treatment. METHODS A network pharmacology approach was used to detect the potential mechanisms of scutellarin in AP and IS treatment by target prediction, protein-protein interaction (PPI) data collection, network construction, network analysis, and enrichment analysis. Furthermore, molecular docking simulation was employed to analyze the interaction between scutellarin and core targets. RESULTS Two networks were established, including a disease-target network and a PPI network of scutellarin targets against AP and IS. Network analysis showed that 14 targets, namely, AKT1, VEGFA, JUN, ALB, MTOR, ESR1, MAPK8, HSP90AA1, NOS3, SERPINE1, FGA, F2, FOXO3, and STAT1, might be the therapeutic targets of scutellarin in AP and IS. Among them, NOS3 and F2 were recognized as the core targets. Additionally, molecular docking simulation confifirmed that scutellarin exhibited a relatively high potential for binding to the active sites of NOS3 and F2. Furthermore, enrichment analysis indicated that scutellarin might exert a therapeutic role in both AP and IS by regulating several important pathways, such as coagulation cascades, mitogen-activated protein kinase (MAPK) signaling pathway, phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway, Toll-like receptor signaling pathway, hypoxia inducible factor-1 (HIF-1) signaling pathway, forkhead box O (FoxO) signaling pathway, tumor necrosis factor (TNF) signaling pathway, adipocytokine signaling pathway, insulin signaling pathway, insulin resistance, and estrogen signaling pathway. CONCLUSIONS The shared underlying mechanisms of scutellarin on AP and IS treatment might be strongly associated with its vasorelaxant, anticoagulant, anti-inflammatory, and antioxidative effects as well as its effect on improving lipid metabolism.
Collapse
|
4
|
Bibli SI, Papapetropoulos A, Iliodromitis EK, Daiber A, Randriamboavonjy V, Steven S, Brouckaert P, Chatzianastasiou A, Kypreos KE, Hausenloy DJ, Fleming I, Andreadou I. Nitroglycerine limits infarct size through S-nitrosation of cyclophilin D: a novel mechanism for an old drug. Cardiovasc Res 2020; 115:625-636. [PMID: 30165375 DOI: 10.1093/cvr/cvy222] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
AIMS Nitroglycerine (NTG) given prior to an ischaemic insult exerts cardioprotective effects. However, whether administration of an acute low dose of NTG in a clinically relevant manner following an ischaemic episode limits infarct size, has not yet been explored. METHODS AND RESULTS Adult mice were subjected to acute myocardial infarction in vivo and then treated with vehicle or low-dose NTG prior to reperfusion. This treatment regimen minimized myocardial infarct size without affecting haemodynamic parameters but the protective effect was absent in mice rendered tolerant to the drug. Mechanistically, NTG was shown to nitrosate and inhibit cyclophilin D (CypD), and NTG administration failed to limit infarct size in CypD knockout mice. Additional experiments revealed lack of the NTG protective effect following genetic (knockout mice) or pharmacological inhibition (L-NAME treatment) of the endothelial nitric oxide synthase (eNOS). The protective effect of NTG was attributed to preservation of the eNOS dimer. Moreover, NTG retained its cardioprotective effects in a model of endothelial dysfunction (ApoE knockout) by preserving CypD nitrosation. Human ischaemic heart biopsies revealed reduced eNOS activity and exhibited reduced CypD nitrosation. CONCLUSION Low-dose NTG given prior to reperfusion reduces myocardial infarct size by preserving eNOS function, and the subsequent eNOS-dependent S-nitrosation of CypD, inhibiting cardiomyocyte necrosis. This novel pharmacological action of NTG warrants confirmation in clinical studies, although our data in human biopsies provide promising preliminary results.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece.,Institute for Vascular Signaling, Goethe University, Theodor Stern Kai 7, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Papapetropoulos
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| | - Efstathios K Iliodromitis
- Faculty of Medicine, Second Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Daiber
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.,University Medical Center of Mainz, Center for Cardiology, Cardiology I, Molecular Cardiology, Mainz, Germany
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signaling, Goethe University, Theodor Stern Kai 7, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Sebastian Steven
- University Medical Center of Mainz, Center for Cardiology, Cardiology I, Molecular Cardiology, Mainz, Germany.,University Medical Center of Mainz, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Molecular Biomedical Research, VIB, Ghent, Belgium
| | - Athanasia Chatzianastasiou
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| | - Kyriakos E Kypreos
- Department of Pharmacology, University of Patras Medical School, Patras, Greece
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore.,National Heart Research Institute Singapore, National Heart Centre, Singapore.,Yong Loo Lin School of Medicine, National University Singapore, Singapore.,The Hatter Cardiovascular Institute, University College London, London, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London, UK.,Department of Cardiology, Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - Ingrid Fleming
- Institute for Vascular Signaling, Goethe University, Theodor Stern Kai 7, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Ioanna Andreadou
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| |
Collapse
|
5
|
Song M, Li L, Lei Y, Sun X. NOS3 Deletion in Cav1 Deficient Mice Decreases Drug Sensitivity to a Nitric Oxide Donor and Two Nitric Oxide Synthase Inhibitors. Invest Ophthalmol Vis Sci 2020; 60:4002-4007. [PMID: 31560766 DOI: 10.1167/iovs.19-27582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose This study aims to investigate the pharmacologic consequence of genetic deletion of nitric oxide synthase 3 (NOS3) in caveolin 1 (Cav1)-/- mice (double knockout [DKO]) in response to a nitric oxide (NO) donor and two NOS inhibitors. Methods NO donor sodium nitroprusside (SNP; 10-40 mg/mL), NOS inhibitor L-NG-nitroarginine methyl ester (L-NAME; 10-200 μM), and cavtratin (10-75 μM ) was administered topically to the eye while the contralateral eyes were vehicle controls. Intraocular pressure (IOP) was measured in both eyes by tonometry. Cyclic guanosine monophosphate (cGMP) level in outflow tissue was measured by ELISA assay. Protein expression were analyzed by western blot. Results Inducible NOS (iNOS) expression significantly increased in the DKO mice compared with the wild type (WT), Cav1 knockout (Cav1 KO), and NOS3 KO mice. In contrast to WT, Cav1 KO and NOS3 KO mice, SNP concentration of up to 30 mg/mL did not significantly affect IOP in DKO mice. However, higher concentration (40 mg/mL) SNP significantly reduced IOP by 14% (n = 8, P < 0.01). Similarly, only 200 μM L-NAME produced a significant increase in IOP (n = 10, P < 0.05). Cavtratin did not significantly change IOP in DKO and NOS3 KO mice. cGMP activity in DKO mice was significantly lower than Cav1 KO mice (n = 4, P < 0.05). Conclusions In conclusion, our results demonstrated that genetic deletion of NOS3 in Cav1 deficient mice resulted in reduced sensitivity to the NO donor SNP and the two NOS inhibitors possibly due to compromised NOS and cGMP activity.
