301
|
Rho kinase II interference by small hairpin RNA ameliorates 1‑methyl‑4‑phenyl‑1,2,3,6‑tetrahydropyridine‑induced parkinsonism in mice. Mol Med Rep 2016; 14:4947-4956. [PMID: 27840922 PMCID: PMC5355756 DOI: 10.3892/mmr.2016.5889] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 05/12/2016] [Indexed: 02/03/2023] Open
Abstract
Novel therapeutic targets are required for the treatment of Parkinson's disease (PD). Previous studies suggest that the Rho/Rho-associated, coiled-coil-containing protein kinases (ROCKs) signaling pathway may be a promising therapeutic target in PD. To elucidate the importance of ROCKII in the pathogenesis of dopaminergic (DA) neuron loss and to investigate the efficacy of ROCK inhibitors in PD, ROCKII expression in the substantia nigra (SN) of mice was silenced through the injection of a lentivirus-based small hairpin RNA system. Empty lentivirus vectors served as controls. Mice were subsequently challenged with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The expression levels and activity of ROCKII were elevated in tyrosine hydroxylase-positive neurons and in cluster of differentiation (CD) 11b-positive microglia within the SN of MPTP-treated mice, which was accompanied by an increased level of expression of inducible nitric oxide synthase (iNOS) and activation of the Toll-like receptor (TLR)2/nuclear factor (NF)-κB signaling pathway in M1 microglia. ROCKII interference (RI) significantly improved movement disorder and attenuated DA neuron loss induced by MPTP. In addition, RI inhibited the activation of M1 microglia in the SN, exhibiting reduced activity of the TLR2/NF-κB signaling pathway and decreased expression levels of iNOS and inflammatory factors, including interleukin (IL)-1β and IL-6. The results of the present study verify that ROCKII participates in the loss of DA neurons induced by MPTP and suggest that ROCKII inhibition may be a promising therapeutic target for PD.
Collapse
|
302
|
Rodent Gymnastics: Neurobehavioral Assays in Ischemic Stroke. Mol Neurobiol 2016; 54:6750-6761. [PMID: 27752994 DOI: 10.1007/s12035-016-0195-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 10/04/2016] [Indexed: 10/20/2022]
Abstract
Despite years of research, most preclinical trials on ischemic stroke have remained unsuccessful owing to poor methodological and statistical standards leading to "translational roadblocks." Various behavioral tests have been established to evaluate traits such as sensorimotor function, cognitive and social interactions, and anxiety-like and depression-like behavior. A test's validity is of cardinal importance as it influences the chance of a successful translation of preclinical results to clinical settings. The mission of choosing a behavioral test for a particular project is, therefore, imperative and the present review aims to provide a structured way to evaluate rodent behavioral tests with implications in ischemic stroke.
Collapse
|
303
|
Ma J, Zhang L, He G, Tan X, Jin X, Li C. Transcutaneous auricular vagus nerve stimulation regulates expression of growth differentiation factor 11 and activin-like kinase 5 in cerebral ischemia/reperfusion rats. J Neurol Sci 2016; 369:27-35. [DOI: 10.1016/j.jns.2016.08.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 01/09/2023]
|
304
|
3K3A-activated protein C stimulates postischemic neuronal repair by human neural stem cells in mice. Nat Med 2016; 22:1050-5. [PMID: 27548576 DOI: 10.1038/nm.4154] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 06/24/2016] [Indexed: 12/13/2022]
Abstract
Activated protein C (APC) is a blood protease with anticoagulant activity and cell-signaling activities mediated by the activation of protease-activated receptor 1 (F2R, also known as PAR1) and F2RL1 (also known as PAR3) via noncanonical cleavage. Recombinant variants of APC, such as the 3K3A-APC (Lys191-193Ala) mutant in which three Lys residues (KKK191-193) were replaced with alanine, and/or its other mutants with reduced (>90%) anticoagulant activity, engineered to reduce APC-associated bleeding risk while retaining normal cell-signaling activity, have shown benefits in preclinical models of ischemic stroke, brain trauma, multiple sclerosis, amyotrophic lateral sclerosis, sepsis, ischemic and reperfusion injury of heart, kidney and liver, pulmonary, kidney and gastrointestinal inflammation, diabetes and lethal body radiation. On the basis of proof-of-concept studies and an excellent safety profile in humans, 3K3A-APC has advanced to clinical trials as a neuroprotectant in ischemic stroke. Recently, 3K3A-APC has been shown to stimulate neuronal production by human neural stem and progenitor cells (NSCs) in vitro via a PAR1-PAR3-sphingosine-1-phosphate-receptor 1-Akt pathway, which suggests the potential for APC-based treatment as a strategy for structural repair in the human central nervous (CNS) system. Here we report that late postischemic treatment of mice with 3K3A-APC stimulates neuronal production by transplanted human NSCs, promotes circuit restoration and improves functional recovery. Thus, 3K3A-APC-potentiated neuronal recruitment from engrafted NSCs might offer a new approach to the treatment of stroke and related neurological disorders.
Collapse
|
305
|
Wang P, He Y, Li D, Han R, Liu G, Kong D, Hao J. Class I PI3K inhibitor ZSTK474 mediates a shift in microglial/macrophage phenotype and inhibits inflammatory response in mice with cerebral ischemia/reperfusion injury. J Neuroinflammation 2016; 13:192. [PMID: 27549161 PMCID: PMC4994222 DOI: 10.1186/s12974-016-0660-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 07/14/2016] [Indexed: 01/21/2023] Open
Abstract
Background Microglia/macrophages play a critical role in the inflammatory and immune processes of cerebral ischemia/reperfusion injury. Since microglia/macrophages can reversibly shift their phenotype toward either a “detrimental” or a “restorative” state in the injured central nervous system (CNS), compounds mediate that shift which could inhibit inflammation and restore the ability to alleviate cerebral ischemia/reperfusion injury would have therapeutic potential. Methods Transient middle cerebral artery occlusion was induced in male C57BL/6 mice. Mice were randomly separated into a sham-operated group, a control group, and a ZSTK474-treated group. We investigated the effect of ZSTK474 by assessing neurological deficits, infarct volume, and histopathological changes. We then determined the potential mechanism by immunofluorescent staining, quantitative real-time polymerase chain reaction (PCR), and Western blot analysis. The Tukey’s test or Mann–Whitney U test was used to compare differences among the groups. Results ZSTK474 alleviated neurological deficits and reduced infarct volume in the cerebral ischemia/reperfusion injury model. Presumably, ZSTK474 shifted the phenotype of microglia/macrophages to a restorative state, since this treatment decreased the secretion of pro-inflammatory factors and advanced the secretion of anti-inflammatory factors. These neuroprotective properties of ZSTK474 may be mediated by the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin complex 1 (mTORC1) pathway. Conclusions ZSTK474 can mediate a shift in microglia/macrophage phenotype and inhibit the inflammatory response in cerebral ischemia reperfusion injury of mice. These effects appeared to ensue via the PI3K/AKT/mTORC1 pathway. Therefore, ZSTK474 may represent a therapeutic intervention with potential for circumventing the catastrophic aftermath of ischemic stroke. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0660-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Po Wang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurology, Baotou Central Hospital, Baotou, Inner Mongolia, 014000, China
| | - Yating He
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Daojing Li
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ranran Han
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Guiyou Liu
- Genome Analysis Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300000, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Junwei Hao
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, China. .,Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
306
|
Spencer B, Williams S, Rockenstein E, Valera E, Xin W, Mante M, Florio J, Adame A, Masliah E, Sierks MR. α-synuclein conformational antibodies fused to penetratin are effective in models of Lewy body disease. Ann Clin Transl Neurol 2016; 3:588-606. [PMID: 27606342 PMCID: PMC4999592 DOI: 10.1002/acn3.321] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/12/2016] [Accepted: 04/29/2016] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Progressive accumulation of α-synuclein (α-syn) has been associated with Parkinson's disease (PD) and Dementia with Lewy body (DLB). The mechanisms through which α-syn leads to neurodegeneration are not completely clear; however, the formation of various oligomeric species have been proposed to play a role. Antibody therapy has shown effectiveness at reducing α-syn accumulation in the central nervous system (CNS); however, most of these studies have been conducted utilizing antibodies that recognize both monomeric and higher molecular weight α-syn. In this context, the main objective of this study was to investigate the efficacy of immunotherapy with single-chain antibodies (scFVs) against specific conformational forms of α-syn fused to a novel brain penetrating sequence. METHOD We screened various scFVs against α-syn expressed from lentiviral vectors by intracerebral injections in an α-syn tg model. The most effective scFVs were fused to the cell-penetrating peptide penetratin to enhance transport across the blood-brain barrier, and lentiviral vectors were constructed and tested for efficacy following systemic delivery intraperitoneal into α-syn tg mice. RESULT Two scFVs (D5 and 10H) selectively targeted different α-syn oligomers and reduced the accumulation of α-syn and ameliorated functional deficits when delivered late in disease development; however, only one of the antibodies (D5) was also effective when delivered early in disease development. These scFVs were also utilized in an enzyme-linked immunosorbent assay (ELISA) assay to monitor the effects of immunotherapy on α-syn oligomers in brain and plasma. INTERPRETATION The design and targeting of antibodies for specific species of α-syn oligomers is crucial for therapeutic immunotherapy and might be of relevance for the treatment of Lewy body disease.
Collapse
Affiliation(s)
- Brian Spencer
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | | | | | - Elvira Valera
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Wei Xin
- Department of Chemical EngineeringArizona State UniversityTempeArizona
| | - Michael Mante
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Jazmin Florio
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Anthony Adame
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Eliezer Masliah
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
- Department of PathologyUniversity of CaliforniaSan DiegoCalifornia
| | - Michael R. Sierks
- Department of Chemical EngineeringArizona State UniversityTempeArizona
| |
Collapse
|
307
|
Tetramethylpyrazine Nitrone Improves Neurobehavioral Functions and Confers Neuroprotection on Rats with Traumatic Brain Injury. Neurochem Res 2016; 41:2948-2957. [DOI: 10.1007/s11064-016-2013-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 11/26/2022]
|
308
|
Wang JY, Huang YN, Chiu CC, Tweedie D, Luo W, Pick CG, Chou SY, Luo Y, Hoffer BJ, Greig NH, Wang JY. Pomalidomide mitigates neuronal loss, neuroinflammation, and behavioral impairments induced by traumatic brain injury in rat. J Neuroinflammation 2016; 13:168. [PMID: 27353053 PMCID: PMC4924242 DOI: 10.1186/s12974-016-0631-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/16/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a global health concern that typically causes emotional disturbances and cognitive dysfunction. Secondary pathologies following TBI may be associated with chronic neurodegenerative disorders and an enhanced likelihood of developing dementia-like disease in later life. There are currently no approved drugs for mitigating the acute or chronic effects of TBI. METHODS The effects of the drug pomalidomide (Pom), an FDA-approved immunomodulatory agent, were evaluated in a rat model of moderate to severe TBI induced by controlled cortical impact. Post-TBI intravenous administration of Pom (0.5 mg/kg at 5 or 7 h and 0.1 mg/kg at 5 h) was evaluated on functional and histological measures that included motor function, fine more coordination, somatosensory function, lesion volume, cortical neurodegeneration, neuronal apoptosis, and the induction of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). RESULTS Pom 0.5 mg/kg administration at 5 h, but not at 7 h post-TBI, significantly mitigated the TBI-induced injury volume and functional impairments, neurodegeneration, neuronal apoptosis, and cytokine mRNA and protein induction. To evaluate underlying mechanisms, the actions of Pom on neuronal survival, microglial activation, and the induction of TNF-α were assessed in mixed cortical cultures following a glutamate challenge. Pom dose-dependently ameliorated glutamate-mediated cytotoxic effects on cell viability and reduced microglial cell activation, significantly attenuating the induction of TNF-α. CONCLUSIONS Post-injury treatment with a single Pom dose within 5 h significantly reduced functional impairments in a well-characterized animal model of TBI. Pom decreased the injury lesion volume, augmented neuronal survival, and provided anti-inflammatory properties. These findings strongly support the further evaluation and optimization of Pom for potential use in clinical TBI.
