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
|
Small molecule inhibiting microglial nitric oxide release could become a potential treatment for neuroinflammation. PLoS One 2023; 18:e0278325. [PMID: 36745631 PMCID: PMC9901772 DOI: 10.1371/journal.pone.0278325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 11/14/2022] [Indexed: 02/07/2023] Open
Abstract
Microglia are the immune effector cells of the central nervous system (CNS) and react to pathologic events with a complex process including the release of nitric oxide (NO). NO is a free radical, which is toxic for all cells at high concentrations. To target an exaggerated NO release, we tested a library of 16 544 chemical compounds for their effect on lipopolysaccharide (LPS)-induced NO release in cell line and primary neonatal microglia. We identified a compound (C1) which significantly reduced NO release in a dose-dependent manner, with a low IC50 (252 nM) and no toxic side effects in vitro or in vivo. Target finding strategies such as in silico modelling and mass spectroscopy hint towards a direct interaction between C1 and the nitric oxide synthase making C1 a great candidate for specific intra-cellular interaction with the NO producing machinery.
Collapse
|
3
|
Zullig LL, Sung AD, Khouri MG, Jazowski S, Shah NP, Sitlinger A, Blalock DV, Whitney C, Kikuchi R, Bosworth HB, Crowley MJ, Goldstein KM, Klem I, Oeffinger KC, Dent S. Cardiometabolic Comorbidities in Cancer Survivors. JACC CardioOncol 2022; 4:149-165. [PMID: 35818559 PMCID: PMC9270612 DOI: 10.1016/j.jaccao.2022.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/28/2022] Open
Abstract
There are nearly 17 million cancer survivors in the United States, including those who are currently receiving cancer therapy with curative intent and expected to be long-term survivors, as well as those with chronic cancers such as metastatic disease or chronic lymphocytic leukemia, who will receive cancer therapy for many years. Current clinical practice guidelines focus on lifestyle interventions, such as exercise and healthy eating habits, but generally do not address management strategies for clinicians or strategies to increase adherence to medications. We discuss 3 cardiometabolic comorbidities among cancer survivors and present the prevalence of comorbidities prior to a cancer diagnosis, treatment of comorbidities during cancer therapy, and management considerations of comorbidities in long-term cancer survivors or those on chronic cancer therapy. Approaches to support medication adherence and potential methods to enhance a team approach to optimize care of the individual with cancer across the continuum of disease are discussed. Cancer survivors are at increased risk for several chronic conditions, including hypertension, dyslipidemia, and diabetes. Determining optimal management of comorbidities for patients with cancer is critical. A multidisciplinary care approach is recommended throughout the continuum of active cancer treatment and survivorship. Survivorship research should focus on medication adherence and coordination of care.
Collapse
Affiliation(s)
- Leah L. Zullig
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Address for correspondence: Dr Leah Zullig, Duke University, 411 West Chapel Hill Street, Suite 600, Durham, North Carolina 27701, USA. @LeahZullig
| | - Anthony D. Sung
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Michel G. Khouri
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Shelley Jazowski
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA
| | - Nishant P. Shah
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Andrea Sitlinger
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Dan V. Blalock
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
| | - Colette Whitney
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
- Cascades East–Oregon Health and Science University, Klamath Falls, Oregon, USA
| | - Robin Kikuchi
- Keck School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Hayden B. Bosworth
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
| | - Matthew J. Crowley
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Karen M. Goldstein
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Igor Klem
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kevin C. Oeffinger
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Susan Dent
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
4
|
Ates N, Caglayan A, Balcikanli Z, Sertel E, Beker MC, Dilsiz P, Caglayan AB, Celik S, Dasdelen MF, Caglayan B, Yigitbasi T, Ozbek H, Doeppner TR, Hermann DM, Kilic E. Phosphorylation of PI3K/Akt at Thr308, but not MAPK kinase, mediates lithium-induced neuroprotection against cerebral ischemia in mice. Exp Neurol 2022; 351:113996. [PMID: 35122865 DOI: 10.1016/j.expneurol.2022.113996] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/31/2021] [Accepted: 01/27/2022] [Indexed: 11/30/2022]
Abstract
Lithium, in addition to its effect on acute and long-term bipolar disorder, is involved in neuroprotection after ischemic stroke. Yet, its mechanism of action is still poorly understood, which was only limited to its modulatory effect on GSK pathway. Therefore, we initially analyzed the dose-dependent effects of lithium on neurological deficits, infarct volume, brain edema and blood-brain barrier integrity, along with neuronal injury and survival in mice subjected to focal cerebral ischemia. Thereafter, we investigated the involvement of the PI3K/Akt and MEK signal transduction pathways and their components. Our observations revealed that 2 mmol/kg lithium significantly improved post-ischemic brain tissue survival. Although, 2 mmol/kg lithium had no negative effect on brain microcirculation, 5 and 20 mmol/kg lithium reduced brain perfusion. Furthermore, supratherapeutic dose of lithium in 20 mmol/kg lead to animal death. In addition, improvement of brain perfusion with L-arginine, did not change the effect of 5 mmol/kg lithium on brain injury. Additionally, post-stroke blood-brain barrier leakage, hemodynamic impairment and apoptosis have been reversed by lithium treatment. Interestingly, lithium-induced neuroprotection was associated with increased phosphorylation of Akt at Thr308 and suppressed GSK-3β phosphorylation at Ser9 residue. Lithium upregulated Erk-2 and downregulated JNK-2 phosphorylation. To distinguish whether neuroprotective effects of lithium are modulated by PI3K/Akt or MEK, we sequentially blocked these pathways and demonstrated that the neuroprotective activity of lithium persisted during MEK/ERK inhibition, whereas PI3K/Akt inhibition abolished neuroprotection. Collectively, we demonstrated lithium exerts its post-stroke neuroprotective activity via the PI3K/Akt pathway, specifically via Akt phosphorylation at Thr308, but not via MEK/ERK.
Collapse
Affiliation(s)
- Nilay Ates
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Pharmacology, Istanbul, Turkey
| | - Aysun Caglayan
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Zeynep Balcikanli
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Elif Sertel
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Mustafa Caglar Beker
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Pelin Dilsiz
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Pharmacology, Istanbul, Turkey
| | - Ahmet Burak Caglayan
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Süleyman Celik
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Muhammed Furkan Dasdelen
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Berrak Caglayan
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, International School of Medicine, Dept. of Medical Biology, Istanbul, Turkey
| | - Türkan Yigitbasi
- Istanbul Medipol University, Faculty of Medicine, Dept. of Biochemistry, Istanbul, Turkey
| | - Hanefi Ozbek
- Istanbul Medipol University, Faculty of Medicine, Dept. of Pharmacology, Istanbul, Turkey
| | - Thorsten Roland Doeppner
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
| | - Dirk Matthias Hermann
- University Hospital Essen, University of Duisburg-Essen, Department of Neurology, Essen, Germany
| | - Ertugrul Kilic
- Istanbul Medipol University, Regenerative and Restorative Medical Research Center, Istanbul, Turkey; Istanbul Medipol University, Faculty of Medicine, Dept. of Physiology, Istanbul, Turkey.
| |
Collapse
|
5
|
Therapeutic potential of nitric oxide synthase inhibitor from natural sources for the treatment of ischemic stroke. Saudi J Biol Sci 2022; 29:984-991. [PMID: 35197767 PMCID: PMC8848027 DOI: 10.1016/j.sjbs.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/03/2021] [Accepted: 10/03/2021] [Indexed: 11/28/2022] Open
Abstract
Nitric oxide (NO) is one of the major signalling molecules in the mammalian body playing critical role in regulation of blood pressure, cardiovascular disease including stroke, immune activation, neuronal and cell communication. Moreover, hyper production of NO by the activity of nitric oxide synthase (NOS) involved in neuropathic pain, neurodegenerative disorders and stroke. Hence, the search on small molecules from the natural sources for the inhibition of NOS is desirable in therapeutic point of view. The elevated level of NO caused by NOS enzyme become a novel target in finding new inhibitors from natural sources as antistroke agents. The present study focuses on the molecular docking of quercetin and its analogues against NOS. The active site of the enzyme was docked with the ligand and pharmacological properties were analysed. From this result, we suggest the therapeutic property of quercetin and its analogues against NOS.
Collapse
|
6
|
Bonnin P, Kubis N, Charriaut-Marlangue C. Collateral Supply in Preclinical Cerebral Stroke Models. Transl Stroke Res 2021; 13:512-527. [PMID: 34797519 PMCID: PMC9232412 DOI: 10.1007/s12975-021-00969-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 02/01/2023]
Abstract
Enhancing the collateral blood supply during the acute phase of cerebral ischemia may limit both the extension of the core infarct, by rescuing the penumbra area, and the degree of disability. Many imaging techniques have been applied to rodents in preclinical studies, to evaluate the magnitude of collateral blood flow and the time course of responses during the early phase of ischemic stroke. The collateral supply follows several different routes at the base of the brain (the circle of Willis) and its surface (leptomeningeal or pial arteries), corresponding to the proximal and distal collateral pathways, respectively. In this review, we describe and illustrate the cerebral collateral systems and their modifications following pre-Willis or post-Willis occlusion in rodents. We also review the potential pharmaceutical agents for stimulating the collateral blood supply tested to date. The time taken to establish a collateral blood flow supply through the leptomeningeal anastomoses differs between young and adult animals and between different species and genetic backgrounds. Caution is required when transposing preclinical findings to humans, and clinical trials must be performed to check the added value of pharmacological agents for stimulating the collateral blood supply at appropriate time points. However, collateral recruitment appears to be a rapid, beneficial, endogenous mechanism that can be stimulated shortly after artery occlusion. It should be considered a treatment target for use in addition to recanalization strategies.