Collapse
Affiliation(s)
- Maomao Song
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liping Li
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Lei
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinghuai Sun
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
6
|
An YM, Li YJ, Zhang CL, Cong X, Gao YS, Wu LL, Dou D. Decreased PKG transcription mediated by PI3K/Akt/FoxO1 pathway is involved in the development of nitroglycerin tolerance. Biochem Biophys Res Commun 2018; 508:1195-1201. [PMID: 30554658 DOI: 10.1016/j.bbrc.2018.12.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 12/10/2018] [Indexed: 12/27/2022]
Abstract
Phosphoinositide 3-kinase (PI3K)/Akt plays a pivotal role in the vascular response. The present study is to determine whether PI3K/Akt pathway in vascular smooth muscle cells is involved in nitroglycerin (NTG) tolerance and the underlying mechanism. Nitrate tolerance of porcine coronary arteries in vitro was induced by incubation of NTG (10-5 M) for 24 h. Nitrate tolerance in vivo was obtained by subcutaneous injection of mice with NTG (20 mg kg-1, tid, 3 days) and the aortas were used. Protein levels of total and phosphorylated Akt, forkhead box protein O1 (FoxO1), and cGMP-dependent protein kinase (PKG) were determined by western blot analysis. Isometric vessel tension was recorded by organ chamber technique. PKG mRNA was determined by real-time PCR. The cellular translocation of FoxO1 was observed by immunofluorescence. Reactive oxygen species (ROS) level was measured by DHE staining. The vascular relaxation to NTG was significantly inhibited in in vivo and in vitro NTG tolerant arteries. Meanwhile, the protein level of phosphorylated Akt at Ser473 was increased in the tolerant arteries. The attenuated relaxation and the augmented Akt-p were ameliorated by LY294002, a specific inhibitor of PI3K. The protein and mRNA expression of PKG were significantly down-regulated in NTG tolerant arteries, which were reversed by LY294002. The level of phosphorylated FoxO1 at Ser256 and its translocation from the nucleus to the cytosol were both increased in NTG tolerance and were also inhibited by LY294002. ROS production was significantly increased in NTG tolerant arteries, which was not be affected by LY294002 but inhibited by N-acetyl-L-cysteine. In conclusion, the present study suggests that PI3K/Akt in vascular smooth muscle is involved in the development of NTG tolerance via inhibiting PKG transcription and the effect is mediated by FoxO1.
Collapse
Affiliation(s)
- Yuan-Ming An
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yan-Jing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Cheng-Lin Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yuan-Sheng Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
| | - Dou Dou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| |
Collapse
|
7
|
Zhou K, Parker JD. The role of vascular endothelium in nitroglycerin-mediated vasodilation. Br J Clin Pharmacol 2018; 85:377-384. [PMID: 30378151 DOI: 10.1111/bcp.13804] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/23/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022] Open
Abstract
AIMS Nitroglycerin (or glyceryl trinitrate, GTN) has been long considered an endothelium-independent vasodilator because GTN vasodilation is intact in the absence of the endothelium and in the presence of endothelial dysfunction. However, in animal and in vitro models, GTN has been shown to stimulate the release of certain endothelium-derived vasodilators such as nitric oxide (NO) and prostacyclin (PGI2 ). In addition, chronic GTN therapy leads to endothelial dysfunction. In this series of experiments, we explored how GTN might interact with the vascular endothelium in normal humans, without cardiovascular disease or risk factors associated with abnormalities in vascular function. METHODS We examined the effect of inhibition of NO, PGI2 , and epoxyeicosatrienoic acids (EETs, a class of endothelium-derived hyperpolarizing factor) on GTN-mediated vasodilation. We measured arterial blood flow responses to brachial artery infusions of GTN in the absence and presence of L-NMMA (n = 13), ketorolac (n = 14) and fluconazole (n = 16), which are inhibitors of endothelium-derived NO, PGI2 and EETs, respectively, in healthy volunteers. RESULTS Our results demonstrate that inhibition of endothelium-dependent vasodilator mechanisms does not alter forearm resistance vessel responses to GTN. CONCLUSION We conclude that GTN-mediated dilation of forearm resistance vessels is largely independent of vascular endothelium.