Collapse
Affiliation(s)
- Jin-Ya Wang
- />Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
| | - Ya-Ni Huang
- />Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
- />Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Chong-Chi Chiu
- />Department of General Surgery, Chi Mei Medical Center, Tainan and Liouying, Taiwan
| | - David Tweedie
- />Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Weiming Luo
- />Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Chaim G. Pick
- />Department of Anatomy and Anthropology, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Szu-Yi Chou
- />Graduate Program on Neuroregeneration, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu Luo
- />Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH USA
| | - Barry J. Hoffer
- />Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH USA
| | - Nigel H. Greig
- />Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Jia-Yi Wang
- />Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
- />Department of Physiology, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
| |
Collapse
|
309
|
Wali B, Sayeed I, Guthrie DB, Natchus MG, Turan N, Liotta DC, Stein DG. Evaluating the neurotherapeutic potential of a water-soluble progesterone analog after traumatic brain injury in rats. Neuropharmacology 2016; 109:148-158. [PMID: 27267687 DOI: 10.1016/j.neuropharm.2016.05.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/06/2016] [Accepted: 05/24/2016] [Indexed: 01/03/2023]
Abstract
The poor aqueous solubility of progesterone (PROG) limits its potential use as a therapeutic agent. We designed and tested EIDD-1723, a novel water-soluble analog of PROG with >100-fold higher solubility than that of native PROG, as candidate for development as a field-ready treatment for traumatic brain injury (TBI). The pharmacokinetic effects of EIDD-1723 on morphological and functional outcomes in rats with bilateral cortical impact injury were evaluated. Following TBI, 10-mg/kg doses of EIDD-1723 or PROG were given intramuscularly (i.m.) at 1, 6 and 24 h post-injury, then daily for the next 6 days, with tapering of the last 2 treatments. Rats were tested pre-injury to establish baseline performance on grip strength and sensory neglect, and then retested at 4, 9 and 21 days post-TBI. Spatial learning was evaluated from days 11-17 post-TBI. At 22 days post-injury, rats were perfused and brains extracted and processed for lesion size. For the edema assay the animals were killed and brains removed at 24 h post-injury. EIDD-1723 significantly reduced cerebral edema and improved recovery from motor, sensory and spatial learning deficits as well as, or better than, native PROG. Pharmacokinetic investigation after a single i.m. injection in rats revealed that EIDD-1723 was rapidly converted to the active metabolite EIDD-036, demonstrating first-order elimination kinetics and ability to cross the blood-brain barrier. Our results suggest that EIDD-1723 represents a substantial advantage over current PROG formulations because it overcomes storage, formulation and delivery limitations of PROG and can thereby reduce the time between injury and treatment.
Collapse
Affiliation(s)
- Bushra Wali
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA 30322, USA.
| | - Iqbal Sayeed
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA 30322, USA
| | - David B Guthrie
- Emory Institute for Drug Development/Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Michael G Natchus
- Emory Institute for Drug Development/Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Nefize Turan
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dennis C Liotta
- Emory Institute for Drug Development/Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Donald G Stein
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
310
|
Llovera G, Hofmann K, Roth S, Salas-Pérdomo A, Ferrer-Ferrer M, Perego C, Zanier ER, Mamrak U, Rex A, Party H, Agin V, Fauchon C, Orset C, Haelewyn B, De Simoni MG, Dirnagl U, Grittner U, Planas AM, Plesnila N, Vivien D, Liesz A. Results of a preclinical randomized controlled multicenter trial (pRCT): Anti-CD49d treatment for acute brain ischemia. Sci Transl Med 2016; 7:299ra121. [PMID: 26246166 DOI: 10.1126/scitranslmed.aaa9853] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous treatments have been reported to provide a beneficial outcome in experimental animal stroke models; however, these treatments (with the exception of tissue plasminogen activator) have failed in clinical trials. To improve the translation of treatment efficacy from bench to bedside, we have performed a preclinical randomized controlled multicenter trial (pRCT) to test a potential stroke therapy under circumstances closer to the design and rigor of a clinical randomized control trial. Anti-CD49d antibodies, which inhibit the migration of leukocytes into the brain, were previously investigated in experimental stroke models by individual laboratories. Despite the conflicting results from four positive and one inconclusive preclinical studies, a clinical trial was initiated. To confirm the preclinical results and to test the feasibility of conducting a pRCT, six independent European research centers investigated the efficacy of anti-CD49d antibodies in two distinct mouse models of stroke in a centrally coordinated, randomized, and blinded approach. The results pooled from all research centers revealed that treatment with CD49d-specific antibodies significantly reduced both leukocyte invasion and infarct volume after the permanent distal occlusion of the middle cerebral artery, which causes a small cortical infarction. In contrast, anti-CD49d treatment did not reduce lesion size or affect leukocyte invasion after transient proximal occlusion of the middle cerebral artery, which induces large lesions. These results suggest that the benefits of immune-targeted approaches may depend on infarct severity and localization. This study supports the feasibility of performing pRCTs.
Collapse
Affiliation(s)
- Gemma Llovera
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Feodor-Lynen-Straße 17, 81377 Munich, Germany. Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Kerstin Hofmann
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Feodor-Lynen-Straße 17, 81377 Munich, Germany. Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Feodor-Lynen-Straße 17, 81377 Munich, Germany. Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Angelica Salas-Pérdomo
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain. Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Maura Ferrer-Ferrer
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain. Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlo Perego
- Neuroscience Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milan, Italy
| | - Elisa R Zanier
- Neuroscience Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milan, Italy
| | - Uta Mamrak
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Feodor-Lynen-Straße 17, 81377 Munich, Germany. Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Andre Rex
- Department of Experimental Neurology and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin; German Center for Neurodegenerative Diseases (DZNE) and German Center for Cardiovascular Diseases (DZHK), Berlin sites; Excellence Cluster NeuroCure, 10117 Berlin, Germany
| | - Hélène Party
- INSERM, UMR-S U919, Université de Caen Basse-Normandie, team Serine Proteases and Pathophysiology of the neurovascular Unit, GIP Cyceron, F-14074 Caen Cedex, France
| | - Véronique Agin
- INSERM, UMR-S U919, Université de Caen Basse-Normandie, team Serine Proteases and Pathophysiology of the neurovascular Unit, GIP Cyceron, F-14074 Caen Cedex, France
| | - Claudine Fauchon
- Experimental Stroke Research Platform (ESRP), IBiSA platform, Centre Universitaire de Ressources Biologiques (CURB), Université de Caen Basse-Normandie, F-14074 Caen Cedex, France
| | - Cyrille Orset
- INSERM, UMR-S U919, Université de Caen Basse-Normandie, team Serine Proteases and Pathophysiology of the neurovascular Unit, GIP Cyceron, F-14074 Caen Cedex, France. Experimental Stroke Research Platform (ESRP), IBiSA platform, Centre Universitaire de Ressources Biologiques (CURB), Université de Caen Basse-Normandie, F-14074 Caen Cedex, France
| | - Benoît Haelewyn
- INSERM, UMR-S U919, Université de Caen Basse-Normandie, team Serine Proteases and Pathophysiology of the neurovascular Unit, GIP Cyceron, F-14074 Caen Cedex, France. Experimental Stroke Research Platform (ESRP), IBiSA platform, Centre Universitaire de Ressources Biologiques (CURB), Université de Caen Basse-Normandie, F-14074 Caen Cedex, France
| | - Maria-Grazia De Simoni
- Neuroscience Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milan, Italy
| | - Ulrich Dirnagl
- Department of Experimental Neurology and Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin; German Center for Neurodegenerative Diseases (DZNE) and German Center for Cardiovascular Diseases (DZHK), Berlin sites; Excellence Cluster NeuroCure, 10117 Berlin, Germany
| | - Ulrike Grittner
- Department of Biostatistics and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain. Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Feodor-Lynen-Straße 17, 81377 Munich, Germany. Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Denis Vivien
- INSERM, UMR-S U919, Université de Caen Basse-Normandie, team Serine Proteases and Pathophysiology of the neurovascular Unit, GIP Cyceron, F-14074 Caen Cedex, France. Experimental Stroke Research Platform (ESRP), IBiSA platform, Centre Universitaire de Ressources Biologiques (CURB), Université de Caen Basse-Normandie, F-14074 Caen Cedex, France
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Feodor-Lynen-Straße 17, 81377 Munich, Germany. Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
311
|
Lee S, Mattingly A, Lin A, Sacramento J, Mannent L, Castel MN, Canolle B, Delbary-Gossart S, Ferzaz B, Morganti JM, Rosi S, Ferguson AR, Manley GT, Bresnahan JC, Beattie MS. A novel antagonist of p75NTR reduces peripheral expansion and CNS trafficking of pro-inflammatory monocytes and spares function after traumatic brain injury. J Neuroinflammation 2016; 13:88. [PMID: 27102880 PMCID: PMC4840857 DOI: 10.1186/s12974-016-0544-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/10/2016] [Indexed: 12/17/2022] Open
Abstract
Background Traumatic brain injury (TBI) results in long-term neurological deficits, which may be mediated in part by pro-inflammatory responses in both the injured brain and the circulation. Inflammation may be involved in the subsequent development of neurodegenerative diseases and post-injury seizures. The p75 neurotrophin receptor (p75NTR) has multiple biological functions, affecting cell survival, apoptotic cell death, axonal growth, and degeneration in pathological conditions. We recently found that EVT901, a novel piperazine derivative that inhibits p75NTR oligomerization, is neuroprotective, reduces microglial activation, and improves outcomes in two models of TBI in rats. Since TBI elicits both CNS and peripheral inflammation, we used a mouse model of TBI to examine whether EVT901 would affect peripheral immune responses and trafficking to the injured brain. Methods Cortical contusion injury (CCI)-TBI of the sensory/motor cortex was induced in C57Bl/6 wild-type mice and CCR2+/RFP heterozygote transgenic mice, followed by treatment with EVT901, a selective antagonist of p75NTR, or vehicle by i.p. injection at 4 h after injury and then daily for 7 days. Brain and blood were collected at 1 and 6 weeks after injury. Flow cytometry and histological analysis were used to determine peripheral immune responses and trafficking of peripheral immune cells into the lesion site at 1 and 6 weeks after TBI. A battery of behavioral tests administered over 6 weeks was used to evaluate neurological outcome, and stereological estimation of brain tissue volume at 6 weeks was used to assess tissue damage. Finally, multivariate principal components analysis (PCA) was used to evaluate the relationships between inflammatory events, EVT901 treatment, and neurological outcomes. Results EVT901 is neuroprotective in mouse CCI-TBI and dramatically reduced the early trafficking of CCR2+ and pro-inflammatory monocytes into the lesion site. EVT901 reduced the number of CD45highCD11b+ and CD45highF4/80+ cells in the injured brain at 6 weeks. TBI produced a significant increase in peripheral pro-inflammatory monocytes (Ly6Cint-high pro-inflammatory monocytes), and this peripheral effect was also blocked by EVT901 treatment. Further, we found that blocking p75NTR with EVT901 reduces the expansion of pro-inflammatory monocytes, and their response to LPS in vitro, supporting the idea that there is a peripheral EVT901 effect that blunts inflammation. Further, 1 week of EVT901 blocks the expansion of pro-inflammatory monocytes in the circulation after TBI, reduces the number of multiple subsets of pro-inflammatory monocytes that enter the injury site at 1 and 6 weeks post-injury, and is neuroprotective, as it was in the rat. Conclusions Together, these findings suggest that p75NTR signaling participates in the production of the peripheral pro-inflammatory response to CNS injury and implicates p75NTR as a part of the pro-inflammatory cascade. Thus, the neuroprotective effects of p75NTR antagonists might be due to a combination of central and peripheral effects, and p75NTR may play a role in the production of peripheral inflammation in addition to its many other biological roles. Thus, p75NTR may be a therapeutic target in human TBI. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0544-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sangmi Lee
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Aaron Mattingly
- Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA
| | - Amity Lin
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Jeffrey Sacramento
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Leda Mannent
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | - Marie-Noelle Castel
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | - Benoit Canolle
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | | | - Badia Ferzaz
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | - Josh M Morganti
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Susanna Rosi
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA.,Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA.,Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA
| | - Geoffrey T Manley
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Jacqueline C Bresnahan
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Michael S Beattie
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA. .,Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA.