Collapse
Affiliation(s)
- Philippe Bonnin
- APHP, Physiologie Clinique - Explorations Fonctionnelles, Hôpital Lariboisiere, Université de Paris, 2 rue Ambroise Paré, F-75010, Paris, France. .,INSERM U1148, LVTS, Hôpital Bichat, Université de Paris, F-75018, Paris, France.
| | - Nathalie Kubis
- APHP, Physiologie Clinique - Explorations Fonctionnelles, Hôpital Lariboisiere, Université de Paris, 2 rue Ambroise Paré, F-75010, Paris, France.,INSERM U1148, LVTS, Hôpital Bichat, Université de Paris, F-75018, Paris, France
| | | |
Collapse
|
7
|
Casili G, Ardizzone A, Basilotta R, Lanza M, Filippone A, Paterniti I, Esposito E, Campolo M. The Protective Role of Prolyl Oligopeptidase (POP) Inhibition in Kidney Injury Induced by Renal Ischemia-Reperfusion. Int J Mol Sci 2021; 22:11886. [PMID: 34769337 PMCID: PMC8584363 DOI: 10.3390/ijms222111886] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022] Open
Abstract
Ischemia/reperfusion injury (IRI) is a complex pathophysiological process characterized by blood circulation disorder caused by various factors, such as traumatic shock, surgery, organ transplantation, and thrombus. Severe metabolic dysregulation and tissue structure destruction are observed upon restoration of blood flow to the ischemic tissue. The kidney is a highly perfused organ, sensitive to ischemia and reperfusion injury, and the incidence of renal IRI has high morbidity and mortality. Several studies showed that infiltration of inflammatory cells, apoptosis, and angiogenesis are important mechanisms involved in renal IRI. Despite advances in research, effective therapies for renal IRI are lacking. Recently it has been demonstrated the role of KYP2047, a selective inhibitor of prolyl oligopeptidase (POP), in the regulation of inflammation, apoptosis, and angiogenesis. Thus, this research focused on the role of POP in kidney ischemia/reperfusion (KI/R). An in vivo model of KI/R was performed and mice were subjected to KYP2047 treatment (intraperitoneal, 0.5, 1 and 5 mg/kg). Histological analysis, Masson's trichrome and periodic acid shift (PAS) staining, immunohistochemical and Western blots analysis, real-time PCR (RT-PCR) and ELISA were performed on kidney samples. Moreover, serum creatinine and blood urea nitrogen (BUN) were quantified. POP-inhibition by KYP2047 treatment, only at the doses of 1 and 5 mg/kg, significantly reduced renal injury and collagen amount, regulated inflammation through canonical and non-canonical NF-κB pathway, and restored renal function. Moreover, KYP2047 modulated angiogenesis markers, such as TGF-β and VEGF, also slowing down apoptosis. Interestingly, treatment with KYP2047 modulated PP2A activity. Thus, these findings clarified the role of POP inhibition in AKI, also offering novel therapeutic target for renal injury after KI/R.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31-98166 Messina, Italy; (G.C.); (A.A.); (R.B.); (M.L.); (A.F.); (I.P.); (M.C.)
| | | |
Collapse
|
8
|
Wierońska JM, Cieślik P, Kalinowski L. Nitric Oxide-Dependent Pathways as Critical Factors in the Consequences and Recovery after Brain Ischemic Hypoxia. Biomolecules 2021; 11:biom11081097. [PMID: 34439764 PMCID: PMC8392725 DOI: 10.3390/biom11081097] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Brain ischemia is one of the leading causes of disability and mortality worldwide. Nitric oxide (NO•), a molecule that is involved in the regulation of proper blood flow, vasodilation, neuronal and glial activity constitutes the crucial factor that contributes to the development of pathological changes after stroke. One of the early consequences of a sudden interruption in the cerebral blood flow is the massive production of reactive oxygen and nitrogen species (ROS/RNS) in neurons due to NO• synthase uncoupling, which leads to neurotoxicity. Progression of apoptotic or necrotic neuronal damage activates reactive astrocytes and attracts microglia or lymphocytes to migrate to place of inflammation. Those inflammatory cells start to produce large amounts of inflammatory proteins, including pathological, inducible form of NOS (iNOS), which generates nitrosative stress that further contributes to brain tissue damage, forming vicious circle of detrimental processes in the late stage of ischemia. S-nitrosylation, hypoxia-inducible factor 1α (HIF-1α) and HIF-1α-dependent genes activated in reactive astrocytes play essential roles in this process. The review summarizes the roles of NO•-dependent pathways in the early and late aftermath of stroke and treatments based on the stimulation or inhibition of particular NO• synthases and the stabilization of HIF-1α activity.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Paulina Cieślik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Biobank Fahrenheit BBMRI.pl, Medical University of Gdansk, Debinki Street 7, 80-211 Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Debinki Street 7, 80-211 Gdansk, Poland
- BioTechMed Center/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza 11/12, 80-223 Gdansk, Poland
- Correspondence: ; Tel.: +48-58-349-1182
| |
Collapse
|
9
|
Pendharkar AV, Smerin D, Gonzalez L, Wang EH, Levy S, Wang S, Ishizaka S, Ito M, Uchino H, Chiang T, Cheng MY, Steinberg GK. Optogenetic Stimulation Reduces Neuronal Nitric Oxide Synthase Expression After Stroke. Transl Stroke Res 2021; 12:347-356. [PMID: 32661768 PMCID: PMC7925487 DOI: 10.1007/s12975-020-00831-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/23/2023]
Abstract
Post-stroke optogenetic stimulation has been shown to enhance neurovascular coupling and functional recovery. Neuronal nitric oxide synthase (nNOS) has been implicated as a key regulator of the neurovascular response in acute stroke; however, its role in subacute recovery remains unclear. We investigated the expression of nNOS in stroke mice undergoing optogenetic stimulation of the contralesional lateral cerebellar nucleus (cLCN). We also examined the effects of nNOS inhibition on functional recovery using a pharmacological inhibitor targeting nNOS. Optogenetically stimulated stroke mice demonstrated significant improvement on the horizontal rotating beam task at post-stroke days 10 and 14. nNOS mRNA and protein expression was significantly and selectively decreased in the contralesional primary motor cortex (cM1) of cLCN-stimulated mice. The nNOS expression in cM1 was negatively correlated with improved recovery. nNOS inhibitor (ARL 17477)-treated stroke mice exhibited a significant functional improvement in speed at post-stroke day 10, when compared to stroke mice receiving vehicle (saline) only. Our results show that optogenetic stimulation of cLCN and systemic nNOS inhibition both produce functional benefits after stroke, and suggest that nNOS may play a maladaptive role in post-stroke recovery.
Collapse
Affiliation(s)
- Arjun V Pendharkar
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Smerin
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Lorenzo Gonzalez
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric H Wang
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Sabrina Levy
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephanie Wang
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Shunsuke Ishizaka
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Masaki Ito
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Haruto Uchino
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Terrance Chiang
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle Y Cheng
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA.
| | - Gary K Steinberg
- Department of Neurosurgery and Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
10
|
Aliena-Valero A, Baixauli-Martín J, Castelló-Ruiz M, Torregrosa G, Hervás D, Salom JB. Effect of uric acid in animal models of ischemic stroke: A systematic review and meta-analysis. J Cereb Blood Flow Metab 2021; 41:707-722. [PMID: 33210575 PMCID: PMC7983496 DOI: 10.1177/0271678x20967459] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Addition of uric acid (UA) to thrombolytic therapy, although safe, showed limited efficacy in improving patients' stroke outcome, despite alleged neuroprotective effects of UA in preclinical research. This systematic review assessed the effects of UA on brain structural and functional outcomes in animal models of ischemic stroke. We searched Medline, Embase and Web of Science to identify 16 and 14 eligible rodent studies for qualitative and quantitative synthesis, respectively. Range of evidence met 10 of a possible 13 STAIR criteria. Median (Q1, Q3) quality score was 7.5 (6, 10) on the CAMARADES 15-item checklist. For each outcome, we used standardised mean difference (SMD) as effect size and random-effects modelling. Meta-analysis showed that UA significantly reduced infarct size (SMD: -1.18; 95% CI [-1.47, -0.88]; p < 0.001), blood-brain barrier (BBB) impairment/oedema (SMD: -0.72; 95% CI [-0.97, -0.48]; p < 0.001) and neurofunctional deficit (SMD: -0.98; 95% CI [-1.32, -0.63]; p < 0.001). Overall, there was low to moderate between-study heterogeneity and sizeable publication bias. In conclusion, published rodent data suggest that UA improves outcome following ischemic stroke by reducing infarct size, improving BBB integrity and ameliorating neurofunctional condition. Specific recommendations are given for further high-quality preclinical research required to better inform clinical research.
Collapse
Affiliation(s)
- Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | | | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Valencia, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | - David Hervás
- Unidad de Bioestadística, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
11
|
Synergistic Network Pharmacology for Traditional Chinese Medicine Liangxue Tongyu Formula in Acute Intracerebral Hemorrhagic Stroke. Neural Plast 2021; 2021:8874296. [PMID: 33727915 PMCID: PMC7936909 DOI: 10.1155/2021/8874296] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/22/2020] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Background Nowadays, acute intracerebral hemorrhage stroke (AICH) still causes higher mortality. Liangxue Tongyu Formula (LXTYF), originating from a traditional Chinese medicine (TCM) prescription, is widely used as auxiliary treatment for AICH. Objective To dig into the multicomponent, multitarget, and multipathway mechanism of LXTYF on treating AICH via network pharmacology and RNA-seq. Methods Network pharmacology analysis was used by ingredient collection, target exploration and prediction, network construction, and Gene Ontology (GO) and KEGG analysis, with the Cytoscape software and ClusterProfiler package in R. The RNA-seq data of the AICH-rats were analyzed for differential expression and functional enrichments. Herb-Compound-Target-Pathway (H-C-T-P) network was shown to clarify the mechanism of LXTYF for AICH. Results 76 active ingredients (quercetin, Alanine, kaempferol, etc.) of LXTYF and 376 putative targets to alleviate AICH (PTGS2, PTGS1, ESR1, etc.) were successfully identified. The protein-protein interaction (PPI) network indicated the important role of STAT3. The functional enrichment of GO and KEGG pathway showed that LXTYF is most likely to influence MAPK and PI3K-Akt signaling pathways for AICH treatment. From the RNA-seq of AICH-rats, 583 differential mRNAs were identified and 14 of them were consistent with the putative targets of LXTYF for AICH treatment. The KEGG pathway enrichment also implied that the MAPK signaling pathway was the most correlated one among all the related signaling pathways. Many important targets with expression changes of LXTYF for AICH treatment and their related pathways are great markers of antioxidation, anti-inflammatory, antiapoptosis, and lowering blood pressure, which indicated that LXTYF may play mutiroles in the mechanisms for AICH treatment. Conclusion The LXTYF attenuates AICH partially by antioxidation, anti-inflammatory, and antiapoptosis and lowers blood pressure roles through regulating the targets involved MAPK, calcium, apoptosis, and TNF signaling pathway, which provide notable clues for further experimental validation.
