1
|
AlRuwaili R, Al-Kuraishy HM, Alruwaili M, Khalifa AK, Alexiou A, Papadakis M, Saad HM, Batiha GES. The potential therapeutic effect of phosphodiesterase 5 inhibitors in the acute ischemic stroke (AIS). Mol Cell Biochem 2024; 479:1267-1278. [PMID: 37395897 PMCID: PMC11116240 DOI: 10.1007/s11010-023-04793-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/16/2023] [Indexed: 07/04/2023]
Abstract
Acute ischemic stroke (AIS) is a focal neurological disorder that accounts for 85% of all stroke types, due to occlusion of cerebral arteries by thrombosis and emboli. AIS is also developed due to cerebral hemodynamic abnormality. AIS is associated with the development of neuroinflammation which increases the severity of AIS. Phosphodiesterase enzyme (PDEs) inhibitors have neuro-restorative and neuroprotective effects against the development of AIS through modulation of the cerebral cyclic adenosine monophosphate (cAMP)/cyclic guanosine monophosphate (cGMP)/nitric oxide (NO) pathway. PDE5 inhibitors through mitigation of neuroinflammation may decrease the risk of long-term AIS-induced complications. PDE5 inhibitors may affect the hemodynamic properties and coagulation pathway which are associated with thrombotic complications in AIS. PDE5 inhibitors reduce activation of the pro-coagulant pathway and improve the microcirculatory level in patients with hemodynamic disturbances in AIS. PDE5 inhibitors mainly tadalafil and sildenafil improve clinical outcomes in AIS patients through the regulation of cerebral perfusion and cerebral blood flow (CBF). PDE5 inhibitors reduced thrombomodulin, P-selectin, and tissue plasminogen activator. Herein, PDE5 inhibitors may reduce activation of the pro-coagulant pathway and improve the microcirculatory level in patients with hemodynamic disturbances in AIS. In conclusion, PDE5 inhibitors may have potential roles in the management of AIS through modulation of CBF, cAMP/cGMP/NO pathway, neuroinflammation, and inflammatory signaling pathways. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Raed AlRuwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Kasr El-Ainy School of Medicine, Cairo University, El Manial, Cairo, 11562, Egypt
- Lecturer of Medical Pharmacology, Nahda Faculty of Medicine, Beni Suef, Egypt
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matrouh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
2
|
Medicarpin isolated from Radix Hedysari ameliorates brain injury in a murine model of cerebral ischemia. J Food Drug Anal 2021; 29:581-605. [PMID: 35649147 PMCID: PMC9931010 DOI: 10.38212/2224-6614.3377] [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: 03/26/2021] [Accepted: 08/12/2021] [Indexed: 11/18/2022] Open
Abstract
The development of effective post-stroke therapy is highly demanded. Medicarpin is a key active component of a famous Chinese herbal prescription used for post-stroke treatment in Taiwan; however, little is known about its biological effects and mechanisms of action. Herein, we implemented a murine model of cerebral ischemic/reperfusional injury-related stroke to elucidate medicarpin's neuroprotective effect. In male ICR mice 24 h after stroke induction, treatment with medicarpin (0.5 and 1.0 mg/kg, i.v.) markedly enhanced the survival rates, improved moving distance and walking area coverage, reduced brain infarction, and preserved the blood-brain barrier, supporting medicarpin's protective effect on stroke-induced injury. Immunohistochemistry analysis further revealed that medicarpin treatment decreased the expression/activation of p65NF-κB and caspase 3, especially near the infarct cortex, while promoting the expression of neurogenesis-associated proteins, including doublecortin (DCX), brain-derived neurotrophic factor (BDNF), and tyrosine receptor kinase B (TrkB). These changes of expression levels were accompanied by GSK-3 inactivation and β-catenin upregulation. Notably, pretreatment with LY294002, a PI3K inhibitor, abolished the aforementioned beneficial effects of medicarpin, illustrating an essential role of PI3K/Akt activation in medicarpin's neuroprotective and reparative activities. In vitro studies revealed that medicarpin displayed strong anti-inflammatory activity by reducing nitric oxide (NO) production in lipopolysaccharide-stimulated microglial cells (BV2) with an IC50 around 5 ±1 (μM) and anti-apoptotic activity in neuronal cells (N2A) subjected to oxygen-glucose deprivation with an IC50 around 13 ± 2 (μM). Collectively, this is the first report to demonstrate that medicarpin, isolated from Radix Hedysari, ameliorates ischemic brain injury through its anti-inflammatory microglia/NO), anti-apoptotic (neuronal cells/OGD) and neuroprotective effects by activating the PI3K/Akt-dependent GSK-3 inactivation for upregulating β-catenin, which in turn decreases the expression/activation of p65NF-κB and caspase 3 and promotes the expression of neurogenic (DCX, BDNF, TrkB) and neuroprotective (Bcl2) factors in the brain.
Collapse
|
3
|
El Alaoui MZ, Guy A, Khalki L, Limami Y, Benomar A, Zaid N, Cherrah Y, Mekhfi H, Cadi R, Zaid Y. [Current antiplatelet agents, new inhibitors and therapeutic targets]. Med Sci (Paris) 2020; 36:348-357. [PMID: 32356711 DOI: 10.1051/medsci/2020061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular diseases are the leading cause of deaths in the world. Platelets play a major role in the occurrence of these diseases and the development of antiplatelet drugs is a priority in the fight against cardiovascular diseases-associated mortality. Aspirin and thienopyridine-based P2Y12 inhibitors are the main drugs currently used. These molecules target the initiation of platelets activation and are responsible for a moderate inhibitory action. Other antiplatelet agents, as glycoprotein (GP) IIb/IIIa antagonists, inhibit platelet aggregation independently of initial activation-associated pathways, but are responsible for increased hemorrhagic events. Regarding each antiplatelet agent's specific characteristics, the prescription of these drugs must take into account the type of cardiovascular event, the age of the patient, the past medical history, and the potential hemorrhagic adverse events. Thus, there is a need for the development of new molecules with a more targeted effect, maintaining optimal efficiency but with a reduction of the hemorrhagic risk, which is the principal limitation of these treatments.
Collapse
Affiliation(s)
| | - Alexandre Guy
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Maroc
| | - Loubna Khalki
- Research Center of Mohammed VI University of Health Sciences, Casablanca, Maroc
| | - Youness Limami
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Ali Benomar
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Nabil Zaid
- Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Maroc
| | - Yahia Cherrah
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Hassan Mekhfi
- Laboratory of Physiology, Genetic and Ethnopharmacology, Faculty of Sciences, Mohammed the First University, Oujda, Maroc
| | - Rachida Cadi
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Maroc
| | - Younes Zaid
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc - Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Maroc
| |
Collapse
|
4
|
Wang YH, Chern CM, Liou KT, Kuo YH, Shen YC. Ergostatrien-7,9(11),22-trien-3β-ol from Antrodia camphorata ameliorates ischemic stroke brain injury via downregulation of p65NF-κ-B and caspase 3, and activation of Akt/GSK3/catenin-associated neurogenesis. Food Funct 2019; 10:4725-4738. [PMID: 31304955 DOI: 10.1039/c9fo00908f] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Antrodia camphorata is a well-known traditional Chinese mushroom used as a functional food and nutraceutical in Taiwan and China. The aim of this study was to explore the protective effects and mechanism(s) of the ethyl acetate crude extract of A. camphorata (EtOAc-AC) and its active constituent ergostatrien-7,9(11),22-trien-3β-ol (EK100) in an acute ischemic stroke (AIS) murine model. Treating mice with induced AIS injury by using EtOAc-AC (0.3-0.6 g kg-1, p.o.) and EK100 (60 and 120 mg kg-1, p.o.) 2 h after AIS induction significantly increased the tracking distance and reduced brain infarction. Both EtOAc-AC and EK-100 reduced the expression levels of p65NF-κB and caspase 3 near the peri-infarct cortex and promoted the expression of neurogenesis-associated protein doublecortin (DCX) near the hippocampus, accompanied by glycogen synthase kinase 3 (GSK-3) inhibition and β-catenin upregulation. Signaling pathway analysis revealed that the advantageous effects of EtOAc-AC and EK-100 involved triggering the activation of PI3K/Akt and inhibition of GSK-3. Our findings suggest that EtOAc-AC and its active constituent EK100 display anti-inflammatory and anti-apoptotic activities. Both EtOAc-AC and EK100 reduce ischemic brain injury by decreasing p65NF-κB and caspase 3 expression, and they promote neurogenesis (DCX) and neuroprotection (Bcl2) by activating the PI3k/Akt-associated GSK3 inhibition and β-catenin activation.