| |
Collapse
|
312
|
Min H, Jang YH, Cho IH, Yu SW, Lee SJ. Alternatively activated brain-infiltrating macrophages facilitate recovery from collagenase-induced intracerebral hemorrhage. Mol Brain 2016; 9:42. [PMID: 27094968 PMCID: PMC4837536 DOI: 10.1186/s13041-016-0225-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 04/14/2016] [Indexed: 12/22/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is one of the major causes of stroke. After onset of ICH, massive infiltration of macrophages is detected in the peri-hematoma regions. Still, the function of these macrophages in ICH has not been completely elucidated. Results In a collagenase-induced ICH model, CX3CR1+ macrophages accumulated in the peri-hematoma region. Characterization of these macrophages revealed expression of alternatively activated (M2) macrophage markers. In the macrophage-depleted mice, ICH-induced brain lesion volume was larger and neurological deficits were more severe compared to those of control mice, indicating a protective role of these macrophages in ICH. In the ICH-injured brain, mannose receptor-expressing macrophages increased at a delayed time point after ICH, indicating M2 polarization of the brain-infiltrating macrophages in the brain microenvironment. To explore this possibility, bone marrow-derived macrophages (BMDM) were co-cultured with mouse brain glial cells and then tested for activation phenotype. Upon co-culture with glia, the number of mannose receptor-positive M2 macrophages was significantly increased. Furthermore, treatment with glia-conditioned media increased the number of BMDM of M2 phenotype. Conclusions In this study, our data suggest that brain-infiltrating macrophages after ICH are polarized to the M2 phenotype by brain glial cells and thereby contribute to recovery from ICH injury.
Collapse
Affiliation(s)
- Hyunjung Min
- Department of Neuroscience and Dental Research Institute, School of Dentistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yong Ho Jang
- Department of Neuroscience and Dental Research Institute, School of Dentistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Oriental Medicine, Kyung Hee University, Seoul, 02447, Korea
| | - Seong-Woon Yu
- Department of Brain Science, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, Korea
| | - Sung Joong Lee
- Department of Neuroscience and Dental Research Institute, School of Dentistry, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
313
|
β-Dystroglycan cleavage by matrix metalloproteinase-2/-9 disturbs aquaporin-4 polarization and influences brain edema in acute cerebral ischemia. Neuroscience 2016; 326:141-157. [PMID: 27038751 DOI: 10.1016/j.neuroscience.2016.03.055] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 03/19/2016] [Accepted: 03/23/2016] [Indexed: 01/31/2023]
Abstract
Dystroglycan (DG) is widely expressed in various tissues, and throughout the cerebral microvasculature. It consists of two subunits, α-DG and β-DG, and the cleavage of the latter by matrix metalloproteinase (MMP)-2 and -9 underlies a number of physiological and pathological processes. However, the involvement of MMP-2/-9-mediated β-DG cleavage in cerebral ischemia remains uncertain. In astrocytes, DG is crucial for maintaining the polarization of aquaporin-4 (AQP4), which plays a role in the regulation of cytotoxic and vasogenic edema. The present study aimed to explore the effects of MMP-2/-9-mediated β-DG cleavage on AQP4 polarization and brain edema in acute cerebral ischemia. A model of cerebral ischemia was established via permanent middle cerebral artery occlusion (pMCAO) in male C57BL/6 mice. Western blotting, real-time polymerase chain reaction (PCR), immunohistochemical staining, immunofluorescent staining, electron microscopy, and light microscopy were used. Captopril was applied as a selective MMP-2/-9 inhibitor. Recombinant mouse MMP (rmMMP)-2 and -9 were used in an in vitro cleavage experiment. The present study demonstrated evidence of β-DG cleavage by MMP-2/-9 in pMCAO mouse brains; this cleavage was implicated in AQP4 redistribution and brain edema in cerebral ischemia. In addition, captopril exacerbated cytotoxic edema and ameliorated vasogenic edema at 24h after pMCAO, and alleviated brain edema and neurological deficit at 48h and 72h. In conclusion, this study provides novel insight into the effects of MMP-2/-9-mediated β-DG cleavage in acute cerebral ischemia. Such findings might facilitate the development of a therapeutic strategy for the optimization of MMP-2/-9 targeted treatment in cerebral ischemia.
Collapse
|
314
|
Fagoe ND, Attwell CL, Eggers R, Tuinenbreijer L, Kouwenhoven D, Verhaagen J, Mason MRJ. Evaluation of Five Tests for Sensitivity to Functional Deficits following Cervical or Thoracic Dorsal Column Transection in the Rat. PLoS One 2016; 11:e0150141. [PMID: 26934672 PMCID: PMC4775041 DOI: 10.1371/journal.pone.0150141] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 02/09/2016] [Indexed: 02/02/2023] Open
Abstract
The dorsal column lesion model of spinal cord injury targets sensory fibres which originate from the dorsal root ganglia and ascend in the dorsal funiculus. It has the advantages that fibres can be specifically traced from the sciatic nerve, verifiably complete lesions can be performed of the labelled fibres, and it can be used to study sprouting in the central nervous system from the conditioning lesion effect. However, functional deficits from this type of lesion are mild, making assessment of experimental treatment-induced functional recovery difficult. Here, five functional tests were compared for their sensitivity to functional deficits, and hence their suitability to reliably measure recovery of function after dorsal column injury. We assessed the tape removal test, the rope crossing test, CatWalk gait analysis, and the horizontal ladder, and introduce a new test, the inclined rolling ladder. Animals with dorsal column injuries at C4 or T7 level were compared to sham-operated animals for a duration of eight weeks. As well as comparing groups at individual timepoints we also compared the longitudinal data over the whole time course with linear mixed models (LMMs), and for tests where steps are scored as success/error, using generalized LMMs for binomial data. Although, generally, function recovered to sham levels within 2–6 weeks, in most tests we were able to detect significant deficits with whole time-course comparisons. On the horizontal ladder deficits were detected until 5–6 weeks. With the new inclined rolling ladder functional deficits were somewhat more consistent over the testing period and appeared to last for 6–7 weeks. Of the CatWalk parameters base of support was sensitive to cervical and thoracic lesions while hind-paw print-width was affected by cervical lesion only. The inclined rolling ladder test in combination with the horizontal ladder and the CatWalk may prove useful to monitor functional recovery after experimental treatment in this lesion model.
Collapse
Affiliation(s)
- Nitish D Fagoe
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Callan L Attwell
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ruben Eggers
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Lizz Tuinenbreijer
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Dorette Kouwenhoven
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands.,Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Matthew R J Mason
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
315
|
Wali B, Ishrat T, Stein DG, Sayeed I. Progesterone improves long-term functional and histological outcomes after permanent stroke in older rats. Behav Brain Res 2016; 305:46-56. [PMID: 26921692 DOI: 10.1016/j.bbr.2016.02.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/16/2016] [Accepted: 02/21/2016] [Indexed: 12/22/2022]
Abstract
Previous studies have shown progesterone to be beneficial in animal models of central nervous system injury, but less is known about its longer-term sustained effects on recovery of function following stroke. We evaluated progesterone's effects on a panel of behavioral tests up to 8 weeks after permanent middle cerebral artery occlusion (pMCAO). Male Sprague-Dawley rats 12m.o. were subjected to pMCAO and, beginning 3h post-pMCAO, given intraperitoneal injections of progesterone (8mg/kg) or vehicle, followed by subcutaneous injections at 8h and then every 24h for 7 days, with tapering of the last 2 treatments. The rats were then tested on functional recovery at 3, 6 and 8 weeks post-stroke. We observed that progesterone-treated animals showed attenuation of infarct volume and improved functional outcomes at 8 weeks after stroke on grip strength, sensory neglect, motor coordination and spatial navigation tests. Progesterone treatments significantly improved motor deficits in the affected limb on a number of gait parameters. Glial fibrillary acidic protein expression was increased in the vehicle group and considerably lowered in the progesterone group at 8 weeks post-stroke. With repeated post-stroke testing, sensory neglect and some aspects of spatial learning performance showed spontaneous recovery, but on gait and grip-strength measres progesterone given only in the acute stage of stroke (first 7 days) showed sustained beneficial effects on all other measures of functional recovery up to 8 weeks post-stroke.
Collapse
Affiliation(s)
- Bushra Wali
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA, USA.
| | - Tauheed Ishrat
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA, USA.
| | - Donald G Stein
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA, USA.
| | - Iqbal Sayeed
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA, USA.
| |
Collapse
|
316
|
Quaegebeur A, Segura I, Schmieder R, Verdegem D, Decimo I, Bifari F, Dresselaers T, Eelen G, Ghosh D, Davidson SM, Schoors S, Broekaert D, Cruys B, Govaerts K, De Legher C, Bouché A, Schoonjans L, Ramer MS, Hung G, Bossaert G, Cleveland DW, Himmelreich U, Voets T, Lemmens R, Bennett CF, Robberecht W, De Bock K, Dewerchin M, Ghesquière B, Fendt SM, Carmeliet P. Deletion or Inhibition of the Oxygen Sensor PHD1 Protects against Ischemic Stroke via Reprogramming of Neuronal Metabolism. Cell Metab 2016; 23:280-91. [PMID: 26774962 PMCID: PMC4880550 DOI: 10.1016/j.cmet.2015.12.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/30/2015] [Accepted: 12/11/2015] [Indexed: 01/08/2023]
Abstract
The oxygen-sensing prolyl hydroxylase domain proteins (PHDs) regulate cellular metabolism, but their role in neuronal metabolism during stroke is unknown. Here we report that PHD1 deficiency provides neuroprotection in a murine model of permanent brain ischemia. This was not due to an increased collateral vessel network. Instead, PHD1(-/-) neurons were protected against oxygen-nutrient deprivation by reprogramming glucose metabolism. Indeed, PHD1(-/-) neurons enhanced glucose flux through the oxidative pentose phosphate pathway by diverting glucose away from glycolysis. As a result, PHD1(-/-) neurons increased their redox buffering capacity to scavenge oxygen radicals in ischemia. Intracerebroventricular injection of PHD1-antisense oligonucleotides reduced the cerebral infarct size and neurological deficits following stroke. These data identify PHD1 as a regulator of neuronal metabolism and a potential therapeutic target in ischemic stroke.