Collapse
|
12
|
Dao VTV, Elbatreek MH, Fuchß T, Grädler U, Schmidt HHHW, Shah AM, Wallace A, Knowles R. Nitric Oxide Synthase Inhibitors into the Clinic at Last. Handb Exp Pharmacol 2021; 264:169-204. [PMID: 32797331 DOI: 10.1007/164_2020_382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The 1998 Nobel Prize in Medicine and Physiology for the discovery of nitric oxide, a nitrogen containing reactive oxygen species (also termed reactive nitrogen or reactive nitrogen/oxygen species) stirred great hopes. Clinical applications, however, have so far pertained exclusively to the downstream signaling of cGMP enhancing drugs such as phosphodiesterase inhibitors and soluble guanylate cyclase stimulators. All clinical attempts, so far, to inhibit NOS have failed even though preclinical models were strikingly positive and clinical biomarkers correlated perfectly. This rather casts doubt on our current way of target identification in drug discovery in general and our way of patient stratification based on correlating but not causal biomarkers or symptoms. The opposite, NO donors, nitrite and enhancing NO synthesis by eNOS/NOS3 recoupling in situations of NO deficiency, are rapidly declining in clinical relevance or hold promise but need yet to enter formal therapeutic guidelines, respectively. Nevertheless, NOS inhibition in situations of NO overproduction often jointly with enhanced superoxide (or hydrogen peroxide production) still holds promise, but most likely only in acute conditions such as neurotrauma (Stover et al., J Neurotrauma 31(19):1599-1606, 2014) and stroke (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019). Conversely, in chronic conditions, long-term inhibition of NOS might be too risky because of off-target effects on eNOS/NOS3 in particular for patients with cardiovascular risks or metabolic and renal diseases. Nitric oxide synthases (NOS) and their role in health (green) and disease (red). Only neuronal/type 1 NOS (NOS1) has a high degree of clinical validation and is in late stage development for traumatic brain injury, followed by a phase II safety/efficacy trial in ischemic stroke. The pathophysiology of NOS1 (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016) is likely to be related to parallel superoxide or hydrogen peroxide formation (Kleinschnitz et al., J Cereb Blood Flow Metab 1508-1512, 2016; Casas et al., Proc Natl Acad Sci U S A 114(46):12315-12320, 2017; Casas et al., Proc Natl Acad Sci U S A 116(14):7129-7136, 2019) leading to peroxynitrite and protein nitration, etc. Endothelial/type 3 NOS (NOS3) is considered protective only and its inhibition should be avoided. The preclinical evidence for a role of high-output inducible/type 2 NOS (NOS2) isoform in sepsis, asthma, rheumatic arthritis, etc. was high, but all clinical development trials in these indications were neutral despite target engagement being validated. This casts doubt on the role of NOS2 in humans in health and disease (hence the neutral, black coloring).
Collapse
Affiliation(s)
- Vu Thao-Vi Dao
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Thomas Fuchß
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Ulrich Grädler
- Takeda GmbH (former Nycomed/Altana Pharma), Konstanz, Germany
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht, The Netherlands
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Alan Wallace
- Health and Life Sciences, Coventry University, Coventry, UK
| | - Richard Knowles
- Knowles Consulting Ltd., The Stevenage Bioscience Catalyst, Stevenage, UK.
| |
Collapse
|
13
|
Underly RG, Shih AY. Rapid, Nitric Oxide Synthesis-Dependent Activation of MMP-9 at Pericyte Somata During Capillary Ischemia in vivo. Front Physiol 2021; 11:619230. [PMID: 33505320 PMCID: PMC7830159 DOI: 10.3389/fphys.2020.619230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/10/2020] [Indexed: 11/28/2022] Open
Abstract
Nitric oxide serves essential roles in normal vascular physiology, but paradoxically contributes to vascular pathology in disease. During brain ischemia, aberrant nitric oxide levels can cause cellular injury through induction of nitrosative/oxidative stress and post-translational activation of matrix-metalloproteinase-9 (MMP-9). We recently demonstrated that brain pericyte somata were associated with very early and localized MMP-9 activation along capillaries during cerebral ischemia, leading to focal blood-brain barrier disruption. Here, we tested whether this effect was dependent upon nitric oxide production. In vivo two-photon imaging was used to directly visualize MMP9 activity using a FITC-gelatin probe and leakage of intravenous dye during photothrombotically induced capillary ischemia. Results showed that the NOS inhibitor, L-NIL, at concentrations affecting both iNOS and constitutive NOS isoforms, attenuated capillary leakage at pericyte soma-specific locations and substantially reduced FITC-gelatin cleavage. We also found that combined administration of L-NIL and anisomycin, an inhibitor of protein synthesis, led to near complete elimination of FITC-gelatin cleavage and vascular leakage. These results indicate that both nitric oxide synthase and new protein synthesis are involved in the rapid activation of MMP-9 at somata of capillary pericytes during ischemia.
Collapse
Affiliation(s)
- Robert G Underly
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
| | - Andy Y Shih
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States.,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States.,Department of Bioengineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
14
|
Kaminski N, Köster C, Mouloud Y, Börger V, Felderhoff-Müser U, Bendix I, Giebel B, Herz J. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Reduce Neuroinflammation, Promote Neural Cell Proliferation and Improve Oligodendrocyte Maturation in Neonatal Hypoxic-Ischemic Brain Injury. Front Cell Neurosci 2020; 14:601176. [PMID: 33362471 PMCID: PMC7758466 DOI: 10.3389/fncel.2020.601176] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Neonatal encephalopathy caused by hypoxia-ischemia (HI) is a major cause of childhood mortality and disability. Stem cell-based regenerative therapies seem promising to prevent long-term neurological deficits. Our previous work in neonatal HI revealed an unexpected interaction between mesenchymal stem/stromal cells (MSCs) and the brains' microenvironment leading to an altered therapeutic efficiency. MSCs are supposed to mediate most of their therapeutic effects in a paracrine mode via extracellular vesicles (EVs), which might be an alternative to cell therapy. In the present study, we investigated the impact of MSC-EVs on neonatal HI-induced brain injury. Methods: Nine-day-old C57BL/6 mice were exposed to HI through ligation of the right common carotid artery followed by 1 h hypoxia (10% oxygen). MSC-EVs were injected intraperitoneally 1, 3, and 5 days after HI. One week after HI, brain injury was evaluated by regional neuropathological scoring, atrophy measurements and immunohistochemistry to assess effects on neuronal, oligodendrocyte and vessel densities, proliferation, oligodendrocyte maturation, myelination, astro-, and microglia activation. Immunohistochemistry analyses were complemented by mRNA expression analyses for a broad set of M1/M2- and A1/A2-associated molecules and neural growth factors. Results: While total neuropathological scores and tissue atrophy were not changed, MSC-EVs significantly protected from HI-induced striatal tissue loss and decreased micro- and astroglia activation. MSC-EVs lead to a significant downregulation of the pro-inflammatory cytokine TNFa, accompanied by a significant upregulation of the M2 marker YM-1 and the anti-inflammatory cytokine TGFb. MSC-EVs significantly decreased astrocytic expression of the A1 marker C3, concomitant with an increased expression of neural growth factors (i.e., BDNF, VEGF, and EGF). These alterations were associated with an increased neuronal and vessel density, coinciding with a significant increase of proliferating cells in the neurogenic sub-ventricular zone juxtaposed to the striatum. MSC-EV-mediated neuroprotection went along with a significant improvement of oligodendrocyte maturation and myelination. Conclusion: The present study demonstrates that MSC-EVs mediate anti-inflammatory effects, promote regenerative responses and improve key developmental processes in the injured neonatal brain. The present results suggest different cellular target mechanisms of MSC-EVs, preventing secondary HI-induced brain injury. MSC-EV treatment may be a promising alternative to risk-associated cell therapies in neonatal brain injury.
Collapse
Affiliation(s)
- Nicole Kaminski
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christian Köster
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Yanis Mouloud
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Verena Börger
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Müser
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, Neonatology and Experimental Perinatal Neurosciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
15
|
Cerebral Vasodilator Property of Poly(ADP-Ribose) Polymerase Inhibitor (PJ34) in the Neonatal and Adult Mouse Is Mediated by the Nitric Oxide Pathway. Int J Mol Sci 2020; 21:ijms21186569. [PMID: 32911782 PMCID: PMC7555622 DOI: 10.3390/ijms21186569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/28/2020] [Accepted: 09/04/2020] [Indexed: 12/29/2022] Open
Abstract
The poly(ADP-ribose) polymerase (PARP) inhibitor PJ34 has been reported to improve endothelial dysfunction in the peripheral system. We addressed the role of PJ34 on the vascular tone and vasoreactivity during development in the mouse brain. Blood flows were measured in the basilar trunk using ultrasonography. Cerebral vasoreactivity or vasodilation reserve was estimated as a percentage increase in mean blood flow velocities (mBFV) recorded under normoxia-hypercapnia in control and after PJ34 administration. Non-selective and selective eNOS and nNOS inhibitors were used to evaluate the role of NO-pathway into the hemodynamic effects of PJ34. PJ34 increased mBFVs from 15.8 ± 1.6 to 19.1 ± 1.9 cm/s (p = 0.0043) in neonatal, from 14.6 ± 1.4 to 16.1 ± 0.9 cm/s (p = 0.0049) in adult, and from 15.7 ± 1.7 to 17.5 ± 2.0 cm/s (p = 0.0024) in aged mice 48 h after administration. These PJ34 values were similar to those measured in age-matched control mice under normoxia-hypercapnia. This recruitment was mediated through the activation of constitutive NO synthases in both the neonatal (38.2 ± 6.7 nmol/min/mg protein) and adult (31.5 ± 4.4 nmol/min/mg protein) brain, as compared to age-matched control brain (6.9 ± 0.4 and 6.3 ± 0.7 nmol/min/mg protein), respectively. In addition, quite selective eNOS inhibitor was able to inhibit the recruitment. PJ34 by itself is able to increase cerebral blood flow through the NO-pathway activation at least over 48 h after a single administration.
Collapse
|
16
|
Beard DJ, Li Z, Schneider AM, Couch Y, Cipolla MJ, Buchan AM. Rapamycin Induces an eNOS (Endothelial Nitric Oxide Synthase) Dependent Increase in Brain Collateral Perfusion in Wistar and Spontaneously Hypertensive Rats. Stroke 2020; 51:2834-2843. [PMID: 32772681 DOI: 10.1161/strokeaha.120.029781] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND PURPOSE Rapamycin is a clinically approved mammalian target of rapamycin inhibitor that has been shown to be neuroprotective in animal models of stroke. However, the mechanism of rapamycin-induced neuroprotection is still being explored. Our aims were to determine if rapamycin improved leptomeningeal collateral perfusion, to determine if this is through eNOS (endothelial nitric oxide synthase)-mediated vessel dilation and to determine if rapamycin increases immediate postreperfusion blood flow. METHODS Wistar and spontaneously hypertensive rats (≈14 weeks old, n=22 and n=15, respectively) were subjected to ischemia by middle cerebral artery occlusion (90 and 120 minutes, respectively) with or without treatment with rapamycin at 30-minute poststroke. Changes in middle cerebral artery and collateral perfusion territories were measured by dual-site laser Doppler. Reactivity to rapamycin was studied using isolated and pressurized leptomeningeal anastomoses. Brain injury was measured histologically or with triphenyltetrazolium chloride staining. RESULTS In Wistar rats, rapamycin increased collateral perfusion (43±17%), increased reperfusion cerebral blood flow (16±8%) and significantly reduced infarct volume (35±6 versus 63±8 mm3, P<0.05). Rapamycin dilated leptomeningeal anastomoses by 80±9%, which was abolished by nitric oxide synthase inhibition. In spontaneously hypertensive rats, rapamycin increased collateral perfusion by 32±25%, reperfusion cerebral blood flow by 44±16%, without reducing acute infarct volume 2 hours postreperfusion. Reperfusion cerebral blood flow was a stronger predictor of brain damage than collateral perfusion in both Wistar and spontaneously hypertensive rats. CONCLUSIONS Rapamycin increased collateral perfusion and reperfusion cerebral blood flow in both Wistar and comorbid spontaneously hypertensive rats that appeared to be mediated by enhancing eNOS activation. These findings suggest that rapamycin may be an effective acute therapy for increasing collateral flow and as an adjunct therapy to thrombolysis or thrombectomy to improve reperfusion blood flow.