Collapse
Affiliation(s)
- Kangbin Zhou
- Department Pharmacology and Toxicology, the University of Toronto
| | - John D Parker
- Department Pharmacology and Toxicology, the University of Toronto.,Division of Cardiology, Department of Medicine, Sinai Health System and the Peter Munk Cardiac Centre, University Health Network, Toronto.,The Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto
| |
Collapse
|
8
|
Schulz JM, Al-Khazraji BK, Shoemaker JK. Sodium nitroglycerin induces middle cerebral artery vasodilatation in young, healthy adults. Exp Physiol 2018; 103:1047-1055. [PMID: 29766604 PMCID: PMC6099468 DOI: 10.1113/ep087022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/08/2018] [Indexed: 01/03/2023]
Abstract
NEW FINDINGS What is the central question of this study? Nitric oxide causes dilatation in peripheral vessels; however, whether nitric oxide affects basal cerebral artery dilatation has not been explored. What is the main finding and its importance? This study demonstrated that vasodilatation occurs in the right middle cerebral artery in response to exogenous nitric oxide. However, blood velocity decreased and, therefore, overall cerebral blood flow remained unchanged. This study provides new insight into the role of nitric oxide in cerebral blood flow control. ABSTRACT Recent evidence indicates that basal cerebral conduit vessels dilate with hypercapnia, with a nitric oxide (NO) mechanism explaining one way in which parenchymal cerebral arterioles dilate. However, whether NO affects basal cerebral artery dilatation remains unknown. This study quantified the effect of an exogenous NO donor [sodium nitroglycerin (NTG); 0.4 mg sublingual spray] on the right middle cerebral artery (rMCA) cross-sectional area (CSA), blood velocity and overall blood flow. Measures of vessel CSA (7 T magnetic resonance imaging) and MCA blood velocity (transcranial Doppler ultrasound) were made at baseline (BL) and after exogenous NTG or placebo (PLO) administration in young, healthy individuals (n = 10, two males, age range 20-23 years). The CSA increased in the rMCA [BL, 5.2 ± 1.2 mm2 ; PLO, 5.4 ± 1.5 mm2 ; NTG, 6.6 ± 1.5 mm2 , P < 0.05; mean ± SD]. Concurrently, rMCA blood velocity decreased from BL during NTG compared with PLO (BL, 67 ± 10 cm s-1 ; PLO, 62 ± 10 cm s-1 ; NTG, 59 ± 9.3 cm s-1 , P < 0.05; mean ± SD]. However, total MCA blood flow did not change with NTG or PLO [BL, 221 ± 37.4 ml min-1 ; PLO, 218 ± 35.0 ml min-1 ; NTG, 213 ± 46.4 ml min-1 ). Therefore, exogenous NO mediates a dilatory response in the rMCA, but not in its downstream vascular bed.
Collapse
Affiliation(s)
- Jenna M Schulz
- School of Physical Therapy, Department of Health Sciences, Western University, London, ON, Canada
| | - Baraa K Al-Khazraji
- School of Kinesiology, Department of Health Sciences, Western University, London, ON, Canada
| | - J Kevin Shoemaker
- Department of Physiology and Pharmacology, Western University, London, ON, Canada.,School of Kinesiology, Department of Health Sciences, Western University, London, ON, Canada
| |
Collapse
|
9
|
Li H, Liao Y, Gao L, Zhuang T, Huang Z, Zhu H, Ge J. Coronary Serum Exosomes Derived from Patients with Myocardial Ischemia Regulate Angiogenesis through the miR-939-mediated Nitric Oxide Signaling Pathway. Am J Cancer Res 2018; 8:2079-2093. [PMID: 29721064 PMCID: PMC5928872 DOI: 10.7150/thno.21895] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 02/09/2018] [Indexed: 12/19/2022] Open
Abstract
Rationale: Angiogenesis is a crucial step towards tissue repair and regeneration after ischemia. The role of circulating exosomes in angiogenic signal transduction has not been well elucidated. Thus, this study aims to investigate the effects of coronary serum exosomes from patients with myocardial ischemia on angiogenesis and to elucidate the underlying mechanisms. Methods and Results: The patients were enrolled according to the inclusion and exclusion criteria. Coronary blood was obtained from the angiography catheter. Serum exosomes were purified and characterized by their specific morphology and surface markers. In vitro analysis showed that compared to exosomes from healthy controls (con-Exo), exosomes from patients with myocardial ischemia (isc-Exo) enhanced endothelial cell proliferation, migration and tube formation. In a mouse hind-limb ischemia model, blood perfusion and histological staining demonstrated that isc-Exo significantly promoted blood flow recovery and enhanced neovascularization compared to con-Exo. Further, we revealed that cardiomyocytes, but not cardiac fibroblasts or endothelial cells, were initiated to release exosomes under ischemic stress; cardiomyocytes might be the source of bioactive exosomes in coronary serum. In addition, microarray analysis indicated that miR-939-5p was significantly down-regulated in isc-Exo. By knockdown and overexpression analyses, we found that miR-939-5p regulated angiogenesis by targeting iNOS. miR-939-5p inhibited both iNOS's expression and its activity, attenuated endothelial NO production, and eventually impaired angiogenesis. Conclusions: Exosomes derived from patients with myocardial ischemia promote angiogenesis via the miR-939-iNOS-NO pathway. Our study highlights that coronary serum exosomes serve as an important angiogenic messenger in patients suffering from myocardial ischemia.