Collapse
Affiliation(s)
- Annelies Quaegebeur
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Inmaculada Segura
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Roberta Schmieder
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Vesalius Research Center, VIB, Leuven, Belgium
| | - Dries Verdegem
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium; Metabolomics Expertise Center, Vesalius Research Center, VIB, Leuven, Belgium
| | - Ilaria Decimo
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Francesco Bifari
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Tom Dresselaers
- Biomedical MRI/Mosaic, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Debapriva Ghosh
- Laboratory of Ion Channel Research and TRP channel research platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Shawn M Davidson
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sandra Schoors
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Dorien Broekaert
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Vesalius Research Center, VIB, Leuven, Belgium
| | - Bert Cruys
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Kristof Govaerts
- Biomedical MRI/Mosaic, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Carla De Legher
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Ann Bouché
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Luc Schoonjans
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Matt S Ramer
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium; International Collaboration on Repair Discoveries, the University of British Columbia, Vancouver, Canada
| | - Gene Hung
- Isis Pharmaceuticals, Carlsbad, CA 92008, USA
| | - Goele Bossaert
- Leuven Statistics Research Centre (LStat), University of Leuven, Leuven, Belgium
| | - Don W Cleveland
- Ludwig Institute for Cancer Research, Department of Medicine and Neuroscience, University of California, San Diego, La Jolla, CA 92093, USA
| | - Uwe Himmelreich
- Biomedical MRI/Mosaic, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research and TRP channel research platform Leuven, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Robin Lemmens
- Laboratory of Neurobiology, Vesalius Research Center, VIB, Leuven, Belgium; Experimental Neurology (Department of Neurosciences) and Leuven Research Institute for Neuroscience and Disease (LIND), University of Leuven, Leuven, Belgium; Neurology, University Hospitals Leuven, Leuven, Belgium
| | | | - Wim Robberecht
- Laboratory of Neurobiology, Vesalius Research Center, VIB, Leuven, Belgium; Experimental Neurology (Department of Neurosciences) and Leuven Research Institute for Neuroscience and Disease (LIND), University of Leuven, Leuven, Belgium; Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Katrien De Bock
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Expertise Center, Vesalius Research Center, VIB, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Vesalius Research Center, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular link, Department of Oncology, University of Leuven, Leuven, Belgium; Laboratory of Angiogenesis and Neurovascular link, Vesalius Research Center, VIB, Leuven, Belgium.
| |
Collapse
|
317
|
Ejaz S, Emmrich JV, Sitnikov SL, Hong YT, Sawiak SJ, Fryer TD, Aigbirhio FI, Williamson DJ, Baron JC. Normobaric hyperoxia markedly reduces brain damage and sensorimotor deficits following brief focal ischaemia. Brain 2016; 139:751-64. [PMID: 26767570 DOI: 10.1093/brain/awv391] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/16/2015] [Indexed: 01/02/2023] Open
Abstract
'True' transient ischaemic attacks are characterized not only clinically, but also radiologically by a lack of corresponding changes on magnetic resonance imaging. During a transient ischaemic attack it is assumed that the affected tissue is penumbral but rescued by early spontaneous reperfusion. There is, however, evidence from rodent studies that even brief focal ischaemia not resulting in tissue infarction can cause extensive selective neuronal loss associated with long-lasting sensorimotor impairment but normal magnetic resonance imaging. Selective neuronal loss might therefore contribute to the increasingly recognized cognitive impairment occurring in patients with transient ischaemic attacks. It is therefore relevant to consider treatments to reduce brain damage occurring with transient ischaemic attacks. As penumbral neurons are threatened by markedly constrained oxygen delivery, improving the latter by increasing arterial O2 content would seem logical. Despite only small increases in arterial O2 content, normobaric oxygen therapy experimentally induces significant increases in penumbral O2 pressure and by such may maintain the penumbra alive until reperfusion. Nevertheless, the effects of normobaric oxygen therapy on infarct volume in rodent models have been conflicting, although duration of occlusion appeared an important factor. Likewise, in the single randomized trial published to date, early-administered normobaric oxygen therapy had no significant effect on clinical outcome despite reduced diffusion-weighted imaging lesion growth during therapy. Here we tested the hypothesis that normobaric oxygen therapy prevents both selective neuronal loss and sensorimotor deficits in a rodent model mimicking true transient ischaemic attack. Normobaric oxygen therapy was applied from the onset and until completion of 15 min distal middle cerebral artery occlusion in spontaneously hypertensive rats, a strain representative of the transient ischaemic attack-prone population. Whereas normoxic controls showed normal magnetic resonance imaging but extensive cortical selective neuronal loss associated with microglial activation (present both at Day 14 in vivo and at Day 28 post-mortem) and marked and long-lasting sensorimotor deficits, normobaric oxygen therapy completely prevented sensorimotor deficit (P < 0.02) and near-completely Day 28 selective neuronal loss (P < 0.005). Microglial activation was substantially reduced at Day 14 and completely prevented at Day 28 (P = 0.002). Our findings document that normobaric oxygen therapy administered during ischaemia nearly completely prevents the neuronal death, microglial inflammation and sensorimotor impairment that characterize this rodent true transient ischaemic attack model. Taken together with the available literature, normobaric oxygen therapy appears a promising therapy for short-lasting ischaemia, and is attractive clinically as it could be started at home in at-risk patients or in the ambulance in subjects suspected of transient ischaemic attack/early stroke. It may also be a straightforward adjunct to reperfusion therapies, and help prevent subtle brain damage potentially contributing to long-term cognitive and sensorimotor impairment in at-risk populations.
Collapse
Affiliation(s)
- Sohail Ejaz
- 1 Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Julius V Emmrich
- 1 Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, UK 2 Department of Neurology, Charité - Universitätsmedizin Berlin, Germany
| | - Sergey L Sitnikov
- 1 Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Young T Hong
- 3 Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Stephen J Sawiak
- 3 Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Tim D Fryer
- 3 Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Franklin I Aigbirhio
- 3 Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, UK
| | - David J Williamson
- 3 Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Jean-Claude Baron
- 1 Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, UK 4 INSERM U894, Hôpital Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
318
|
Analysis of Small Ischemic Lesions in the Examinees of a Brain Dock and Neurological Examination of Animals Subjected to Cortical or Basal Ganglia Photothrombotic Infarction. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:93-7. [PMID: 26463929 DOI: 10.1007/978-3-319-18497-5_16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We analyzed cases of small brain ischemic lesions found in examinees of a brain dock (neurological health screening center). Small cerebral infarction was found in 17 % of the examinees (733 cases). White matter lesions were found in 24 %. Infarctions were located in the cortex or subcortical white matter in 31 % and in the basal ganglia in 44 % of cases. Infratentorial infarction was found in 1.6 %. We have developed an animal model of small infarction in the cortex or basal ganglia induced by photothrombosis in rodents. Sprague-Dawley rats or Mongolian gerbils were anesthetized and photothrombotic infarction was induced in the left caudate nucleus or parietal cortex by light exposure via an optic fiber and intravenous Rose Bengal dye injection. Histological examination revealed development of a small spherical infarction surrounding the tip of the optic fiber. The lesion turned to a cyst by 6 weeks after lesioning. Neurological deficits were found in animals both with cortical and caudate infarction. Behavioral changes in an open field test differed with the lesion site. Neurological deficits were sustained longer in animals with larger infarctions. Thus, photothrombotic infarction is useful for analyzing location-dependent and size-dependent neurological and neuropathological changes after cerebral infarction.
Collapse
|
319
|
Hiu T, Farzampour Z, Paz JT, Wang EHJ, Badgely C, Olson A, Micheva KD, Wang G, Lemmens R, Tran KV, Nishiyama Y, Liang X, Hamilton SA, O'Rourke N, Smith SJ, Huguenard JR, Bliss TM, Steinberg GK. Enhanced phasic GABA inhibition during the repair phase of stroke: a novel therapeutic target. Brain 2015; 139:468-80. [PMID: 26685158 PMCID: PMC4805083 DOI: 10.1093/brain/awv360] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/20/2015] [Indexed: 11/13/2022] Open
Abstract
Ischaemic stroke is the leading cause of severe long-term disability yet lacks drug therapies that promote the repair phase of recovery. This repair phase of stroke occurs days to months after stroke onset and involves brain remapping and plasticity within the peri-infarct zone. Elucidating mechanisms that promote this plasticity is critical for the development of new therapeutics with a broad treatment window. Inhibiting tonic (extrasynaptic) GABA signalling during the repair phase was reported to enhance functional recovery in mice suggesting that GABA plays an important function in modulating brain repair. While tonic GABA appears to suppress brain repair after stroke, less is known about the role of phasic (synaptic) GABA during the repair phase. We observed an increase in postsynaptic phasic GABA signalling in mice within the peri-infarct cortex specific to layer 5; we found increased numbers of α1 receptor subunit-containing GABAergic synapses detected using array tomography, and an associated increased efficacy of spontaneous and miniature inhibitory postsynaptic currents in pyramidal neurons. Furthermore, we demonstrate that enhancing phasic GABA signalling using zolpidem, a Food and Drug Administration (FDA)-approved GABA-positive allosteric modulator, during the repair phase improved behavioural recovery. These data identify potentiation of phasic GABA signalling as a novel therapeutic strategy, indicate zolpidem’s potential to improve recovery, and underscore the necessity to distinguish the role of tonic and phasic GABA signalling in stroke recovery.
Collapse
Affiliation(s)
- Takeshi Hiu
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Zoya Farzampour
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Jeanne T Paz
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA 4 Gladstone Institute of Neurological Disease and University of California, San Francisco San Francisco, CA USA
| | - Eric Hou Jen Wang
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Corrine Badgely
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Andrew Olson
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Kristina D Micheva
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Gordon Wang
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Robin Lemmens
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA 6 KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology; VIB - Vesalius Research Center University Hospitals Leuven, Department of Neurology, B-3000 Leuven, Belgium
| | - Kevin V Tran
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Yasuhiro Nishiyama
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Xibin Liang
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Scott A Hamilton
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Nancy O'Rourke
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Stephen J Smith
- 5 Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - John R Huguenard
- 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Tonya M Bliss
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA
| | - Gary K Steinberg
- 1 Department of Neurosurgery, Stanford University, Stanford, CA, USA 2 Stanford Stroke Centre, Palo Alto, CA 94304, USA 3 Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| |
Collapse
|
320
|
Plasma L5 levels are elevated in ischemic stroke patients and enhance platelet aggregation. Blood 2015; 127:1336-45. [PMID: 26679863 DOI: 10.1182/blood-2015-05-646117] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 12/02/2015] [Indexed: 12/30/2022] Open
Abstract
L5, the most electronegative and atherogenic subfraction of low-density lipoprotein (LDL), induces platelet activation. We hypothesized that plasma L5 levels are increased in acute ischemic stroke patients and examined whether lectin-like oxidized LDL receptor-1 (LOX-1), the receptor for L5 on endothelial cells and platelets, plays a critical role in stroke. Because amyloid β (Aβ) stimulates platelet aggregation, we studied whether L5 and Aβ function synergistically to induce prothrombotic pathways leading to stroke. Levels of plasma L5, serum Aβ, and platelet LOX-1 expression were significantly higher in acute ischemic stroke patients than in controls without metabolic syndrome (P < .01). In mice subjected to focal cerebral ischemia, L5 treatment resulted in larger infarction volumes than did phosphate-buffered saline treatment. Deficiency or neutralizing of LOX-1 reduced infarct volume up to threefold after focal cerebral ischemia in mice, illustrating the importance of LOX-1 in stroke injury. In human platelets, L5 but not L1 (the least electronegative LDL subfraction) induced Aβ release via IκB kinase 2 (IKK2). Furthermore, L5+Aβ synergistically induced glycoprotein IIb/IIIa receptor activation; phosphorylation of IKK2, IκBα, p65, and c-Jun N-terminal kinase 1; and platelet aggregation. These effects were blocked by inhibiting IKK2, LOX-1, or nuclear factor-κB (NF-κB). Injecting L5+Aβ shortened tail-bleeding time by 50% (n = 12; P < .05 vs L1-injected mice), which was prevented by the IKK2 inhibitor. Our findings suggest that, through LOX-1, atherogenic L5 potentiates Aβ-mediated platelet activation, platelet aggregation, and hemostasis via IKK2/NF-κB signaling. L5 elevation may be a risk factor for cerebral atherothrombosis, and downregulating LOX-1 and inhibiting IKK2 may be novel antithrombotic strategies.