Collapse
Affiliation(s)
- Daniel J Beard
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, United Kingdom (D.J.B., A.M.S., Y.C., A.M.B.)
- School of Biomedical Science and Pharmacy, The University of Newcastle, Australia (D.J.B.)
| | - Zhaojin Li
- Department of Neurological Sciences, The University of Vermont, Burlington (Z.L., M.J.C.)
| | - Anna M Schneider
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, United Kingdom (D.J.B., A.M.S., Y.C., A.M.B.)
| | - Yvonne Couch
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, United Kingdom (D.J.B., A.M.S., Y.C., A.M.B.)
| | - Marilyn J Cipolla
- Department of Neurological Sciences, The University of Vermont, Burlington (Z.L., M.J.C.)
| | - Alastair M Buchan
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, United Kingdom (D.J.B., A.M.S., Y.C., A.M.B.)
| |
Collapse
|
17
|
Ma S, Liu X, Cheng B, Jia Z, Hua H, Xin Y. Chemical characterization of polysaccharides isolated from scrophularia ningpoensis and its protective effect on the cerebral ischemia/reperfusin injury in rat model. Int J Biol Macromol 2019; 139:955-966. [DOI: 10.1016/j.ijbiomac.2019.08.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/12/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022]
|
18
|
Strohm E, Herzner G, Ruther J, Kaltenpoth M, Engl T. Nitric oxide radicals are emitted by wasp eggs to kill mold fungi. eLife 2019; 8:e43718. [PMID: 31182189 PMCID: PMC6559793 DOI: 10.7554/elife.43718] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/05/2019] [Indexed: 12/11/2022] Open
Abstract
Detrimental microbes caused the evolution of a great diversity of antimicrobial defenses in plants and animals. Insects developing underground seem particularly threatened. Here we show that the eggs of a solitary digger wasp, the European beewolf Philanthus triangulum, emit large amounts of gaseous nitric oxide (NO⋅) to protect themselves and their provisions, paralyzed honeybees, against mold fungi. We provide evidence that a NO-synthase (NOS) is involved in the generation of the extraordinary concentrations of nitrogen radicals in brood cells (~1500 ppm NO⋅ and its oxidation product NO2⋅). Sequencing of the beewolf NOS gene revealed no conspicuous differences to related species. However, due to alternative splicing, the NOS-mRNA in beewolf eggs lacks an exon near the regulatory domain. This preventive external application of high doses of NO⋅ by wasp eggs represents an evolutionary key innovation that adds a remarkable novel facet to the array of functions of the important biological effector NO⋅.
Collapse
Affiliation(s)
- Erhard Strohm
- Evolutionary Ecology Group, Institute of ZoologyUniversity of RegensburgRegensburgGermany
| | - Gudrun Herzner
- Evolutionary Ecology Group, Institute of ZoologyUniversity of RegensburgRegensburgGermany
| | - Joachim Ruther
- Chemical Ecology Group, Institute of ZoologyUniversity of RegensburgRegensburgGermany
| | - Martin Kaltenpoth
- Evolutionary Ecology Group, Institute of ZoologyUniversity of RegensburgRegensburgGermany
- Insect Symbiosis Research GroupMax Planck Institute for Chemical EcologyJenaGermany
| | - Tobias Engl
- Evolutionary Ecology Group, Institute of ZoologyUniversity of RegensburgRegensburgGermany
- Insect Symbiosis Research GroupMax Planck Institute for Chemical EcologyJenaGermany
| |
Collapse
|
19
|
Chaturvedi P, Mehrotra V, Saxena Y, Manna S. Correlation of Serum Nitric Oxide (NO) with Glasgow Coma Scale (GCS) in Acute Ischemic Stroke Patient: A Study in North India. Indian J Clin Biochem 2018; 33:322-327. [PMID: 30072832 DOI: 10.1007/s12291-017-0677-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 06/26/2017] [Indexed: 11/28/2022]
Abstract
Nitric oxide (NO) is one of the key players in the pathogenesis of ischemic stroke. Limited reports are available about the serum level of NO and their correlation with Glasgow Coma Scale (GCS) score in acute (<24 h) ischemic stroke (AIS) patients. A hospital based, cross sectional study was done in North Indian population to find out correlation of serum NO and GCS in AIS patients. 50 patients of AIS and 25 healthy controls were chosen for the study. Serum NO level was measured by ELISA and GCS scores were assessed by a neurologist. Pearson correlation coefficients were analyzed to look for the relationship between NO and GCS. Statistically highly significant elevation in mean serum NO level was observed in cases as compared to controls (p < 0.01). A negative correlation of NO levels with neurological score of GCS r (48) = -0.144, p > 0.05 was seen. This indicates oxidative stress in acute ischemic stroke may be the result of imbalance in oxidant/antioxidant homeostasis.
Collapse
Affiliation(s)
- Priti Chaturvedi
- Department of Biochemistry, Swami Rama Himalyan University, Dehradun, Uttarakhand India
| | - Vinit Mehrotra
- Department of Biochemistry, Swami Rama Himalyan University, Dehradun, Uttarakhand India
| | - Yogesh Saxena
- 2Department of Physiology, Swami Rama Himalayan University, Dehradun, Uttarakhand India
| | - Soumen Manna
- 2Department of Physiology, Swami Rama Himalayan University, Dehradun, Uttarakhand India
| |
Collapse
|
20
|
Nash KM, Schiefer IT, Shah ZA. Development of a reactive oxygen species-sensitive nitric oxide synthase inhibitor for the treatment of ischemic stroke. Free Radic Biol Med 2018; 115:395-404. [PMID: 29275014 PMCID: PMC11970191 DOI: 10.1016/j.freeradbiomed.2017.12.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 01/25/2023]
Abstract
Ischemic stroke is caused by a blockage of cerebral blood flow resulting in neuronal and glial hypoxia leading to inflammatory and reactive oxygen species (ROS)-mediated cell death. Nitric oxide (NO) formed by NO synthase (NOS) is known to be protective in ischemic stroke, however NOS has been shown to 'uncouple' under oxidative conditions to instead produce ROS. Nitrones are antioxidant molecules that are shown to trap ROS to then decompose and release NO. In this study, the nitrone 5 was designed such that its decomposition product is a NOS inhibitor, 6, effectively leading to NOS inhibition specifically at the site of ROS production. The ability of 5 to spin-trap radicals and decompose to 6 was observed using EPR and LC-MS/MS. The pro-drug concept was tested in vitro by measuring cell viability and 6 formation in SH-SY5Y cells subjected to oxygen glucose deprivation (OGD). 5 was found to be more efficacious and more potent than PBN, and was able to increase phospho-Akt while reducing nitrotyrosine and cleaved caspase-3 levels. 6 treatment, but not 5, was found to decrease NO production in LPS-stimulated microglia. Doppler flowmetry on anesthetized mice showed increased cerebral blood flow upon intravenous administration of 1mg/kg of 5, but a return to baseline upon administration of 10mg/kg, likely due to its dual nature of antioxidant/NO-donor and NOS-inhibition. Mice treated with 5 after permanent ischemia exhibited a >30% reduction in infarct volume, and higher formation of 6 in ischemic tissue resulting in region specific effects limited to the infarct area.
Collapse
Affiliation(s)
- Kevin M Nash
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, USA
| | - Isaac T Schiefer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA.
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA.
| |
Collapse
|
21
|
Bhowmick S, Drew KL. Arctic ground squirrel resist peroxynitrite-mediated cell death in response to oxygen glucose deprivation. Free Radic Biol Med 2017; 113:203-211. [PMID: 28962873 PMCID: PMC5699938 DOI: 10.1016/j.freeradbiomed.2017.09.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/01/2017] [Accepted: 09/25/2017] [Indexed: 12/13/2022]
Abstract
Cerebral ischemia-reperfusion (I/R) injury initiates a cascade of events, generating nitric oxide (NO) and superoxide(O2•-) to form peroxynitrite (ONOO-), a potent oxidant. Arctic ground squirrels (AGS; Urocitellus parryii) show high tolerance to I/R injury. However, the underlying mechanism remains elusive. We hypothesize that tolerance to I/R modeled in an acute hippocampal slice preparation in AGS is modulated by reduced oxidative and nitrative stress. Hippocampal slices (400µm) from rat and AGS were subjected to oxygen glucose deprivation (OGD) using a novel microperfusion technique. Slices were exposed to NO, O2.- donors with and without OGD; pretreatment with inhibitors of NO, O2.- and ONOO- followed by OGD. Perfusates collected every 15min were analyzed for LDH release, a marker of cell death. 3-nitrotyrosine (3NT) and 4-hydroxynonenal (4HNE) were measured to assess oxidative and nitrative stress. Results show that NO/O2.- alone is not sufficient to cause ischemic-like cell death, but with OGD enhances cell death more in rat than in AGS. A NOS inhibitor, SOD mimetic and ONOO- inhibitor attenuates OGD injury in rat but has no effect in AGS. Rats also show a higher level of 3NT and 4HNE with OGD than AGS suggesting the greater level of injury in rat is via formation of ONOO-.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, AK, USA; Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - Kelly L Drew
- Department of Chemistry and Biochemistry, University of Alaska Fairbanks, Fairbanks, AK, USA; Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, USA.
| |
Collapse
|
22
|
Abstract
Stroke is considered to be an acute cerebrovascular disease, including ischemic stroke and hemorrhagic stroke. The high incidence and poor prognosis of stroke suggest that it is a highly disabling and highly lethal disease which can pose a serious threat to human health. Nitric oxide (NO), a common gas in nature, which is often thought as a toxic gas, because of its intimate relationship with the pathological processes of many diseases, especially in the regulation of blood flow and cell inflammation. However, recent years have witnessed an increased interest that NO plays a significant and positive role in stroke as an essential gas signal molecule. In view of the fact that the neuroprotective effect of NO is closely related to its concentration, cell type and time, only in the appropriate circumstances can NO play a protective effect. The purpose of this review is to summarize the roles of NO in ischemic stroke and hemorrhagic stroke.
