101
|
Jiang Z, Alamuri TT, Muir ER, Choi DW, Duong TQ. Longitudinal multiparametric MRI study of hydrogen-enriched water with minocycline combination therapy in experimental ischemic stroke in rats. Brain Res 2020; 1748:147122. [DOI: 10.1016/j.brainres.2020.147122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
|
102
|
Ronaldson PT, Davis TP. Regulation of blood-brain barrier integrity by microglia in health and disease: A therapeutic opportunity. J Cereb Blood Flow Metab 2020; 40:S6-S24. [PMID: 32928017 PMCID: PMC7687032 DOI: 10.1177/0271678x20951995] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The blood-brain barrier (BBB) is a critical regulator of CNS homeostasis. It possesses physical and biochemical characteristics (i.e. tight junction protein complexes, transporters) that are necessary for the BBB to perform this physiological role. Microvascular endothelial cells require support from astrocytes, pericytes, microglia, neurons, and constituents of the extracellular matrix. This intricate relationship implies the existence of a neurovascular unit (NVU). NVU cellular components can be activated in disease and contribute to dynamic remodeling of the BBB. This is especially true of microglia, the resident immune cells of the brain, which polarize into distinct proinflammatory (M1) or anti-inflammatory (M2) phenotypes. Current data indicate that M1 pro-inflammatory microglia contribute to BBB dysfunction and vascular "leak", while M2 anti-inflammatory microglia play a protective role at the BBB. Understanding biological mechanisms involved in microglia activation provides a unique opportunity to develop novel treatment approaches for neurological diseases. In this review, we highlight characteristics of M1 proinflammatory and M2 anti-inflammatory microglia and describe how these distinct phenotypes modulate BBB physiology. Additionally, we outline the role of other NVU cell types in regulating microglial activation and highlight how microglia can be targeted for treatment of disease with a focus on ischemic stroke and Alzheimer's disease.
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Pharmacology, College of Medicine University of Arizona, Tucson, AZ, USA
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine University of Arizona, Tucson, AZ, USA
| |
Collapse
|
103
|
Sharma T, Airao V, Buch P, Vaishnav D, Parmar S. Sesamol protects hippocampal CA1 neurons and reduces neuronal infarction in global model of cerebral ischemia in rats. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
104
|
Yang Y, Torbey MT. Angiogenesis and Blood-Brain Barrier Permeability in Vascular Remodeling after Stroke. Curr Neuropharmacol 2020; 18:1250-1265. [PMID: 32691713 PMCID: PMC7770645 DOI: 10.2174/1570159x18666200720173316] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/27/2020] [Accepted: 07/11/2020] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis, the growth of new blood vessels, is a natural defense mechanism helping to restore oxygen and nutrient supply to the affected brain tissue following an ischemic stroke. By stimulating vessel growth, angiogenesis may stabilize brain perfusion, thereby promoting neuronal survival, brain plasticity, and neurologic recovery. However, therapeutic angiogenesis after stroke faces challenges: new angiogenesis-induced vessels have a higher than normal permeability, and treatment to promote angiogenesis may exacerbate outcomes in stroke patients. The development of therapies requires elucidation of the precise cellular and molecular basis of the disease. Microenvironment homeostasis of the central nervous system is essential for its normal function and is maintained by the blood-brain barrier (BBB). Tight junction proteins (TJP) form the tight junction (TJ) between vascular endothelial cells (ECs) and play a key role in regulating the BBB permeability. We demonstrated that after stroke, new angiogenesis-induced vessels in peri-infarct areas have abnormally high BBB permeability due to a lack of major TJPs in ECs. Therefore, promoting TJ formation and BBB integrity in the new vessels coupled with speedy angiogenesis will provide a promising and safer treatment strategy for improving recovery from stroke. Pericyte is a central neurovascular unite component in vascular barriergenesis and are vital to BBB integrity. We found that pericytes also play a key role in stroke-induced angiogenesis and TJ formation in the newly formed vessels. Based on these findings, in this article, we focus on regulation aspects of the BBB functions and describe cellular and molecular special features of TJ formation with an emphasis on role of pericytes in BBB integrity during angiogenesis after stroke.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| | - Michel T Torbey
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| |
Collapse
|
105
|
Choi JH, Poli S, Chen M, Nguyen TN, Saver JL, Matouk C, Pile-Spellman J. Selective Brain Hypothermia in Acute Ischemic Stroke: Reperfusion Without Reperfusion Injury. Front Neurol 2020; 11:594289. [PMID: 33281733 PMCID: PMC7691595 DOI: 10.3389/fneur.2020.594289] [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: 08/13/2020] [Accepted: 10/15/2020] [Indexed: 01/19/2023] Open
Abstract
In acute ischemic stroke, early recanalization of the occluded artery is crucial for best outcome to be achieved. Recanalization aims at restoring blood flow to the ischemic tissue (reperfusion) and is achieved with pharmacological thrombolytic drugs, endovascular thrombectomy (EVT) devices, or both. The introduction of modern endovascular devices has led to tremendous anatomical and clinical success with rates of substantial reperfusion exceeding 80% and proven clinical benefit in patients with anterior circulation large vessel occlusions (LVOs). However, not every successful reperfusion procedure leads to the desired clinical outcome. In fact, the rate of non-disabled outcome at 3 months with current EVT treatment is ~1 out of 4. A constraint upon better outcomes is that reperfusion, though resolving ischemic stress, may not restore the anatomic structures and metabolic functions of ischemic tissue to their baseline states. In fact, ischemia triggers a complex cascade of destructive mechanisms that can sometimes be exacerbated rather than alleviated by reperfusion therapy. Such reperfusion injury may cause infarct progression, intracranial hemorrhage, and unfavorable outcome. Therapeutic hypothermia has been shown to have a favorable impact on the molecular elaboration of ischemic injury, but systemic hypothermia is limited by slow speed of attaining target temperatures and clinical complications. A novel approach is endovascular delivery of hypothermia to cool the affected brain tissue selectively and rapidly with tight local temperature control, features not available with systemic hypothermia devices. In this perspective article, we discuss the possible benefits of adjunctive selective endovascular brain hypothermia during interventional stroke treatment.
Collapse
Affiliation(s)
- Jae H. Choi
- Neurovascular Center, Neurological Surgery, P.C., Lake Success, NY, United States
- Hybernia Medical, LLC, New Rochelle, NY, United States
| | - Sven Poli
- Department of Neurology & Stroke, Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Michael Chen
- Stroke Center, Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| | - Thanh N. Nguyen
- Interventional Neurology/Neuroradiology, Boston University School of Medicine, Boston, MA, United States
| | - Jeffrey L. Saver
- Comprehensive Stroke Center and Department of Neurology, University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Charles Matouk
- Neurovascular Surgery, Department of Neurosurgery, Yale University-New Haven Hospital, New Haven, CT, United States
| | - John Pile-Spellman
- Neurovascular Center, Neurological Surgery, P.C., Lake Success, NY, United States
- Hybernia Medical, LLC, New Rochelle, NY, United States
| |
Collapse
|
106
|
Shoaib M, Becker LB. A walk through the progression of resuscitation medicine. Ann N Y Acad Sci 2020; 1507:23-36. [PMID: 33040363 DOI: 10.1111/nyas.14507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/03/2020] [Accepted: 09/14/2020] [Indexed: 01/11/2023]
Abstract
Cardiac arrest (CA) is a sudden and devastating disease process resulting in more deaths in the United States than many cancers, metabolic diseases, and even car accidents. Despite such a heavy mortality burden, effective treatments have remained elusive. The past century has been productive in establishing the guidelines for resuscitation, known as cardiopulmonary resuscitation (CPR), as well as developing a scientific field whose aim is to elucidate the underlying mechanisms of CA and develop therapies to save lives. CPR has been successful in reinitiating the heart after arrest, enabling a survival rate of approximately 10% in out-of-hospital CA. Although current advanced resuscitation methods, including hypothermia and extracorporeal membrane oxygenation, have improved survival in some patients, they are unlikely to significantly improve the national survival rate any further without a paradigm shift. Such a change is possible with sustained efforts in the basic and clinical sciences of resuscitation and their implementation. This review seeks to discuss the current landscape in resuscitation medicine-how we got here and where we are going.
Collapse
Affiliation(s)
- Muhammad Shoaib
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York.,The Feinstein Institutes for Medical Research, Manhasset, New York
| | - Lance B Becker
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York.,The Feinstein Institutes for Medical Research, Manhasset, New York.,Department of Emergency Medicine, North Shore University Hospital, Northwell Health, Manhasset, New York
| |
Collapse
|
107
|
Valent A, Maïer B, Chabanne R, Degos V, Lapergue B, Lukaszewicz AC, Mazighi M, Gayat E. Anaesthesia and haemodynamic management of acute ischaemic stroke patients before, during and after endovascular therapy. Anaesth Crit Care Pain Med 2020; 39:859-870. [PMID: 33039657 DOI: 10.1016/j.accpm.2020.05.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/16/2020] [Accepted: 05/26/2020] [Indexed: 11/29/2022]
Abstract
Endovascular therapy (EVT) is now standard of care for eligible patients with acute ischaemic stroke caused by large vessel occlusion in the anterior circulation. EVT can be performed with general anaesthesia (GA) or with monitored anaesthesia care, involving local anaesthesia with or without conscious sedation (LA/CS). Controversies remain regarding the optimal choice of anaesthetic strategy and observational studies suggested poorer functional outcome and higher mortality in patients treated under GA, essentially because of its haemodynamic consequences and the delay to put patients under GA. However, these studies are limited by selection bias, the most severe patients being more likely to receive GA and recent randomised trials and meta-analysis showed that protocol-based GA compared with LA/CS is significantly associated with less disability at 3 months. Unlike for intravenous thrombolysis, few data exist to guide management of blood pressure (BP) before and during EVT, but arterial hypotension should be avoided as long as the occlusion persists. BP targets following EVT should probably be adapted to the degree of recanalisation and the extent of ischaemia. Lower BP levels may be warranted to prevent reperfusion injuries even if prospective haemodynamic management evaluations after EVT are lacking.
Collapse
Affiliation(s)
- Arnaud Valent
- Department of Anaesthesiology and Critical Care, Lariboisière Hospital, DMU Parabol, AP-HP Nord & University of Paris, Paris, France; UMR-S 942 MASCOT, Inserm, France
| | - Benjamin Maïer
- Interventional Neuroradiology, Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
| | - Russell Chabanne
- Department of Perioperative Medicine, University Hospital of Clermont-Ferrand, Clermont-Ferrand Cedex, France
| | - Vincent Degos
- Department of Anaesthesia and Critical Care, Pitié Salpêtrière Hospital, AP-HP-SU, Paris, France, Groupe recherche clinique BIOSFAST, Sorbonne University, Paris, France
| | - Bertrand Lapergue
- Stroke Centre Neurology Division, Hôpital Foch, 92150, Suresnes, France
| | - Anne-Claire Lukaszewicz
- Service d'Anesthésie Réanimation, Hôpital Neurologique, Hospices Civils de Lyon, Bron, France; EA 7426 PI3 (Pathophysiology of Injury-induced Immunosuppression), Hospices Civils de Lyon/Université de Lyon/bioMérieux, Hôpital E. Herriot, Lyon cedex 03, France
| | - Mikael Mazighi
- Department of Neurology and Stroke Centre, Lariboisière Hospital, AP-HP, Paris University, Sorbonne Paris Cité, Paris, France; Département Hospitalo-Universistaire Neurovasc, Paris, France
| | - Etienne Gayat
- Department of Anaesthesiology and Critical Care, Lariboisière Hospital, DMU Parabol, AP-HP Nord & University of Paris, Paris, France; UMR-S 942 MASCOT, Inserm, France.
| |
Collapse
|
108
|
Lipid Emulsion Improves Functional Recovery in an Animal Model of Stroke. Int J Mol Sci 2020; 21:ijms21197373. [PMID: 33036206 PMCID: PMC7582956 DOI: 10.3390/ijms21197373] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/29/2020] [Accepted: 10/03/2020] [Indexed: 12/16/2022] Open
Abstract
Stroke is a life-threatening condition that leads to the death of many people around the world. Reperfusion injury after ischemic stroke is a recurrent problem associated with various surgical procedures that involve the removal of blockages in the brain arteries. Lipid emulsion was recently shown to attenuate ischemic reperfusion injury in the heart and to protect the brain from excitotoxicity. However, investigations on the protective mechanisms of lipid emulsion against ischemia in the brain are still lacking. This study aimed to determine the neuroprotective effects of lipid emulsion in an in vivo rat model of ischemic reperfusion injury through middle cerebral artery occlusion (MCAO). Under sodium pentobarbital anesthesia, rats were subjected to MCAO surgery and were administered with lipid emulsion through intra-arterial injection during reperfusion. The experimental animals were assessed for neurological deficit wherein the brains were extracted at 24 h after reperfusion for triphenyltetrazolium chloride staining, immunoblotting and qPCR. Neuroprotection was found to be dosage-dependent and the rats treated with 20% lipid emulsion had significantly decreased infarction volumes and lower Bederson scores. Phosphorylation of Akt and glycogen synthase kinase 3-β (GSK3-β) were increased in the 20% lipid-emulsion treated group. The Wnt-associated signals showed a marked increase with a concomitant decrease in signals of inflammatory markers in the group treated with 20% lipid emulsion. The protective effects of lipid emulsion and survival-related expression of genes such as Akt, GSK-3β, Wnt1 and β-catenin were reversed by the intra-peritoneal administration of XAV939 through the inhibition of the Wnt/β-catenin signaling pathway. These results suggest that lipid emulsion has neuroprotective effects against ischemic reperfusion injury in the brain through the modulation of the Wnt signaling pathway and may provide potential insights for the development of therapeutic targets.