Collapse
|
10
|
El-Gharabawy RM, Ahmed AS, Al-Najjar AH. Cataract induction by administration of nitroglycerin in cardiac patients through imbalance in redox status. Ther Clin Risk Manag 2016; 12:1487-1496. [PMID: 27729797 PMCID: PMC5045900 DOI: 10.2147/tcrm.s114469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Purpose The objective of this study was to evaluate the role of nitroglycerin in the pathogenesis of cataract. Design Prospective study. Patient and methods This study was performed in adults from tertiary Saudi Arabian hospitals (34 males and 26 females in each group, aged from 40 to 60 years), who were divided into four groups with an equal number of subjects (control group, cardiac group, idiopathic cataract group, and a group of cardiac patients using nitroglycerin and with cataracts). Fasting glucose concentrations, blood glycated hemoglobin levels, lipid profiles, and levels of nitrite, conjugated dienes (CD), thiobarbituric acid reactive substances (TBARS), superoxide dismutase (SOD), and reduced glutathione (GSH) were determined. Results Treatment of cardiac patients with nitroglycerin produced an imbalance in their systemic redox status, leading to the development of cataracts, which was reflected by a significant increase in the levels of nitrite, CD, and TBARS and a significant decrease in SOD activity and GSH, compared with idiopathic cataract patients. The results of correlation studies and multiple regression analysis revealed a significant positive correlation between different biochemical parameters (GSH, SOD, TBARS, CD, and nitrite) in the blood and lens in both idiopathic cataract patients and cardiac patients treated with nitroglycerin. Conclusion The study points to the relative and predictive effects of nitric oxide derived from nitroglycerin in the development of cataract in the presence of the oxidative stress induced by nitroglycerin treatment.
Collapse
Affiliation(s)
- Rehab M El-Gharabawy
- Pharmacology and Toxicology Department, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia; Pharmacology and Toxicology Department, College of Pharmacy, Tanta University, Tanta
| | - Amira S Ahmed
- Pharmacology and Toxicology Department, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia; Hormone Department, National Research Center, Giza, Egypt
| | - Amal H Al-Najjar
- Pharmacy Services Department, Security Forces Hospital, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
11
|
Nanoliposomal Nitroglycerin Exerts Potent Anti-Inflammatory Effects. Sci Rep 2015; 5:16258. [PMID: 26584637 PMCID: PMC4653649 DOI: 10.1038/srep16258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/13/2015] [Indexed: 12/22/2022] Open
Abstract
Nitroglycerin (NTG) markedly enhances nitric oxide (NO) bioavailability. However, its ability to mimic the anti-inflammatory properties of NO remains unknown. Here, we examined whether NTG can suppress endothelial cell (EC) activation during inflammation and developed NTG nanoformulation to simultaneously amplify its anti-inflammatory effects and ameliorate adverse effects associated with high-dose NTG administration. Our findings reveal that NTG significantly inhibits human U937 cell adhesion to NO-deficient human microvascular ECs in vitro through an increase in endothelial NO and decrease in endothelial ICAM-1 clustering, as determined by NO analyzer, microfluorimetry, and immunofluorescence staining. Nanoliposomal NTG (NTG-NL) was formulated by encapsulating NTG within unilamellar lipid vesicles (DPhPC, POPC, Cholesterol, DHPE-Texas Red at molar ratio of 6:2:2:0.2) that were ~155 nm in diameter and readily uptaken by ECs, as determined by dynamic light scattering and quantitative fluorescence microscopy, respectively. More importantly, NTG-NL produced a 70-fold increase in NTG therapeutic efficacy when compared with free NTG while preventing excessive mitochondrial superoxide production associated with high NTG doses. Thus, these findings, which are the first to reveal the superior therapeutic effects of an NTG nanoformulation, provide the rationale for their detailed investigation for potentially superior vascular normalization therapies.
Collapse
|
12
|
Sukhatme V, Bouche G, Meheus L, Sukhatme VP, Pantziarka P. Repurposing Drugs in Oncology (ReDO)-nitroglycerin as an anti-cancer agent. Ecancermedicalscience 2015; 9:568. [PMID: 26435741 PMCID: PMC4583240 DOI: 10.3332/ecancer.2015.568] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Indexed: 01/30/2023] Open
Abstract
Nitroglycerin (NTG), a drug that has been in clinical use for more than a century, has a range of actions which make it of particular interest in an oncological setting. It is generally accepted that the main mechanism of action of NTG is via the production of nitric oxide (NO), which improves cardiac oxygenation via multiple mechanisms including improved blood flow (vasodilation), decreased platelet aggregation, increased erythrocyte O2 release and decreased mitochondrial utilization of oxygen. Its vasoactive properties mean that it has the potential to exploit more fully the enhanced permeability and retention effect in delivering anti-cancer drugs to tumour tissues. Moreover NTG can reduce HIF-1α levels in hypoxic tumour tissues and this may have anti-angiogenic, pro-apoptotic and anti-efflux effects. Additionally NTG may enhance anti-tumour immunity. Pre-clinical and clinical data on these anti-cancer properties of NTG are summarised and discussed. While there is evidence of a positive action as a monotherapy in prostate cancer, there are mixed results in NSCLC where initially positive results have yet to be fully replicated. Based on the evidence presented, a case is made that further exploration of the clinical benefits that may accrue to cancer patients is warranted. Additionally, it is proposed that NTG may synergise with a number of other drugs, including other repurposed drugs, and these are discussed in the supplementary material appended to this paper.