Collapse
Affiliation(s)
- Zhou-Qing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ru-Tao Mou
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Dong-Xia Feng
- Department of Scott & White Clinic-Temple, Temple, TX, USA
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
23
|
Salehpour M, Khodagholi F, Zeinaddini Meymand A, Nourshahi M, Ashabi G. Exercise training with concomitant nitric oxide synthase inhibition improved anxiogenic behavior, spatial cognition, and BDNF/P70S6 kinase activation in 20-month-old rats. Appl Physiol Nutr Metab 2017; 43:45-53. [PMID: 28854337 DOI: 10.1139/apnm-2017-0313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This study aimed to investigate the effect of exercise and nitric oxide synthase (NOS) inhibition on memory, anxiety, and protein levels of brain-derived neurotrophic factor (BDNF) and P70S6 kinase (P70S6K). Twenty-month-old rats were divided into 6 groups: a control group, 2 groups treated with l-nitro-arginine methyl ester (L-NAME) (25 or 100 mg/kg) for 63 days, 2 groups treated with L-NAME (25 or 100 mg/kg) for 63 days plus 2 months of exercise, and 1 group treated with exercise. Behavioral tests were conducted to determine the anxiolytic and memory-improving role of exercise and NOS inhibition. BDNF, P70S6K, and cleaved caspase-3 protein levels in the hippocampus and prefrontal cortex were evaluated by Western blotting. Exercise and L-NAME (25 mg/kg) or their combination had an anxiolytic effect and improved spatial memory in old rats compared with the control or exercised group, respectively. Exercise and treatment with a low dose of L-NAME (25 mg/kg) each increased BDNF and P70S6K in the hippocampus and prefrontal cortex compared with levels in control rats. In comparison with exercise alone, co-treatment with exercise and a low dose of L-NAME (25 mg/kg) also increased BDNF and P70S6K in the hippocampus. The neuronal level of cleaved caspase-3 was reduced in the L-NAME (25 mg/kg) + exercise group compared with the exercised group. The L-NAME (100 mg/kg) + exercise treatment had no positive behavioral or molecular effects compared with exercise alone. The protective role of NOS inhibition and aerobic exercise against aging is probably modulated via BDNF and P70S6K in the brain.
Collapse
Affiliation(s)
- Mojtaba Salehpour
- a Department of Sport Physiology, Faculty of Sport Sciences, Shahid Rajaee Teacher Training University, PO box 16875-163, Tehran, Iran
| | - Fariba Khodagholi
- b NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, PO box 19615-1178, Tehran, Iran
| | - Arman Zeinaddini Meymand
- c Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, PO box 19615-1178, Tehran, Iran
| | - Maryam Nourshahi
- d Department of Sport Physiology, Faculty of Physical Education and Sport Sciences, Shahid Beheshti University, PO box 19839-6113, Tehran, Iran
| | - Ghorbangol Ashabi
- e Department of Physiology, Faculty of Medicine, Tehran University of Medical Sciences, PO box 141761-3151, Tehran, Iran
| |
Collapse
|
24
|
|
25
|
Vogt L, Reichlin TS, Nathues C, Würbel H. Authorization of Animal Experiments Is Based on Confidence Rather than Evidence of Scientific Rigor. PLoS Biol 2016; 14:e2000598. [PMID: 27911892 PMCID: PMC5135031 DOI: 10.1371/journal.pbio.2000598] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/03/2016] [Indexed: 11/20/2022] Open
Abstract
Accumulating evidence indicates high risk of bias in preclinical animal research, questioning the scientific validity and reproducibility of published research findings. Systematic reviews found low rates of reporting of measures against risks of bias in the published literature (e.g., randomization, blinding, sample size calculation) and a correlation between low reporting rates and inflated treatment effects. That most animal research undergoes peer review or ethical review would offer the possibility to detect risks of bias at an earlier stage, before the research has been conducted. For example, in Switzerland, animal experiments are licensed based on a detailed description of the study protocol and a harm-benefit analysis. We therefore screened applications for animal experiments submitted to Swiss authorities (n = 1,277) for the rates at which the use of seven basic measures against bias (allocation concealment, blinding, randomization, sample size calculation, inclusion/exclusion criteria, primary outcome variable, and statistical analysis plan) were described and compared them with the reporting rates of the same measures in a representative sub-sample of publications (n = 50) resulting from studies described in these applications. Measures against bias were described at very low rates, ranging on average from 2.4% for statistical analysis plan to 19% for primary outcome variable in applications for animal experiments, and from 0.0% for sample size calculation to 34% for statistical analysis plan in publications from these experiments. Calculating an internal validity score (IVS) based on the proportion of the seven measures against bias, we found a weak positive correlation between the IVS of applications and that of publications (Spearman's rho = 0.34, p = 0.014), indicating that the rates of description of these measures in applications partly predict their rates of reporting in publications. These results indicate that the authorities licensing animal experiments are lacking important information about experimental conduct that determines the scientific validity of the findings, which may be critical for the weight attributed to the benefit of the research in the harm-benefit analysis. Similar to manuscripts getting accepted for publication despite poor reporting of measures against bias, applications for animal experiments may often be approved based on implicit confidence rather than explicit evidence of scientific rigor. Our findings shed serious doubt on the current authorization procedure for animal experiments, as well as the peer-review process for scientific publications, which in the long run may undermine the credibility of research. Developing existing authorization procedures that are already in place in many countries towards a preregistration system for animal research is one promising way to reform the system. This would not only benefit the scientific validity of findings from animal experiments but also help to avoid unnecessary harm to animals for inconclusive research.
Collapse
Affiliation(s)
- Lucile Vogt
- Division of Animal Welfare, Veterinary Public Health Institute, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Thomas S. Reichlin
- Division of Animal Welfare, Veterinary Public Health Institute, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Christina Nathues
- Division of VPH-Epidemiology, Veterinary Public Health Institute, Vetsuisse Faculty, University of Bern, Liebefeld, Switzerland
| | - Hanno Würbel
- Division of Animal Welfare, Veterinary Public Health Institute, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
26
|
Abstract
An adverse outcome is still encountered in 45% of full-term neonates with perinatal asphyxia who are treated with moderate hypothermia. At present pharmacologic therapies are developed to be added to hypothermia. In the present article, these potential neuroprotective interventions are described based on the molecular pathways set in motion during fetal hypoxia and following reoxygenation and reperfusion after birth. These pathways include excessive production of excitotoxins with subsequent over-stimulation of NMDA receptors and calcium influx in neuronal cells, excessive production of reactive oxygen and nitrogen species, activation of inflammation leading to inappropriate apoptosis, and loss of neurotrophic factors. Possibilities for pharmacologic combination therapy, where each drug will be administered based on the optimal point of time in the cascade of destructive molecular reactions, may further reduce brain damage due to perinatal asphyxia.
Collapse
Affiliation(s)
- Frank van Bel
- Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands.
| | - Floris Groenendaal
- Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| |
Collapse
|
27
|
Chang CZ, Wu SC. 4'-O-β-D-Glucosyl-5-O-Methylvisamminol, A Natural Histone H3 Phosphorylation Epigenetic Suppressor, Exerts a Neuroprotective Effect Through PI3K/Akt Signaling Pathway on Focal Cerebral Ischemia in Rats. World Neurosurg 2016; 89:474-88. [PMID: 26868427 DOI: 10.1016/j.wneu.2016.01.061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND A bursting inflammation has been observed that compromises neurologic function in patients who experience stroke. We sought to examine the neuroprotective efficacy of 4'-O-β-D-glucosyl-5-O-methylvisamminol (OGOMV), a novel histone H3 phosphorylation epigenetic suppressor) in a transient middle cerebral artery occlusion (tMCAO). METHODS A rodent tMCAO model was used. Administration with 400 μg/kg/day OGOMV was initiated 12 hours before (prevention) and 1 hour after animals were subjected to tMCAO (reversal). The cerebral cortex was harvested to examine protein kinase B (PI3D/Akt), 5-bromo-2'-deoxyuridine (Western blot), and caspases (reverse-transcription polymerase chain reaction). In addition, cerebrospinal fluid samples were collected to examine interleukin 1-β, interleukin-6, monocyte chemoattractant protein-1, and tumor necrosis factor-α (reverse-transcription polymerase chain reaction). RESULTS Cortical 5-bromo-2'-deoxyuridine and phospho-PI3D/Akt were reduced in tMCAO animals, compared with the healthy controls but increased in the OGOMV treatment and prevention groups. Activated cortical caspase-3,-6, and -9a as well as increased IL-1β and TNF-α levels were observed in the tMCAO animals (P < 0.05). Both prevention and treatment with OGOMV significantly reduced cleaved caspase-3 and -9a groups, but no significant change in caspase-6 was noted. Perifosine, an Akt inhibitor, was added to reduce the bioexpression of phospho-P13D/Akt, and Bcl-2 level and increased cleaved caspase-9a level in both OGOMV prevention and treatment tMCAO groups (P > 0.05). CONCLUSION Our study suggests that OGOMV could exert a neuroprotective effect by inhibiting the P13D/Akt protein, attenuating inflammation, and cleaved caspase-3- and -9a-related apoptosis. This study also lends credence to support the notion that the prevention of OGOMV could attenuate proinflammatory cytokine mRNA and late-onset caspases in tMCAO and merits further study.
Collapse
Affiliation(s)
- Chih-Zen Chang
- Department of Surgery, Faculty of Medicine, School of Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Surgery, Kaohsiung Municipal Ta Tung Hospital, Kaohsiung, Taiwan.
| | - Shu-Chuan Wu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
28
|
Li Y, Liu K, Kang ZM, Sun XJ, Liu WW, Mao YF. Helium preconditioning protects against neonatal hypoxia-ischemia via nitric oxide mediated up-regulation of antioxidases in a rat model. Behav Brain Res 2015; 300:31-7. [PMID: 26675888 DOI: 10.1016/j.bbr.2015.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/29/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
Abstract
This study aimed to investigate the role of nitric oxide (NO) in the neuroprotective effects of helium preconditioning (He-PC) in a neonatal hypoxia/ischemia (HI) rat model. Seven-day old rat pups were divided into normal control group, He-PC group, HI group, He-PC+HI group, L-NAME+HI group and L-NAME+He-PC+HI group. HI was induced by exposure to 80% oxygen for 90 min. He-PC was conducted with 70% helium-30% oxygen for three 5-min periods. Three hours after He-PC, animals in control group and He-PC group were sacrificed, and the brain was collected for the detection of NO content. At 24h after HI, animals in control group, HI group, He-PC+HI group, and L-NAME+He-PC+HI group were sacrificed, and the brain was collected for detection of infarct ratio, antioxidases (SOD, HO-1 and Nrf2), DNA binding activity of Nrf2 and TUNEL staining. Three weeks later, the neurological function and brain atrophy were determined. Results showed pretreatment with L-NAME alone failed to exert protective effect on HI. He-PC significantly increased NO content, reduced the brain infarct area, increased anti-oxidases expression and DNA binding activity of Nrf2, decreased the apoptotic cells, and improved the neurological function and brain atrophy. In addition, this protection was markedly inhibited by L-NAME (a non-selective NOS inhibitor). These findings suggest that the He-PC may induce NO production to activate Nrf2, exerting neuroprotective effect on neonatal HI.