Collapse
|
109
|
The Usefulness of Quantitative Analysis of Blood-Brain Barrier Disruption Measured Using Contrast-Enhanced Magnetic Resonance Imaging to Predict Neurological Prognosis in Out-of-Hospital Cardiac Arrest Survivors: A Preliminary Study. J Clin Med 2020; 9:jcm9093013. [PMID: 32962022 PMCID: PMC7564654 DOI: 10.3390/jcm9093013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
We aimed to evaluate neurological outcomes associated with blood-brain barrier (BBB) disruption using contrast-enhanced magnetic resonance imaging (CE-MRI) in out-of-hospital cardiac arrest (OHCA) survivors. This retrospective observational study involved OHCA survivors who had undergone CE-MRI for prognostication. Qualitative and quantitative analyses were performed using the presence of BBB disruption (pBD) and the BBB disruption score (sBD) in CE-MRI scans, respectively. For the sBD, 1 point was assigned for each area of BBB disruption, and 6 points were assigned when an absence of intracranial blood flow due to severe brain oedema was confirmed. The primary outcome was poor neurological outcome at 3 months (defined as cerebral performance categories 3-5). We analysed 46 CE-MRI brain scans (27 patients). Of these, 15 (55.6%) patients had poor neurological outcomes. Poor neurological outcome group patients showed a significantly higher proportion of pBD than those in the good neurological outcome group (22 (88%) vs. 6 (28.6%) patients, respectively, p < 0.001) and a higher sBD (5.0 (4.0-5.0) vs. 0.0 (0.0-1.0) patients, p < 0.001). Poor neurological outcome predictions showed that the sBD had a significantly better prognostic performance (area under the curve (AUC) 0.95, 95% confidence interval (CI) 0.84-0.99) than the pBD (AUC 0.80, 95% CI 0.65-0.90). The sBD cut-off value was >1 point (sensitivity, 96.0%; specificity, 81.0%). The sBD is a highly predictive and sensitive marker of 3-month poor neurological outcome in OHCA survivors. Multicentre prospective studies are required to determine the generalisability of these results.
Collapse
|
110
|
Ravindran AV, Killingsworth MC, Bhaskar S. Cerebral collaterals in acute ischaemia: Implications for acute ischaemic stroke patients receiving reperfusion therapy. Eur J Neurosci 2020; 53:1238-1261. [PMID: 32871623 DOI: 10.1111/ejn.14955] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/21/2022]
Abstract
The cerebral collaterals play an important role in penumbral tissue sustenance after an acute ischaemic stroke. Recent studies have demonstrated the potential role of collaterals in the selection of acute ischaemic stroke patients eligible for reperfusion therapy. However, the understanding of the significance and evidence around the role of collateral status in predicting outcomes in acute ischaemic stroke patients treated with reperfusion therapy is still unclear. Moreover, the use of pre-treatment collaterals in patient selection and prognosis is relatively underappreciated in clinical settings. A focused review of the literature was performed on the various methods of collateral evaluation and the role of collateral status in acute ischaemic stroke patients receiving reperfusion therapy. We discuss the methods of evaluating pre-treatment collaterals in clinical settings. The patient selection based on collateral status as well as the prognostic and therapeutic value of collaterals in acute ischaemic stroke, in settings of intravenous thrombolysis or endovascular therapy alone, and bridge therapy, are summarized. Recommendations for future research and possible pharmacological intervention strategies aimed at collateral enhancement are also discussed. Collaterals may play an important role in identifying acute ischaemic stroke patients who are likely to benefit from endovascular treatment in an extended time window. Future neuroscientific efforts to better improve our understanding of the role of collaterals in acute ischaemia as well as clinical studies to delineate its role in patient selection and acute stroke prognosis are warranted.
Collapse
Affiliation(s)
- Abina Vishni Ravindran
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia.,Neurovascular Imaging Laboratory, Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.,Thrombolysis and Endovascular WorkFLOw Network (TEFLON), Sydney, NSW, Australia
| | - Murray C Killingsworth
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia.,NSW Brain Clot Bank, NSW Health Statewide Biobank and NSW Health Pathology, Sydney, NSW, Australia.,Correlative Microscopy Facility, Ingham Institute for Applied Medical Research and Department of Anatomical Pathology, NSW Health Pathology and Liverpool Hospital, Liverpool, NSW, Australia
| | - Sonu Bhaskar
- South Western Sydney Clinical School, University of New South Wales (UNSW), Sydney, NSW, Australia.,Department of Neurology & Neurophysiology, Liverpool Hospital & South West Sydney Local Health District (SWSLHD), Sydney, NSW, Australia.,Neurovascular Imaging Laboratory, Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.,Stroke & Neurology Research Group, Ingham Institute for Applied Medical Research, Sydney, NSW, Australia.,NSW Brain Clot Bank, NSW Health Statewide Biobank and NSW Health Pathology, Sydney, NSW, Australia.,Thrombolysis and Endovascular WorkFLOw Network (TEFLON), Sydney, NSW, Australia
| |
Collapse
|
111
|
The Mitochondria-targeted Peptide, Bendavia, Attenuated Ischemia/Reperfusion-induced Stroke Damage. Neuroscience 2020; 443:110-119. [DOI: 10.1016/j.neuroscience.2020.07.044] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023]
|
112
|
Won J, Yi KS, Choi CH, Jeon CY, Seo J, Kim K, Yeo HG, Park J, Kim YG, Jin YB, Koo BS, Lim KS, Lee S, Kim KJ, Choi WS, Park SH, Kim YH, Huh JW, Lee SR, Cha SH, Lee Y. Assessment of Hand Motor Function in a Non-human Primate Model of Ischemic Stroke. Exp Neurobiol 2020; 29:300-313. [PMID: 32921642 PMCID: PMC7492846 DOI: 10.5607/en20023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/07/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke results from arterial occlusion and can cause irreversible brain injury. A non-human primate (NHP) model of ischemic stroke was previously developed to investigate its pathophysiology and for efficacy testing of therapeutic candidates; however, fine motor impairment remains to be well-characterized. We evaluated hand motor function in a cynomolgus monkey model of ischemic stroke. Endovascular transient middle cerebral artery occlusion (MCAO) with an angiographic microcatheter induced cerebral infarction. In vivo magnetic resonance imaging mapped and measured the ischemia-induced infarct lesion. In vivo diffusion tensor imaging (DTI) of the stroke lesion to assess the neuroplastic changes and fiber tractography demonstrated three-dimensional patterns in the corticospinal tract 12 weeks after MCAO. The hand dexterity task (HDT) was used to evaluate fine motor movement of upper extremity digits. The HDT was modified for a home cage-based training system, instead of conventional chair restraint training. The lesion was localized in the middle cerebral artery territory, including the sensorimotor cortex. Maximum infarct volume was exhibited over the first week after MCAO, which progressively inhibited ischemic core expansion, manifested by enhanced functional recovery of the affected hand over 12 weeks after MCAO. The total performance time decreased with increasing success rate for both hands on the HDT. Compensatory strategies and retrieval failure improved in the chronic phase after stroke. Our findings demonstrate the recovery of fine motor skill after stroke, and outline the behavioral characteristics and features of functional disorder of NHP stroke model, providing a basis for assessing hand motor function after stroke.
Collapse
Affiliation(s)
- Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kyung Sik Yi
- Department of Radiology, Chungbuk National University Hospital, Cheongju 28644, Korea
| | - Chi-Hoon Choi
- Department of Radiology, Chungbuk National University Hospital, Cheongju 28644, Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jincheol Seo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Keonwoo Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Hyeon-Gu Yeo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113 Korea
| | - Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Yu Gyeong Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113 Korea
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Bon-Sang Koo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource & Research Center, KRIBB, Cheongju 28116 Korea
| | - Sangil Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Ki Jin Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Won Seok Choi
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Sung-Hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113 Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113 Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113 Korea
| | - Sang-Hoon Cha
- Department of Radiology, Chungbuk National University Hospital, Cheongju 28644, Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113 Korea
| |
Collapse
|
113
|
Souza Monteiro de Araújo D, De Logu F, Adembri C, Rizzo S, Janal MN, Landini L, Magi A, Mattei G, Cini N, Pandolfo P, Geppetti P, Nassini R, Calaza KDC. TRPA1 mediates damage of the retina induced by ischemia and reperfusion in mice. Cell Death Dis 2020; 11:633. [PMID: 32801314 PMCID: PMC7429961 DOI: 10.1038/s41419-020-02863-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022]
Abstract
Oxidative stress is implicated in retinal cell injury associated with glaucoma and other retinal diseases. However, the mechanism by which oxidative stress leads to retinal damage is not completely understood. Transient receptor potential ankyrin 1 (TRPA1) is a redox-sensitive channel that, by amplifying the oxidative stress signal, promotes inflammation and tissue injury. Here, we investigated the role of TRPA1 in retinal damage evoked by ischemia (1 hour) and reperfusion (I/R) in mice. In wild-type mice, retinal cell numbers and thickness were reduced at both day-2 and day-7 after I/R. By contrast, mice with genetic deletion of TRPA1 were protected from the damage seen in their wild-type littermates. Daily instillation of eye drops containing two different TRPA1 antagonists, an oxidative stress scavenger, or a NADPH oxidase-1 inhibitor also protected the retinas of C57BL/6J mice exposed to I/R. Mice with genetic deletion of the proinflammatory TRP channels, vanilloid 1 (TRPV1) or vanilloid 4 (TRPV4), were not protected from I/R damage. Surprisingly, genetic deletion or pharmacological blockade of TRPA1 also attenuated the increase in the number of infiltrating macrophages and in the levels of the oxidative stress biomarker, 4-hydroxynonenal, and of the apoptosis biomarker, active caspase-3, evoked by I/R. These findings suggest that TRPA1 mediates the oxidative stress burden and inflammation that result in murine retinal cell death. We also found that TRPA1 (both mRNA and protein) is expressed by human retinal cells. Thus, it is possible that inhibition of a TRPA1-dependent pathway could also attenuate glaucoma-related retinal damage.
Collapse
Affiliation(s)
- Daniel Souza Monteiro de Araújo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil.,Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Chiara Adembri
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Stanislao Rizzo
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), Division of Ophthalmology, University of Florence, Florence, Italy
| | - Malvin N Janal
- Department of Epidemiology and Health Promotion, New York University College of Dentistry, New York, NY, USA
| | - Lorenzo Landini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Alberto Magi
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Gianluca Mattei
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Nicoletta Cini
- General Laboratory, Careggi University Hospital, Florence, Italy
| | - Pablo Pandolfo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| | - Karin da Costa Calaza
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
114
|
Kim HK, Lee JJ, Choi G, Sung B, Kim YH, Baek AR, Kim S, Song H, Kim M, Cho AE, Lee GH, Moon S, Kang MK, Lee JJ, Chang Y. Gadolinium-Based Neuroprognostic Magnetic Resonance Imaging Agents Suppress COX-2 for Prevention of Reperfusion Injury after Stroke. J Med Chem 2020; 63:6909-6923. [PMID: 32545964 DOI: 10.1021/acs.jmedchem.0c00285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Advancements in recanalization therapies have rendered reperfusion injury an important challenge for stroke management. It is essential to work toward effective therapeutics that protect the ischemic brain from reperfusion injury. Here, we report a new concept of neuroprognostic agents, which combine molecular diagnostic imaging and targeted neuroprotection for treatment of reperfusion injury after stroke. These neuroprognostic agents are inflammation-targeted gadolinium compounds conjugated with nonsteroidal anti-inflammatory drugs (NSAIDs). Our results demonstrated that gadolinium-based MRI contrast agents conjugated with NSAIDs suppressed the increase in cyclooxygenase-2 (COX-2) levels, ameliorated glial activation, and neuron damage that are phenotypic for stroke by mitigating neuroinflammation, which prevented reperfusion injury. In addition, this study showed that the neuroprognostic agents are promising T1 molecular MRI contrast agents for detecting precise reperfusion injury locations at the molecular level. Our results build on this new concept of neuroprognostics as a novel management strategy for ischemia-reperfusion injury, combining neuroprotection and molecular diagnostics.