Collapse
Affiliation(s)
- Yea-Hwey Wang
- National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | | | | | | | | |
Collapse
|
5
|
Wang YH, Liou KT, Tsai KC, Liu HK, Yang LM, Chern CM, Shen YC. GSK-3 inhibition through GLP-1R allosteric activation mediates the neurogenesis promoting effect of P7C3 after cerebral ischemic/reperfusional injury in mice. Toxicol Appl Pharmacol 2018; 357:88-105. [PMID: 30189238 DOI: 10.1016/j.taap.2018.08.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/22/2022]
Abstract
An aminopropyl carbazole compound, P7C3, has been shown to be a potent neurogenesis promoting agent; however, its fundamental signaling action has yet to be elucidated. A cerebral ischemic/reperfusional (CI/R) injury model in mice was implemented to elucidate the neuronal protective mechanism(s) of P7C3. Treating CI/R mice using P7C3 (50-100 μg/kg, i.v.) significantly improved tracking distance and walking behavior, and reduced brain damage. Specifically, P7C3 promoted the expression of neurogenesis-associated proteins, including doublecortin, beta tubulin III (β-tub3), adam11 and adamts20, near the peri-infarct cortex, accompanied by glycogen synthase kinase 3 (GSK-3) inhibition and β-catenin upregulation. The application of a specific inhibitor against glucagon-like peptide 1 receptor (GLP-1R), exendin(9-39), revealed that the beneficial effects of P7C3 involved triggering the activation of GLP-1R-associated PKA/Akt signaling. P7C3 elicited the GLP-1R-dependent intracellular cAMP increment and the insulin secretion in cellular models. Surface plasmon resonance assay of P7C3 showed a Kd value of 0.53 μM for GLP-1R binding, and the docking of P7C3 to the putative active site on GLP-1R was successfully predicted by molecular modeling. Our findings indicate that P7C3 promotes the expression of neurogenesis proteins by activation of the cAMP/PKA-dependent and Akt/GSK3-associated β-catenin through positive allosteric stimulation of GLP-1R. Within the P7C3 class of neuroprotective molecules, this mechanism appears to be unique to the prototypical P7C3 molecule, as other active derivatives such as P7C2-A20 and P7C3-S243 they do not engage this same pathway and have been shown to work by nicotinamide phosphoribosyltransferase (NAMPT) stimulation.
Collapse
Affiliation(s)
- Yea-Hwey Wang
- National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Kuo-Tong Liou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Chinese Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; Ph.D. Program for Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan
| | - Hui-Kang Liu
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; Ph.D. Program for the Clinical Drug Discovery from Botanical Herbs, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Li-Ming Yang
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; School of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Chang-Ming Chern
- Division of Neurovascular Disease, Neurological Institute, Taipei Veterans General Hospital, Taiwan; Taipei Municipal Gan-Dau Hospital, Taiwan; Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei City, Taiwan
| | - Yuh-Chiang Shen
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, Taiwan; National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan; Ph.D. Program for the Clinical Drug Discovery from Botanical Herbs, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan.
| |
Collapse
|
6
|
Mathematical Modelling of Nitric Oxide/Cyclic GMP/Cyclic AMP Signalling in Platelets. Int J Mol Sci 2018; 19:ijms19020612. [PMID: 29462984 PMCID: PMC5855834 DOI: 10.3390/ijms19020612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 02/04/2023] Open
Abstract
Platelet activation contributes to normal haemostasis but also to pathologic conditions like stroke and cardiac infarction. Signalling by cGMP and cAMP inhibit platelet activation and are therefore attractive targets for thrombosis prevention. However, extensive cross-talk between the cGMP and cAMP signalling pathways in multiple tissues complicates the selective targeting of their activities. We have used mathematical modelling based on experimental data from the literature to quantify the steady state behaviour of nitric oxide (NO)/cGMP/cAMP signalling in platelets. The analysis provides an assessment of NO-induced cGMP synthesis and PKG activation as well as cGMP-mediated cAMP and PKA activation though modulation of phosphodiesterase (PDE2 and 3) activities. Both one- and two-compartment models of platelet cyclic nucleotide signalling are presented. The models provide new insight for understanding how NO signalling to cGMP and indirectly cAMP, can inhibit platelet shape-change, the initial step of platelet activation. Only the two-compartment models could account for the experimental observation that NO-mediated PKA activation can occur when the bulk platelet cAMP level is unchanged. The models revealed also a potential for hierarchical interplay between the different platelet phosphodiesterases. Specifically, the models predict, unexpectedly, a strong effect of pharmacological inhibitors of cGMP-specific PDE5 on the cGMP/cAMP cross-talk. This may explain the successful use of weak PDE5-inhibitors, such as dipyridamole, in anti-platelet therapy. In conclusion, increased NO signalling or PDE5 inhibition are attractive ways of increasing cGMP-cAMP cross-talk selectively in platelets.
Collapse
|
7
|
Chien MY, Chuang CH, Chern CM, Liou KT, Liu DZ, Hou YC, Shen YC. Salvianolic acid A alleviates ischemic brain injury through the inhibition of inflammation and apoptosis and the promotion of neurogenesis in mice. Free Radic Biol Med 2016; 99:508-519. [PMID: 27609227 DOI: 10.1016/j.freeradbiomed.2016.09.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/11/2016] [Accepted: 09/04/2016] [Indexed: 12/18/2022]
Abstract
Salvianolic acid A (SalA), a chemical type of caffeic acid trimer, has drawn great attention for its potent bioactivities against ischemia-induced injury both in vitro and in vivo. In this study, we evaluated SalA's protective effects against acute ischemic stroke by inducing middle cerebral artery occlusion/reperfusion (MCAO) injuries in mice. Treatment of the mice with SalA (50 and 100μg/kg, i.v.) at 2h after MCAO enhanced their survival rate, improved their moving activity, and ameliorated the severity of brain infarction and apoptosis seen in the mice by diminishing pathological changes such as the extensive breakdown of the blood-brain barrier (BBB), nitrosative stress, and the activation of an inflammatory transcriptional factor p65 nuclear factor-kappa B (NF-κB) and a pro-apoptotic kinase p25/Cdk5. SalA also intensively limited cortical infarction and promoted the expression of neurogenesis protein near the peri-infarct cortex and subgranular zone of the hippocampal dentate gyrus by compromising the activation of GSK3β and p25/Cdk5, which in turn upregulated β-catenin, doublecortin (DCX), and Bcl-2, most possibly through the activation of PI3K/Akt signaling via the upregulation of brain-derived neurotrophic factor. We conclude that SalA blocks inflammatory responses by impairing NF-κB signaling, thereby limiting inflammation/nitrosative stress and preserving the integrity of the BBB; SalA also concomitantly promotes neurogenesis-related protein expression by compromising GSK3β/Cdk5 activity to enhance the expression levels of β-catenin/DCX and Bcl-2 for neuroprotection.
Collapse
Affiliation(s)
- Mei-Yin Chien
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan; Ko Da Pharmaceutical Co., Taoyuan, Taiwan
| | | | - Chang-Ming Chern
- Division of Neurovascular Disease, Neurological Institute, Taipei Veterans General Hospital & Taipei Municipal Gan-Dau Hospital, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Kou-Tong Liou
- Department of Combat Sports and Chinese Martial Arts, Chinese Culture University, Taipei, Taiwan
| | - Der-Zen Liu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Center for General Education, Hsuan Chuang University, Hsinchu, Taiwan.
| | - Yu-Chang Hou
- Department of Chinese Medicine, Taoyuan General Hospital, Ministry of Health and Welfare, Taiwan; Department of Bioscience Technology, Chuan-Yuan Christian University, Taoyuan, Taiwan
| | - Yuh-Chiang Shen
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Institute of Biomedical Sciences, National Chung-Hsing University, Taichung, Taiwan; National Taipei University of Nursing and Health Science, Taipei, Taiwan.
| |
Collapse
|
8
|
Ingberg E, Dock H, Theodorsson E, Theodorsson A, Ström JO. Method parameters' impact on mortality and variability in mouse stroke experiments: a meta-analysis. Sci Rep 2016; 6:21086. [PMID: 26876353 PMCID: PMC4753409 DOI: 10.1038/srep21086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Although hundreds of promising substances have been tested in clinical trials,
thrombolysis currently remains the only specific pharmacological treatment for
ischemic stroke. Poor quality, e.g. low statistical power, in the preclinical
studies has been suggested to play an important role in these failures. Therefore,
it would be attractive to use animal models optimized to minimize unnecessary
mortality and outcome variability, or at least to be able to power studies more
exactly by predicting variability and mortality given a certain experimental setup.