Collapse
Affiliation(s)
- Vidula Sukhatme
- GlobalCures, Inc, Newton MA 02459, USA
- Corresponding authors
- Lead authors
| | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc, Newton MA 02459, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
- The George Pantziarka TP53 Trust, London KT1 2JP, UK
- Corresponding authors
- Lead authors
| |
Collapse
|
13
|
Mao M, Varadarajan S, Fukai T, Bakhshi FR, Chernaya O, Dudley SC, Minshall RD, Bonini MG. Nitroglycerin tolerance in caveolin-1 deficient mice. PLoS One 2014; 9:e104101. [PMID: 25158065 PMCID: PMC4144835 DOI: 10.1371/journal.pone.0104101] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/06/2014] [Indexed: 11/18/2022] Open
Abstract
Nitrate tolerance developed after persistent nitroglycerin (GTN) exposure limits its clinical utility. Previously, we have shown that the vasodilatory action of GTN is dependent on endothelial nitric oxide synthase (eNOS/NOS3) activity. Caveolin-1 (Cav-1) is known to interact with NOS3 on the cytoplasmic side of cholesterol-enriched plasma membrane microdomains (caveolae) and to inhibit NOS3 activity. Loss of Cav-1 expression results in NOS3 hyperactivation and uncoupling, converting NOS3 into a source of superoxide radicals, peroxynitrite, and oxidative stress. Therefore, we hypothesized that nitrate tolerance induced by persistent GTN treatment results from NOS3 dysfunction and vascular toxicity. Exposure to GTN for 48-72 h resulted in nitrosation and depletion (>50%) of Cav-1, NOS3 uncoupling as measured by an increase in peroxynitrite production (>100%), and endothelial toxicity in cultured cells. In the Cav-1 deficient mice, NOS3 dysfunction was accompanied by GTN tolerance (>50% dilation inhibition at low GTN concentrations). In conclusion, GTN tolerance results from Cav-1 modification and depletion by GTN that causes persistent NOS3 activation and uncoupling, preventing it from participating in GTN-medicated vasodilation.
Collapse
Affiliation(s)
- Mao Mao
- Department of Medicine-Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Sudhahar Varadarajan
- Department of Medicine-Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Tohru Fukai
- Department of Medicine-Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Farnaz R. Bakhshi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Olga Chernaya
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Samuel C. Dudley
- Department of Medicine-Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Marcelo G. Bonini
- Department of Medicine-Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
14
|
Endothelial barrier protection by local anesthetics: ropivacaine and lidocaine block tumor necrosis factor-α-induced endothelial cell Src activation. Anesthesiology 2014; 120:1414-28. [PMID: 24525631 DOI: 10.1097/aln.0000000000000174] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Pulmonary endothelial barrier dysfunction mediated in part by Src-kinase activation plays a crucial role in acute inflammatory disease. Proinflammatory cytokines, such as tumor necrosis factor-α (TNFα), activate Src via phosphatidylinositide 3-kinase/Akt-dependent nitric oxide generation, a process initiated by recruitment of phosphatidylinositide 3-kinase regulatory subunit p85 to TNF-receptor-1. Because amide-linked local anesthetics have well-established anti-inflammatory effects, the authors hypothesized that ropivacaine and lidocaine attenuate inflammatory Src signaling by disrupting the phosphatidylinositide 3-kinase-Akt-nitric oxide pathway, thus blocking Src-dependent neutrophil adhesion and endothelial hyperpermeability. METHODS Human lung microvascular endothelial cells, incubated with TNFα in the absence or presence of clinically relevant concentrations of ropivacaine and lidocaine, were analyzed by Western blot, probing for phosphorylated/activated Src, endothelial nitric oxide synthase, Akt, intercellular adhesion molecule-1, and caveolin-1. The effect of ropivacaine on TNFα-induced nitric oxide generation, co-immunoprecipitation of TNF-receptor-1 with p85, neutrophil adhesion, and endothelial barrier disruption were assessed. RESULTS Ropivacaine and lidocaine attenuated TNFα-induced Src activation (half-maximal inhibitory concentration [IC50] = 8.611 × 10 M for ropivacaine; IC50 = 5.864 × 10 M for lidocaine) and endothelial nitric oxide synthase phosphorylation (IC50 = 7.572 × 10 M for ropivacaine; IC50 = 6.377 × 10 M for lidocaine). Akt activation (n = 7; P = 0.006) and stimulus-dependent binding of TNF-receptor-1 and p85 (n = 6; P = 0.043) were blocked by 1 nM of ropivacaine. TNFα-induced neutrophil adhesion and disruption of endothelial monolayers via Src-dependent intercellular adhesion molecule-1- and caveolin-1-phosphorylation, respectively, were also attenuated. CONCLUSIONS Ropivacaine and lidocaine effectively blocked inflammatory TNFα signaling in endothelial cells by attenuating p85 recruitment to TNF-receptor-1. The resultant decrease in Akt, endothelial nitric oxide synthase, and Src phosphorylation reduced neutrophil adhesion and endothelial hyperpermeability. This novel anti-inflammatory "side-effect" of ropivacaine and lidocaine may provide therapeutic benefit in acute inflammatory disease.