Collapse
Affiliation(s)
- Hee-Kyung Kim
- BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, 41944 Daegu, Korea.,Institute of Biomedical Engineering Research, Kyungpook National University, 41944 Daegu, Korea
| | - Jung-Jin Lee
- Department of R & D Center, Myungmoon Bio. Co., Hwaseong, 18622 Gyeonggi-do, Korea
| | - Garam Choi
- Department of R & D Center, Myungmoon Bio. Co., Hwaseong, 18622 Gyeonggi-do, Korea.,Department of Medical & Biological Engineering, Kyungpook National University, 41944 Daegu, Korea
| | - Bokyung Sung
- Department of Medical & Biological Engineering, Kyungpook National University, 41944 Daegu, Korea
| | - Yeoun-Hee Kim
- Department of R & D Center, Myungmoon Bio. Co., Hwaseong, 18622 Gyeonggi-do, Korea
| | - Ah Rum Baek
- Department of Medical & Biological Engineering, Kyungpook National University, 41944 Daegu, Korea
| | - Soyeon Kim
- Department of Medical & Biological Engineering, Kyungpook National University, 41944 Daegu, Korea
| | - Huijin Song
- Institute of Biomedical Engineering Research, Kyungpook National University, 41944 Daegu, Korea
| | - Minsup Kim
- Department of Bioinformatics, Korea University, 30019 Sejong, Korea
| | - Art E Cho
- Department of Bioinformatics, Korea University, 30019 Sejong, Korea
| | - Gang Ho Lee
- Department of Chemistry, Kyungpook National University, 41566 Daegu, Korea
| | - Sungjun Moon
- Department of Radiology, Yeungnam University Medical Center, 42415 Daegu, Korea
| | - Min-Kyoung Kang
- Laboratory Animal Center, KBIO Osong Medical Innovation Foundation, 28160 Osong, Korea
| | - Jae Jun Lee
- Laboratory Animal Center, KBIO Osong Medical Innovation Foundation, 28160 Osong, Korea
| | - Yongmin Chang
- Department of Medical & Biological Engineering, Kyungpook National University, 41944 Daegu, Korea.,Department of Radiology, Kyungpook National University Hospital, 41944 Daegu, Korea.,Department of Molecular Medicine, School of Medicine, Kyungpook National University, 41944 Daegu, Korea
| |
Collapse
|
115
|
Ye S, Hananya N, Green O, Chen H, Zhao AQ, Shen J, Shabat D, Yang D. A Highly Selective and Sensitive Chemiluminescent Probe for Real‐Time Monitoring of Hydrogen Peroxide in Cells and Animals. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005429] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sen Ye
- Morningside Laboratory for Chemical Biology and Department of Chemistry The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI) The University of Hong Kong Pokfulam Road Hong Kong P. R. China
| | - Nir Hananya
- School of Chemistry Faculty of Exact Sciences Tel Aviv University Tel Aviv 69978 Israel
| | - Ori Green
- School of Chemistry Faculty of Exact Sciences Tel Aviv University Tel Aviv 69978 Israel
| | - Hansen Chen
- School of Chinese Medicine The University of Hong Kong Pokfulam Road Hong Kong P. R. China
| | - Angela Qian Zhao
- Morningside Laboratory for Chemical Biology and Department of Chemistry The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI) The University of Hong Kong Pokfulam Road Hong Kong P. R. China
| | - Jiangang Shen
- School of Chinese Medicine The University of Hong Kong Pokfulam Road Hong Kong P. R. China
| | - Doron Shabat
- School of Chemistry Faculty of Exact Sciences Tel Aviv University Tel Aviv 69978 Israel
| | - Dan Yang
- Morningside Laboratory for Chemical Biology and Department of Chemistry The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI) The University of Hong Kong Pokfulam Road Hong Kong P. R. China
| |
Collapse
|
116
|
Chiu PC, Liou HC, Ling TY, Shen LJ. Development of a Neuroprotective Erythropoietin Modified with a Novel Carrier for the Blood-Brain Barrier. Neurotherapeutics 2020; 17:1184-1196. [PMID: 32144722 PMCID: PMC7609523 DOI: 10.1007/s13311-020-00845-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Extremely high doses of erythropoietin (EPO) has been used for neuroprotection in ischemia-reperfusion brain injury to deliver sufficient amounts of EPO across the blood-brain barrier (BBB); however, harmful outcomes were observed afterward. We aimed to test the ability of HBHAc (heparin-binding haemagglutinin adhesion c), an intracellular delivery peptide for macromolecules, as an EPO carrier across the BBB. The cellular internalization and transcytosis ability of HBHAc-modified EPO (EPO-HBHAc) were evaluated in bEnd.3 cells and in the bEnd.3/CTX TNA2 co-culture BBB model, respectively. Subsequently, the NMDA-induced-toxicity model and ischemia-reperfusion rat model were used to understand the neuronal protective activity of EPO-HBHAc. The biodistribution of EPO-HBHAc was demonstrated in rats by the quantification of EPO-HBHAc in the brain, plasma, and organs by ELISA. Our results demonstrate that EPO-HBHAc exhibited significantly higher cellular internalization in dose- and time-dependent manners and better transcytosis ability than EPO. In addition, the transported EPO-HBHAc in the co-culture transwell system maintained the neuronal protective activity when primary rat cortical neurons underwent NMDA-induced toxicity. The calculated cerebral infarction area of rats treated with EPO-HBHAc was significantly reduced compared to that of rats treated with EPO (29.9 ± 7.0% vs 48.9 ± 7.9%) 24 h after occlusion in 3VO rat experiments. Moreover, the EPO amount in both CSF and damaged cortex from the EPO-HBHAc group was 4.0-fold and 3.0-fold higher than the EPO group, respectively. These results suggest that HBHAc would be a favorable tool for EPO brain delivery and would further extend the clinical applications of EPO in neuroprotection.
Collapse
Affiliation(s)
- Po-Chuan Chiu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Houng-Chi Liou
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Jiuan Shen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
117
|
Ye S, Hananya N, Green O, Chen H, Zhao AQ, Shen J, Shabat D, Yang D. A Highly Selective and Sensitive Chemiluminescent Probe for Real-Time Monitoring of Hydrogen Peroxide in Cells and Animals. Angew Chem Int Ed Engl 2020; 59:14326-14330. [PMID: 32472602 DOI: 10.1002/anie.202005429] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/26/2020] [Indexed: 12/25/2022]
Abstract
Selective and sensitive molecular probes for hydrogen peroxide (H2 O2 ), which plays diverse roles in oxidative stress and redox signaling, are urgently needed to investigate the physiological and pathological effects of H2 O2 . A lack of reliable tools for in vivo imaging has hampered the development of H2 O2 mediated therapeutics. By combining a specific tandem Payne/Dakin reaction with a chemiluminescent scaffold, H2 O2 -CL-510 was developed as a highly selective and sensitive probe for detection of H2 O2 both in vitro and in vivo. A rapid 430-fold enhancement of chemiluminescence was triggered directly by H2 O2 without any laser excitation. Arsenic trioxide induced oxidative damage in leukemia was successfully detected. In particular, cerebral ischemia-reperfusion injury-induced H2 O2 fluxes were visualized in rat brains using H2 O2 -CL-510, providing a new chemical tool for real-time monitoring of H2 O2 dynamics in living animals.
Collapse
Affiliation(s)
- Sen Ye
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | - Nir Hananya
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ori Green
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | - Angela Qian Zhao
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| | - Doron Shabat
- School of Chemistry, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Dan Yang
- Morningside Laboratory for Chemical Biology and Department of Chemistry, The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), The University of Hong Kong, Pokfulam Road, Hong Kong, P. R. China
| |
Collapse
|
118
|
Kang BS, Choi BY, Kho AR, Lee SH, Hong DK, Jeong JH, Kang DH, Park MK, Suh SW. An Inhibitor of the Sodium-Hydrogen Exchanger-1 (NHE-1), Amiloride, Reduced Zinc Accumulation and Hippocampal Neuronal Death after Ischemia. Int J Mol Sci 2020; 21:ijms21124232. [PMID: 32545865 PMCID: PMC7352629 DOI: 10.3390/ijms21124232] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Acidosis in the brain plays an important role in neuronal injury and is a common feature of several neurological diseases. It has been reported that the sodium–hydrogen exchanger-1 (NHE-1) is a key mediator of acidosis-induced neuronal injury. It modulates the concentration of intra- and extra-cellular sodium and hydrogen ions. During the ischemic state, excessive sodium ions enter neurons and inappropriately activate the sodium–calcium exchanger (NCX). Zinc can also enter neurons through voltage-gated calcium channels and NCX. Here, we tested the hypothesis that zinc enters the intracellular space through NCX and the subsequent zinc accumulation induces neuronal cell death after global cerebral ischemia (GCI). Thus, we conducted the present study to confirm whether inhibition of NHE-1 by amiloride attenuates zinc accumulation and subsequent hippocampus neuronal death following GCI. Mice were subjected to GCI by bilateral common carotid artery (BCCA) occlusion for 30 min, followed by restoration of blood flow and resuscitation. Amiloride (10 mg/kg, intraperitoneally (i.p.)) was immediately injected, which reduced zinc accumulation and neuronal death after GCI. Therefore, the present study demonstrates that amiloride attenuates GCI-induced neuronal injury, likely via the prevention of intracellular zinc accumulation. Consequently, we suggest that amiloride may have a high therapeutic potential for the prevention of GCI-induced neuronal death.
Collapse
Affiliation(s)
- Beom Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - A Ra Kho
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Dae Ki Hong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Jeong Hyun Jeong
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Dong Hyeon Kang
- Department of Medical Science, College of Medicine, Hallym University, Chuncheon 24252, Korea;
| | - Min Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (B.S.K.); (B.Y.C.); (A.R.K.); (S.H.L.); (D.K.H.); (J.H.J.); (M.K.P.)