Collapse
|
321
|
Abstract
A multifunctional microRNA, miR-155, has been recently recognized as an important modulator of numerous biological processes. In our previous in vitro studies, miR-155 was identified as a potential regulator of the endothelial morphogenesis. The present study demonstrates that in vivo inhibition of miR-155 supports cerebral vasculature after experimental stroke. Intravenous injections of a specific miR-155 inhibitor were initiated at 48 h after mouse distal middle cerebral artery occlusion (dMCAO). Microvasculature in peri-infarct area, infarct size, and animal functional recovery were assessed at 1, 2, and 3 weeks after dMCAO. Using in vivo two-photon microscopy, we detected improved blood flow and microvascular integrity in the peri-infarct area of miR-155 inhibitor-injected mice. Electron microscopy revealed that, in contrast to the control group, these animals demonstrated well preserved capillary tight junctions (TJs). Western blot analysis data indicate that improved TJ integrity in the inhibitor-injected animals could be associated with stabilization of the TJ protein ZO-1 and mediated by the miR-155 target protein Rheb. MRI analysis showed significant (34%) reduction of infarct size in miR-155 inhibitor-injected animals at 21 d after dMCAO. Reduced brain injury was confirmed by electron microscopy demonstrating decreased neuronal damage in the peri-infarct area of stroke. Preservation of brain tissue was reflected in efficient functional recovery of inhibitor-injected animals. Based on our findings, we propose that in vivo miR-155 inhibition after ischemia supports brain microvasculature, reduces brain tissue damage, and improves the animal functional recovery. Significance statement: In the present study, we investigated an effect of the in vivo inhibition of a microRNA, miR-155, on brain recovery after experimental cerebral ischemia. To our knowledge, this is the first report describing the efficiency of intravenous anti-miRNA injections in a mouse model of ischemic stroke. The role of miRNAs in poststroke revascularization has been unexplored and in vivo regulation of miRNAs during the subacute phase of stroke has not yet been proposed. Our investigation introduces a new and unexplored approach to cerebral regeneration: regulation of poststroke angiogenesis and recovery through direct modulation of specific miRNA activity. We expect that our findings will lead to the development of novel strategies for regulating neurorestorative processes in the postischemic brain.
Collapse
|
322
|
Enriched Endogenous Omega-3 Polyunsaturated Fatty Acids Protect Cortical Neurons from Experimental Ischemic Injury. Mol Neurobiol 2015; 53:6482-6488. [DOI: 10.1007/s12035-015-9554-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 11/18/2015] [Indexed: 10/22/2022]
|
323
|
Khan M, Dhammu TS, Matsuda F, Annamalai B, Dhindsa TS, Singh I, Singh AK. Targeting the nNOS/peroxynitrite/calpain system to confer neuroprotection and aid functional recovery in a mouse model of TBI. Brain Res 2015; 1630:159-70. [PMID: 26596859 DOI: 10.1016/j.brainres.2015.11.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/04/2015] [Accepted: 11/07/2015] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) derails nitric oxide (NO)-based anti-inflammatory and anti-excitotoxicity mechanisms. NO is consumed by superoxide to form peroxynitrite, leading to decreased NO bioavailability for S-nitrosoglutathione (GSNO) synthesis and regulation of neuroprotective pathways. Neuronal peroxynitrite is implicated in neuronal loss and functional deficits following TBI. Using a contusion mouse model of TBI, we investigated mechanisms for the opposed roles of GSNO versus peroxynitrite for neuroprotection and functional recovery. TBI was induced by controlled cortical impact (CCI) in adult male mice. GSNO treatment at 2h after CCI decreased the expression levels of phospho neuronal nitric oxide synthase (pnNOS), alpha II spectrin degraded products, and 3-NT, while also decreasing the activities of nNOS and calpains. Treatment of TBI with FeTPPS, a peroxynitrite scavenger, had effects similar to GSNO treatment. GSNO treatment of TBI also reduced neuronal degeneration and improved neurobehavioral function in a two-week TBI study. In a cell free system, SIN-1 (a peroxynitrite donor and 3-nitrotyrosinating agent) increased whereas GSNO (an S-nitrosylating agent) decreased calpain activity, and these activities were reversed by, respectively, FeTPPS and mercuric chloride, a cysteine-NO bond cleaving agent. These data indicate that peroxynitrite-mediated activation and GSNO-mediated inhibition of the deleterious nNOS/calpain system play critical roles in the pathobiology of neuronal protection and functional recovery in TBI disease. Given GSNO׳s safety record in other diseases, its neuroprotective efficacy and promotion of functional recovery in this TBI study make low-dose GSNO a potential candidate for preclinical evaluation.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States..
| | - Tajinder S Dhammu
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States..
| | - Fumiyo Matsuda
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States.; School of Health Science, Kagoshima University, Kagoshima, Japan.
| | | | - Tejbir Singh Dhindsa
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States..
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States..
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| |
Collapse
|
324
|
Jung WB, Im GH, Chung JJ, Ahn SY, Jeon TY, Chang YS, Park WS, Kim JH, Kim KS, Lee JH. Neuroplasticity for spontaneous functional recovery after neonatal hypoxic ischemic brain injury in rats observed by functional MRI and diffusion tensor imaging. Neuroimage 2015; 126:140-50. [PMID: 26589335 DOI: 10.1016/j.neuroimage.2015.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/09/2015] [Accepted: 11/13/2015] [Indexed: 01/24/2023] Open
Abstract
For infants and children, an incredible resilience from injury is often observed. There is growing evidence that functional recovery after brain injury might well be a consequence of the reorganization of the neural network as a process of neuroplasticity. We demonstrate the presence of neuroplasticity at work in spontaneous recovery after neonatal hypoxic ischemic (HI) injury, by elucidating a precise picture in which such reorganization takes place using functional MRI techniques. For all 12 siblings, 6 rats were subjected to severe HI brain injury and 6 rats underwent sham operation only. Severe HI brain injury was induced to postnatal day 7 (p7) Sprague-Dawley rats according to the Rice-Vannucci model (right carotid artery occlusion followed by 150min of hypoxia with 8% O2 and 92% of N2). Brain activation maps along with anatomical and functional connectivity maps related to the sensory motor function were obtained at adult (p63) using blood oxygen level dependent (BOLD)-functional MRI (fMRI), resting state-functional MRI (rs-fMRI) and diffusion tensor imaging (DTI); each of these MRI data was related to sensory motor functional outcome. In-depth investigation of the functional MRI data revealed: 1) intra-hemispheric expansion of BOLD signal activation in the contralesional undamaged hemisphere for ipsilesional forepaw stimuli to include the M2 and Cg1 in addition to the S1 and M1 wide spreading in the anterior and posterior directions, 2) inter-hemispheric transfer of BOLD signal activation for contralesional forepaw stimuli, normally routed to the injured hemisphere, to analogous sites in the contralesional undamaged hemisphere, localized newly to the M1 and M2 with a reduced portion of the S1, 3) inter-hemispheric axonal disconnection and axonal rewiring within the undamaged hemisphere as shown through DTI, and 4) increased functional interactions within the cingulate gyrus in the HI injured rats as shown through rs-fMRI. The BOLD signal amplitudes as well as DTI and rs-fMRI data well correlate with behavioral tests (tape to remove). We found that function normally utilizing what would be the injured hemisphere is transferred to the uninjured hemisphere, and functionality of the uninjured hemisphere remains not untouched but is also rewired in an expansion corresponding to the newly formed sensorimotor function from both the contralesional and the ipsilesional sides. The conclusion drawn from the data in our current study is that enhanced motor function in the contralesional hemisphere governs both the normal and damaged sides, indicating that active plasticity with brain laterality was spontaneously generated to overcome functional loss and established autonomously through normal experience via modification of neural circuitry for neonatal HI injured brain.
Collapse
Affiliation(s)
- Won-Beom Jung
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea; Department of Global Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Geun Ho Im
- Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, South Korea
| | - Julius Juhyun Chung
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea; Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul 06351, South Korea
| | - So-Yoon Ahn
- Department of Pediatrics Division of Neonatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Tae Yeon Jeon
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Yun Sil Chang
- Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul 06351, South Korea; Department of Pediatrics Division of Neonatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Won Soon Park
- Department of Pediatrics Division of Neonatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Ji Hye Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Ki-Soo Kim
- Department of Pediatrics Division of Neonatology, Asan Medical Center, University of Ulsan School of Medicine, Seoul 05535, South Korea
| | - Jung Hee Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea; Department of Global Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea; Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul 06351, South Korea.
| |
Collapse
|
325
|
Pharmacologically-induced neurovascular uncoupling is associated with cognitive impairment in mice. J Cereb Blood Flow Metab 2015; 35:1871-81. [PMID: 26174328 PMCID: PMC4635246 DOI: 10.1038/jcbfm.2015.162] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/23/2015] [Accepted: 05/29/2015] [Indexed: 01/15/2023]
Abstract
There is increasing evidence that vascular risk factors, including aging, hypertension, diabetes mellitus, and obesity, promote cognitive impairment; however, the underlying mechanisms remain obscure. Cerebral blood flow (CBF) is adjusted to neuronal activity via neurovascular coupling (NVC) and this mechanism is known to be impaired in the aforementioned pathophysiologic conditions. To establish a direct relationship between impaired NVC and cognitive decline, we induced neurovascular uncoupling pharmacologically in mice by inhibiting the synthesis of vasodilator mediators involved in NVC. Treatment of mice with the epoxygenase inhibitor N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide (MSPPOH), the NO synthase inhibitor l-NG-Nitroarginine methyl ester (L-NAME), and the COX inhibitor indomethacin decreased NVC by over 60% mimicking the aging phenotype, which was associated with significantly impaired spatial working memory (Y-maze), recognition memory (Novel object recognition), and impairment in motor coordination (Rotarod). Blood pressure (tail cuff) and basal cerebral perfusion (arterial spin labeling perfusion MRI) were unaffected. Thus, selective experimental disruption of NVC is associated with significant impairment of cognitive and sensorimotor function, recapitulating neurologic symptoms and signs observed in brain aging and pathophysiologic conditions associated with accelerated cerebromicrovascular aging.
Collapse
|
326
|
Potey C, Ouk T, Petrault O, Petrault M, Berezowski V, Salleron J, Bordet R, Gautier S. Early treatment with atorvastatin exerts parenchymal and vascular protective effects in experimental cerebral ischaemia. Br J Pharmacol 2015; 172:5188-98. [PMID: 26289767 DOI: 10.1111/bph.13285] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/06/2015] [Accepted: 08/10/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE From the clinical and experimental data available, statins appear to be interesting drug candidates for preventive neuroprotection in ischaemic stroke. However, their acute protective effect is, as yet, unconfirmed. EXPERIMENTAL APPROACH Male C57Bl6/JRj mice were subjected to middle cerebral artery occlusion and treated acutely with atorvastatin (10-20 mg·kg(-1) day(-1) ; 24 or 72 h). Functional recovery (neuroscore, forelimb gripping strength and adhesive removal test) was assessed during follow-up and lesion volume measured at the end. Vasoreactivity of the middle cerebral artery (MCA), type IV collagen and FITC-dextran distribution were evaluated to assess macrovascular and microvascular protection. Activated microglia, leucocyte adhesion and infiltration were chosen as markers of inflammation. KEY RESULTS Acute treatment with atorvastatin provided parenchymal and cerebral protection only at the higher dose of 20 mg·kg(-1) ·day(-1) . In this treatment group, functional recovery was ameliorated, and lesion volumes were reduced as early as 24 h after experimental stroke. This was associated with vascular protection as endothelial function of the MCA and the density and patency of the microvascular network were preserved. Acute atorvastatin administration also induced an anti-inflammatory effect in association with parenchymal and vascular mechanisms; it reduced microglial activation, and decreased leucocyte adhesion and infiltration. CONCLUSIONS AND IMPLICATIONS Acute atorvastatin provides global cerebral protection, but only at the higher dose of 20 mg·kg(-1) ·day(-1) ; this was associated with a reduction in inflammation in both vascular and parenchymal compartments. Our results suggest that atorvastatin could also be beneficial when administered early after stroke.