The possible combinations of methodological parameters are innumerous, and an
experimental comparison of them all is therefore not feasible. As an alternative
approach, we extracted data from 334 experimental mouse stroke articles and, using a
hypothesis-driven meta-analysis, investigated the method parameters’
impact on infarct size variability and mortality. The use of Swiss and C57BL6 mice
as well as permanent occlusion of the middle cerebral artery rendered the lowest
variability of the infarct size while the emboli methods increased variability. The
use of Swiss mice increased mortality. Our study offers guidance for researchers
striving to optimize mouse stroke models.
Collapse
Affiliation(s)
- Edvin Ingberg
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Hua Dock
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Elvar Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Annette Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Division of Neuro and Inflammation Science, Department of Clinical and Experimental Medicine, Linköping University, Department of Neurosurgery, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Sweden
| | - Jakob O Ström
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro, Sweden.,School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
9
|
Puri N, Mohey V, Singh M, Kaur T, Pathak D, Buttar HS, Singh AP. Dipyridamole attenuates ischemia reperfusion induced acute kidney injury through adenosinergic A1 and A2A receptor agonism in rats. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:361-8. [PMID: 26728617 DOI: 10.1007/s00210-015-1206-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/21/2015] [Indexed: 01/16/2023]
Abstract
Dipyridamole (DYP) is an anti-platelet agent with marked vasodilator, anti-oxidant, and anti-inflammatory activity. The present study investigated the role of adenosine receptors in DYP-mediated protection against ischemia reperfusion-induced acute kidney injury (AKI) in rats. The rats were subjected to bilateral renal ischemia for 40 min followed by reperfusion for 24 h. The renal damage induced by ischemia reperfusion injury (IRI) was assessed by measuring creatinine clearance, blood urea nitrogen, uric acid, plasma potassium, fractional excretion of sodium, and microproteinuria in rats. The oxidative stress in renal tissues was assessed by quantification of thiobarbituric acid-reactive substances, superoxide anion generation, and reduced glutathione level. The hematoxylin-eosin staining was carried out to observe histopathological changes in renal tissues. DYP (10 and 30 mg/kg, intraperitoneal, i.p.) was administered 30 min before subjecting the rats to renal IRI. In separate groups, caffeine (50 mg/kg, i.p.), an adenosinergic A1 and A2A receptor antagonist was administered with and without DYP treatment before subjecting the rats to renal IRI. The ischemia reperfusion-induced AKI was demonstrated by significant changes in serum as well as urinary parameters, enhanced oxidative stress, and histopathological changes in renal tissues. The administration of DYP demonstrated protection against AKI. The prior treatment with caffeine abolished DYP-mediated reno-protection suggesting role of A1 and A2A adenosine receptors in DYP-mediated reno-protection in rats. It is concluded that adenosine receptors find their definite involvement in DYP-mediated anti-oxidative and reno-protective effect against ischemia reperfusion-induced AKI.
Collapse
Affiliation(s)
- Nikkita Puri
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Vinita Mohey
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Manjinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Tajpreet Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India.,Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Devendra Pathak
- Department of Veterinary Anatomy, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Harpal Singh Buttar
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India.,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India.
| |
Collapse
|
10
|
Luo F, Le NB, Mills T, Chen NY, Karmouty-Quintana H, Molina JG, Davies J, Philip K, Volcik KA, Liu H, Xia Y, Eltzschig HK, Blackburn MR. Extracellular adenosine levels are associated with the progression and exacerbation of pulmonary fibrosis. FASEB J 2015; 30:874-83. [PMID: 26527068 DOI: 10.1096/fj.15-274845] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/19/2015] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis is a devastating lung disease with limited treatment options. The signaling molecule adenosine is produced in response to injury and serves a protective role in early stages of injury and is detrimental during chronic stages of disease such as seen in lung conditions such as pulmonary fibrosis. Understanding the association of extracellular adenosine levels and the progression of pulmonary fibrosis is critical for designing adenosine based approaches to treat pulmonary fibrosis. The goal of this study was to use various models of experimental lung fibrosis to understand when adenosine levels are elevated during pulmonary fibrosis and whether these elevations were associated with disease progression and severity. To accomplish this, extracellular adenosine levels, defined as adenosine levels found in bronchioalveolar lavage fluid, were determined in mouse models of resolvable and progressive pulmonary fibrosis. We found that relative bronchioalveolar lavage fluid adenosine levels are progressively elevated in association with pulmonary fibrosis and that adenosine levels diminish in association with the resolution of lung fibrosis. In addition, treatment of these models with dipyridamole, an inhibitor of nucleoside transporters that potentiates extracellular adenosine levels, demonstrated that the resolution of lung fibrosis is blocked by the failure of adenosine levels to subside. Furthermore, exacerbating adenosine levels led to worse fibrosis in a progressive fibrosis model. Increased adenosine levels were associated with elevation of IL-6 and IL-17, which are important inflammatory cytokines in pulmonary fibrosis. These results demonstrate that extracellular adenosine levels are closely associated with the progression of experimental pulmonary fibrosis and that this signaling pathway may mediate fibrosis by regulating IL-6 and IL-17 production.
Collapse
Affiliation(s)
- Fayong Luo
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Ngoc-Bao Le
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Tingting Mills
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Ning-Yuan Chen
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Harry Karmouty-Quintana
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Jose G Molina
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Jonathan Davies
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Kemly Philip
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Kelly A Volcik
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Hong Liu
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Yang Xia
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Holger K Eltzschig
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| | - Michael R Blackburn
- *Department of Biochemistry and Molecular Biology, University of Texas Medical School at Houston, Houston, Texas, USA; Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA; and Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
11
|
Potey C, Ouk T, Petrault O, Petrault M, Berezowski V, Salleron J, Bordet R, Gautier S. Early treatment with atorvastatin exerts parenchymal and vascular protective effects in experimental cerebral ischaemia. Br J Pharmacol 2015; 172:5188-98. [PMID: 26289767 DOI: 10.1111/bph.13285] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/06/2015] [Accepted: 08/10/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE From the clinical and experimental data available, statins appear to be interesting drug candidates for preventive neuroprotection in ischaemic stroke. However, their acute protective effect is, as yet, unconfirmed. EXPERIMENTAL APPROACH Male C57Bl6/JRj mice were subjected to middle cerebral artery occlusion and treated acutely with atorvastatin (10-20 mg·kg(-1) day(-1) ; 24 or 72 h). Functional recovery (neuroscore, forelimb gripping strength and adhesive removal test) was assessed during follow-up and lesion volume measured at the end. Vasoreactivity of the middle cerebral artery (MCA), type IV collagen and FITC-dextran distribution were evaluated to assess macrovascular and microvascular protection. Activated microglia, leucocyte adhesion and infiltration were chosen as markers of inflammation. KEY RESULTS Acute treatment with atorvastatin provided parenchymal and cerebral protection only at the higher dose of 20 mg·kg(-1) ·day(-1) . In this treatment group, functional recovery was ameliorated, and lesion volumes were reduced as early as 24 h after experimental stroke. This was associated with vascular protection as endothelial function of the MCA and the density and patency of the microvascular network were preserved. Acute atorvastatin administration also induced an anti-inflammatory effect in association with parenchymal and vascular mechanisms; it reduced microglial activation, and decreased leucocyte adhesion and infiltration. CONCLUSIONS AND IMPLICATIONS Acute atorvastatin provides global cerebral protection, but only at the higher dose of 20 mg·kg(-1) ·day(-1) ; this was associated with a reduction in inflammation in both vascular and parenchymal compartments. Our results suggest that atorvastatin could also be beneficial when administered early after stroke.