Collapse
Affiliation(s)
- Y Li
- Department of Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No 1665 Kongjiang Road, Yangpu District, Shanghai 200092, China
| | - K Liu
- Department of Diving and Hyperbaric Medicine, Secondary Military Medical University, No 800, Xiangyin Road, Yangpu District, Shanghai 200433, China
| | - Z M Kang
- Department of Diving and Hyperbaric Medicine, Secondary Military Medical University, No 800, Xiangyin Road, Yangpu District, Shanghai 200433, China
| | - X J Sun
- Department of Diving and Hyperbaric Medicine, Secondary Military Medical University, No 800, Xiangyin Road, Yangpu District, Shanghai 200433, China
| | - W W Liu
- Department of Diving and Hyperbaric Medicine, Secondary Military Medical University, No 800, Xiangyin Road, Yangpu District, Shanghai 200433, China.
| | - Y F Mao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No 1665 Kongjiang Road, Yangpu District, Shanghai 200092, China.
| |
Collapse
|
29
|
Lapi D, Vagnani S, Sapio D, Mastantuono T, Boscia F, Pignataro G, Penna C, Pagliaro P, Colantuoni A. Effects of bone marrow mesenchymal stem cells (BM-MSCs) on rat pial microvascular remodeling after transient middle cerebral artery occlusion. Front Cell Neurosci 2015; 9:329. [PMID: 26379500 PMCID: PMC4548191 DOI: 10.3389/fncel.2015.00329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/10/2015] [Indexed: 11/13/2022] Open
Abstract
Previous studies have shown that the pial microcirculation remodeling improves neurological outcome after middle cerebral artery occlusion (MCAO), accompanied by higher expression of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS), modulating in vivo angiogenesis. This study was aimed to assess the effects of bone marrow mesenchymal stem cells (BM-MSCs) infused after MCAO on rat pial microcirculation. Animals were subjected to 2 h MCAO followed by BM-MSCs infusion into internal carotid artery. Pial microcirculation was observed at different reperfusion times by fluorescence microscopy. Geometric characteristics of arteriolar networks, permeability increase, leukocyte adhesion, perfused capillary density, VEGF, and endothelial nitric oxide synthase (e-NOS) expression were evaluated. Green fluorescent protein (GFP)-BM-MSCs were used to evaluate their distribution and cell phenotype development during reperfusion. BM-MSCs stimulated a geometric rearrangement of pial networks with formation of new anastomotic vessels sprouting from preexistent arterioles in the penumbra at 7-14-28 days of reperfusion. At the same time VEGF and eNOS expression increased. GFP-BM-MSCs appear to be involved in endothelial and smooth muscle cell programming in the infarcted area. In conclusion, transient MCAO induced pial vascular remodeling characterized by arteriolar anastomotic arcades (originated from preexistent arterioles in penumbra area) able to overlap the ischemic core supplying blood to the neuronal tissue. BM-MSCs appear to accelerate angiogenic processes facilitating new vessel formation; this mechanism was promoted by an increase in VEGF and eNOS expression.
Collapse
Affiliation(s)
- Dominga Lapi
- Department of Clinical Medicine and Surgery, "Federico II" University Medical School Naples, Italy
| | - Sabrina Vagnani
- Rheumatology Unit, Department of Internal Medicine, University of Pisa Pisa, Italy
| | - Daniela Sapio
- Department of Clinical Medicine and Surgery, "Federico II" University Medical School Naples, Italy
| | - Teresa Mastantuono
- Department of Clinical Medicine and Surgery, "Federico II" University Medical School Naples, Italy
| | - Francesca Boscia
- Department of Neuroscience, Reproductive and Odontostomatologic Sciences, "Federico II" University Medical School Naples, Italy
| | - Giuseppe Pignataro
- Department of Neuroscience, Reproductive and Odontostomatologic Sciences, "Federico II" University Medical School Naples, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino Orbassano, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino Orbassano, Italy
| | - Antonio Colantuoni
- Department of Clinical Medicine and Surgery, "Federico II" University Medical School Naples, Italy
| |
Collapse
|
30
|
Khan M, Dhammu TS, Matsuda F, Singh AK, Singh I. Blocking a vicious cycle nNOS/peroxynitrite/AMPK by S-nitrosoglutathione: implication for stroke therapy. BMC Neurosci 2015; 16:42. [PMID: 26174015 PMCID: PMC4502912 DOI: 10.1186/s12868-015-0179-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Stroke immediately sets into motion sustained excitotoxicity and calcium dysregulation, causing aberrant activity in neuronal nitric oxide synthase (nNOS) and an imbalance in the levels of nitric oxide (NO). Drugs targeting nNOS-originated toxicity may therefore reduce stroke-induced damage. Recently, we observed that a redox-modulating agent of the NO metabolome, S-nitrosoglutathione (GSNO), confers neurovascular protection by reducing the levels of peroxynitrite, a product of aberrant NOS activity. We therefore investigated whether GSNO-mediated neuroprotection and improved neurological functions depend on blocking nNOS/peroxynitrite-associated injurious mechanisms using a rat model of cerebral ischemia reperfusion (IR). RESULTS IR increased the activity of nNOS, the levels of neuronal peroxynitrite and phosphorylation at Ser(1412) of nNOS. GSNO treatment of IR animals decreased IR-activated nNOS activity and neuronal peroxynitrite levels by reducing nNOS phosphorylation at Ser(1412). The Ser(1412) phosphorylation is associated with increased nNOS activity. Supporting the notion that nNOS activity and peroxynitrite are deleterious following IR, inhibition of nNOS by its inhibitor 7-nitroindazole or reducing peroxynitrite by its scavenger FeTPPS decreased IR injury. GSNO also decreased the activation of AMP Kinase (AMPK) and its upstream kinase LKB1, both of which were activated in IR brain. AMPK has been implicated in nNOS activation via Ser(1412) phosphorylation. To determine whether AMPK activation is deleterious in the acute phase of IR, we treated animals after IR with AICAR (an AMPK activator) and compound c (an AMPK inhibitor). While AICAR potentiated, compound c reduced the IR injury. CONCLUSIONS Taken together, these results indicate an injurious nNOS/peroxynitrite/AMPK cycle following stroke, and GSNO treatment of IR inhibits this vicious cycle, resulting in neuroprotection and improved neurological function. GSNO is a natural component of the human body, and its exogenous administration to humans is not associated with any known side effects. Currently, the FDA-approved thrombolytic therapy suffers from a lack of neuronal protective activity. Because GSNO provides neuroprotection by ameliorating stroke's initial and causative injuries, it is a candidate of translational value for stroke therapy.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Tajinder S Dhammu
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Fumiyo Matsuda
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA. .,School of Health Science, Kagoshima University, Kagoshima, Japan.
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA. .,Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
31
|
Barros-Miñones L, Orejana L, Goñi-Allo B, Suquía V, Hervías I, Aguirre N, Puerta E. Modulation of the ASK1-MKK3/6-p38/MAPK signalling pathway mediates sildenafil protection against chemical hypoxia caused by malonate. Br J Pharmacol 2015. [PMID: 23186227 DOI: 10.1111/bph.12071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE PD5 inhibitors have recently been reported to exert beneficial effects against ischaemia-reperfusion injury in several organs. However, there are few studies regarding their neuroprotective effects in brain ischaemia. The present study was designed to assess the effects of sildenafil against chemical hypoxia induced by malonate. Intrastriatal injection of malonate produces energy depletion and striatal lesions similar to that seen in cerebral ischaemia through mechanisms that involve generation of reactive oxygen species (ROS). EXPERIMENTAL APPROACH Volume lesion was analysed by cytochrome oxidase histochemistry. Generation of reactive species was determined by in situ visualization of superoxide production and nitrotyrosine measurement. Protein levels were determined by Western blot after subcellular fractionation. KEY RESULTS Sildenafil, given 30 min before malonate, significantly decreased the lesion volume in the rat. This protective effect cannot be attributed to any effect on ROS production but to the inhibition of downstream pathways. Thus, malonate induced the activation of apoptosis signal-regulating kinase-1 (ASK1) and two MAPK kinases, MKK3/6 and MKK7, which lead to an increased phosphorylation of JNK and p38 MAPK, effects that were blocked by sildenafil. Selective inhibitors of p38 and JNK (SB203580 or SP600125, respectively) were used in combination with malonate in order to evaluate the plausible implication of these pathways in the protection afforded by sildenafil. While inhibition of p38 provided a significant protection against malonate-induced neurotoxicity, inhibition of JNK did not. CONCLUSIONS AND IMPLICATIONS Sildenafil protects against the chemical hypoxia induced by malonate through the regulation of the ASK1-MKK3/6-p38/MAPK signalling pathway.
Collapse
Affiliation(s)
- L Barros-Miñones
- Department of Pharmacology, School of Pharmacy, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
32
|
Arandarcikaite O, Jokubka R, Borutaite V. Neuroprotective effects of nitric oxide donor NOC-18 against brain ischemia-induced mitochondrial damages: role of PKG and PKC. Neurosci Lett 2015; 586:65-70. [DOI: 10.1016/j.neulet.2014.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/01/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
|
33
|
Protective Actions of Ghrelin on Global Cerebral Ischemia-Induced Memory Deficits. NEUROPHYSIOLOGY+ 2014. [DOI: 10.1007/s11062-014-9454-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
34
|
Towiwat P, Phattanarudee S, Maher TJ, Ally A. Modulation of inducible nitric oxide synthase (iNOS) expression and cardiovascular responses during static exercise following iNOS antagonism within the ventrolateral medulla. Mol Cell Biochem 2014; 398:185-94. [PMID: 25234194 DOI: 10.1007/s11010-014-2218-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/12/2014] [Indexed: 10/24/2022]
Abstract
Previous reports indicate that inducible nitric oxide synthase (iNOS) blockade within the rostral ventrolateral medulla (RVLM) and caudal ventrolateral medulla (CVLM) differentially modulated cardiovascular responses, medullary glutamate, and GABA concentrations during static skeletal muscle contraction. In the current study, we determined the role of iNOS antagonism within the RVLM and CVLM on cardiovascular responses and iNOS protein expression during the exercise pressor reflex in anesthetized rats. Following 120 min of bilateral microdialysis of a selective iNOS antagonist, aminoguanidine (AGN; 10 µM), into the RVLM, the pressor responses were attenuated by 72 % and changes in heart rate were reduced by 38 % during a static muscle contraction. Furthermore, western blot analysis of iNOS protein abundance within the RVLM revealed a significant attenuation when compared to control animals. In contrast, bilateral administration of AGN (10 µM) into the CVLM augmented the increases in mean arterial pressure by 60 % and potentiated changes in heart rate by 61 % during muscle contractions, but did not alter expression of the iNOS protein within the CVLM. These results demonstrate that iNOS protein expression within the ventrolateral medulla is differentially regulated by iNOS blockade that may, in part, contribute to the modulation of cardiovascular responses during static exercise.