Collapse
Affiliation(s)
- C Potey
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - T Ouk
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - O Petrault
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - M Petrault
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - V Berezowski
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - J Salleron
- EA2694 - Biostatistics Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - R Bordet
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| | - S Gautier
- U1171 - Medical Pharmacology Department, Faculty of Medicine, University of Lille 2 - Lille University Hospital, Lille, France
| |
Collapse
|
327
|
Korhonen P, Kanninen KM, Lehtonen Š, Lemarchant S, Puttonen KA, Oksanen M, Dhungana H, Loppi S, Pollari E, Wojciechowski S, Kidin I, García-Berrocoso T, Giralt D, Montaner J, Koistinaho J, Malm T. Immunomodulation by interleukin-33 is protective in stroke through modulation of inflammation. Brain Behav Immun 2015; 49:322-36. [PMID: 26111431 DOI: 10.1016/j.bbi.2015.06.013] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/05/2015] [Accepted: 06/18/2015] [Indexed: 01/19/2023] Open
Abstract
Cerebral stroke induces massive Th1-shifted inflammation both in the brain and the periphery, contributing to the outcome of stroke. A Th1-type response is neurotoxic whereas a Th2-type response is accompanied by secretion of anti-inflammatory cytokines, such as interleukin-4 (IL-4). Interleukin-33 (IL-33) is a cytokine known to induce a shift towards the Th2-type immune response, polarize macrophages/microglia towards the M2-type, and induce production of anti-inflammatory cytokines. We found that the plasma levels of the inhibitory IL-33 receptor, sST2, are increased in human stroke and correlate with a worsened stroke outcome, suggesting an insufficient IL-33-driven Th2-type response. In mouse, peripheral administration of IL-33 reduced stroke-induced cell death and improved the sensitivity of the contralateral front paw at 5days post injury. The IL-33-treated mice had increased levels of IL-4 in the spleen and in the peri-ischemic area of the cortex. Neutralization of IL-4 by administration of an IL-4 antibody partially prevented the IL-33-mediated protection. IL-33 treatment also reduced astrocytic activation in the peri-ischemic area and increased the number of Arginase-1 immunopositive microglia/macrophages at the lesion site. In human T-cells, IL-33 treatment induced IL-4 secretion, and the conditioned media from IL-33-exposed T-cells reduced astrocytic activation. This study demonstrates that IL-33 is protective against ischemic insult by induction of IL-4 secretion and may represent a novel therapeutic approach for the treatment of stroke.
Collapse
Affiliation(s)
- Paula Korhonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Katja M Kanninen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Šárka Lehtonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sighild Lemarchant
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Katja A Puttonen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Minna Oksanen
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Hiramani Dhungana
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sanna Loppi
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Eveliina Pollari
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Sara Wojciechowski
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Iurii Kidin
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Dolors Giralt
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Joan Montaner
- Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain
| | - Jari Koistinaho
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland.
| | - Tarja Malm
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| |
Collapse
|
328
|
Li H, Xie Y, Zhang N, Yu Y, Zhang Q, Ding S. Disruption of IP₃R2-mediated Ca²⁺ signaling pathway in astrocytes ameliorates neuronal death and brain damage while reducing behavioral deficits after focal ischemic stroke. Cell Calcium 2015; 58:565-76. [PMID: 26433454 DOI: 10.1016/j.ceca.2015.09.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 12/12/2022]
Abstract
Inositol trisphosphate receptor (IP3R)-mediated intracellular Ca(2+) increase is the major Ca(2+) signaling pathway in astrocytes in the central nervous system (CNS). Ca(2+) increases in astrocytes have been found to modulate neuronal function through gliotransmitter release. We previously demonstrated that astrocytes exhibit enhanced Ca(2+) signaling in vivo after photothrombosis (PT)-induced ischemia, which is largely due to the activation of G-protein coupled receptors (GPCRs). The aim of this study is to investigate the role of astrocytic IP3R-mediated Ca(2+) signaling in neuronal death, brain damage and behavior outcomes after PT. For this purpose, we conducted experiments using homozygous type 2 IP3R (IP3R2) knockout (KO) mice. Histological and immunostaining studies showed that IP3R2 KO mice were indeed deficient in IP3R2 in astrocytes and exhibited normal brain cytoarchitecture. IP3R2 KO mice also had the same densities of S100β+ astrocytes and NeuN+ neurons in the cortices, and exhibited the same glial fibrillary acidic protein (GFAP) and glial glutamate transporter (GLT-1) levels in the cortices and hippocampi as compared with wild type (WT) mice. Two-photon (2-P) imaging showed that IP3R2 KO mice did not exhibit ATP-induced Ca(2+) waves in vivo in the astrocytic network, which verified the disruption of IP3R-mediated Ca(2+) signaling in astrocytes of these mice. When subject to PT, IP3R2 KO mice had smaller infarction than WT mice in acute and chronic phases of ischemia. IP3R2 KO mice also exhibited less neuronal apoptosis, reactive astrogliosis, and tissue loss than WT mice. Behavioral tests, including cylinder, hanging wire, pole and adhesive tests, showed that IP3R2 KO mice exhibited reduced functional deficits after PT. Collectively, our study demonstrates that disruption of astrocytic Ca(2+) signaling by deleting IP3R2s has beneficial effects on neuronal and brain protection and functional deficits after stroke. These findings reveal a novel non-cell-autonomous neuronal and brain protective function of astrocytes in ischemic stroke, whereby suggest that the astrocytic IP3R2-mediated Ca(2+) signaling pathway might be a promising target for stroke therapy.
Collapse
Affiliation(s)
- Hailong Li
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211, United States; Department of Bioengineering, University of Missouri-Columbia, MO 65211, United States
| | - Yicheng Xie
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211, United States; Department of Bioengineering, University of Missouri-Columbia, MO 65211, United States
| | - Nannan Zhang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211, United States
| | - Yang Yu
- Department of Bioengineering, University of Missouri-Columbia, MO 65211, United States
| | - Qiao Zhang
- Department of Bioengineering, University of Missouri-Columbia, MO 65211, United States
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211, United States; Department of Bioengineering, University of Missouri-Columbia, MO 65211, United States.
| |
Collapse
|
329
|
Chen D, Lee J, Gu X, Wei L, Yu SP. Intranasal Delivery of Apelin-13 Is Neuroprotective and Promotes Angiogenesis After Ischemic Stroke in Mice. ASN Neuro 2015; 7:7/5/1759091415605114. [PMID: 26391329 PMCID: PMC4580122 DOI: 10.1177/1759091415605114] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Apelin is a peptide originally isolated from bovine stomach tissue extracts and identified as an endogenous ligand of the APJ receptor; recent work showed that apelin ameliorates the ischemic injury in the heart and the brain. Being an analogue to the angiotensin II receptor, the apelin/APJ signaling may mediate angiogenesis process. We explored the noninvasive intranasal brain delivery method and investigated therapeutic effects of apelin-13 in a focal ischemic stroke model of mice. Intranasal administration of apelin-13 (4 mg/kg) was given 30 min after the onset of stroke and repeated once daily. Three days after stroke, mice received apelin-13 had significantly reduced infarct volume and less neuronal death in the penumbra. Western blot analyses showed upregulated levels of apelin, apelin receptor APLNR, and Bcl-2 and decreased caspase-3 activation in the apelin-13-treated brain. The proinflammatory cytokines tumor necrosis factor-alpha, interleukin-1β, and chemokine monocyte chemoattractant protein-1 mRNA increased in the ischemic brain, which were significantly attenuated by apelin-13. Apelin-13 remarkably reduced microglia recruitment and activation in the penumbra according to morphological features of Iba-1-positive cells 3 days after ischemia. Apelin-13 significantly increased the expression of angiogenic factor vascular endothelial growth factor and matrix metalloproteinase-9 14 days after stroke. Angiogenesis illustrated by collagen IV + /5-bromo-2'-deoxyuridin + colabeled cells was significantly increased by the apelin-13 treatment 21 days after stroke. Finally, apelin-13 promoted the local cerebral blood flow restoration and long-term functional recovery. This study demonstrates a noninvasive intranasal delivery of apelin-13 after stroke, suggesting that the reduced inflammatory activities, decreased cell death, and increased angiogenesis contribute to the therapeutic benefits of apelin-13.
Collapse
Affiliation(s)
- Dongdong Chen
- Deptartment of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, USA
| | - Jinhwan Lee
- Deptartment of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, USA
| | - Xiaohuan Gu
- Deptartment of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, USA
| | - Ling Wei
- Deptartment of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, USA Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Shan Ping Yu
- Deptartment of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affair Medical Center, Decatur, GA, USA
| |
Collapse
|
330
|
Wei X, Zhang B, Cheng L, Chi M, Deng L, Pan H, Yao X, Wang G. Hydrogen sulfide induces neuroprotection against experimental stroke in rats by down-regulation of AQP4 via activating PKC. Brain Res 2015; 1622:292-9. [PMID: 26168888 DOI: 10.1016/j.brainres.2015.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/22/2015] [Accepted: 07/03/2015] [Indexed: 12/23/2022]
Abstract
Hydrogen sulfide (H2S) is now known as an important neuromodulator in the central nervous system. The aim of the current study was to investigate whether exogenous H2S gas can attenuate brain edema induced by experimental stroke and to clarify the potential mechanisms. Rats underwent 2-h middle cerebral artery occlusion (MCAO) and received 40 ppm or 80 ppm H2S inhalation for 3h at the beginning of reperfusion. The effects of H2S were investigated by evaluating neurological function, infarct size, brain edema volume, and aquaporin4 (AQP4) protein expression at 24h after reperfusion. Moreover, to explore the possible mechanisms for the neuroprotective effects of H2S, protein kinase C (PKC) activity was detected and a PKC inhibitor, Go6983, was used via intracerebral ventricular injection. Our results showed that 40 ppm or 80 ppm H2S inhalation significantly reduced neurological deficits, infarct size, and brain edema after MCAO. The expression of AQP4 in the peri-infarct area of brain was also inhibited after inhalation of H2S. PKC was activated by H2S treatment and the PKC inhibitor attenuated the neuroprotection of H2S with an increased AQP4 expression at the same time. In conclusion, H2S inhalation attenuates brain edema, reduces infarct volume, and improves neurologic function in a rat experimental stroke model. The therapeutic benefits of H2S inhalation are associated with down-regulation of AQP4 expression via activating PKC.