Collapse
Affiliation(s)
- C Potey
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - T Ouk
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - O Petrault
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - M Petrault
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - V Berezowski
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - J Salleron
- EA2694 - Biostatistics Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - R Bordet
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - S Gautier
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| |
Collapse
|
12
|
Interleukin-6 mediates enhanced thrombus development in cerebral arterioles following a brief period of focal brain ischemia. Exp Neurol 2015; 271:351-7. [PMID: 26054883 DOI: 10.1016/j.expneurol.2015.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/29/2015] [Accepted: 06/03/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The cerebral microvasculature is rendered more vulnerable to thrombus formation following a brief (5.0 min) period of focal ischemia. This study examined the contribution of interleukin-6 (IL-6), a neuroprotective and prothrombotic cytokine produced by the brain, to transient ischemia-induced thrombosis in cerebral arterioles. APPROACH & RESULTS The middle cerebral artery of C57BL/6J mice was occluded for 5 min, followed by 24h of reperfusion (MCAo/R). Intravital fluorescence microscopy was used to monitor thrombus development in cerebral arterioles induced by light/dye photoactivation. Thrombosis was quantified as the time of onset of platelet aggregation on the vessel wall and the time for complete blood flow cessation. MCAo/R in wild type (WT) mice yielded an acceleration of thrombus formation that was accompanied by increased IL-6 levels in plasma and in post-ischemic brain tissue. The exaggerated thrombosis response to MCAo/R was blunted in WT mice receiving an IL-6 receptor-blocking antibody and in IL-6 deficient (IL-6(-/-)) mice. Bone marrow chimeras, produced by transplanting IL-6(-/-) marrow into WT recipients, did not exhibit protection against MCAo/R-induced thrombosis. CONCLUSIONS The increased vulnerability of the cerebral vasculature to thrombus development after MCAo/R is mediated by IL-6, which is likely derived from brain cells rather than circulating blood cells. These findings suggest that anti-IL-6 therapy may reduce the likelihood of cerebral thrombus development after a transient ischemic attack.
Collapse
|
13
|
Zhao J, Zhang X, Dong L, Wen Y, Cui L. The many roles of statins in ischemic stroke. Curr Neuropharmacol 2014; 12:564-74. [PMID: 25977681 PMCID: PMC4428028 DOI: 10.2174/1570159x12666140923210929] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/29/2014] [Accepted: 09/22/2014] [Indexed: 12/22/2022] Open
Abstract
Stroke is the third leading cause of human death. Endothelial dysfunction, thrombogenesis, inflammatory and oxidative stress damage, and angiogenesis play an important role in cerebral ischemic pathogenesis and represent a target for prevention and treatment. Statins have been found to improve endothelial function, modulate thrombogenesis, attenuate inflammatory and oxidative stress damage, and facilitate angiogenesis far beyond lowering cholesterol levels. Statins have also been proved to significantly decrease cardiovascular risk and to improve clinical outcome. Could statins be the new candidate agent for the prevention and therapy in ischemic stroke? In recent years, a vast expansion in the understanding of the pathophysiology of ischemic stroke and the pleiotropic effects of statins has occurred and clinical trials involving statins for the prevention and treatment of ischemic stroke have begun. These facts force us to revisit ischemic stroke and consider new strategies for prevention and treatment. Here, we survey the important developments in the non-lipid dependent pleiotropic effects and clinical effects of statins in ischemic stroke.
Collapse
Affiliation(s)
- Jingru Zhao
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei 050051, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei collaborative innovation center for cerebro-vascular disease, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory for Neurology, Shijiazhuang, Hebei 050000, China
| | - Lipeng Dong
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | | | - Lili Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei collaborative innovation center for cerebro-vascular disease, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
14
|
Cui M, Ding H, Chen F, Zhao Y, Yang Q, Dong Q. Mdivi-1 Protects Against Ischemic Brain Injury via Elevating Extracellular Adenosine in a cAMP/CREB-CD39-Dependent Manner. Mol Neurobiol 2014; 53:240-253. [PMID: 25428621 DOI: 10.1007/s12035-014-9002-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/12/2014] [Indexed: 01/15/2023]
Abstract
This study aimed to examine whether the neuroprotective effects of Mdivi-1 are attributable to extracellular ATP and adenosine. Mdivi-1 was administered prior to or post middle cerebral artery occlusion (MCAO). The extracellular adenosine was measured by in vivo microdialysis and high-pressure liquid chromatography (HPLC) in MCAO mouse model. Western blot was done to determine the influence of Mdivi-1 on the expression of CD39 and CREB phosphorylation both in vivo and in the cultured astrocytes. Intracellular cAMP and protein kinase A (PKA) activity were detected in primary astrocytes. Results showed that Mdivi-1 significantly reduced infarct volume and neurological scores when administered either prior to or post MCAO. Interestingly, pretreatment with Mdivi-1 resulted in marked increase of extracellular adenosine and concomitant decrease in ATP. The expression of CD39, but not CD73, was upregulated by Mdivi-1, which was associated with the elevated phosphorylated cAMP response element-binding protein (CREB), a transcription factor potentially regulating CD39 expression. In primary astrocytes, Mdivi-1 treatment induced increases in intracellular cAMP, PKA activity and CREB phosphorylation, and PKA-specific inhibitor completely reversed Mdivi-1-induced CD39 expression. Our results demonstrate that Mdivi-1 protects against ischemic brain injury through increasing extracellular adenosine, a process involving elevated CD39 expression that is likely modulated by cAMP/PKA/CREB cascade. Figure Potential mechanisms by which Mdivi-1 mediates the neuroprotection on cerebral ischemic stroke. Results from the present study indicate that Mdivi-1 protects against ischemic brain injury through increasing extracellular adenosine, a process involving elevated CD39 expression that is likely modulated by the cAMP/PKA/CREB cascades.
Collapse
Affiliation(s)
- Mei Cui
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.
| | - Hongyan Ding
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Fangzhe Chen
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Yanxin Zhao
- Department of Neurology, The 10th People's Hospital, Tongji University, Shanghai, China
| | - Qi Yang
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.
| |
Collapse
|
15
|
Tang YH, Vital S, Russell J, Seifert H, Senchenkova E, Granger DN. Transient ischemia elicits a sustained enhancement of thrombus development in the cerebral microvasculature: effects of anti-thrombotic therapy. Exp Neurol 2014; 261:417-23. [PMID: 25058045 DOI: 10.1016/j.expneurol.2014.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/26/2014] [Accepted: 07/10/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE While transient ischemic attack (TIA) is a well-known harbinger of ischemic stroke, the mechanisms that link TIA to subsequent strokes remain poorly understood. The overall aim of this study was to determine whether: 1) brief periods of transient cerebral ischemia render this tissue more vulnerable to thrombus development and 2) antiplatelet agents used in TIA patients alter ischemia-induced thrombogenesis. APPROACH & RESULTS The middle cerebral artery of C57BL/6 mice was occluded for 2.5-10min, followed by reperfusion periods of 1-28days. Intravital microscopy was used to monitor thrombus development in cerebral microvessels induced by light/dye photoactivation. Thrombosis was quantified as the time to platelet aggregation on the vessel wall and the time for complete blood flow cessation. While brief periods of cerebral ischemia were not associated with neurological deficits or brain infarction (evaluated after 1day), it yielded a pronounced and prolonged (up to 28days) acceleration of thrombus formation, compared to control (sham) mice. This prothrombotic phenotype was not altered by pre- and/or post-treatment of mice with either aspirin (A), clopidogrel (C), dipyridamole (D), or atorvastatin (S), or with A+D+S. CONCLUSIONS The increased vulnerability of the cerebral vasculature to thrombus development after a brief period of transient ischemia can be recapitulated in a murine model. Antiplatelet or antithrombotic agents used in patients with TIA show no benefit in this mouse model of brief transient ischemia.