Collapse
|
15
|
Bonini MG, Consolaro MEL, Hart PC, Mao M, de Abreu ALP, Master AM. Redox control of enzymatic functions: The electronics of life's circuitry. IUBMB Life 2014; 66:167-181. [PMID: 24668617 DOI: 10.1002/iub.1258] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/06/2014] [Indexed: 12/22/2022]
Abstract
The field of redox biology has changed tremendously over the past 20 years. Formerly regarded as bi-products of the aerobic metabolism exclusively involved in tissue damage, reactive oxygen species (ROS) are now recognized as active participants of cell signaling events in health and in disease. In this sense, ROS and the more recently defined reactive nitrogen species (RNS) are, just like hormones and second messengers, acting as fundamental orchestrators of cell signaling pathways. The chemical modification of enzymes by ROS and RNS (that result in functional enzymatic alterations) accounts for a considerable fraction of the transient and persistent perturbations imposed by variations in oxidant levels. Upregulation of ROS and RNS in response to stress is a common cellular response that foments adaptation to a variety of physiologic alterations (hypoxia, hyperoxia, starvation, and cytokine production). Frequently, these are beneficial and increase the organisms' resistance against subsequent acute stress (preconditioning). Differently, the sustained ROS/RNS-dependent rerouting of signaling produces irreversible alterations in cellular functioning, often leading to pathogenic events. Thus, the duration and reversibility of protein oxidations define whether complex organisms remain "electronically" healthy. Among the 20 essential amino acids, four are particularly susceptible to oxidation: cysteine, methionine, tyrosine, and tryptophan. Here, we will critically review the mechanisms, implications, and repair systems involved in the redox modifications of these residues in proteins while analyzing well-characterized prototypic examples. Occasionally, we will discuss potential consequences of amino acid oxidation and speculate on the biologic necessity for such events in the context of adaptative redox signaling. © 2014 IUBMB Life, 66(3):167-181, 2014.
Collapse
Affiliation(s)
- Marcelo G Bonini
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa, Parana, Brazil
| | - Marcia E L Consolaro
- Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa, Parana, Brazil
| | - Peter C Hart
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Mao Mao
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andre Luelsdorf Pimenta de Abreu
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Programa de Biociencias Aplicadas a Farmacia (PBF), Universidade Estadual de Maringa, Maringa, Parana, Brazil
| | - Alyssa M Master
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
16
|
Greco R, Mangione AS, Siani F, Blandini F, Vairetti M, Nappi G, Sandrini G, Buzzi MG, Tassorelli C. Effects of CGRP receptor antagonism in nitroglycerin-induced hyperalgesia. Cephalalgia 2013; 34:594-604. [DOI: 10.1177/0333102413517776] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 11/28/2013] [Indexed: 11/16/2022]
Abstract
Background The release of calcitonin gene-related peptide (CGRP) from trigeminal nerves plays a central role in the pathophysiology of migraine and clinical evidence shows an antimigraine effect for CGRP receptor antagonists. Systemic administration of nitroglycerin (NTG), a nitrovasodilator, consistently provokes spontaneous-like migraine attacks in migraine sufferers; in the rat, systemic NTG induces a condition of hyperalgesia, probably through the activation of cerebral/spinal structures involved in nociceptive transmission. Aim The aim of this article is to test the analgesic effect of the CGRP receptor antagonist MK-8825 in two animal models of pain that may be relevant for migraine: the tail flick test and the formalin test performed during NTG-induced hyperalgesia. Results MK-8825 showed analgesic activity when administered alone at both the tail flick test and the formalin test. Furthermore, the CGRP antagonist proved effective in counteracting NTG-induced hyperalgesia in both tests. MK-8825 indeed reduced the nociceptive behavior when administered either simultaneously or prior to (30–60 minutes before) NTG. Conclusion These data suggest that MK-8825 may represent a potential therapeutic tool for the treatment of migraine.