- Correspondence: ; Tel.: +82-10-8573-6364
| |
Collapse
|
119
|
Zhang Z, Xu C, Hao J, Zhang M, Wang Z, Yin T, Lin K, Liu W, Jiang Q, Li Z, Wang D, Mao Z, Tong H, Zhang L. Beneficial consequences of Lupeol on middle cerebral artery-induced cerebral ischemia in the rat involves Nrf2 and P38 MAPK modulation. Metab Brain Dis 2020; 35:841-848. [PMID: 32212043 DOI: 10.1007/s11011-020-00565-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/06/2020] [Indexed: 10/24/2022]
Abstract
Lupeol has been reported to exhibit anti-inflammatory and anti-tumor activities in many diseases, but its potential effects in cerebral ischemia injury have not been studied to date. In this work we present evidence for a beneficial effect of lupeol in a rat model of middle cerebral artery occlusion (MCAO) followed by reperfusion (MCAO/R) injury and provide some histological and biochemical evidence for its mechanism of action. A cerebral MCAO rat model was established by vascular occlusion for 2 h, followed by 24 h reperfusion period. The infarct volume, neurological deficits, and brain water content were compared with animals treated during reperfusion with different concentrations of lupeol. Macroscopic parameters, cell viability, pro-inflammatory factors generation, as well as oxidative stress parameters and associated apoptotic signaling cascades were evaluated. Treatment with lupeol significantly reduced the cerebral infarct volume and water content and recovered neuro behavioral functions in affected rats. Lupeol treatment down-regulated the expression of oxidative stress and inflammation factors. In addition, lupeol activated Nrf2, suppressed caspase-3 activity, reduced BAX/Bcl-2 ratio and inhibited phosphorylation of p38 MAPK. The data suggest that lupeol may exert protective effects against cerebral ischemia by suppressing oxidative stress and reduction of inflammation factors possible via activation of nuclear transcription factors and inhibition of cell death pathways.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Chongfu Xu
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Jiheng Hao
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Meng Zhang
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Zidong Wang
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Tengkun Yin
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Kai Lin
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Weidong Liu
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Qunlong Jiang
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Zhongchen Li
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Dan Wang
- Department of Ultrasound, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China
| | - Zhiqi Mao
- Department of Neurosurgery, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing City, 100853, People's Republic of China
| | - Huaiyu Tong
- Department of Neurosurgery, Chinese PLA General Hospital, No.28 Fuxing Road, Haidian District, Beijing City, 100853, People's Republic of China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng people's hospital, No. 67 Dongchang West Road, Liaocheng City, Shandong Province, 252000, People's Republic of China.
| |
Collapse
|
120
|
Lin YH, Liu HM. Update on cerebral hyperperfusion syndrome. J Neurointerv Surg 2020; 12:788-793. [PMID: 32414892 PMCID: PMC7402457 DOI: 10.1136/neurintsurg-2019-015621] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023]
Abstract
Cerebral hyperperfusion syndrome (CHS) is a clinical syndrome following a revascularization procedure. In the past decade, neurointerventional surgery has become a standard procedure to treat stenotic or occluded cerebral vessels in both acute and chronic settings, as well as endovascular thrombectomy in acute ischemic stroke. This review aims to summarize relevant recent studies regarding the epidemiology, diagnosis, and management of CHS as well as to highlight areas of uncertainty. Extracranial and intracranial cerebrovascular diseases in acute and chronic conditions are considered. The definition and diagnostic criteria of CHS are diverse. Although impaired cerebrovascular autoregulation plays a major role in the pathophysiology of CHS, the underlying mechanism is still not fully understood. Its clinical characteristics vary in different patients. The current findings on clinical and radiological presentation, pathophysiology, incidence, and risk factors are based predominantly on carotid angioplasty and stenting studies. Hemodynamic assessment using imaging modalities is the main form of diagnosis although the criteria are distinct, but it is helpful for patient selection before an elective revascularization procedure is conducted. After endovascular thrombectomy, a diagnosis of CHS is even more complex, and physicians should consider concomitant reperfusion injury. Management and preventative measures, including intensive blood pressure control before, during, and after revascularization procedures and staged angioplasty, are discussed in detail.
Collapse
Affiliation(s)
- Yen-Heng Lin
- Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Hon-Man Liu
- Radiology, National Taiwan University, Taipei, Taiwan .,Medical Imaging, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 24352, Taiwan
| |
Collapse
|
121
|
Argon Inhalation for 24 Hours After Onset of Permanent Focal Cerebral Ischemia in Rats Provides Neuroprotection and Improves Neurologic Outcome. Crit Care Med 2020; 47:e693-e699. [PMID: 31094741 DOI: 10.1097/ccm.0000000000003809] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES We tested the hypothesis that prolonged inhalation of 70% argon for 24 hours after in vivo permanent or temporary stroke provides neuroprotection and improves neurologic outcome and overall recovery after 7 days. DESIGN Controlled, randomized, double-blinded laboratory study. SETTING Animal research laboratories. SUBJECTS Adult Wistar male rats (n = 110). INTERVENTIONS Rats were subjected to permanent or temporary focal cerebral ischemia via middle cerebral artery occlusion, followed by inhalation of 70% argon or nitrogen in 30% oxygen for 24 hours. On postoperative day 7, a 48-point neuroscore and histologic lesion size were assessed. MEASUREMENTS AND MAIN RESULTS After argon inhalation for 24 hours immediately following "severe permanent ischemia" induction, neurologic outcome (neuroscore, p = 0.034), overall recovery (body weight, p = 0.02), and infarct volume (total infarct volume, p = 0.0001; cortical infarct volume, p = 0.0003; subcortical infarct volume, p = 0.0001) were significantly improved. When 24-hour argon treatment was delayed for 2 hours after permanent stroke induction or until after postischemic reperfusion treatment, neurologic outcomes remained significantly improved (neuroscore, p = 0.043 and p = 0.014, respectively), as was overall recovery (body weight, p = 0.015), compared with nitrogen treatment. However, infarct volume and 7-day mortality were not significantly reduced when argon treatment was delayed. CONCLUSIONS Neurologic outcome (neuroscore), overall recovery (body weight), and infarct volumes were significantly improved after 24-hour inhalation of 70% argon administered immediately after severe permanent stroke induction. Neurologic outcome and overall recovery were also significantly improved even when argon treatment was delayed for 2 hours or until after reperfusion.
Collapse
|
122
|
Malçok ÜA, Aras AB, Şehitoğlu MH, Akman T, Yüksel Y. Therapeutic effects of syringaldehyde on spinal cord ischemia in rabbits. Saudi Med J 2020; 41:341-350. [PMID: 32291420 PMCID: PMC7841612 DOI: 10.15537/smj.2020.4.24993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 02/04/2020] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES To investigate the effects of syringaldehyde (SA) on the antioxidant and oxidant system in spinal cord ischemia (SCI). METHODS These study and experiments were conducted at Medical Research Center, Çanakkale Onsekiz Mart University, Çanakkale, Turkey, between 2014-2018. Eighteen New Zealand White adult male rabbits were randomly divided into 3 groups (n=6). Superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), myeloperoxidase (MPO) activities, and malondialdehyde (MDA) levels were measured in the spinal cord tissues. Degenerated neurons, hemorrhage and in ammatory cell migration in the spinal cord were investigated histopathologically. Expressions of neuronal nitric oxide synthase (nNOS), caspase-3, and nuclear factor-κB (NF-κB) were evaluated immunohistochemically. Clinically, it was evaluated with modified Tarlov score. RESULTS Biochemically, there was an expected decrease in SOD, CAT, and GPx enzyme activities in ischemia groups, there was also an increase in MPO activity at the same time. When the enzyme activities spinal cord ischemia/ reperfusion (SCI/R)+SA, control and SCI/R groups were compared, the difference was found to be statistically significant (p less than 0.05). Glutathione peroxidase enzyme activity levels were very low in ischemia group compared to the significant increase in the SA group (p less than 0.05). Histopathologically, when SCI/R and SCI/R+SA groups were compared, there were statistically significant differences in the number of degenerative neurons and amount of hemorrhage; this comparison shows the significance of treatment in terms of inflammatory cell migration (p less than 0.05). The expressions of nNOS, caspase-3, and NF-κB were found significantly increased in SCI/R group compared to the control group (p less than 0.05). Syringaldehyde treatment decreased nNOS, caspase-3, and NF-κB expressions immunohistochemically. Clinical evaluation showed improvement in the SA-treated group. CONCLUSION Syringaldehyde therapy administered for protective purposes may reduce oxidative stress, degenerative changes and in ammatory cell migration in the ischemic spinal cord.Saudi Med J 2020; Vol. 41 (4): 341-350doi: 10.15537/smj.2020.4.24993 How to cite this article:Malçok UA, Aras AB, Şehitoğlu MH, Akman T, Yüksel Y. Therapeutic effects of syringaldehyde on spinal cord ischemia in rabbits. Saudi Med J 2020; Vol. 41: 341-350. doi: 10.15537/smj.2020.4.24993.
Collapse
Affiliation(s)
- Ümit A Malçok
- Department of Neurosurgery, School of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey. E-mail.
| | | | | | | | | |
Collapse
|
123
|
Zhang X, Shen R, Shu Z, Zhang Q, Chen Z. S100A12 promotes inflammation and apoptosis in ischemia/reperfusion injury via ERK signaling
in vitro
study using PC12 cells. Pathol Int 2020; 70:403-412. [DOI: 10.1111/pin.12924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 03/11/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Xiang Zhang
- Department of Neurosurgery, Shanghai Tenth People's HospitalTongji University Shanghai China
| | - Rui Shen
- Department of Neurosurgery, Shanghai Tenth People's HospitalTongji University Shanghai China
| | - Zhongwen Shu
- Department of Neurosurgery, Shanghai Tenth People's HospitalTongji University Shanghai China
| | - Quanbin Zhang
- Department of Neurosurgery, Shanghai Tenth People's HospitalTongji University Shanghai China
| | - Zuoquan Chen
- Department of Neurosurgery, Shanghai Tenth People's HospitalTongji University Shanghai China
| |
Collapse
|
124
|
Blood Pressure Management Following Acute Ischemic Stroke: A Review of Primary Literature. Crit Care Nurs Q 2020; 43:109-121. [PMID: 32084057 DOI: 10.1097/cnq.0000000000000297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Elevated blood pressure is common in patients with acute ischemic stroke. While this may occur secondary to the body's own response to preserve cerebral blood flow, elevated blood pressure may also increase the risk of hemorrhagic transformation. Current guidelines recommend various blood pressure goals based upon multiple factors, including thresholds specific to certain treatment interventions. Despite these guidelines, there is limited evidence to support specific blood pressure targets, and variability in clinical practice is common. The purpose of this review was to discuss blood pressure management in adult patients with acute ischemic stroke, focusing on appropriate targets in the setting of alteplase administration, mechanical thrombectomy, and hemorrhagic transformation.
Collapse
|
125
|
Electroacupuncture Pretreatment Elicits Neuroprotection Against Cerebral Ischemia-Reperfusion Injury in Rats Associated with Transient Receptor Potential Vanilloid 1-Mediated Anti-Oxidant Stress and Anti-Inflammation. Inflammation 2020; 42:1777-1787. [PMID: 31190106 DOI: 10.1007/s10753-019-01040-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Electroacupuncture (EA) pretreatment, electrical stimulation using metal needle at specific acupoints in advance, possesses the potential to prevent cerebral ischemia-reperfusion injury (CIRI). Transient receptor potential vanilloid 1 (TRPV-1) has been indicated to take part in cerebral protection of EA; however, the detailed mechanisms remain unclear. The aim of this study was to investigate whether neuroprotection of EA pretreatment against CIRI is associated with TRPV-1 and explore the underlying mechanisms. Middle cerebral artery occlusion (MCAO) was performed to induce CIRI after EA pretreatment at Baihui (GV20), bilateral Shenshu (BL23), and Sanyinjiao (SP6) acupoints in rats. Neurological deficit scores, infarct volumes, oxidative stress damage, inflammatory cytokine production, MAPK signaling activation, and the expression of TRPV-1 were assessed. EA pretreatment lowered neurological deficit scores, reduced infarct volumes, impeded oxidative stress injury, inhibited inflammatory cytokine production, curbed P38 phosphorylation, and suppressed TRPV-1 expression in MCAO rats. Attributing to inhibition of TRPV-1 expression, AMG-517 (TRPV-1 antagonist) showed the synergistic effect with EA pretreatment on the neuroprotection against ischemia-reperfusion injury. However, TRPV-1 agonists capsaicin significantly abrogated the neuroprotective effects of EA pretreatment in MCAO rats accompanying enhancement of TRPV-1 expression. These findings indicated EA pretreatment exerted neuroprotection in rats with cerebral ischemia-reperfusion injury, which at least partially were associated with TRPV1-mediated anti-oxidant stress and anti-inflammation via inhibiting P38 MAPK activation.
Collapse
|
126
|
Zhao W, Wu C, Dornbos D, Li S, Song H, Wang Y, Ding Y, Ji X. Multiphase adjuvant neuroprotection: A novel paradigm for improving acute ischemic stroke outcomes. Brain Circ 2020; 6:11-18. [PMID: 32166195 PMCID: PMC7045534 DOI: 10.4103/bc.bc_58_19] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/29/2019] [Accepted: 01/17/2020] [Indexed: 12/24/2022] Open
Abstract
While several large pivotal clinical trials recently revealed a substantial benefit of endovascular thrombectomy for acute ischemic stroke (AIS) caused by large-vessel occlusion, many patients still experience mediocre prognosis. Enlargement of the ischemic core, failed revascularization, incomplete reperfusion, distal embolization, and secondary reperfusion injury substantially impact the salvaging of brain tissue and the functional outcomes of AIS. Here, we propose novel concept of “Multiphase Adjuvant Neuroprotection” as a new paradigm that may help guide our search for adjunctive treatments to be used together with thrombectomy. The premise of multiphase adjuvant neuroprotection is based on the diverse and potentially nonoverlapping pathophysiologic mechanisms that are triggered before, during, and after thrombectomy therapies. Before thrombectomy, strategies should focus on preventing the growth of the ischemic core; during thrombectomy, improving recanalization while reducing distal embolization and maximizing reperfusion are of significant importance; after reperfusion, strategies should focus on seeking targets to reduce secondary reperfusion injury. The concept of multiphase adjuvant neuroprotection, wherein different strategies are employed throughout the various phases of clinical care, might provide a paradigm to minimize the final infarct size and improve functional outcome in AIS patients treated with thrombectomy. With the success of thrombectomy in selected AIS patients, there is now an opportunity to revisit stroke neuroprotection. Notably, if the underlying mechanisms of these neuroprotective strategies are identified, their role in the distinct phases will provide further avenues to improve patient outcomes of AIS.