Collapse
Affiliation(s)
- Ya Hui Tang
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Shantel Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Janice Russell
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Hilary Seifert
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Elena Senchenkova
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - D Neil Granger
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
16
|
Pandyra A, Mullen PJ, Kalkat M, Yu R, Pong JT, Li Z, Trudel S, Lang KS, Minden MD, Schimmer AD, Penn LZ. Immediate utility of two approved agents to target both the metabolic mevalonate pathway and its restorative feedback loop. Cancer Res 2014; 74:4772-82. [PMID: 24994712 DOI: 10.1158/0008-5472.can-14-0130] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
New therapies are urgently needed for hematologic malignancies, especially in patients with relapsed acute myelogenous leukemia (AML) and multiple myeloma. We and others have previously shown that FDA-approved statins, which are used to control hypercholesterolemia and target the mevalonate pathway (MVA), can trigger tumor-selective apoptosis. Our goal was to identify other FDA-approved drugs that synergize with statins to further enhance the anticancer activity of statins in vivo. Using a screen composed of other FDA approved drugs, we identified dipyridamole, used for the prevention of cerebral ischemia, as a potentiator of statin anticancer activity. The statin-dipyridamole combination was synergistic and induced apoptosis in multiple myeloma and AML cell lines and primary patient samples, whereas normal peripheral blood mononuclear cells were not affected. This novel combination also decreased tumor growth in vivo. Statins block HMG-CoA reductase (HMGCR), the rate-limiting enzyme of the MVA pathway. Dipyridamole blunted the feedback response, which upregulates HMGCR and HMG-CoA synthase 1 (HMGCS1) following statin treatment. We further show that dipyridamole inhibited the cleavage of the transcription factor required for this feedback regulation, sterol regulatory element-binding transcription factor 2 (SREBF2, SREBP2). Simultaneously targeting the MVA pathway and its restorative feedback loop is preclinically effective against hematologic malignancies. This work provides strong evidence for the immediate evaluation of this novel combination of FDA-approved drugs in clinical trials.
Collapse
Affiliation(s)
- Aleksandra Pandyra
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Peter J Mullen
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Manpreet Kalkat
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Rosemary Yu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Janice T Pong
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Zhihua Li
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Suzanne Trudel
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Karl S Lang
- Institute of Immunology, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Mark D Minden
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Linda Z Penn
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
17
|
Chern CM, Wang YH, Liou KT, Hou YC, Chen CC, Shen YC. 2-Methoxystypandrone ameliorates brain function through preserving BBB integrity and promoting neurogenesis in mice with acute ischemic stroke. Biochem Pharmacol 2014; 87:502-14. [DOI: 10.1016/j.bcp.2013.11.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/20/2013] [Accepted: 11/22/2013] [Indexed: 11/15/2022]
|
18
|
Yan BC, Park JH, Shin BN, Ahn JH, Kim IH, Lee JC, Yoo KY, Hwang IK, Choi JH, Park JH, Lee YL, Suh HW, Jun JG, Kwon YG, Kim YM, Kwon SH, Her S, Kim JS, Hyun BH, Kim CK, Cho JH, Lee CH, Won MH. Neuroprotective effect of a new synthetic aspirin-decursinol adduct in experimental animal models of ischemic stroke. PLoS One 2013; 8:e74886. [PMID: 24073226 PMCID: PMC3779249 DOI: 10.1371/journal.pone.0074886] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 08/07/2013] [Indexed: 02/04/2023] Open
Abstract
Stroke is the second leading cause of death. Experimental animal models of cerebral ischemia are widely used for researching mechanisms of ischemic damage and developing new drugs for the prevention and treatment of stroke. The present study aimed to comparatively investigate neuroprotective effects of aspirin (ASA), decursinol (DA) and new synthetic aspirin-decursinol adduct (ASA-DA) against transient focal and global cerebral ischemic damage. We found that treatment with 20 mg/kg, not 10 mg/kg, ASA-DA protected against ischemia-induced neuronal death after transient focal and global ischemic damage, and its neuroprotective effect was much better than that of ASA or DA alone. In addition, 20 mg/kg ASA-DA treatment reduced the ischemia-induced gliosis and maintained antioxidants levels in the corresponding injury regions. In brief, ASA-DA, a new synthetic drug, dramatically protected neurons from ischemic damage, and neuroprotective effects of ASA-DA may be closely related to the attenuation of ischemia-induced gliosis and maintenance of antioxidants.
Collapse
Affiliation(s)
- Bing Chun Yan
- Institute of Integrative traditional & western Medicine,Medical College, Yangzhou University, Yangzhou, China
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Bich Na Shin
- Department of Physiology, College of Medicine and Institute of Neurodegeneration and Neuroregeneration, Hallym University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Ki-Yeon Yoo
- Department of Oral Anatomy, College of Dentistry, Gangneung-Wonju National University, Gangneung, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong Ho Park
- Division of Applied Chemistry and Biotechnology, Hanbat National University, Daejeon, South Korea
| | - Yun Lyul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Hong-Won Suh
- Department of Pharmacology and Institute of Natural Medicine, College of Medicine Hallym University, Chuncheon, South Korea
| | - Jong-Gab Jun
- Department of Chemistry and Institute of Applied Chemistry, Hallym University, Chuncheon, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Young-Myeong Kim
- Vascular System Research Center and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Seung-Hae Kwon
- Division of Analytical Bio-imaging, Chuncheon Center, Korea Basic Science Institute, Chuncheon, Kangwon, South Korea
| | - Song Her
- Division of Analytical Bio-imaging, Chuncheon Center, Korea Basic Science Institute, Chuncheon, Kangwon, South Korea
| | - Jin Su Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Byung-Hwa Hyun
- Laboratory Animal Center, OSONG Medical Innovation Foundation, Osong, South Korea
| | - Chul-Kyu Kim
- Laboratory Animal Center, OSONG Medical Innovation Foundation, Osong, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Choong Hyun Lee
- Department of Anatomy and Physiology, College of Pharmacy, Dankook University, Cheonan, South Korea
- * E-mail: (MHW); (CHL)
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
- * E-mail: (MHW); (CHL)
| |
Collapse
|
19
|
Eckle T, Hughes K, Ehrentraut H, Brodsky KS, Rosenberger P, Choi DS, Ravid K, Weng T, Xia Y, Blackburn MR, Eltzschig HK. Crosstalk between the equilibrative nucleoside transporter ENT2 and alveolar Adora2b adenosine receptors dampens acute lung injury. FASEB J 2013; 27:3078-89. [PMID: 23603835 DOI: 10.1096/fj.13-228551] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The signaling molecule adenosine has been implicated in attenuating acute lung injury (ALI). Adenosine signaling is terminated by its uptake through equilibrative nucleoside transporters (ENTs). We hypothesized that ENT-dependent adenosine uptake could be targeted to enhance adenosine-mediated lung protection. To address this hypothesis, we exposed mice to high-pressure mechanical ventilation to induce ALI. Initial studies demonstrated time-dependent repression of ENT1 and ENT2 transcript and protein levels during ALI. To examine the contention that ENT repression represents an endogenous adaptive response, we performed functional studies with the ENT inhibitor dipyridamole. Dipyridamole treatment (1 mg/kg; EC50=10 μM) was associated with significant increases in ALI survival time (277 vs. 395 min; P<0.05). Subsequent studies in gene-targeted mice for Ent1 or Ent2 revealed a selective phenotype in Ent2(-/-) mice, including attenuated pulmonary edema and improved gas exchange during ALI in conjunction with elevated adenosine levels in the bronchoalveolar fluid. Furthermore, studies in genetic models for adenosine receptors implicated the A2B adenosine receptor (Adora2b) in mediating ENT-dependent lung protection. Notably, dipyridamole-dependent attenuation of lung inflammation was abolished in mice with alveolar epithelial Adora2b gene deletion. Our newly identified crosstalk pathway between ENT2 and alveolar epithelial Adora2b in lung protection during ALI opens possibilities for combined therapies targeted to this protein set.