Collapse
Affiliation(s)
- R Greco
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
| | - AS Mangione
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
| | - F Siani
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, “C. Mondino” National Neurological Institute, Italy
| | - F Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, “C. Mondino” National Neurological Institute, Italy
| | - M Vairetti
- Department of Internal Medicine and Therapeutics, Pharmacology and Toxicology Unit, University of Pavia, Italy
| | - G Nappi
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
| | - G Sandrini
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
- IRCCS Santa Lucia Foundation, Italy
| | - MG Buzzi
- Department of Brain and Behaviour, University of Pavia, Italy
| | - C Tassorelli
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, “C. Mondino” National Neurological Institute, Italy
- IRCCS Santa Lucia Foundation, Italy
| |
Collapse
|
17
|
Bakhshi FR, Mao M, Shajahan AN, Piegeler T, Chen Z, Chernaya O, Sharma T, Elliott WM, Szulcek R, Bogaard HJ, Comhair S, Erzurum S, van Nieuw Amerongen GP, Bonini MG, Minshall RD. Nitrosation-dependent caveolin 1 phosphorylation, ubiquitination, and degradation and its association with idiopathic pulmonary arterial hypertension. Pulm Circ 2013; 3:816-30. [PMID: 25006397 PMCID: PMC4070841 DOI: 10.1086/674753] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/18/2013] [Indexed: 01/15/2023] Open
Abstract
In the present study, we tested the hypothesis that chronic inflammation and oxidative/nitrosative stress induce caveolin 1 (Cav-1) degradation, providing an underlying mechanism of endothelial cell activation/dysfunction and pulmonary vascular remodeling in patients with idiopathic pulmonary arterial hypertension (IPAH). We observed reduced Cav-1 protein despite increased Cav-1 messenger RNA expression and also endothelial nitric oxide synthase (eNOS) hyperphosphorylation in human pulmonary artery endothelial cells (PAECs) from patients with IPAH. In control human lung endothelial cell cultures, tumor necrosis factor α-induced nitric oxide (NO) production and S-nitrosation (SNO) of Cav-1 Cys-156 were associated with Src displacement and activation, Cav-1 Tyr-14 phosphorylation, and destabilization of Cav-1 oligomers within 5 minutes that could be blocked by eNOS or Src inhibition. Prolonged stimulation (72 hours) with NO donor DETANONOate reduced oligomerized and total Cav-1 levels by 40%-80%, similar to that observed in IPAH patient-derived PAECs. NO donor stimulation of endothelial cells for >72 hours, which was associated with sustained Src activation and Cav-1 phosphorylation, ubiquitination, and degradation, was blocked by NOS inhibitor L-NAME, Src inhibitor PP2, and proteosomal inhibitor MG132. Thus, chronic inflammation, sustained eNOS and Src signaling, and Cav-1 degradation may be important causal factors in the development of IPAH by promoting PAEC dysfunction/activation via sustained oxidative/nitrosative stress.
Collapse
Affiliation(s)
- Farnaz R. Bakhshi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mao Mao
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ayesha N. Shajahan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tobias Piegeler
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Zhenlong Chen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Olga Chernaya
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tiffany Sharma
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - W. Mark Elliott
- Pulmonary Division, James Hogg Research Centre Biobank, University of British Columbia, Vancouver, Canada
| | - Robert Szulcek
- Department of Physiology, Institute for Cardiovascular Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
- Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Suzy Comhair
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Geerten P. van Nieuw Amerongen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Physiology, Institute for Cardiovascular Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Marcelo G. Bonini
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
18
|
Ying WZ, Aaron KJ, Sanders PW. Transforming growth factor-β regulates endothelial function during high salt intake in rats. Hypertension 2013; 62:951-6. [PMID: 24041947 DOI: 10.1161/hypertensionaha.113.01835] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Previous studies have demonstrated that an increase in dietary NaCl (salt) intake stimulated endothelial cells to produce transforming growth factor-β (TGF-β). The intent of the present study was to determine the functional significance of increased TGF-β on endothelial cell function. Young Sprague-Dawley rats were fed diets containing 0.3 or 8.0% NaCl for 2 days before treatment with a specific inhibitor of the TGF-β receptor I/activin receptor-like kinase 5 kinase, or vehicle for another 2 days. At day 4 of study, endothelial phosphorylated Smad2 (S465/467) increased and phosphatase and tensin homologue deleted on chromosome 10 (PTEN) levels decreased in the high-salt-treated rats. In addition, phosphorylated Akt (S473) and phosphorylation of the endothelial isoform of NO synthase (NOS3) at S1177 increased. Treatment with the TGF-β receptor I/activin receptor-like kinase 5 inhibitor reduced Smad2 phosphorylation to levels observed in rats on the low-salt diet and prevented the downstream signaling events induced by the high-salt diet. In human umbilical vein endothelial cells, reduction in PTEN levels increased phosphorylated Akt and NOS3. Treatment of macrovascular endothelial cells with TGF-β1 increased phosphorylated NOS3 and the concentration of NO metabolites in the medium but had no effect on either of these variables in cells pretreated with small interfering RNA directed against PTEN. Thus, during high salt intake, an increase in TGF-β directly promoted a reduction in endothelial PTEN levels, which in turn regulated Akt activation and NOS3 phosphorylation. This effect closes a feedback loop that potentially mitigates the effect of TGF-β on the vasculature.
Collapse
Affiliation(s)
- Wei-Zhong Ying
- Division of Nephrology/Department of Medicine, 642 Lyons-Harrison Research Bldg, 1530 Third Ave, South, University of Alabama at Birmingham, Birmingham, AL 35294-0007.
| | | | | |
Collapse
|
19
|
Yang Z, Li J, Kong J, Wu S. Impairment of vascular endothelial function following reperfusion therapy in patients with acute myocardial infarction. J Int Med Res 2013; 41:1074-8. [PMID: 23771713 DOI: 10.1177/0300060513487650] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective To investigate vascular endothelial impairment as a result of reperfusion therapy in patients with acute myocardial infarction (AMI). Methods Patients with AMI underwent reperfusion therapy (percutaneous cardiac intervention [PCI] or thrombolytic therapy) or conservative drug therapy. Healthy control subjects were recruited. Endothelial impairment was assessed via endothelial nitric oxide (NO) synthase (eNOS), NO and endothelin-1 (ET-1) levels, 24 h after reperfusion or on enrolment, as appropriate. Results Patients who underwent PCI ( n = 47) or thrombolytic therapy ( n = 45) had significantly lower eNOS and NO levels, and higher ET-1 levels than those who received conservative drug therapy ( n = 46). All patient groups had significantly lower eNOS and NO levels, and higher ET-1 levels, than controls ( n = 45). There was a significant positive correlation between eNOS and NO, as well as significant negative correlations between eNOS/ET-1 and NO/ET-1 in all four groups. Conclusions Patients with AMI who underwent reperfusion therapy displayed low eNOS activity. This may result in impairment of endothelial function via downregulation of NO and upregulation of ET-1.