Collapse
Affiliation(s)
- Wenbo Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chuanjie Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - David Dornbos
- Department of Neurological Surgery, Semmes-Murphey Clinic and University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haiqing Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuping Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
127
|
Williams EI, Betterton RD, Davis TP, Ronaldson PT. Transporter-Mediated Delivery of Small Molecule Drugs to the Brain: A Critical Mechanism That Can Advance Therapeutic Development for Ischemic Stroke. Pharmaceutics 2020; 12:pharmaceutics12020154. [PMID: 32075088 PMCID: PMC7076465 DOI: 10.3390/pharmaceutics12020154] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Ischemic stroke is the 5th leading cause of death in the United States. Despite significant improvements in reperfusion therapies, stroke patients still suffer from debilitating neurocognitive deficits. This indicates an essential need to develop novel stroke treatment paradigms. Endogenous uptake transporters expressed at the blood-brain barrier (BBB) provide an excellent opportunity to advance stroke therapy via optimization of small molecule neuroprotective drug delivery to the brain. Examples of such uptake transporters include organic anion transporting polypeptides (OATPs in humans; Oatps in rodents) and organic cation transporters (OCTs in humans; Octs in rodents). Of particular note, small molecule drugs that have neuroprotective properties are known substrates for these transporters and include 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (i.e., statins) for OATPs/Oatps and 1-amino-3,5-dimethyladamantane (i.e., memantine) for OCTs/Octs. Here, we review current knowledge on specific BBB transporters that can be targeted for improvement of ischemic stroke treatment and provide state-of-the-art perspectives on the rationale for considering BBB transport properties during discovery/development of stroke therapeutics.
Collapse
|
128
|
Transcriptional activation of antioxidant gene expression by Nrf2 protects against mitochondrial dysfunction and neuronal death associated with acute and chronic neurodegeneration. Exp Neurol 2020; 328:113247. [PMID: 32061629 DOI: 10.1016/j.expneurol.2020.113247] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/04/2020] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
Abstract
Mitochondria are both a primary source of reactive oxygen species (ROS) and a sensitive target of oxidative stress; damage to mitochondria can result in bioenergetic dysfunction and both necrotic and apoptotic cell death. These relationships between mitochondria and cell death are particularly strong in both acute and chronic neurodegenerative disorders. ROS levels are affected by both the production of superoxide and its toxic metabolites and by antioxidant defense mechanisms. Mitochondrial antioxidant activities include superoxide dismutase 2, glutathione peroxidase and reductase, and intramitochondrial glutathione. When intracellular conditions disrupt the homeostatic balance between ROS production and detoxification, a net increase in ROS and an oxidized shift in cellular redox state ensues. Cells respond to this imbalance by increasing the expression of genes that code for proteins that protect against oxidative stress and inhibit cytotoxic oxidation of proteins, DNA, and lipids. If, however, the genomic response to mitochondrial oxidative stress is insufficient to maintain homeostasis, mitochondrial bioenergetic dysfunction and release of pro-apoptotic mitochondrial proteins into the cytosol initiate a variety of cell death pathways, ultimately resulting in potentially lethal damage to vital organs, including the brain. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a translational activating protein that enters the nucleus in response to oxidative stress, resulting in increased expression of numerous cytoprotective genes, including genes coding for mitochondrial and non-mitochondrial antioxidant proteins. Many experimental and some FDA-approved drugs promote this process. Since mitochondria are targets of ROS, it follows that protection against mitochondrial oxidative stress by the Nrf2 pathway of gene expression contributes to neuroprotection by these drugs. This document reviews the evidence that Nrf2 activation increases mitochondrial antioxidants, thereby protecting mitochondria from dysfunction and protecting neural cells from damage and death. New experimental results are provided demonstrating that post-ischemic administration of the Nrf2 activator sulforaphane protects against hippocampal neuronal death and neurologic injury in a clinically-relevant animal model of cardiac arrest and resuscitation.
Collapse
|
129
|
Gad SN, Nofal S, Raafat EM, Ahmed AAE. Lixisenatide Reduced Damage in Hippocampus CA1 Neurons in a Rat Model of Cerebral Ischemia-Reperfusion Possibly Via the ERK/P38 Signaling Pathway. J Mol Neurosci 2020; 70:1026-1037. [PMID: 32040827 DOI: 10.1007/s12031-020-01497-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is a gut-derived peptide that has various physiological actions. One of its main actions is the regulation of blood glucose level when it is elevated as it potentiates insulin release. It is also known that GLP-1 protects neurons from damage caused by neurodegenerative diseases. Lixisenatide is one of the GLP-1 analogues that has a strong affinity to the GLP-1 receptor. Experimental animal studies have shown that it holds a neuroprotective effect in Parkinson, myocardial, and cerebral ischemic disease animal models. The beneficial effect of lixisenatide on the brain after cerebral ischemia-reperfusion (I/R) is not clarified yet; thus, it needs further explanatory studies. Our research is the first to study the effect of lixisenatide on myeloperoxidase (MPO) and toll-like receptors (TLRs)/mitogen-activated protein kinase (MAPK) pathway in a rat model of cerebral I/R. Lixisenatide with 2 doses 0.7 and 7 nmol/kg was given intraperitoneal in 2 different groups for 14 days; then, the bilateral common carotid artery was occluded for 1 h followed by reperfusion for 1 h. Examination of hippocampus CA1 neurons by Nissl stain showed that the number of intact neurons was elevated in the lixisenatide-treated group related to the control group (I/R group). Lixisenatide exhibited neuroprotection action possibly via downregulation of MPO, TLR2/4, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and pP38 and upregulation of phosphorylated extracellular signal-regulated kinase (pERK1/2); thus, this study gives possible link between lixisenatide and TLR/MAPK pathway following cerebral I/R and supports the use of lixisenatide for neuroprotection against stroke.
Collapse
Affiliation(s)
- Salma N Gad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt.
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Eman M Raafat
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Amany A E Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
130
|
Wang Y, Xiao G, He S, Liu X, Zhu L, Yang X, Zhang Y, Orgah J, Feng Y, Wang X, Zhang B, Zhu Y. Protection against acute cerebral ischemia/reperfusion injury by QiShenYiQi via neuroinflammatory network mobilization. Biomed Pharmacother 2020; 125:109945. [PMID: 32028240 DOI: 10.1016/j.biopha.2020.109945] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Cerebral ischemia/reperfusion injury (CI/RI) is a common feature of ischemic stroke, involving a period of impaired blood supply to the brain, followed by the restoration of cerebral perfusion through medical intervention. Although ischemia and reperfusion brain damage is a complex pathological process with an unclear physiological mechanism, more attention is currently focused on the neuroinflammatory response of an ischemia/reperfusion origin, and anti-inflammatory appears to be a potential therapeutic strategy following ischemic stroke. QiShenYiQi (QSYQ), a component-based Chinese medicine with Qi-tonifying and blood-activating property, has pharmacological actions of anti-inflammatory, antioxidant, mitochondrial protectant, anti-apoptosis, and antiplatelet aggregation. We have previously reported that the cardioprotective effect of QSYQ against ischemia/reperfusion injury is via improvement of mitochondrial functional integrity. In this research work, we aimed to investigate the possible mechanism involved in the neuroprotection of QSYQ in mice model of cerebral ischemia/reperfusion injury based on the inflammatory pathway. The cerebral protection was evaluated in the stroke mice after 24 h reperfusion by assessing the neurological deficit, cerebral infarction, brain edema, BBB functionality, and via histopathological assessment. TCM-based network pharmacology method was performed to establish and analyze compound-target-disease & function-pathway network so as to find the possible mechanism linking to the role of QSYQ in CI/RI. In addition, RT-qPCR was used to verify the accuracy of predicted signaling gene expression. As a result, improvement of neurological outcome, reduction of infarct volume and brain edema, a decrease in BBB disruption, and amelioration of histopathological alteration were observed in mice pretreated with QSYQ after experimental stroke surgery. Network pharmacology analysis revealed neuroinflammatory response was associated with the action of QSYQ in CI/RI. RT-qPCR data showed that the mice pretreated with QSYQ could significantly decrease IFNG-γ, IL-6, TNF-α, NF-κB p65, and TLR-4 mRNA levels and increase TGF-β1 mRNA level in the brain compared to the untreated mice after CI/RI (p < 0.05). In conclusion, our study indicated the cerebral protective effect of pretreatment with QSYQ against CI/RI, which may be partly related to its potential to the reduction of neuroinflammatory response in a stroke subject.
Collapse
Affiliation(s)
- Yule Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xinyan Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Lin Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xinyue Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Yiqian Zhang
- State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tianjin Tasly Holding Group Co., Ltd., Tianjin, China
| | - John Orgah
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xiaoying Wang
- Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Boli Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China.
| |
Collapse
|
131
|
Naderi Y, Panahi Y, Barreto GE, Sahebkar A. Neuroprotective effects of minocycline on focal cerebral ischemia injury: a systematic review. Neural Regen Res 2020; 15:773-782. [PMID: 31719236 PMCID: PMC6990777 DOI: 10.4103/1673-5374.268898] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
To review the neuroprotective effects of minocycline in focal cerebral ischemia in animal models. By searching in the databases of PubMed, ScienceDirect, and Scopus, and considering the inclusion and exclusion criteria of the study. Studies were included if focal cerebral ischemia model was performed in mammals and including a control group that has been compared with a minocycline group. Written in languages other than English; duplicate data; in vitro studies and combination of minocycline with other neuroprotective agents were excluded. Neurological function of patients was assessed by National Institute of Health Stroke Scale, modified Rankin Scale, and modified Barthel Index. Neuroprotective effects were assessed by detecting the expression of inflammatory cytokines. We examined 35 papers concerning the protective effects of minocycline in focal cerebral ischemia in animal models and 6 clinical trials which had evaluated the neuroprotective effects of minocycline in ischemic stroke. These studies revealed that minocycline increases the viability of neurons and decreases the infarct volume following cerebral ischemia. The mechanisms that were reported in these studies included anti-inflammatory, antioxidant, as well as anti-apoptotic effects. Minocycline also increases the neuronal regeneration following cerebral ischemia. Minocycline has considerable neuroprotective effects against cerebral ischemia-induced neuronal damages. However, larger clinical trials may be required before using minocycline as a neuroprotective drug in ischemic stroke.
Collapse
Affiliation(s)
- Yazdan Naderi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yunes Panahi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Amirhosein Sahebkar
- Halal Research Center of IRI, FDA, Tehran; Biotechnology Research Center, Pharmaceutical Technology Institute; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
132
|
Mo Z, Tang C, Li H, Lei J, Zhu L, Kou L, Li H, Luo S, Li C, Chen W, Zhang L. Eicosapentaenoic acid prevents inflammation induced by acute cerebral infarction through inhibition of NLRP3 inflammasome activation. Life Sci 2019; 242:117133. [PMID: 31830477 DOI: 10.1016/j.lfs.2019.117133] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/27/2019] [Accepted: 11/29/2019] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Acute cerebral infarction (ACI) is the most common type of acute cerebrovascular diseases resulting in high rate of death and disability. Numerous evidences show that inflammation is the leading cause of ischemic brain injury, thus anti-inflammatory therapy is an attractive candidate for ischemic brain damage. Eicosapentaenoic acid (EPA) exerts anti-inflammatory activity in lots of human inflammatory diseases, whereas its effect in ACI is left to elucidate. METHOD Nlpr3-/- mice, Gpr40-/-; Gpr120-/- mice and mice with right middle cerebral artery occlusion (MCAO) were used to detect NLR family pyrin domain containing 3 (NLRP3) inflammasome activation by Western Blot and the release of proinflammatory cytokines by ELISA. To estimate the acute ischemic condition in vitro, oxygen-glucose deprivation (OGD) was induced in BV2 microglia cells. Transfection of the shRNA targeting GPR40 and GPR120 mRNA into BV2 cells was also assessed. Apoptosis in ischemic cerebral tissues and BV2 cells was detected by terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay and flow cytometry. RESULT Here we show that EPA suppresses ACI-induced inflammatory responses through blocking NLRP3 inflammasome activation. In addition, EPA inhibits NLRP3 inflammasome activation through G protein-coupled receptor 40 (GPR40) and GPR120. Importantly, EPA ameliorates ACI-induced apoptosis. CONCLUSION EPA exerts beneficial effect on ACI-induced inflammation through blocking NLRP3 inflammasome activation by GPR40 and GPR120. Our findings suggest the potential clinical use of EPA in ACI.