Collapse
Affiliation(s)
- Tobias Eckle
- Mucosal Inflammation Program, Department of Anesthesiology, University of Colorado School of Medicine, 12700 E. 19th Ave., Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Efficacy of combined atorvastatin and sildenafil in promoting recovery after ischemic stroke in mice. Am J Phys Med Rehabil 2013; 92:143-50. [PMID: 22854903 DOI: 10.1097/phm.0b013e3182643f1a] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The aim of this study was to test the hypothesis that a combination of atorvastatin and sildenafil promotes recovery in an additive manner after ischemic stroke in mice. DESIGN Adult C57BL/6 mice (n = 67) were subjected to transient middle cerebral artery occlusion. Vehicle-control (H2O), atorvastatin (0.3 mg/kg), sildenafil (0.3 mg/kg), or combined atorvastatin (0.3 mg/kg) and sildenafil (0.3 mg/kg) were administrated via oral gavage daily for 6 days starting 24 hrs after ischemia. Behavioral studies including neurologic score and adhesive removal test were performed before surgery and on postoperative days 1 and 7; cylinder test was performed before surgery and on postoperative day 7. Mice were killed after 7 days and brain slices were stained with hematoxylin and eosin to measure the infarct volume. RESULTS The combination group performed significantly better in the adhesive removal test (mean ± SD) (50 ± 54 secs) as compared with the control group (147 ± 109 secs) (P < 0.05) and to atorvastatin (144 ± 102 secs) (P < 0.05) but did not show statistically significant improvement as compared with sildenafil (107 ± 115 secs) (P = 0.148). There were no significant differences among the groups in neurologic score and cylinder test. There was no significant difference in the infarct volume. CONCLUSIONS The data suggest that combined atorvastatin and sildenafil generates a better functional outcome as compared with atorvastatin-only treatment, but not sildenafil-only treatment, in one of multiple variables tested.
Collapse
|
21
|
Cui M, Bai X, Li T, Chen F, Dong Q, Zhao Y, Liu X. Decreased extracellular adenosine levels lead to loss of hypoxia-induced neuroprotection after repeated episodes of exposure to hypoxia. PLoS One 2013; 8:e57065. [PMID: 23437309 PMCID: PMC3578825 DOI: 10.1371/journal.pone.0057065] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Accepted: 01/17/2013] [Indexed: 12/20/2022] Open
Abstract
Achieving a prolonged neuroprotective state following transient ischemic attacks (TIAs) is likely to effectively reduce the brain damage and neurological dysfunction associated with recurrent stroke. HPC is a phenomenon in which advanced exposure to mild hypoxia reduces the stroke volume produced by a subsequent TIA. However, this neuroprotection is not long-lasting, with the effects reaching a peak after 3 days. Therefore, in this study, we investigated the use of multiple episodes of hypoxic exposure at different time intervals to induce longer-term protection in a mouse stroke model. C57BL/6 mice were subjected to different hypoxic preconditioning protocols: a single episode of HPC or five identical episodes at intervals of 3 days (E3d HPC) or 6 days (E6d HPC). Three days after the last hypoxic exposure, temporary middle cerebral artery occlusion (MCAO) was induced. The effects of these HPC protocols on hypoxia-inducible factor (HIF) regulated gene mRNA expression were measured by quantitative PCR. Changes in extracellular adenosine concentrations, known to exert neuroprotective effects, were also measured using in vivo microdialysis and high pressure liquid chromatography (HPLC). Neuroprotection was provided by E6d HPC but not E3d HPC. HIF-regulated target gene expression increased significantly following all HPC protocols. However, E3d HPC significantly decreased extracellular adenosine and reduced cerebral blood flow in the ischemic region with upregulated expression of the adenosine transporter, equilibrative nucleoside transporter 1 (ENT1). An ENT1 inhibitor, propentofylline increased the cerebral blood flow and re-established neuroprotection in E3d HPC. Adenosine receptor specific antagonists showed that adenosine mainly through A1 receptor mediates HPC induced neuroprotection. Our data indicate that cooperation of HIF-regulated genes and extracellular adenosine is necessary for HPC-induced neuroprotection.
Collapse
Affiliation(s)
- Mei Cui
- Department of Neurology, Huashan hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Abstract
HMG-CoA reductase inhibitors (statins) are associated with improved stroke outcome. This observation has been attributed in part to the palliative effect of statins on cerebral hemodynamics and cerebral autoregulation (CA), which are mediated mainly through the upregulation of endothelium nitric oxide synthase (eNOS). Several animal studies indicate that statin pretreatment enhances cerebral blood flow after ischemic stroke, although this finding is not further supported in clinical settings. Cerebral vasomotor reactivity, however, is significantly improved after long-term statin administration in most patients with severe small vessel disease, aneurysmal subarachnoid hemorrhage, or impaired baseline CA.
Collapse
|
24
|
Terpolilli NA, Moskowitz MA, Plesnila N. Nitric oxide: considerations for the treatment of ischemic stroke. J Cereb Blood Flow Metab 2012; 32:1332-46. [PMID: 22333622 PMCID: PMC3390820 DOI: 10.1038/jcbfm.2012.12] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 01/02/2012] [Accepted: 01/06/2012] [Indexed: 12/21/2022]
Abstract
Some 40 years ago it was recognized by Furchgott and colleagues that the endothelium releases a vasodilator, endothelium-derived relaxing factor (EDRF). Later on, several groups identified EDRF to be a gas, nitric oxide (NO). Since then, NO was identified as one of the most versatile and unique molecules in animal and human biology. Nitric oxide mediates a plethora of physiological functions, for example, maintenance of vascular tone and inflammation. Apart from these physiological functions, NO is also involved in the pathophysiology of various disorders, specifically those in which regulation of blood flow and inflammation has a key role. The aim of the current review is to summarize the role of NO in cerebral ischemia, the most common cause of stroke.
Collapse
Affiliation(s)
- Nicole A Terpolilli
- Department of Neurosurgery, University of
Munich Medical School, Munich, Germany
| | - Michael A Moskowitz
- Neuroscience Center, Massachusetts General
Hospital, Harvard Medical School, Boston,
Massachusetts, USA
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research,
University of Munich Medical School, Munich, Germany
| |
Collapse
|
25
|
Srivastava K, Bath PMW, Bayraktutan U. Current therapeutic strategies to mitigate the eNOS dysfunction in ischaemic stroke. Cell Mol Neurobiol 2012; 32:319-36. [PMID: 22198555 DOI: 10.1007/s10571-011-9777-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 11/29/2011] [Indexed: 12/22/2022]
Abstract
Impairment of endothelial nitric oxide synthase (eNOS) activity is implicated in the pathogenesis of endothelial dysfunction in many diseases including ischaemic stroke. The modulation of eNOS during and/or following ischaemic injury often represents a futile compensatory mechanism due to a significant decrease in nitric oxide (NO) bioavailability coupled with dramatic increases in the levels of reactive oxygen species that further neutralise NO. However, applications of a number of therapeutic agents alone or in combination have been shown to augment eNOS activity under a variety of pathological conditions by potentiating the expression and/or activity of Akt/eNOS/NO pathway components. The list of these therapeutic agents include NO donors, statins, angiotensin-converting enzyme inhibitors, calcium channel blockers, phosphodiesterase-3 inhibitors, aspirin, dipyridamole and ellagic acid. While most of these compounds exhibit anti-platelet properties and are able to up-regulate eNOS expression in endothelial cells and platelets, others suppress eNOS uncoupling and tetrahydrobiopterin (an eNOS stabiliser) oxidation. As the number of therapeutic molecules that modulate the expression and activity of eNOS increases, further detailed research is required to reveal their mode of action in preventing and/or reversing the endothelial dysfunction.