Collapse
Affiliation(s)
- Pasarapa Towiwat
- Department of Pharmacology and Physiology, Chulalongkorn University, Bangkok, 10330, Thailand
| | | | | | | |
Collapse
|
35
|
Peng W, Xing Z, Yang J, Wang Y, Wang W, Huang W. The efficacy of erythropoietin in treating experimental traumatic brain injury: a systematic review of controlled trials in animal models. J Neurosurg 2014; 121:653-64. [PMID: 25036201 DOI: 10.3171/2014.6.jns132577] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECT Erythropoietin (EPO) shows promise as a neuroprotective agent in animal models of traumatic brain injury (TBI). However, clinical trials of the efficacy of EPO treatment in patients with TBI yield conflicting results. The authors conducted a systematic review and meta-analysis to assess the effect of EPO in experimental animal models of TBI, the goal being to inform the design of future clinical trials. METHODS The authors identified eligible studies by searching PubMed, Web of Science, MEDLINE, Embase, and Google Scholar in October 2013. Data were pooled using the random-effects model, and results were reported in terms of standardized mean difference. Statistical heterogeneity was examined using both I(2) and chi-square tests, and the presence of small study effects was investigated with funnel plots and Egger tests. In-depth analyses were performed for lesion volume and neurobehavioral outcome, and the studies' methodological quality was also evaluated. RESULTS Of a total of 290 studies, 13 found an effect of EPO on lesion volume and neurobehavioral outcome. Overall, the methodological quality of the studies was poor, and there was evidence of statistical heterogeneity among the publications as well as small-study effects. However, in-depth analyses showed statistically significant findings in favor of a beneficial effect of EPO after TBI. CONCLUSIONS Despite limitations of this systematic review that may have influenced the findings, the authors conclude that EPO might be beneficial in treating experimental TBI in terms of reducing lesion volume and improving neurobehavioral outcome. However, this review also indicates that more well-designed and well-reported animal studies are needed.
Collapse
Affiliation(s)
- Weijun Peng
- Institute of Integrated Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | | | | | | | | | | |
Collapse
|
36
|
Cerebral endothelial function determined by cerebrovascular reactivity to L-arginine. BIOMED RESEARCH INTERNATIONAL 2014; 2014:601515. [PMID: 24860826 PMCID: PMC4016874 DOI: 10.1155/2014/601515] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 03/27/2014] [Indexed: 12/27/2022]
Abstract
Endothelium forms the inner cellular lining of blood vessels and plays an important role in many physiological functions including the control of vasomotor tone. Cerebral endothelium is probably one of the most specific types but until recently it was impossible to determine its function. In this review, the role of cerebrovascular reactivity to L-arginine (CVR-L-Arg) for assessment of cerebral endothelial function is discussed. L-Arginine induces vasodilatation through enhanced production of nitric oxide (NO) in the cerebral endothelium. Transcranial Doppler sonography is used for evaluation of cerebral blood flow changes. The method is noninvasive, inexpensive, and enables reproducible measurements. CVR-L-Arg has been compared to flow-mediated dilatation as a gold standard for systemic endothelial function and intima-media thickness as a marker for morphological changes. However, it seems to show specific cerebral endothelial function. So far CVR-L-Arg has been used to study cerebral endothelial function in many pathological conditions such as stroke, migraine, etc. In addition CVR-L-Arg has also proven its usefulness in order to show potential improvement after pharmacological interventions. In conclusion CVR-L-Arg is a promising noninvasive research method that could provide means for evaluation of cerebral endothelial function in physiological and pathological conditions.
Collapse
|
37
|
Current preclinical studies on neuroinflammation and changes in blood-brain barrier integrity by MDMA and methamphetamine. Neuropharmacology 2014; 87:125-34. [PMID: 24594477 DOI: 10.1016/j.neuropharm.2014.02.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/05/2014] [Accepted: 02/20/2014] [Indexed: 11/24/2022]
Abstract
The blood-brain barrier (BBB) is essential in the maintenance of brain homeostasis both by preserving normal brain functioning and also by protecting the brain from exposure to a range of potentially harmful substances. This review presents some of the evidence of BBB disruption following exposure to the substituted amphetamines 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy') and methamphetamine (METH), two drugs of abuse which are widely consumed recreationally by younger sectors of the population. Both MDMA and METH have been shown to produce disruption of the BBB as reflected by IgG extravasation and Evans Blue leakage. In particular, METH decreases the expression of basal lamina proteins associated with an increase in matrix metalloproteinase activity. These changes in BBB integrity appear to be related to MDMA-induced activation of the mitogen-activated protein kinase (MAPK) JNK1/2. The consequences of the disruption in the BBB by these two drugs remain to be established, but there is evidence in the literature that, at least in the case of METH, increased matrix metalloproteinase (MMP) activity may be related to increased behavioural sensitization and reward perhaps because of the modification of the passage of the drug into the CNS. In addition, the high incidence of AIDS-related neurologic disease in METH users may also be related to increased entry into the brain of virally derived neurotoxic products. This article is part of the Special Issue entitled 'CNS Stimulants'.
Collapse
|
38
|
Jadhav RS, Ahmed L, Swamy PL, Sanaullah S. Neuroprotective effects of polyhydroxy pregnane glycoside isolated from Wattakaka volubilis (L.f.) Stapf. after middle cerebral artery occlusion and reperfusion in rats. Brain Res 2013; 1515:78-87. [DOI: 10.1016/j.brainres.2013.02.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 02/12/2013] [Accepted: 02/25/2013] [Indexed: 10/27/2022]
|
39
|
Phattanarudee S, Towiwat P, Maher TJ, Ally A. Effects of medullary administration of a nitric oxide precursor on cardiovascular responses and neurotransmission during static exercise following ischemic stroke. Can J Physiol Pharmacol 2013; 91:510-20. [PMID: 23826997 DOI: 10.1139/cjpp-2013-0066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have reported that in rats with a 90 min left middle cerebral artery occlusion (MCAO) and 24 h reperfusion, pressor responses during muscle contractions were attenuated, as were glutamate concentrations in the left rostral ventrolateral medulla (RVLM) and left caudal VLM (CVLM), but gamma-aminobutyric acid (GABA) levels increased in left RVLM and CVLM. This study determined the effects of L-arginine, a nitric oxide (NO) precursor, within the RVLM and (or) CVLM on cardiovascular activity and glutamate/GABA levels during static exercise in left-sided MCAO rats. Microdialysis of L-arginine into left RVLM had a greater attenuation of cardiovascular responses, a larger decrease in glutamate, and a significant increase in GABA levels during muscle contractions in stroke rats. Administration of N(G)-monomethyl-L-arginine, an NO-synthase inhibitor, reversed the effects. In contrast, L-arginine administration into left CVLM evoked a greater potentiation of cardiovascular responses, increased glutamate, and decreased GABA levels during contractions in stroked rats. However, L-arginine administration into both left RVLM and left CVLM elicited responses similar to its infusion into the left RVLM. These results suggest that NO within the RVLM and CVLM modulates cardiovascular responses and glutamate/GABA neurotransmission during static exercise following stroke, and that a RVLM-NO mechanism has a dominant effect in the medullary regulation of cardiovascular function.
Collapse
|
40
|
Charriaut-Marlangue C, Bonnin P, Pham H, Loron G, Leger PL, Gressens P, Renolleau S, Baud O. Nitric oxide signaling in the brain: A new target for inhaled nitric oxide? Ann Neurol 2013; 73:442-8. [DOI: 10.1002/ana.23842] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Revised: 11/24/2012] [Accepted: 12/21/2012] [Indexed: 02/06/2023]
Affiliation(s)
| | | | - Hoa Pham
- Paris Diderot University, Sorbonne Paris Cité, INSERM U676; Paris; France
| | - Gauthier Loron
- Paris Diderot University, Sorbonne Paris Cité, INSERM U676; Paris; France
| | | | | | | | | |
Collapse
|
41
|
Hu S, Cui W, Mak S, Tang J, Choi C, Pang Y, Han Y. Bis(propyl)-cognitin protects against glutamate-induced neuro-excitotoxicity via concurrent regulation of NO, MAPK/ERK and PI3-K/Akt/GSK3β pathways. Neurochem Int 2013; 62:468-77. [DOI: 10.1016/j.neuint.2013.01.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 01/14/2013] [Accepted: 01/19/2013] [Indexed: 02/02/2023]
|
42
|
Awooda HA, Lutfi MF, Sharara GM, Saeed AM. Role of N-Nitro-L-Arginine-Methylester as anti-oxidant in transient cerebral ischemia and reperfusion in rats. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2013; 5:1. [PMID: 23289587 PMCID: PMC3552933 DOI: 10.1186/2040-7378-5-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 01/02/2013] [Indexed: 01/05/2023]
Abstract
Background Previous reports assessing the neuroprotective role of nonselective Nitric Oxide synthase (NOS) inhibitor N-nitro-L-arginine-methylester (L-NAME) following cerebral ischemia/reperfusion are contradictory. The aim of this work was to examine the potential benefits of L-NAME on rats subjected to transient focal cerebral ischemia/reperfusion. Methods The study involved 30 adult male Wistar rats divided into three groups 10 rats in each: First group was sham-operated and served as a control, a ischemia/reperfusion (I/R) group of rats infused with 0.9% normal saline intraperitoneally 15 minutes prior to 30 minutes of left common carotid artery (CCA) occlusion and a test group infused with L-NAME intraperitoneally 15 minutes prior to ischemia. Neurobehavioral assessments were evaluated and quantitative assessment of malondialdehyde (MDA), Nitric oxide (NO) metabolites and total antioxidant capacity (TAC) in both serum and the affected cerebral hemisphere were achieved. Results Rats’ neurological deficit and TAC were significantly decreased while NO and MDA were significantly increased in the I/R compared with the control group (P < 0.001). Alternatively in the L-NAME group, neurological deficit and TAC were significantly improved while NO and MDA were significantly decreased compared to I/R group (P < 0.001). Conclusions L-NAME pretreatment for rats undergoing cerebral ischemia/reperfusion significantly improves neurological deficit while reducing oxidative stress biomarkers in the affected cerebral hemisphere.