Collapse
Affiliation(s)
- Zhihuai Mo
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Chaogang Tang
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China; Department of Neurology, Maoming People's Hospital, 525000, Guangdong, China
| | - Huiqing Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Junjie Lei
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Lingjuan Zhu
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Li Kou
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Hao Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China; Department of Neurology, Maoming People's Hospital, 525000, Guangdong, China
| | - Shijian Luo
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Chunyi Li
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China
| | - Wenli Chen
- Department of Pharmacology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China.
| | - Lei Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Guangdong, China.
| |
Collapse
|
133
|
Kwon JI, Woo CW, Kim KW, Choi Y, Kim ST, Kim YJ, Kang J, Lee DW, Tak E, Kim JK, Jung SC, Kim TH, Woo DC. Does the Apparent Diffusion Coefficient Value Predict Permanent Cerebral Ischemia/Reperfusion Injury in Rats? Acad Radiol 2019; 26:e348-e354. [PMID: 30661976 DOI: 10.1016/j.acra.2018.12.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 10/27/2022]
Abstract
RATIONALE AND OBJECTIVES Variation in tissue damage after cerebral ischemia/reperfusion (I/R) can cause uncertainty in stroke-related studies, which can be reduced if the damage can be predicted early after ischemia by measuring the apparent diffusion coefficient (ADC). We investigated whether ADC measurement in the acute phase can predict permanent cerebral I/R injury. MATERIALS AND METHODS The middle cerebral artery occlusion model was established using the intraluminal suture method to induce 60 minutes of ischemia followed by reperfusion in rats. T2-weighted images and diffusion-weighted images were obtained at 30 minutes and 24 hours after ischemia. Neuronal cell survival was assessed by neuronal nuclei (NeuN) immunofluorescence staining. The correlation between relative ADC (rADC) values at 30 minutes and I/R injury at 24 hours after ischemia was analyzed. Magnetic resonance imaging results were confirmed by histologic analysis. RESULTS The correlation between rADC values at 30 minutes and 24 hours was strong in the ischemic core and peri-infarct region but moderate in the anterior choroidal and hypothalamic region. Histologic analysis revealed that the correlation between rADC values at 30 minutes and the number of NeuN-positive cells at 24 hours was strong in the ischemic core and peri-infarct region but moderate in the anterior choroidal and hypothalamic region. Furthermore, there was a strong positive correlation between the sum of rADC values of three regions at 30 minutes and the infarct volume at 24 hours. CONCLUSION ADC measurement in the acute phase can predict permanent cerebral I/R injury and provide important information for the evaluation of ischemic stroke.
Collapse
|
134
|
Shah FA, Li T, Kury LTA, Zeb A, Khatoon S, Liu G, Yang X, Liu F, Yao H, Khan AU, Koh PO, Jiang Y, Li S. Pathological Comparisons of the Hippocampal Changes in the Transient and Permanent Middle Cerebral Artery Occlusion Rat Models. Front Neurol 2019; 10:1178. [PMID: 31798514 PMCID: PMC6868119 DOI: 10.3389/fneur.2019.01178] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/22/2019] [Indexed: 01/04/2023] Open
Abstract
Ischemic strokes are categorized by permanent or transient obstruction of blood flow, which impedes delivery of oxygen and essential nutrients to brain. In the last decade, the therapeutic window for tPA has increased from 3 to 5-6 h, and a new technique, involving the mechanical removal of the clot (endovascular thrombectomy) to allow reperfusion of the injured area, is being used more often. This last therapeutic approach can be done until 24 h after stroke onset. Due to this fact, more acute ischemic stroke patients are now being recanalized, and so tMCAO is probably the "best" model to address these patients that have a potential good outcome in terms of survival and functional recovery. However, permanent occlusion patients are also important, not only to increase survival rate but also to improve functional outcomes, although these are more difficult to achieve. So, both models are important, and which target different stroke patients in the clinical scenario. Hippocampus has a vital role in memory and cognition, is prone to ischemic induced neurodegeneration. This study was designed to delineate the molecular, pathological, and neurological changes in rat models of t-MCAO, permanent MCAO (pMCAO), and pMCAO with diabetic conditions in hippocampal tissue. Our results showed that these three models showed distinct discrepancies at numerous pathological process, including key signaling molecules involved in neuronal apoptosis, glutamate induced excitotoxicity, neuroinflammation, oxidative stress, and neurotrophic changes. Our result suggests that the two commonly used MCAO models exhibited tremendous differences in terms of neuronal cell loss, glutamate excitotoxic related signaling, synaptic transmission markers, neuron inflammatory and oxidative stress molecules. These differences may reflect the variations in different models, which may provide valuable information for mechanistic and therapeutic inconsistences as experienced in both preclinical models and clinical trials.
Collapse
Affiliation(s)
- Fawad Ali Shah
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.,Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, International University, Islamabad, Pakistan
| | - Tao Li
- Department of Forensic Medicine, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Lina Tariq Al Kury
- College of Natural and Health Sciences, Zayed University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Alam Zeb
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, International University, Islamabad, Pakistan
| | - Shehla Khatoon
- Department of Anatomy, Khyber Medical College, Khyber Medical University, Peshawar, Pakistan
| | - Gongping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Fang Liu
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Campbell Research Institute, Toronto, ON, Canada
| | - Huo Yao
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Arif-Ullah Khan
- Department of Pharmacology, Riphah Institute of Pharmaceutical Sciences, International University, Islamabad, Pakistan
| | - Phil Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju-si, South Korea
| | - Yuhua Jiang
- Cancer Centre, The Second Hospital of Shandong University, Jinan, China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Campbell Research Institute, Toronto, ON, Canada
| |
Collapse
|
135
|
The Roles of GABA in Ischemia-Reperfusion Injury in the Central Nervous System and Peripheral Organs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4028394. [PMID: 31814874 PMCID: PMC6878816 DOI: 10.1155/2019/4028394] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/27/2019] [Accepted: 10/18/2019] [Indexed: 12/30/2022]
Abstract
Ischemia-reperfusion (I/R) injury is a common pathological process, which may lead to dysfunctions and failures of multiple organs. A flawless medical way of endogenous therapeutic target can illuminate accurate clinical applications. γ-Aminobutyric acid (GABA) has been known as a marker in I/R injury of the central nervous system (mainly in the brain) for a long time, and it may play a vital role in the occurrence of I/R injury. It has been observed that throughout cerebral I/R, levels, syntheses, releases, metabolisms, receptors, and transmissions of GABA undergo complex pathological variations. Scientists have investigated the GABAergic enhancers for attenuating cerebral I/R injury; however, discussions on existing problems and mechanisms of available drugs were seldom carried out so far. Therefore, this review would summarize the process of pathological variations in the GABA system under cerebral I/R injury and will cover corresponding probable issues and mechanisms in using GABA-related drugs to illuminate the concern about clinical illness for accurately preventing cerebral I/R injury. In addition, the study will summarize the increasing GABA signals that can prevent I/R injuries occurring in peripheral organs, and the roles of GABA were also discussed correspondingly.
Collapse
|
136
|
Li J, Gu Z, Liu Y, Wang Y, Zhao M. Astilbin attenuates cerebral ischemia/reperfusion injury by inhibiting the TLR4/MyD88/NF-κB pathway. Toxicol Res (Camb) 2019; 8:1002-1008. [PMID: 34055311 PMCID: PMC8142934 DOI: 10.1039/c9tx00222g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/18/2019] [Indexed: 12/25/2022] Open
Abstract
Ischemic stroke is the second most common cause of death worldwide and cerebral ischemia/reperfusion (I/R) injury also leads to serious tissue damage. Astilbin, a natural bioactive flavonoid compound, has been reported to have protective effects on neurological diseases. This study aims to investigate the effects of astilbin on cerebral I/R injury and determine the mechanisms involved. The results demonstrated that, in cerebral I/R rats, astilbin could attenuate I/R injury in the hippocampal region, decreasing the activity of lactate dehydrogenase (LDH) and malondialdehyde (MDA) in the rat brain. Astilbin also inhibited the I/R-induced upregulation of pro-inflammatory mediators (TNFα, IL-1β, IL-6). Similarly, in hypoxia/reperfusion (H/R) treated human neuroblastoma cells, astilbin could increase the cell viability of SH-SY5Y, decrease the activity of LDH and MDA, and inhibit the H/R-induced upregulation of pro-inflammatory mediators. For the mechanism study, western blot results indicated that astilbin could inhibit the expression of Toll-like receptor 4 (TLR4), myeloid differential protein 88 (MYD88) and phosphorylated NF-κB p65 in H/R treated SH-SY5Y cells. The research indicated that astilbin ameliorated cerebral I/R injury partly via the TLR4/MyD88/NF-κB pathway. Astilbin may have potential therapeutic effects on cerebral ischemia.
Collapse
Affiliation(s)
- Jing Li
- Changchun University of Chinese Medicine , Changchun City , Jilin Province 130000 , China . ; Tel: +86-0431-81953783
| | - Zhaowei Gu
- Changchun University of Chinese Medicine , Changchun City , Jilin Province 130000 , China . ; Tel: +86-0431-81953783
| | - Yue Liu
- Changchun University of Chinese Medicine , Changchun City , Jilin Province 130000 , China . ; Tel: +86-0431-81953783
| | - Yu Wang
- Changchun University of Chinese Medicine , Changchun City , Jilin Province 130000 , China . ; Tel: +86-0431-81953783
| | - Min Zhao
- Changchun University of Chinese Medicine , Changchun City , Jilin Province 130000 , China . ; Tel: +86-0431-81953783
| |
Collapse
|
137
|
Gomez J, Wolfe S, Sarwal A. Sonographic Demonstration of a Perfusion-Dependent Stroke with Negative MRI and a Flow-Limiting Stenosis. Neurocrit Care 2019; 32:883-888. [PMID: 31664625 DOI: 10.1007/s12028-019-00837-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Jonathan Gomez
- Department of Neurology, Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| | - Stacey Wolfe
- Department of Neurosurgery, Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Aarti Sarwal
- Department of Neurology, Wake Forest Baptist Medical Center, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| |
Collapse
|
138
|
A novel free radical scavenger, NSP-116, ameliorated the brain injury in both ischemic and hemorrhagic stroke models. J Pharmacol Sci 2019; 141:119-126. [PMID: 31679961 DOI: 10.1016/j.jphs.2019.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/13/2019] [Accepted: 09/24/2019] [Indexed: 12/28/2022] Open
Abstract
Reperfusion injury is a serious problem in ischemic stroke therapy, which leads to neuronal damage and intracranial hemorrhage (ICH). A novel free radical scavenger, NSP-116, has anti-oxidative effect and may ameliorate reperfusion injury. The purpose of this study was to investigate the effects of NSP-116 on both ischemic and hemorrhagic stroke models. First, we assessed whether NSP-116 has protective effects in vitro. Pre-treatment of NSP-116 decreased neuronal cell damage induced by H2O2 or LPS. Moreover, NSP-116 also suppressed mitochondria damage and apoptosis in H2O2-induced neuronal injury model. Based on these results, we used a middle cerebral artery occlusion (MCAO)-induced ischemic stroke model or a collagenase-induced ICH model. Using the MCAO model, we evaluated the cerebral blood flow (CBF), neurological deficit, and infarct volume. Hematoma volume was assessed at 3 days after ICH. In the MCAO model, oral administration of NSP-116 at 30 mg/kg attenuated the reduction of CBF, neurological deficits, and infarct formation. Interestingly, NSP-116 also ameliorated hematoma expansion and neurological deficits in the ICH model. Additionally, pre-treatment of NSP-116 suppressed the brain microvascular endothelial cell death induced by collagenase treatment. Collectively, our findings indicated that oral administration of NSP-116 attenuates both ischemic and hemorrhagic brain injuries after stroke.