Collapse
Affiliation(s)
- Xia Wei
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang 150081, China
| | - Bing Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150086, China
| | - Long Cheng
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150086, China
| | - Meng Chi
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang 150081, China
| | - Lin Deng
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang 150081, China
| | - Hong Pan
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150086, China
| | - Xuan Yao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150086, China
| | - Guonian Wang
- Department of Anesthesiology, Cancer Hospital Affiliated to Harbin Medical University, 150 Haping Road, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
331
|
In vivo PET imaging of the α4β2 nicotinic acetylcholine receptor as a marker for brain inflammation after cerebral ischemia. J Neurosci 2015; 35:5998-6009. [PMID: 25878273 DOI: 10.1523/jneurosci.3670-14.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PET imaging of nicotinic acetylcholine receptors (nAChRs) could become an effective tool for the diagnosis and therapy evaluation of neurologic diseases. Despite this, the role of nAChRs α4β2 receptors after brain diseases such as cerebral ischemia and its involvement in inflammatory reaction is still largely unknown. To investigate this, we performed in parallel in vivo magnetic resonance imaging (MRI) and positron emission tomography (PET) with 2[(18)F]-fluoro-A85380 and [(11)C]PK11195 at 1, 3, 7, 14, 21, and 28 d after middle cerebral artery occlusion (MCAO) in rats. In the ischemic territory, PET with 2[(18)F]-fluoro-A85380 and [(11)C]PK11195 showed a progressive binding increase from days 3-7, followed by a progressive decrease from days 14-28 after cerebral ischemia onset. Ex vivo immunohistochemistry for the nicotinic α4β2 receptor and the mitochondrial translocator protein (18 kDa) (TSPO) confirmed the PET findings and demonstrated the overexpression of α4β2 receptors in both microglia/macrophages and astrocytes from days 7-28 after experimental ischemic stroke. Likewise, the role played by α4β2 receptors on neuroinflammation was supported by the increase of [(11)C]PK11195 binding in ischemic rats treated with the α4β2 antagonist dihydro-β-erythroidine hydrobromide (DHBE) at day 7 after MCAO. Finally, both functional and behavioral testing showed major impaired outcome at day 1 after ischemia onset, followed by a recovery of the sensorimotor function and dexterity from days 21-28 after experimental stroke. Together, these results suggest that the nicotinic α4β2 receptor could have a key role in the inflammatory reaction underlying cerebral ischemia in rats.
Collapse
|
332
|
Bentea E, Demuyser T, Van Liefferinge J, Albertini G, Deneyer L, Nys J, Merckx E, Michotte Y, Sato H, Arckens L, Massie A, Smolders I. Absence of system xc- in mice decreases anxiety and depressive-like behavior without affecting sensorimotor function or spatial vision. Prog Neuropsychopharmacol Biol Psychiatry 2015; 59:49-58. [PMID: 25619129 DOI: 10.1016/j.pnpbp.2015.01.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 01/05/2015] [Accepted: 01/16/2015] [Indexed: 01/22/2023]
Abstract
There is considerable preclinical and clinical evidence indicating that abnormal changes in glutamatergic signaling underlie the development of mood disorders. Astrocytic glutamate dysfunction, in particular, has been recently linked with the pathogenesis and treatment of mood disorders, including anxiety and depression. System xc- is a glial cystine/glutamate antiporter that is responsible for nonvesicular glutamate release in various regions of the brain. Although system xc- is involved in glutamate signal transduction, its possible role in mediating anxiety or depressive-like behaviors is currently unknown. In the present study, we phenotyped adult and aged system xc- deficient mice in a battery of tests for anxiety and depressive-like behavior (open field, light/dark test, elevated plus maze, novelty suppressed feeding, forced swim test, tail suspension test). Concomitantly, we evaluated the sensorimotor function of system xc- deficient mice, using motor and sensorimotor based tests (rotarod, adhesive removal test, nest building test). Finally, due to the presence and potential functional relevance of system xc- in the eye, we investigated the visual acuity of system xc- deficient mice (optomotor test). Our results indicate that loss of system xc- does not affect motor or sensorimotor function, in either adult or aged mice, in any of the paradigms investigated. Similarly, loss of system xc- does not affect basic visual acuity, in either adult or aged mice. On the other hand, in the open field and light/dark tests, and forced swim and tail suspension tests respectively, we could observe significant anxiolytic and antidepressive-like effects in system xc- deficient mice that in certain cases (light/dark, forced swim) were age-dependent. These findings indicate that, under physiological conditions, nonvesicular glutamate release via system xc- mediates aspects of higher brain function related to anxiety and depression, but does not influence sensorimotor function or spatial vision. As such, modulation of system xc- might constitute the basis of innovative interventions in mood disorders.
Collapse
Affiliation(s)
- Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Demuyser
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lauren Deneyer
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Julie Nys
- Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology, KU Leuven, Leuven, Belgium
| | - Ellen Merckx
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yvette Michotte
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hideyo Sato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Technology, Faculty of Medicine, Niigata University, Niigata, Japan
| | - Lutgarde Arckens
- Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology, KU Leuven, Leuven, Belgium
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
333
|
Eckert A, Huang L, Gonzalez R, Kim HS, Hamblin MH, Lee JP. Bystander Effect Fuels Human Induced Pluripotent Stem Cell-Derived Neural Stem Cells to Quickly Attenuate Early Stage Neurological Deficits After Stroke. Stem Cells Transl Med 2015; 4:841-51. [PMID: 26025980 DOI: 10.5966/sctm.2014-0184] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 04/06/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED : Present therapies for stroke rest with tissue plasminogen activator (tPA), the sole licensed antithrombotic on the market; however, tPA's effectiveness is limited in that the drug not only must be administered less than 3-5 hours after stroke but often exacerbates blood-brain barrier (BBB) leakage and increases hemorrhagic incidence. A potentially promising therapy for stroke is transplantation of human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs). To date, the effects of iPSCs on injuries that take place during early stage ischemic stroke have not been well studied. Consequently, we engrafted iPSC-NSCs into the ipsilesional hippocampus, a natural niche of NSCs, at 24 hours after stroke (prior to secondary BBB opening and when inflammatory signature is abundant). At 48 hours after stroke (24 hours after transplant), hiPSC-NSCs had migrated to the stroke lesion and quickly improved neurological function. Transplanted mice showed reduced expression of proinflammatory factors (tumor necrosis factor-α, interleukin 6 [IL-6], IL-1β, monocyte chemotactic protein 1, macrophage inflammatory protein 1α), microglial activation, and adhesion molecules (intercellular adhesion molecule 1, vascular cell adhesion molecule 1) and attenuated BBB damage. We are the first to report that engrafted hiPSC-NSCs rapidly improved neurological function (less than 24 hours after transplant). Rapid hiPSC-NSC therapeutic activity is mainly due to a bystander effect that elicits reduced inflammation and BBB damage. SIGNIFICANCE Clinically, cerebral vessel occlusion is rarely permanent because of spontaneous or thrombolytic therapy-mediated reperfusion. These results have clinical implications indicating a much extended therapeutic window for transplantation of human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs; 24 hours after stroke as opposed to the 5-hour window with tissue plasminogen activator [tPA]). In addition, there is potential for a synergistic effect by combining hiPSC-NSC transplantation with tPA to attenuate stroke's adverse effects.
Collapse
Affiliation(s)
- Auston Eckert
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Lei Huang
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Rodolfo Gonzalez
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Hye-Sun Kim
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Milton H Hamblin
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| | - Jean-Pyo Lee
- Center for Stem Cell Research and Regenerative Medicine, Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA; Sanford-Burnham Institute for Medical Research, Neuroscience, Aging and Stem Cell Research, La Jolla, California, USA; Seoul National University, College of Medicine, Department of Pharmacology, Seoul, Republic of Korea
| |
Collapse
|
334
|
Yang Y, Zhang M, Kang X, Jiang C, Zhang H, Wang P, Li J. Impaired adult hippocampal neurogenesis and cognitive ability in a mouse model of intrastriatal hemorrhage. Neurosci Lett 2015; 599:133-9. [PMID: 26021875 DOI: 10.1016/j.neulet.2015.05.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/19/2015] [Accepted: 05/22/2015] [Indexed: 12/14/2022]
Abstract
Thrombin released by hematoma is an important mediator of the secondary injury of intracerebral hemorrhage (ICH), however, the effect of thrombin on adult neurogenesis and cognitive ability remains elusive. In this study, intrastriatal injection of 0.05 U thrombin didn't affect the neurogenesis at the subgranular zone (SGZ), which was distal to the injection site. 0.1 U thrombin increased the 5-bromo-2-deoxyuridine(+) (BrdU(+), S-phase proliferating cells)/doublecortin(+) (DCX(+), immature neurons) double labelled neurons, but decreased BrdU(+)/NeuN(+) double labelled mature neurons. Higher doses of thrombin (1 U, 2 U, and 5 U) significantly decreased the BrdU(+)/DCX(+) and BrdU(+)/NeuN(+) double labelled cells. After 1 U thrombin injection, cell apoptosis was found at the dentate gyrus of hippocampus at 3-24 h, but not 5 d post-injury. Thrombin infusion (1 U) induced spatial memory deficits in Morris water maze test; whereas, hirudin, the thrombin antagonist, significantly reversed both neurogenesis loss and spatial learning and memory impairment. In conclusion, at least at short term (5 days) after striatum ICH, the effect of high dose of thrombin on neurogenesis of SGZ, and the spatial learning and memory ability, is detrimental.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Neurological, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Meikui Zhang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China.
| | - Xiaoni Kang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Chen Jiang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Huan Zhang
- Department Students Brigade, The Second Military Medical University, No. 800, Xiangyin Road, Shanghai 200433, China
| | - Pei Wang
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| | - Jingjing Li
- Department of Telemedicine Center, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing 100036, China
| |
Collapse
|
335
|
Mysoet J, Dupont E, Bastide B, Canu MH. Role of IGF-1 in cortical plasticity and functional deficit induced by sensorimotor restriction. Behav Brain Res 2015; 290:117-23. [PMID: 25958232 DOI: 10.1016/j.bbr.2015.04.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/15/2022]
Abstract
In the adult rat, sensorimotor restriction by hindlimb unloading (HU) is known to induce impairments in motor behavior as well as a disorganization of somatosensory cortex (shrinkage of the cortical representation of the hindpaw, enlargement of the cutaneous receptive fields, decreased cutaneous sensibility threshold). Recently, our team has demonstrated that IGF-1 level was decreased in the somatosensory cortex of rats submitted to a 14-day period of HU. To determine whether IGF-1 is involved in these plastic mechanisms, a chronic cortical infusion of this substance was performed by means of osmotic minipump. When administered in control rats, IGF-1 affects the size of receptive fields and the cutaneous threshold, but has no effect on the somatotopic map. In addition, when injected during the whole HU period, IGF-1 is interestingly implied in cortical changes due to hypoactivity: the shrinkage of somatotopic representation of hindlimb is prevented, whereas the enlargement of receptive fields is reduced. IGF-1 has no effect on the increase in neuronal response to peripheral stimulation. We also explored the functional consequences of IGF-1 level restoration on tactile sensory discrimination. In HU rats, the percentage of paw withdrawal after a light tactile stimulation was decreased, whereas it was similar to control level in HU-IGF-1 rats. Taken together, the data clearly indicate that IGF-1 plays a key-role in cortical plastic mechanisms and in behavioral alterations induced by a decrease in sensorimotor activity.