Collapse
Affiliation(s)
- Kirtiman Srivastava
- Division of Stroke, Clinical Sciences Building, Nottingham City Hospital Campus, The University of Nottingham, Nottingham, UK.
| | | | | |
Collapse
|
26
|
Chang CC, Wang YH, Chern CM, Liou KT, Hou YC, Peng YT, Shen YC. Prodigiosin inhibits gp91(phox) and iNOS expression to protect mice against the oxidative/nitrosative brain injury induced by hypoxia-ischemia. Toxicol Appl Pharmacol 2011; 257:137-47. [PMID: 21925195 DOI: 10.1016/j.taap.2011.08.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 08/29/2011] [Accepted: 08/29/2011] [Indexed: 01/30/2023]
Abstract
This study aimed to explore the mechanisms by which prodigiosin protects against hypoxia-induced oxidative/nitrosative brain injury induced by middle cerebral artery occlusion/reperfusion (MCAo/r) injury in mice. Hypoxia in vitro was modeled using oxygen-glucose deprivation (OGD) followed by reoxygenation of BV-2 microglial cells. Our results showed that treatment of mice that have undergone MCAo/r injury with prodigiosin (10 and 100μg/kg, i.v.) at 1h after hypoxia ameliorated MCAo/r-induced oxidative/nitrosative stress, brain infarction, and neurological deficits in the mice, and enhanced their survival rate. MCAo/r induced a remarkable production in the mouse brains of reactive oxygen species (ROS) and a significant increase in protein nitrosylation; this primarily resulted from enhanced expression of NADPH oxidase 2 (gp91(phox)), inducible nitric oxide synthase (iNOS), and the infiltration of CD11b leukocytes due to breakdown of blood-brain barrier (BBB) by activation of nuclear factor-kappa B (NF-κB). All these changes were significantly diminished by prodigiosin. In BV-2 cells, OGD induced ROS and nitric oxide production by up-regulating gp91(phox) and iNOS via activation of the NF-κB pathway, and these changes were suppressed by prodigiosin. In conclusion, our results indicate that prodigiosin reduces gp91(phox) and iNOS expression possibly by impairing NF-κB activation. This compromises the activation of microglial and/or inflammatory cells, which then, in turn, mediates prodigiosin's protective effect in the MCAo/r mice.
Collapse
Affiliation(s)
- Chia-Che Chang
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
27
|
Oyama N, Yagita Y, Kawamura M, Sugiyama Y, Terasaki Y, Omura-Matsuoka E, Sasaki T, Kitagawa K. Cilostazol, not aspirin, reduces ischemic brain injury via endothelial protection in spontaneously hypertensive rats. Stroke 2011; 42:2571-7. [PMID: 21799161 DOI: 10.1161/strokeaha.110.609834] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE It is well-established that hypertension leads to endothelial dysfunction in the cerebral artery. Recently, cilostazol has been used for the secondary prevention of ischemic stroke. Among antiplatelet drugs, phosphodiesterase inhibitors including cilostazol have been shown to have protective effects on endothelial cells. The aim of the present study is to investigate the effects of cilostazol and aspirin on endothelial nitric oxide synthase (eNOS) phosphorylation in the cerebral cortex, endothelial function, and infarct size after brain ischemia in spontaneously hypertensive rats (SHR). METHODS Five-week-old male SHR received a 5-week regimen of chow containing 0.1% aspirin, 0.1% cilostazol, 0.3% cilostazol, or the vehicle control. The levels of total and Ser(1177)-phosphorylated eNOS protein in the cerebral cortex were evaluated by Western blot. To assess the contribution of eNOS in maintaining cerebral blood flow, we monitored cerebral blood flow by laser-Doppler flowmetry after L-N(5)-(1-iminoethyl)ornithine infusion. Additionally, we evaluated residual microperfusion using fluorescence-labeled serum protein and infarct size after transient focal brain ischemia. RESULTS In SHR, the blood pressure and heart rate were similar among the groups. Cilostazol-treated SHR had a significantly higher ratio of phospho-eNOS/total eNOS protein than vehicle-treated and aspirin-treated SHR. Treating with cilostazol, but not aspirin, significantly improved cerebral blood flow response to L-N(5)-(1-iminoethyl)ornithine. Cilostazol also increased residual perfusion of the microcirculation and reduced brain damage after ischemia compared to vehicle control and aspirin. CONCLUSIONS These findings indicate that cilostazol, but not aspirin, can attenuate ischemic brain injury by maintaining endothelial function in the cerebral cortex of SHR.
Collapse
Affiliation(s)
- Naoki Oyama
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
García-Bonilla L, Sosti V, Campos M, Penalba A, Boada C, Sumalla M, Hernández-Guillamon M, Rosell A, Montaner J. Effects of acute post-treatment with dipyridamole in a rat model of focal cerebral ischemia. Brain Res 2011; 1373:211-20. [DOI: 10.1016/j.brainres.2010.12.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 12/02/2010] [Accepted: 12/02/2010] [Indexed: 01/18/2023]
|
29
|
d’Esterre CD, Tichauer KM, Aviv RI, Eisert W, Lee TY. Dipyridamole Treatment Prior to Stroke Onset: Examining Post-stroke Cerebral Circulation and Outcome in Rabbits. Transl Stroke Res 2011; 2:186-94. [DOI: 10.1007/s12975-010-0062-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 12/01/2010] [Accepted: 12/22/2010] [Indexed: 11/27/2022]
|
30
|
D’Esterre C, Lee TY. Effect of dipyridamole during acute stroke: exploring antithrombosis and neuroprotective benefits. Ann N Y Acad Sci 2010; 1207:71-5. [DOI: 10.1111/j.1749-6632.2010.05801.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
31
|
Guo S, Stins M, Ning M, Lo EH. Amelioration of inflammation and cytotoxicity by dipyridamole in brain endothelial cells. Cerebrovasc Dis 2010; 30:290-6. [PMID: 20664263 DOI: 10.1159/000319072] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 04/23/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Increasing evidence suggests that beyond its antiplatelet properties, dipyridamole may have pleiotropic effects on other cells within the neurovascular elements of the brain. In this experimental cellular study, we asked whether dipyridamole can ameliorate brain endothelial injury after exposure to inflammatory and metabolic insults. METHODS Human brain endothelial cells were grown in culture, and exposed to TNFalpha (continuously for 20 h) or subjected to oxygen-glucose deprivation (OGD; 6 h of insult followed by 18 h recovery). Expression of ICAM-1, VCAM-1 and PECAM-1 were measured by immunoblotting. Matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) in the conditioned media were quantified via zymography. MTT mitochondrial activity was measured to assess endothelial cell viability. RESULTS Exposure of human brain endothelial cells to TNFalpha (12.5-50 ng/ml) induced a clear increase in protein levels of ICAM-1, VCAM-1 and MMP-9. TNFalpha did not alter PECAM-1. Dipyridamole (1-5 muM) significantly attenuated ICAM-1 and MMP-9 levels after this inflammatory insult. No significant effects of dipyridamole were noted for VCAM-1. Six-hour OGD induced moderate endothelial cell death accompanied by a release of MMP-9. Dipyridamole significantly decreased MMP-9 levels and cell death after this metabolic insult. CONCLUSIONS These results suggest that dipyridamole may ameliorate brain endothelial injury after inflammation and/or metabolic insults. How these putative cellular mechanisms may relate to clinical outcomes and conditions in stroke patients remains to be elucidated.
Collapse
Affiliation(s)
- Shuzhen Guo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass., USA.
| | | | | | | |
Collapse
|
32
|
Abstract
The mechanisms responsible for vascular autoregulation in the brain during changes in mean arterial blood pressure are ambiguous. Potentially, adenosine, a purine nucleoside and potent vasodilator, may be involved as earlier studies have documented an increase in brain adenosine concentrations with cerebral ischemia and hypotension. Consequently, we tested the hypothesis that adenosine is involved in vasodilatation during hypotension within the autoregulatory range (>50 mm Hg) by exposing adenosine 2a receptor (A2aR) knockout and wild type (WT) mice to short (2 to 5 mins) periods of hypotension. We found that autoregulation was significantly (P<0.05) impaired by 29% in A2a knockout mice as compared with WT animals. Furthermore, the A2R antagonist (A2a>A2b:10-85>1), ZM-241385, in a dose (1, 5, 10 mg/kg, intraperitoneally)-related manner, attenuated autoregulation in WT mice. In knockout mice treated with ZM-2413585 (5 and 10 mg/kg), autoregulation was further impaired indicating that A2b receptors also participated in cerebral vasodilatation. Treatment with dipyridamole (1.0 mg/kg) that increases extracellular concentrations of adenosine improved autoregulation in the A2aR knockout mice. We would conclude that adenosine through both A2a and A2b receptors is involved in physiologic vascular regulation during hypotension even within the autoregulatory range.