Collapse
|
139
|
Cavarsan CF, Gorassini MA, Quinlan KA. Animal models of developmental motor disorders: parallels to human motor dysfunction in cerebral palsy. J Neurophysiol 2019; 122:1238-1253. [PMID: 31411933 PMCID: PMC6766736 DOI: 10.1152/jn.00233.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cerebral palsy (CP) is the most common motor disability in children. Much of the previous research on CP has focused on reducing the severity of brain injuries, whereas very few researchers have investigated the cause and amelioration of motor symptoms. This research focus has had an impact on the choice of animal models. Many of the commonly used animal models do not display a prominent CP-like motor phenotype. In general, rodent models show anatomically severe injuries in the central nervous system (CNS) in response to insults associated with CP, including hypoxia, ischemia, and neuroinflammation. Unfortunately, most rodent models do not display a prominent motor phenotype that includes the hallmarks of spasticity (muscle stiffness and hyperreflexia) and weakness. To study motor dysfunction related to developmental injuries, a larger animal model is needed, such as rabbit, pig, or nonhuman primate. In this work, we describe and compare various animal models of CP and their potential for translation to the human condition.
Collapse
Affiliation(s)
- Clarissa F Cavarsan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Monica A Gorassini
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Katharina A Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| |
Collapse
|
140
|
Chao HC, Lee TH, Chiang CS, Yang SY, Kuo CH, Tang SC. Sphingolipidomics Investigation of the Temporal Dynamics after Ischemic Brain Injury. J Proteome Res 2019; 18:3470-3478. [PMID: 31310127 DOI: 10.1021/acs.jproteome.9b00370] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sphingolipids (SPLs) have been proposed as potential therapeutic targets for strokes, but no reports have ever profiled the changes of the entire range of SPLs after a stroke. This study applied sphingolipidomic methods to investigate the temporal and individual changes in the sphingolipidome including the effect of atorvastatin after ischemic brain injury. We conducted sphingolipidomic profiling of mouse brain tissue by liquid chromatography-electrospray ionization tandem mass spectrometry at 3 h and 24 h after 1 h of middle cerebral artery occlusion (MCAO), and SPL levels were compared with those of the Sham control group. At 3 h post-MCAO, ceramides (Cers) exhibited an increase in levels of long-chain Cers but a decrease in very-long-chain Cers. Moreover, sphingosine, the precursor of sphingosine-1-phosphate (S1P), decreased and S1P increased at 3 h after MCAO. In contrast to 3 h, both long-chain and very-long-chain Cers showed an increased trend at 24 h post-MCAO. Most important, the administration of atorvastatin improved the neurological function of the mice and significantly reversed the SPL changes resulting from the ischemic injury. Furthermore, we used plasma samples from nonstroke control and stroke patients at time points of 72 h after a stroke, and found a similar trend of Cers as in the MCAO model. This study successfully elucidated the overall effect of ischemic injury on SPL metabolism with and without atorvastatin treatment. The network of SPL components that change upon ischemic damage may provide novel therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Hsi-Chun Chao
- School of Pharmacy, College of Medicine , National Taiwan University , Taipei 100 , Taiwan.,The Metabolomics Core Laboratory, Center of Genomic Medicine , National Taiwan University , Taipei 100 , Taiwan
| | - Tsung-Heng Lee
- School of Pharmacy, College of Medicine , National Taiwan University , Taipei 100 , Taiwan.,The Metabolomics Core Laboratory, Center of Genomic Medicine , National Taiwan University , Taipei 100 , Taiwan
| | - Chien-Sung Chiang
- Stroke Center and Department of Neurology , National Taiwan University Hospital , Taipei 100 , Taiwan
| | - Sin-Yu Yang
- Stroke Center and Department of Neurology , National Taiwan University Hospital , Taipei 100 , Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine , National Taiwan University , Taipei 100 , Taiwan.,The Metabolomics Core Laboratory, Center of Genomic Medicine , National Taiwan University , Taipei 100 , Taiwan.,Department of Pharmacy , National Taiwan University Hospital , Taipei 100 , Taiwan
| | - Sung-Chun Tang
- Stroke Center and Department of Neurology , National Taiwan University Hospital , Taipei 100 , Taiwan
| |
Collapse
|
141
|
Tetrahydroxystilbene Glucoside Suppresses NAPDH Oxidative Stress to Mitigate Apoptosis and Autophagy Induced by Cerebral Ischemia/Reperfusion Injury in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3913981. [PMID: 31379960 PMCID: PMC6662418 DOI: 10.1155/2019/3913981] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/02/2019] [Accepted: 06/24/2019] [Indexed: 11/18/2022]
Abstract
Tetrahydroxystilbene glucoside (TSG) is the active ingredient extracted from the traditional Chinese medicine Fallopia multiflora, which has extensive pharmacological activities. The current study aimed to observe the neuroprotective mechanism of TSG in the ischemia/reperfusion (I/R) brain injury-induced apoptosis and autophagy from the point of view of oxidative stress (OS). The middle cerebral artery occlusion (MCAO) model was prepared through the suture-occluded method, and TSG was administered through tail vein injection at the time of reperfusion at the doses of 3.0, 6.0, and 12.0 mg/kg. Compared with sham group, the neurological score in I/R mice was increased (P<0.05), along with remarkably elevated cerebral infarct volume (P<0.05); while TSG administration could reduce the neurological score and cerebral infarct volume (P<0.05) and improve the neuronal damage in ischemic cortex and hippocampus (P<0.05). The expression of NOX4, activated caspase-3(9), and Beclin 1 (P<0.05), as well as the LC3BII/I ratio, had been markedly elevated (P<0.05), while TSG administration could effectively suppress the expression of the above-mentioned proteins (P<0.05). In conclusion, TSG shows obvious protection against brain injury in I/R mice, and its mechanism may be related to suppressing the NADPH-induced OS and reducing neuronal apoptosis as well as autophagy.
Collapse
|
142
|
Tao L, Yu Q, Zhao P, Yang Q, Wang B, Yang Y, Kuai J, Ding Q. Preconditioning with hydrogen sulfide ameliorates cerebral ischemia/reperfusion injury in a mouse model of transient middle cerebral artery occlusion. Chem Biol Interact 2019; 310:108738. [PMID: 31283913 DOI: 10.1016/j.cbi.2019.108738] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/26/2019] [Accepted: 07/04/2019] [Indexed: 12/22/2022]
Abstract
Ischemic stroke and reperfusion injury are a common and serve medical situation in the elderly population. H2S is a gas neuromodulator which also possesses anti-oxidant and anti-inflammatory properties, and is found to play neuroprotective effect in neurodegenerative diseases. This study investigated the effect of endogenous and exogenous H2S in a mouse model of ischemic stroke. 129P2-Cbstm1Unc/J mice with heterozygous mutants in H2S generating enzyme cystathionine β-synthase were used to study the effect of endogenous H2S. H2S donor NaHS was used as exogenous H2S. Animals were pretreated with H2S and then subjected to middle cerebral artery occlusion surgery. Behavioral outcome was evaluated by novel object recognition test. Inflammatory cytokines were measured using ELISA. Western blot was used to detect the activation of NF-κB. Aged 129P2-Cbstm1Unc/J mice showed exaggerated inflammation and more severe cognitive impairment after ischemia, while exogenous H2S treatment inhibited inflammation and attenuated behavioral impairment. The anti-inflammatory effect of H2S was mediated by inhibiting NF-κB. Our findings suggest that both endogenous and exogenous H2S are involved in the neuroprotection against ischemia/reperfusion-induced cerebral injury.
Collapse
Affiliation(s)
- Lei Tao
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Qian Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Pin Zhao
- Department of Anesthesiology, Third Hospital of Xi'an, Xi'an, 710018, Shaanxi, China
| | - Qian Yang
- Department of Anesthesiology, Third Hospital of Xi'an, Xi'an, 710018, Shaanxi, China
| | - Binrong Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Yonghui Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Jianke Kuai
- Department of Anesthesiology, Third Hospital of Xi'an, Xi'an, 710018, Shaanxi, China.
| | - Qian Ding
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
143
|
Ding Y, Du J, Cui F, Chen L, Li K. The protective effect of ligustrazine on rats with cerebral ischemia-reperfusion injury via activating PI3K/Akt pathway. Hum Exp Toxicol 2019; 38:1168-1177. [PMID: 31250662 DOI: 10.1177/0960327119851260] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The study was to investigate the effects of ligustrazine on rats with cerebral ischemia-reperfusion (I/R) injury and to explore the potential mechanism. Transient focal cerebral ischemia Wistar rat model was established through middle cerebral artery occlusion. The cerebral I/R injury rats were treated with intraperitoneal injection of ligustrazine (1, 3, and 10 mg/kg). Human amniotic epithelial cells (HAECs) were treated with ligustrazine (1, 10, 100 μM) and PI3K inhibitor wortmannin (100 μM), following oxygen-glucose deprivation (OGD) treatment. The expression levels of protein kinase B (PKB or AKT), phospho-Akt (p-Akt), endothelial nitric oxide synthase (eNOS), and phosphor-eNOS (p-eNOS) in HAECs and brains of rats were measured by Western blot. The levels of nitric oxide (NO) in HAECs were measured by Griess method using NO2-/NO3- Assay Kit. Infarct volume and neurological deficits were evaluated 24 h after reperfusion. The levels of NO, p-Akt/Akt, and p-eNOS/eNOS in HAECs were significantly reduced after OGD, but ligustrazine treatment increased the levels of those factors in a dose-dependent manner, while those increases were reversed by PI3K inhibitor wortmannin. Similarly, p-Akt/Akt and p-eNOS/eNOS in brain tissue of rats with I/R were significantly reduced compared with control group (p < 0.05), but ligustrazine treatment increased the levels of p-Akt and p-eNOS in a dose-dependent manner (p < 0.05), while those increases were also reversed by using wortmannin. Ligustrazine also improved the damage of rat brain tissue caused by I/R, but wortmannin reversed the improvement. Ligustrazine plays a neuroprotective role in rats with cerebral I/R injury through the activation of PI3K/Akt pathway.
Collapse
Affiliation(s)
- Y Ding
- 1 Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - J Du
- 1 Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - F Cui
- 1 Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - L Chen
- 2 Outpatient Operating Room, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - K Li
- 1 Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| |
Collapse
|
144
|
Amaro S, Jiménez-Altayó F, Chamorro Á. Uric acid therapy for vasculoprotection in acute ischemic stroke. Brain Circ 2019; 5:55-61. [PMID: 31334357 PMCID: PMC6611195 DOI: 10.4103/bc.bc_1_19] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/18/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023] Open
Abstract
Uric acid (UA) is a product of the catabolism of purine nucleotides, the principal constituents of DNA, RNA, and cellular energy stores, such as adenosine triphosphate. The main properties of UA include scavenging of hydroxyl radicals, superoxide anion, hydrogen peroxide, and peroxynitrite that make this compound to be the most potent antioxidant in the human plasma. As the result of two silencing mutations in the gene of the hepatic enzyme uricase which degrades UA to allantoin, humans have higher levels of UA than most mammals. However, these levels rapidly decrease following an acute ischemic stroke (AIS), and this decrement has been associated to worse stroke outcomes. This review highlights the safety and potential clinical value of UA therapy in AIS, particularly in patients more exposed to redox-mediated mechanism following the onset of ischemia, such as women, hyperglycemic patients, or patients treated with mechanical thrombectomy. The clinical findings are supported by preclinical data gathered in different laboratories, and in assorted animal species which include male and female individuals or animals harboring comorbidities frequently encountered in patients with AIS, such as hyperglycemia or hypertension. A remarkable finding in these studies is that UA targets its main effects in the brain vasculature since available evidence suggests that does not seem to cross the blood–brain barrier. Altogether, the available data with UA therapy extend the importance of vasculoprotection for effective neuroprotection at the bedside and reinforce the role of endothelial cells after brain ischemia for an increased survival of the whole neurovascular unit.