Collapse
Affiliation(s)
- Zhaoying Yang
- Cadre Ward, First Hospital, Jilin University, Changchun, China
- Cadre Ward, Heilongjiang Provincial Hospital, Harbin, China
| | - Jinliang Li
- Cadre Ward, First Hospital, Jilin University, Changchun, China
- Cadre Ward, Heilongjiang Provincial Hospital, Harbin, China
| | - Jian Kong
- Cadre Ward, First Hospital, Jilin University, Changchun, China
| | - Suisheng Wu
- Cadre Ward, First Hospital, Jilin University, Changchun, China
| |
Collapse
|
20
|
Regulation of cardiac nitric oxide signaling by nuclear β-adrenergic and endothelin receptors. J Mol Cell Cardiol 2013; 62:58-68. [PMID: 23684854 DOI: 10.1016/j.yjmcc.2013.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 04/11/2013] [Accepted: 05/07/2013] [Indexed: 01/19/2023]
Abstract
At the cell surface, βARs and endothelin receptors can regulate nitric oxide (NO) production. β-adrenergic receptors (βARs) and type B endothelin receptors (ETB) are present in cardiac nuclear membranes and regulate transcription. The present study investigated the role of the NO pathway in the regulation of gene transcription by these nuclear G protein-coupled receptors. Nitric oxide production and transcription initiation were measured in nuclei isolated from the adult rat heart. The cell-permeable fluorescent dye 4,5-diaminofluorescein diacetate (DAF2 DA) was used to provide a direct assessment of nitric oxide release. Both isoproterenol and endothelin increased NO production in isolated nuclei. Furthermore, a β3AR-selective agonist, BRL 37344, increased NO synthesis whereas the β1AR-selective agonist xamoterol did not. Isoproterenol increased, whereas ET-1 reduced, de novo transcription. The NO synthase inhibitor l-NAME prevented isoproterenol from increasing either NO production or de novo transcription. l-NAME also blocked ET-1-induced NO-production but did not alter the suppression of transcription initiation by ET-1. Inhibition of the cGMP-dependent protein kinase (PKG) using KT5823 also blocked the ability of isoproterenol to increase transcription initiation. Furthermore, immunoblotting revealed eNOS, but not nNOS, in isolated nuclei. Finally, caged, cell-permeable isoproterenol and endothelin-1 analogs were used to selectively activate intracellular β-adrenergic and endothelin receptors in intact adult cardiomyocytes. Intracellular release of caged ET-1 or isoproterenol analogs increased NO production in intact adult cardiomyocytes. Hence, activation of the NO synthase/guanylyl cyclase/PKG pathway is necessary for nuclear β3ARs to increase de novo transcription. Furthermore, we have demonstrated the potential utility of caged receptor ligands in selectively modulating signaling via endogenous intracellular G protein-coupled receptors.
Collapse
|
21
|
Yoon SH, Kim JW. A Study of the Pathway of Nitric Oxide Production by Nitroglycerin in Trabecular Meshwork Cells. JOURNAL OF THE KOREAN OPHTHALMOLOGICAL SOCIETY 2013. [DOI: 10.3341/jkos.2013.54.9.1429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Suk Hyun Yoon
- Department of Ophthalmology, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - Jae Woo Kim
- Department of Ophthalmology, Catholic University of Daegu School of Medicine, Daegu, Korea
| |
Collapse
|
22
|
Ying WZ, Aaron KJ, Sanders PW. Effect of aging and dietary salt and potassium intake on endothelial PTEN (Phosphatase and tensin homolog on chromosome 10) function. PLoS One 2012; 7:e48715. [PMID: 23144940 PMCID: PMC3492426 DOI: 10.1371/journal.pone.0048715] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 09/28/2012] [Indexed: 11/19/2022] Open
Abstract
Aging promotes endothelial dysfunction, defined as a reduction in bioavailable nitric oxide (NO) produced by the endothelial isoform of nitric oxide synthase (NOS3). This enzyme is critically regulated by phosphorylation by protein kinase B (Akt), which in turn is regulated by the lipid phosphatase, PTEN. The present series of studies demonstrated a reduction in bioavailable NO as the age of rats increased from 1 to 12 months. At 12 months of age, rats no longer demonstrated increases in phosphorylated NOS3 in response to high dietary salt intake. Endothelial cell levels of PTEN increased with age and became refractory to change with increased salt intake. In contrast to the reduction in NO production, endothelial cell production of transforming growth factor-ß (TGF-ß) relative to NO increased progressively with age. In macrovascular endothelial cells, PTEN was regulated in a dose-dependent fashion by TGF-ß, which was further regulated by extracellular [KCl]. When combined with prior studies, the present series of experiments suggested an integral role for PTEN in endothelial cell pathobiology of aging and an important mitigating function of TGF-ß in endothelial PTEN regulation. The findings further supported a role for diet in affecting vascular function through the production of TGF-ß and NO.
Collapse
Affiliation(s)
- Wei-Zhong Ying
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, Center for Free Radical Biology, Center for Aging, and Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Kristal J. Aaron
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, Center for Free Radical Biology, Center for Aging, and Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Paul W. Sanders
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, Center for Free Radical Biology, Center for Aging, and Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Veterans Affairs Medical Center, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|