Collapse
|
33
|
Salat D, Ribosa R, Garcia-Bonilla L, Montaner J. Statin use before and after acute ischemic stroke onset improves neurological outcome. Expert Rev Cardiovasc Ther 2010; 7:1219-30. [PMID: 19814665 DOI: 10.1586/erc.09.52] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The devastating consequences of stroke, both in terms of morbidity and mortality, and the economic implications it has worldwide, coupled with a growing knowledge regarding its pathophysiology, has led to the development of novel therapeutic strategies in recent years that have had an enormous impact on clinical practice and the outcome of stroke patients. While thrombolysis is regarded as the most important of these developments, its relatively narrow therapeutic window and complexity of administration imply that only a minority of stroke patients can benefit from it. Among other strategies, in an attempt to overcome this limitation, research has focused on the development of neuroprotection aimed to salvage ischemic brain tissue by means complementary to reperfusion. Among these approaches, the use of statins stands out, which, by inhibiting the HMG CoA reductase, interfere with the formation of isoprenoid intermediates in the biosynthesis of cholesterol, and have been shown to have a positive impact in both the incidence and outcome of acute stroke. The main results of animal models, observational studies, clinical trials and opinion articles available in the medical literature covering the effect of statins on acute brain ischemia will be reviewed in order to provide in-depth information regarding their mode of action and current evidence regarding their potential benefits for ischemic stroke patients.
Collapse
Affiliation(s)
- David Salat
- Neurovascular Research Laboratory, Neurovascular Unit, Institut de Recerca, Hospital Vall d'Hebron, Pg Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | | | | | | |
Collapse
|
34
|
Venkatesh PK, Pattillo CB, Branch B, Hood J, Thoma S, Illum S, Pardue S, Teng X, Patel RP, Kevil CG. Dipyridamole enhances ischaemia-induced arteriogenesis through an endocrine nitrite/nitric oxide-dependent pathway. Cardiovasc Res 2010; 85:661-70. [PMID: 20061326 DOI: 10.1093/cvr/cvq002] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIMS Anti-platelet agents, such as dipyridamole, have several clinical benefits for peripheral artery disease with the speculation of angiogenic potential that could preserve ischaemic tissue viability, yet the effect of dipyridamole on ischaemic arteriogenesis or angiogenesis is unknown. Here we test the hypothesis that dipyridamole therapy augments arteriolar vessel development and function during chronic ischaemia. METHODS AND RESULTS Mice were treated with 200 mg/kg dipyridamole twice daily to achieve therapeutic plasma levels (0.8-1.2 microg/mL). Chronic hindlimb ischaemia was induced by permanent femoral artery ligation followed by measurement of tissue perfusion using laser Doppler blood flow along with quantification of vascular density, cell proliferation, and activation of nitric oxide (NO) metabolism. Dipyridamole treatment quickly restored ischaemic hindlimb blood flow, increased vascular density and cell proliferation, and enhanced collateral artery perfusion compared with control treatments. The beneficial effects of dipyridamole on blood flow and vascular density were dependent on NO production as dipyridamole did not augment ischaemic tissue reperfusion, vascular density, or endothelial cell proliferation in endothelial NO synthase (eNOS)-deficient mice. Blood and tissue nitrite levels were significantly higher in dipyridamole-treated mice compared with controls and eNOS(-/-) mice, verifying increased NO production that was regulated in a PKA-dependent manner. CONCLUSION Dipyridamole augments nitrite/NO production, leading to enhanced arteriogenesis activity and blood perfusion in ischaemic limbs. Together, these data suggest that dipyridamole can augment ischaemic vessel function and restore blood flow, which may be beneficial in peripheral artery disease.
Collapse
|
35
|
Sawada N, Liao JK. Targeting eNOS and beyond: emerging heterogeneity of the role of endothelial Rho proteins in stroke protection. Expert Rev Neurother 2009; 9:1171-86. [PMID: 19673606 DOI: 10.1586/ern.09.70] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Currently available modalities for the treatment of acute ischemic stroke are aimed at preserving or augmenting cerebral blood flow. Experimental evidence suggests that statins, which show 25-30% reduction of stroke incidence in clinical trials, confer stroke protection by upregulation of eNOS and increasing cerebral blood flow. The upregulation of eNOS by statins is mediated by inhibition of small GTP-binding protein RhoA. Our recent study uncovered a unique role for a Rho-family member Rac1 in stroke protection. Rac1 in endothelium does not affect cerebral blood flow. Instead, inhibition of endothelial Rac1 leads to broad upregulation of the genes relevant to neurovascular protection. Intriguingly, inhibition of endothelial Rac1 enhances neuronal cell survival through endothelium-derived neurotrophic factors, including artemin. This review discusses the emerging therapeutic opportunities to target neurovascular signaling beyond the BBB, with special emphasis on the novel role of endothelial Rac1 in stroke protection.
Collapse
Affiliation(s)
- Naoki Sawada
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Center for Life Sciences, Boston, MA 02115, USA.
| | | |
Collapse
|
36
|
Zhou Q, Liao JK. Statins and cardiovascular diseases: from cholesterol lowering to pleiotropy. Curr Pharm Des 2009; 15:467-78. [PMID: 19199975 DOI: 10.2174/138161209787315684] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Statins are 3-hydroxy-3-methyglutaryl coenzyme A (HMG-CoA) reductase inhibitors, which are prescribed extensively for cholesterol lowering in the primary and secondary prevention of cardiovascular disease. Recent compelling evidence suggests that the beneficial effects of statins may not only be due to their cholesterol lowering effects, but also, to their cholesterol-independent or pleiotropic effects. Through these so-called pleiotropic effects, statins are directly involved in restoring or improving endothelial function, attenuating vascular remodeling, inhibiting vascular inflammatory response, and perhaps, stabilizing atherosclerotic plaques. These cholesterol-independent effects of statins are predominantly due to their ability to inhibit isoprenoid synthesis, the products of which are important lipid attachments for intracellular signaling molecules, such as Rho, Rac and Cdc42. In particular, inhibition of Rho and its downstream target, Rho-associated coiled-coil containing protein kinase (ROCK), has emerged as the principle mechanisms underlying the pleiotropic effects of statins. This review provides an update of statin-mediated vascular effects beyond cholesterol lowering and highlights recent findings from bench to bedside to support the concept of statin pleiotropy.
Collapse
Affiliation(s)
- Qian Zhou
- Vascular Medicine Research Unit, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02139, USA
| | | |
Collapse
|
37
|
Toda N, Ayajiki K, Okamura T. Cerebral Blood Flow Regulation by Nitric Oxide: Recent Advances. Pharmacol Rev 2009; 61:62-97. [DOI: 10.1124/pr.108.000547] [Citation(s) in RCA: 268] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
38
|
Sawada N, Kim HH, Moskowitz MA, Liao JK. Rac1 is a critical mediator of endothelium-derived neurotrophic activity. Sci Signal 2009; 2:ra10. [PMID: 19278959 PMCID: PMC2668716 DOI: 10.1126/scisignal.2000162] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The therapeutic potential of neurotrophic factors has been hampered by their inability to achieve adequate tissue penetration. Brain blood vessels, however, could be an alternative target for neurosalvage therapies by virtue of their close proximity to neurons. Here we show that hemizygous deletion of Rac1 in mouse endothelial cells (ECs) attenuates brain injury and edema after focal cerebral ischemia. Microarray analysis of Rac1(+/-) ECs revealed enrichment of stress response genes, basement membrane components, and neurotrophic factors that could affect neuronal survival. Consistent with these expression profiles, endothelial Rac1 hemizygosity enhanced antioxidative and endothelial barrier capacities and potentiated paracrine neuroprotective activities through the up-regulation of the neurotrophic factor, artemin. Endothelial Rac1, therefore, could be an important therapeutic target for promoting endothelial barrier integrity and neurotrophic activity.
Collapse
Affiliation(s)
- Naoki Sawada
- Vascular Medicine Research, Brigham and Women's Hospital, 65 Landsdowne Street, Room 275, Cambridge, MA 02139, USA
| | - Hyung-Hwan Kim
- Vascular Medicine Research, Brigham and Women's Hospital, 65 Landsdowne Street, Room 275, Cambridge, MA 02139, USA
| | - Michael A. Moskowitz
- Stroke and Neurovascular Regulation Laboratory, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - James K. Liao
- Vascular Medicine Research, Brigham and Women's Hospital, 65 Landsdowne Street, Room 275, Cambridge, MA 02139, USA
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|