Collapse
Affiliation(s)
- Sergi Amaro
- Comprehensive Stroke Center, Hospital Clínic, University of Barcelona, Barcelona, Spain.,Department of Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesc Jiménez-Altayó
- Department de Farmacologia, de Terapèutica i de Toxicologia, Facultat de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ángel Chamorro
- Comprehensive Stroke Center, Hospital Clínic, University of Barcelona, Barcelona, Spain.,Department of Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
145
|
Duan H, Huber M, Ding JN, Huber C, Geng X. Local endovascular infusion and hypothermia in stroke therapy: A systematic review. Brain Circ 2019; 5:68-73. [PMID: 31334359 PMCID: PMC6611196 DOI: 10.4103/bc.bc_9_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/03/2019] [Accepted: 05/16/2019] [Indexed: 01/01/2023] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide, but there are no effective, widely applicable stroke therapies. Systemic hypothermia is an international mainstay of postcardiac arrest care, and the neuroprotective benefits of systemic hypothermia following cerebral ischemia have been proven in clinical trials, but logistical issues hinder clinical acceptance. As a novel solution to these logistical issues, the application of local endovascular infusion of cold saline directly to the infarct site using a microcatheter has been put forth. In small animal models, the procedure has shown incredible neuroprotective promise on the biochemical, structural, and functional levels, and preliminary trials in large animals and humans have been similarly encouraging. In addition, the procedure would be relatively cost-effective and widely applicable. The administration of local endovascular hypothermia in humans is relatively simple, as this is a normal part of endovascular intervention for neuroendovascular surgeons. Therefore, it is expected that this new therapy could easily be added to an angiography suite. However, the neuroprotective efficacy in humans has yet to be determined, which is an end goal of researchers in the field. Given the potentially massive benefits, ease of induction, and cost-effective nature, it is likely that local endovascular hypothermia will become an integral part of endovascular treatment following ischemic stroke. This review outlines relevant research, discusses neuroprotective mechanisms, and discusses possibilities for future directions.
Collapse
Affiliation(s)
- Honglian Duan
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Mitchell Huber
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jessie N Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Christian Huber
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
146
|
Nan D, Jin H, Deng J, Yu W, Liu R, Sun W, Huang Y. Cilostazol ameliorates ischemia/reperfusion-induced tight junction disruption in brain endothelial cells by inhibiting endoplasmic reticulum stress. FASEB J 2019; 33:10152-10164. [PMID: 31184927 DOI: 10.1096/fj.201900326r] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endoplasmic reticulum (ER) stress is essential for brain ischemia/reperfusion (I/R) injury. However, whether it contributes to I/R-induced blood-brain barrier (BBB) injury remains unclear. cilostazol exerts protective effects toward I/R-induced BBB injury, with unclear mechanisms. This study explored the potential role of ER stress in I/R-induced endothelial cell damage and determined whether the therapeutic potential of cilostazol, with respect to I/R-induced endothelial cell damage, is related to inhibition of ER stress. We found that exposing brain endothelial cells (bEnd.3) to oxygen-glucose deprivation/reoxygenation (OGD/R) significantly activated ER stress and diminished the barrier function of cell monolayers; treatment with the ER stress inhibitor 4-phenylbutyric acid (4-PBA) or cilostazol prevented OGD/R-induced ER stress and preserved barrier function. Furthermore, OGD/R induced the expression and secretion of matrix metalloproteinase-9 and nuclear translocation of phosphorylated NF-κB. These changes were partially reversed by 4-PBA or cilostazol treatment. In vivo, 4-PBA or cilostazol significantly attenuated I/R-induced ER stress and ameliorated Evans blue leakage and tight junction loss. These results demonstrate that I/R-induced ER stress participates in BBB disruption. Targeting ER stress could be a useful strategy to protect the BBB from ischemic stroke, and cilostazol is a promising therapeutic agent for this process.-Nan, D., Jin, H., Deng, J., Yu, W., Liu, R., Sun, W., Huang, Y. Cilostazol ameliorates ischemia/reperfusion-induced tight junction disruption in brain endothelial cells by inhibiting endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Ding Nan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Weiwei Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Ran Liu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Weiping Sun
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
147
|
Abstract
Natural killer T (NKT) cells are a unique subset of T lymphocytes with the expression of T cell receptor (TCR) and NK cell lineage receptors. These cells can rapidly release large quantities of cytokines and function as a bridge between innate and adaptive immunity. To date, multiple reports have investigated the role of NKT cells under various pathological conditions, such as cancer, autoimmune disease, and infection. Knowledge about NKT cells in neurological diseases is increasing, albeit limited. Here, we review evidence for the involvement of NKT cells in neurological diseases, and discuss immunotherapeutic potential and future study goals. As the development and function of NKT cells become increasingly well understood, the next few years should yield many new insights into NKT cell function, and mechanistic regulation in neurological disorders.
Collapse
Affiliation(s)
- Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| | - Qi Wan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, China
| |
Collapse
|
148
|
Sun J, Yue F. Suppression of REDD1 attenuates oxygen glucose deprivation/reoxygenation-evoked ischemic injury in neuron by suppressing mTOR-mediated excessive autophagy. J Cell Biochem 2019; 120:14771-14779. [PMID: 31021470 DOI: 10.1002/jcb.28737] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 03/15/2019] [Accepted: 03/22/2019] [Indexed: 12/29/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) typically occurs after mechanical thrombectomy to treat ischemic stroke, generation of reactive oxygen species (ROS) after reperfusion may result in neuronal insult, ultimately leading to disability and death. Regulated in development and DNA damage responses 1 (REDD1) is a conserved stress response protein under various pathogenic conditions. Recent research confirms the controversial role of REDD1 in injury processes. Nevertheless, the role of REDD1 in cerebral I/R remains poorly defined. In the current study, increased expression of REDD1 was observed in neurons exposed to simulated I/R via oxygen glucose deprivation/reoxygenation (OGD/R) treatment. Knockdown of REDD1 enhanced OGD/R-inhibited cell viability, but suppressed lactate dehydrogenase (LDH) release in neurons upon OGD/R. Simultaneously, suppression of REDD1 also antagonized OGD/R-evoked cell apoptosis, Bax expression, and caspase-3 activity. Intriguingly, REDD1 depression abrogated neuronal oxidative stress under OGD/R condition by suppressing ROS, MDA generation, and increasing antioxidant SOD levels. Further mechanism analysis corroborated the excessive activation of autophagy in neurons upon OGD/R with increased expression of autophagy-related LC3 and Beclin-1, but decreased autophagy substrate p62 expression. Notably, REDD1 inhibition reversed OGD/R-triggered excessive neuronal autophagy. More importantly, depression of REDD1 also elevated the expression of p-mTOR. Preconditioning with mTOR inhibitor rapamycin engendered not only a reduction in mTOR activation, but also a reactivation of autophagy in REDD1 knockdown-neurons upon OGD/R. In addition, blocking the mTOR pathway muted the protective roles of REDD1 inhibition against OGD/R-induced neuron injury and oxidative stress. Together these data suggested that REDD1 may regulate I/R-induced oxidative stress injury in neurons by mediating mTOR-autophagy signaling, supporting a promising therapeutic strategy against brain ischemic diseases.
Collapse
Affiliation(s)
- Juguang Sun
- Department of Neurology, Xuzhou City Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu, China
| | - Fenglei Yue
- Department of Neurology, 521 Hospital of Norinco Group in Xi'an, Xi'an, Shaanxi, China
| |
Collapse
|
149
|
Carbone F, Busto G, Padroni M, Bernardoni A, Colagrande S, Dallegri F, Montecucco F, Fainardi E. Radiologic Cerebral Reperfusion at 24 h Predicts Good Clinical Outcome. Transl Stroke Res 2019; 10:178-188. [PMID: 29949087 DOI: 10.1007/s12975-018-0637-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/08/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022]
Abstract
Cerebral reperfusion and arterial recanalization are radiological features of the effectiveness of thrombolysis in acute ischemic stroke (AIS) patients. Here, an investigation of the prognostic role of early recanalization/reperfusion on clinical outcome was performed. In AIS patients (n = 55), baseline computerized tomography (CT) was performed ≤ 8 h from symptom onset, whereas CT determination of reperfusion/recanalization was assessed at 24 h. Multiple linear and logistic regression models were used to correlate reperfusion/recanalization with radiological (i.e., hemorrhagic transformation, ischemic core, and penumbra volumes) and clinical outcomes (assessed as National Institutes of Health Stroke Scale [NIHSS] reduction ≥ 8 points or a NIHSS ≤ 1 at 24 h and as modified Rankin Scale [mRS] < 2 at 90 days). At 24 h, patients achieving radiological reperfusion were n = 24, while the non-reperfused were n = 31. Among non-reperfused, n = 15 patients were recanalized. Radiological reperfusion vs. recanalization was also confirmed by early increased levels of circulating inflammatory biomarkers (i.e., serum osteopontin). In multivariate analysis, ischemic lesion volume reduction was associated with both recanalization (β = 0.265; p = 0.014) and reperfusion (β = 0.461; p < 0.001), but only reperfusion was independently associated with final infarct volume (β = - 0.333; p = 0.007). Only radiological reperfusion at 24 h predicted good clinical response at day 1 (adjusted OR 16.054 [1.423-181.158]; p = 0.025) and 90-day good functional outcome (adjusted OR 25.801 [1.483-448.840]; p = 0.026). At ROC curve analysis the AUC of reperfusion was 0.777 (p < 0.001) for the good clinical response at 24 h and 0.792 (p < 0.001) for 90-day clinical outcome. Twenty-four-hour radiological reperfusion assessed by CT is associated with good clinical response on day 1 and good functional outcome on day 90 in patients with ischemic stroke.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, 6 viale Benedetto XV, 13132, Genoa, Italy.
| | - Giorgio Busto
- Struttura Organizzativa Dipartimentale di Radiodiagnostica 2, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze, Azienda Ospedaliero-Universitaria Careggi, 3 Largo Brambilla, 50134, Florence, Italy
| | - Marina Padroni
- Unità Operativa di Neurologia, Dipartimento di Scienze Biologiche, Psichiatriche e Psicologiche, Università di Ferrara, Arcispedale S. Anna, Ferrara, Italy
| | - Andrea Bernardoni
- Unità Operativa di Neuroradiologia, Dipartimento di Neuroscienze, Azienda Ospedaliero-Universitaria di Ferrara, Arcispedale S. Anna, 203 Corso della Giovecca, 44121, Ferrara, Italy
| | - Stefano Colagrande
- Struttura Organizzativa Dipartimentale di Radiodiagnostica 2, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze, Azienda Ospedaliero-Universitaria Careggi, 3 Largo Brambilla, 50134, Florence, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, 6 viale Benedetto XV, 13132, Genoa, Italy
- Ospedale Policlinico San Martino, 10 Largo Benzi, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, 6 viale Benedetto XV, 13132, Genoa, Italy
- Ospedale Policlinico San Martino, 10 Largo Benzi, 16132, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 9 viale Benedetto XV, 16132, Genoa, Italy
| | - Enrico Fainardi
- Struttura Organizzativa Dipartimentale di Neuroradiologia, Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Università degli Studi di Firenze, Azienda Ospedaliero-Universitaria Careggi, 3 Largo Brambilla, 50134, Florence, Italy
| |
Collapse
|
150
|
Lei JR, Tu XK, Wang Y, Tu DW, Shi SS. Resveratrol downregulates the TLR4 signaling pathway to reduce brain damage in a rat model of focal cerebral ischemia. Exp Ther Med 2019; 17:3215-3221. [PMID: 30936996 DOI: 10.3892/etm.2019.7324] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/23/2018] [Indexed: 12/21/2022] Open
Abstract
Previous studies have demonstrated that inflammation and disruption of the blood-brain barrier (BBB) are important pathological processes during focal cerebral ischemia. Therefore, the present study evaluated the neuroprotective effects of resveratrol against brain damage, inflammation and BBB disruption in rats with focal cerebral ischemia and assessed the potential underlying molecular mechanisms. Sprague-Dawley rats underwent cerebral ischemia/reperfusion (IR) and then received intraperitoneal resveratrol (10 and 100 mg/kg) 2 h following the onset of ischemia. Following 24 h of ischemia, neurological deficit scores, cerebral infarctions, morphological characteristics, cerebral water content, myeloperoxidase (MPO) activity and Evans blue extravasation were assessed. Additionally, the protein expression levels of Toll-like receptor 4 (TLR4) and nuclear factor (NF)-κB p65 were detected using western blot analyses, the mRNA expression levels of cyclooxygenase-2 (COX-2) and matrix metalloproteinase-9 (MMP-9) were examined by reverse-transcription polymerase chain reaction, and tumor necrosis factor (TNF)-α and interleukin (IL)-1β blood levels were determined by ELISA. Resveratrol significantly reduced neurological deficit scores, cerebral infarct sizes, neuronal injury, MPO activity and EB content. Cerebral ischemia increased the expression levels of TLR4, NF-κB p65, COX-2, MMP-9, TNF-α and IL-1β, but all of these factors were reduced by resveratrol. In conclusion, the present data suggest that resveratrol reduces inflammation, BBB disruption and brain damage in rats following focal cerebral ischemia. Additionally, the neuroprotective effects of resveratrol against cerebral ischemia may be associated with downregulation of the TLR4 pathway.
Collapse
Affiliation(s)
- Jun-Rong Lei
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China.,Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Xian-Kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yang Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - De-Wen Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|