Collapse
Affiliation(s)
- Hiba A Awooda
- Department of Physiology - Faculty of Medicine and Heath Sciences, Alneelain University, Khartoum, Sudan.
| | | | | | | |
Collapse
|
43
|
Pekcec A, Yigitkanli K, Jung JE, Pallast S, Xing C, Antipenko A, Minchenko M, Nikolov DB, Holman TR, Lo EH, van Leyen K. Following experimental stroke, the recovering brain is vulnerable to lipoxygenase-dependent semaphorin signaling. FASEB J 2012; 27:437-45. [PMID: 23070608 DOI: 10.1096/fj.12-206896] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recovery from stroke is limited, in part, by an inhibitory environment in the postischemic brain, but factors preventing successful remodeling are not well known. Using cultured cortical neurons from mice, brain endothelial cells, and a mouse model of ischemic stroke, we show that signaling from the axon guidance molecule Sema3A via eicosanoid second messengers can contribute to this inhibitory environment. Either 90 nM recombinant Sema3A, or the 12/15-lipoxygenase (12/15-LOX) metabolites 12-HETE and 12-HPETE at 300 nM, block axon extension in neurons compared to solvent controls, and decrease tube formation in endothelial cells. The Sema3A effect is reversed by inhibiting 12/15-LOX, and neurons derived from 12/15-LOX-knockout mice are insensitive to Sema3A. Following middle cerebral artery occlusion to induce stroke in mice, immunohistochemistry shows both Sema3A and 12/15-LOX are increased in the cortex up to 2 wk. To determine whether a Sema3A-dependent damage pathway is activated following ischemia, we injected recombinant Sema3A into the striatum. Sema3A alone did not cause injury in normal brains. But when injected into postischemic brains, Sema3A increased cortical damage by 79%, and again, this effect was reversed by 12/15-LOX inhibition. Our findings suggest that blocking the semaphorin pathway should be investigated as a therapeutic strategy to improve stroke recovery.
Collapse
Affiliation(s)
- Anton Pekcec
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Muñetón-Gómez VC, Doncel-Pérez E, Fernandez AP, Serrano J, Pozo-Rodrigálvarez A, Vellosillo-Huerta L, Taylor JS, Cardona-Gómez GP, Nieto-Sampedro M, Martínez-Murillo R. Neural differentiation of transplanted neural stem cells in a rat model of striatal lacunar infarction: light and electron microscopic observations. Front Cell Neurosci 2012; 6:30. [PMID: 22876219 PMCID: PMC3410634 DOI: 10.3389/fncel.2012.00030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 07/12/2012] [Indexed: 12/14/2022] Open
Abstract
The increased risk and prevalence of lacunar stroke and Parkinson's disease (PD) makes the search for better experimental models an important requirement for translational research. In this study we assess ischemic damage of the nigrostriatal pathway in a model of lacunar stroke evoked by damaging the perforating arteries in the territory of the substantia nigra (SN) of the rat after stereotaxic administration of endothelin-1 (ET-1), a potent vasoconstrictor peptide. We hypothesized that transplantation of neural stem cells (NSCs) with the capacity of differentiating into diverse cell types such as neurons and glia, but with limited proliferation potential, would constitute an alternative and/or adjuvant therapy for lacunar stroke. These cells showed neuritogenic activity in vitro and a high potential for neural differentiation. Light and electron microscopy immunocytochemistry was used to characterize GFP-positive neurons derived from the transplants. 48 h after ET-1 injection, we characterized an area of selective degeneration of dopaminergic neurons within the nigrostriatal pathway characterized with tissue necrosis and glial scar formation, with subsequent behavioral signs of Parkinsonism. Light microscopy showed that grafted cells within the striatal infarction zone differentiated with a high yield into mature glial cells (GFAP-positive) and neuron types present in the normal striatum. Electron microscopy revealed that NSCs-derived neurons integrated into the host circuitry establishing synaptic contacts, mostly of the asymmetric type. Astrocytes were closely associated with normal small-sized blood vessels in the area of infarct, suggesting a possible role in the regulation of the blood brain barrier and angiogenesis. Our results encourage the use of NSCs as a cell-replacement therapy for the treatment of human vascular Parkinsonism.
Collapse
Affiliation(s)
- Vilma C Muñetón-Gómez
- Neurovascular Research Group, Department of Molecular, Cellular, and Developmental Neurobiology, Spanish Council for Scientific Research (CSIC), Instituto Cajal Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
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
|
46
|
Mirabelli-Badenier M, Braunersreuther V, Lenglet S, Galan K, Veneselli E, Viviani GL, Mach F, Montecucco F. Pathophysiological role of inflammatory molecules in paediatric ischaemic brain injury. Eur J Clin Invest 2012; 42:784-94. [PMID: 22248042 DOI: 10.1111/j.1365-2362.2012.02640.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Ischaemic stroke is one of the major causes of death and lifelong disability also in the paediatric population. Strong scientific effort has been put to clarify the pathophysiology of this disease in adults. However, only few studies have been performed in children. Preliminary results indicate that pathophysiological processes might differently affect the poststroke neuronal injury in neonates as compared to children. During the neural development, selective molecular mechanisms might be differently triggered by an ischaemic insult, thus potentially resulting in defined postischaemic clinical outcomes. Basic research studies in neonatal animal models of cerebral ischaemia have recently shown a potential role of soluble inflammatory molecules (such as cytokines, chemokines and oxidants) as pivotal players of neuronal injury in both perinatal and childhood ischaemic stroke. Although larger clinical trials are still needed to confirm these preliminary results, the potential benefits of selective treatments targeting inflammation in perinatal asphyxia encephalopathy might represent a promising investigation field in the near future. In this review, we will update evidence on the pathophysiological role of soluble inflammatory mediators in neonatal and childhood ischaemic stroke. Recent evidence on potential anti-inflammatory treatments to improve paediatric stroke prognosis will be discussed.
Collapse
|
47
|
Hussein SA, Omayma AR, Elwakil A. Biochemical Abnormalities in Brain Tissues during Acute Brain Stroke Induced in Wistar Rats. JOURNAL OF MEDICAL SCIENCES 2012. [DOI: 10.3923/jms.2012.121.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
48
|
Pozo-Rodrigálvarez A, Gradillas A, Serrano J, Fernández AP, Martínez-Murillo R, Pérez-Castells J. New synthesis and promising neuroprotective role in experimental ischemic stroke of ONO-1714. Eur J Med Chem 2012; 54:439-46. [PMID: 22703704 DOI: 10.1016/j.ejmech.2012.05.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/16/2012] [Accepted: 05/22/2012] [Indexed: 12/21/2022]
Abstract
In an experimental permanent stroke model, we report here the contribution of ONO-1714 to brain damage prevention. Daily drug administration, twenty-one days prior to and two days after an experimental infarct, was performed by using mini-osmotic pumps (ALZET). Infarct volumes were assessed by image analysis of sequential coronal brain 1 mm(3) sections stained following the 2,3,5-triphenyltetrazolium chloride histological staining technique. Results of this study provide evidence of a significant reduction of the brain lesion size, suggesting ONO-1714 as a potential neuroprotective agent in stroke patients. ONO-1714 was prepared in our laboratory following a procedure which resulted in the supply of the desired compound in an easy and excellent yield.
Collapse
Affiliation(s)
- Andrea Pozo-Rodrigálvarez
- Neurovascular Research Group, Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal (CSIC), Av. Doctor Arce 37, 28002 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
49
|
Gelderblom M, Leypoldt F, Lewerenz J, Birkenmayer G, Orozco D, Ludewig P, Thundyil J, Arumugam TV, Gerloff C, Tolosa E, Maher P, Magnus T. The flavonoid fisetin attenuates postischemic immune cell infiltration, activation and infarct size after transient cerebral middle artery occlusion in mice. J Cereb Blood Flow Metab 2012; 32:835-43. [PMID: 22234339 PMCID: PMC3345911 DOI: 10.1038/jcbfm.2011.189] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The development of the brain tissue damage in ischemic stroke is composed of an immediate component followed by an inflammatory response with secondary tissue damage after reperfusion. Fisetin, a flavonoid, has multiple biological effects, including neuroprotective and antiinflammatory properties. We analyzed the effects of fisetin on infarct size and the inflammatory response in a mouse model of stroke, temporary middle cerebral artery occlusion, and on the activation of immune cells, murine primary and N9 microglial and Raw264.7 macrophage cells and human macrophages, in an in vitro model of inflammatory immune cell activation by lipopolysaccharide (LPS). Fisetin not only protected brain tissue against ischemic reperfusion injury when given before ischemia but also when applied 3 hours after ischemia. Fisetin also prominently inhibited the infiltration of macrophages and dendritic cells into the ischemic hemisphere and suppressed the intracerebral immune cell activation as measured by intracellular tumor necrosis factor α (TNFα) production. Fisetin also inhibited LPS-induced TNFα production and neurotoxicity of macrophages and microglia in vitro by suppressing nuclear factor κB activation and JNK/Jun phosphorylation. Our findings strongly suggest that the fisetin-mediated inhibition of the inflammatory response after stroke is part of the mechanism through which fisetin is neuroprotective in cerebral ischemia.
Collapse
Affiliation(s)
- Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bonnin P, Leger PL, Villapol S, Deroide N, Gressens P, Pocard M, Renolleau S, Baud O, Charriaut-Marlangue C. Dual action of NO synthases on blood flow and infarct volume consecutive to neonatal focal cerebral ischemia. Exp Neurol 2012; 236:50-7. [PMID: 22531298 DOI: 10.1016/j.expneurol.2012.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 03/23/2012] [Accepted: 04/04/2012] [Indexed: 11/15/2022]
Abstract
Research into neonatal ischemic brain damage is impeded by the lack of a complete understanding of the initial hemodynamic mechanisms resulting in a lesion, particularly that of NO-mediated vascular mechanisms. In a neonatal stroke rat model, we recently show that collateral recruitment contributes to infarct size variability. Non-specific and selective NO synthase (NOS) inhibition was evaluated on cerebral blood-flow changes and outcome in a P7 rat model of arterial occlusion (left middle cerebral artery electrocoagulation with 50 min occlusion of both common carotid arteries). Blood-flow changes were measured by using ultrasound imaging with sequential Doppler recordings in both internal carotid arteries and basilar trunk. Cortical perfusion was measured by using laser Doppler flowmetry. We showed that global NOS inhibition significantly reduced collateral support and cortical perfusion (collateral failure), and worsened the ischemic injury in both gender. Conversely, endothelial NOS inhibition increased blood-flows and aggravated volume lesion in males, whereas in females blood-flows did not change and infarct lesion was significantly reduced. These changes were associated with decreased phosphorylation of neuronal NOS at Ser(847) in males and increased phosphorylation in females at 24h, respectively. Neuronal NOS inhibition also increased blood-flows in males but not in females, and did not significantly change infarct volumes compared to their respective PBS-treated controls. In conclusion, both nNOS and eNOS appear to play a key role in modulating arterial blood flow during ischemia mainly in male pups with subsequent modifications in infarct lesion.
Collapse
Affiliation(s)
- Philippe Bonnin
- Univ Paris Diderot, Sorbonne Paris Cité, AP-HP, Hôpital Lariboisière, Physiologie Clinique - Explorations-Fonctionnelles, 75010 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|