Collapse
Affiliation(s)
- Julien Mysoet
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| | - Erwan Dupont
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| | - Bruno Bastide
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| | - Marie-Hélène Canu
- Laboratoire « Activité Physique, Muscle et Santé », EA 4488, IFR 114, Université Lille 1, Sciences et Technologies, F-59650 Villeneuve d'Ascq, France; Unité de Recherche Pluridisciplinaire Sport, Santé, Société (URePSSS), Université Lille Nord de France, F-59000 Lille, France.
| |
Collapse
|
336
|
Yu J, Zhu H, Gattoni-Celli S, Taheri S, Kindy MS. Dietary supplementation of GrandFusion(®) mitigates cerebral ischemia-induced neuronal damage and attenuates inflammation. Nutr Neurosci 2015; 19:290-300. [PMID: 25879584 DOI: 10.1179/1476830515y.0000000021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Dietary supplementation of fruits and vegetables has been the main stay for nutritional benefit and overall well-being. GrandFusion(®) is a nutritional supplement that contains the natural nutrients from whole fruits and vegetables that include complex nutrients and phytonutrients that contain anti-oxidant, anti-inflammatory, and neuroprotective properties. METHODS In this study, C57BL/6 mice were fed a diet supplemented with GrandFusion(®) for 2 months prior to 1 hour of ischemia induced by occlusion of the middle cerebral artery (MCAo) followed by various times of reperfusion. Mice were subjected to MCAo for 1 hour and then at various times following reperfusion, animals were assessed for behavioral outcomes (open field testing, rotarod, and adhesive test removal), and infarct volumes (cresyl violet and triphenyltetrazolium chloride). In addition, to determine the potential mechanisms associated with treatment, the brain tissue was examined for changes in oxidative stress and inflammatory markers. RESULTS The GrandFusion(®) diet was able to show a significant protection from infarct damage in the brain and an improvement in neurological outcomes. The diet did not alter heart rate, blood pressure, pO2, pCO2, or pH. In addition, the diet mitigated inflammation by reducing microglial and astrocytic activation following ischemia and reperfusion and limiting oxidative stress. DISCUSSION The study demonstrates the neuroprotective effect of a diet rich in fruits and vegetables that contain anti-oxidant and anti-inflammatory against the impact of cerebral ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Jin Yu
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA
| | - Hong Zhu
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA
| | - Sebastiano Gattoni-Celli
- b Department of Radiation Oncology , Medical University of South Carolina , Charleston , SC , USA.,c Ralph H. Johnson VA Medical Center , Charleston , SC , USA
| | - Saeid Taheri
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA
| | - Mark Stephen Kindy
- a Department of Regenerative Medicine and Cell Biology , Medical University of South Carolina , Charleston , SC , USA.,c Ralph H. Johnson VA Medical Center , Charleston , SC , USA
| |
Collapse
|
337
|
Bentea E, Van der Perren A, Van Liefferinge J, El Arfani A, Albertini G, Demuyser T, Merckx E, Michotte Y, Smolders I, Baekelandt V, Massie A. Nigral proteasome inhibition in mice leads to motor and non-motor deficits and increased expression of Ser129 phosphorylated α-synuclein. Front Behav Neurosci 2015; 9:68. [PMID: 25873870 PMCID: PMC4379937 DOI: 10.3389/fnbeh.2015.00068] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 02/27/2015] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disorder characterized by motor and non-motor disturbances. Various pathogenic pathways drive disease progression including oxidative stress, mitochondrial dysfunction, α-synuclein aggregation and impairment of protein degradation systems. Dysfunction of the ubiquitin-proteasome system in the substantia nigra of Parkinson's disease patients is believed to be one of the causes of protein aggregation and cell death associated with this disorder. Lactacystin, a potent inhibitor of the proteasome, was previously delivered to the nigrostriatal pathway of rodents to model nigrostriatal degeneration. Although lactacystin-treated animals develop parkinsonian motor impairment, it is currently unknown whether they also develop non-motor symptoms characteristic of this disorder. In order to further describe the proteasome inhibition model of Parkinson's disease, we characterized the unilateral lactacystin model, performed by stereotaxic injection of the toxin in the substantia nigra of mice. We studied the degree of neurodegeneration and the behavioral phenotype 1 and 3 weeks after lactacystin lesion both in terms of motor impairment, as well as non-motor symptoms. We report that unilateral administration of 3 μg lactacystin to the substantia nigra of mice leads to partial (~40%) dopaminergic cell loss and concurrent striatal dopamine depletion, accompanied by increased expression of Ser129-phosphorylated α-synuclein. Behavioral characterization of the model revealed parkinsonian motor impairment, as well as signs of non-motor disturbances resembling early stage Parkinson's disease including sensitive and somatosensory deficits, anxiety-like behavior, and perseverative behavior. The consistent finding of good face validity, together with relevant construct validity, warrant a further evaluation of proteasome inhibition models of Parkinson's disease in pre-clinical research and validation of therapeutic targets.
Collapse
Affiliation(s)
- Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Anke Van der Perren
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Anissa El Arfani
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Thomas Demuyser
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Ellen Merckx
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Yvette Michotte
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel Brussels, Belgium
| |
Collapse
|
338
|
Zhang L, Zhang RL, Jiang Q, Ding G, Chopp M, Zhang ZG. Focal embolic cerebral ischemia in the rat. Nat Protoc 2015; 10:539-47. [PMID: 25741989 DOI: 10.1038/nprot.2015.036] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Animal models of focal cerebral ischemia are well accepted for investigating the pathogenesis and potential treatment strategies for human stroke. Occlusion of the middle cerebral artery (MCA) with an endovascular filament is a widely used model to induce focal cerebral ischemia. However, this model is not amenable to thrombolytic therapies. As thrombolysis with recombinant tissue plasminogen activator (rtPA) is a standard of care within 4.5 h of human stroke onset, suitable animal models that mimic cellular and molecular mechanisms of thrombosis and thrombolysis of stroke are required. By occluding the MCA with a fibrin-rich allogeneic clot, we previously developed an embolic model of MCA occlusion in the rat, which recapitulates the key components of thrombotic development and of thrombolytic therapy of rtPA observed from human ischemic stroke. Here we describe in detail the surgical procedures of our model, including preparing emboli from rat donors. These procedures can be typically completed within ∼30 min, and they are highly adaptable to other strains of rats, as well as mice, in both sexes. Thus, this model provides a powerful tool for translational stroke research.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Rui Lan Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- 1] Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA. [2] Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
339
|
Wei ZZ, Gu X, Ferdinand A, Lee JH, Ji X, Ji XM, Yu SP, Wei L. Intranasal Delivery of Bone Marrow Mesenchymal Stem Cells Improved Neurovascular Regeneration and Rescued Neuropsychiatric Deficits after Neonatal Stroke in Rats. Cell Transplant 2015; 24:391-402. [DOI: 10.3727/096368915x686887] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neonatal stroke is a major cause of mortality and long-term morbidity in infants and children. Currently, very limited therapeutic strategies are available to protect the developing brain against ischemic damage and promote brain repairs for pediatric patients. Moreover, children who experienced neonatal stroke often have developmental social behavior problems. Cellular therapy using bone marrow mesenchymal stem cells (BMSCs) has emerged as a regenerative therapy after stroke. In the present investigation, neonatal stroke of postnatal day 7 (P7) rat pups was treated with noninvasive and brain-specific intranasal delivery of BMSCs at 6 h and 3 days after stroke (1 × 106cells/animal). Prior to transplantation, BMSCs were subjected to hypoxic preconditioning to enhance their tolerance and regenerative properties. The effects on regenerative activities and stroke-induced sensorimotor and social behavioral deficits were specifically examined at P24 of juvenile age. The BMSC treatment significantly reduced infarct size and blood-brain barrier disruption, promoted angiogenesis, neurogenesis, neurovascular repair, and improved local cerebral blood flow in the ischemic cortex. BMSC-treated rats showed better sensorimotor and olfactory functional recovery than saline-treated animals, measured by the adhesive removal test and buried food finding test. In social behavioral tests, we observed functional and social behavioral deficits in P24 rats subjected to stroke at P7, while the BMSC treatment significantly improved the performance of stroke animals. Overall, intranasal BMSC transplantation after neonatal stroke shows neuroprotection and great potential as a regenerative therapy to enhance neurovascular regeneration and improve functional recovery observed at the juvenile stage of development.
Collapse
Affiliation(s)
- Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Anwar Ferdinand
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaoya Ji
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Xun Ming Ji
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
340
|
Lee JH, Wei ZZ, Chen D, Gu X, Wei L, Yu SP. A neuroprotective role of the NMDA receptor subunit GluN3A (NR3A) in ischemic stroke of the adult mouse. Am J Physiol Cell Physiol 2015; 308:C570-7. [PMID: 25652449 DOI: 10.1152/ajpcell.00353.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/02/2015] [Indexed: 11/22/2022]
Abstract
GluN3A or NR3A is a developmentally regulated N-methyl-d-aspartate receptor (NMDAR) subunit, showing a unique inhibitory role that decreases NMDAR current and the receptor-mediated Ca(2+) influx. In the neonatal brain, GluN3A is shown to associate with synaptic maturation and spine formation and plays a neuroprotective role. Its functional role in the adult brain, however, is largely unknown. We tested the hypothesis that, disrespecting the relatively lower expression level of GluN3A in the adult brain, this inhibitory NMDAR subunit shows a protective action against ischemia-induced brain injury. In littermate wild-type (WT) and GluN3A knockout (KO) mice, focal cerebral ischemia was induced by permanent occlusion of right distal branches of the middle cerebral artery (MCA) plus 10-min ligation of both common carotid arteries (CCAs). Twenty-four hours after focal cerebral ischemia, the infarction volume assessed using 2,3,5-triphenyltetrazolium chloride (TTC) staining was significantly larger in GluN3A KO mice compared with WT mice. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining demonstrated enhanced cell death in GluN3A KO mice. Moreover, the deletion of GluN3A hindered sensorimotor functional recovery after stroke. It is suggested that, although the expression level is relatively lower in the adult brain, GluN3A is still a noteworthy regulator in ischemia-induced excitotoxicity and brain injury.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia; and
| | - Zheng Z Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia; and Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Dongdong Chen
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia; and Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia; and Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia; and Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia; and Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| |
Collapse
|
341
|
Zheng Y, Zhong D, Chen H, Ma S, Sun Y, Wang M, Liu Q, Li G. Pivotal role of cerebral interleukin-23 during immunologic injury in delayed cerebral ischemia in mice. Neuroscience 2015; 290:321-31. [PMID: 25637493 DOI: 10.1016/j.neuroscience.2015.01.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/18/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
Abstract
BACKGROUND Interleukin-23 (IL-23) is required for T helper 17 (Th17) cell responses and IL-17 production in ischemic stroke. We previously showed that the IL-23/IL-17 axis aggravates immune injury after cerebral infarction in mice. However, IL-23 might activate other cytokines and transcription factor forkhead box P3 (Foxp3) production in cerebral ischemia. We aimed to determine whether IL-23p19 knockdown prevents cerebral ischemic injury by reducing ischemic-induced inflammation. METHODS Ischemic stroke models were established by permanent middle cerebral arterial occlusion (pMCAO) in male C57BL/6 mice. In vivo gene knockdown was achieved by intravenous delivery of lentiviral vectors (LVs) encoding IL-23p19 short hairpin RNA (LV-IL-23p19 shRNA). Enzyme-linked immunoassay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR) confirmed inhibitory efficiency. Behavioral deficits were evaluated by adhesive-removal somatic-sensory test. Brain infarct volume was measured at day 5 after pMCAO by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Expression of IL-17, IL-4, interferon (IFN)-γ and Foxp3 in ischemic brain tissues were detected by qRT-PCR and Western blotting, respectively. Additionally, immunohistochemical staining located cytokines in ischemic brain tissues. RESULTS RNA interference knockdown of IL-23p19 resulted in improved neurological function and reduced infarct volume. IL-23p19 knockdown suppressed IL-17 gene and protein expression. Moreover, IL-23p19 deficiency enhanced IFN-γ and Foxp3 expressions in delayed cerebral ischemic mice, and did not impact IL-4 expression. Immunohistochemical staining showed that IL-17, IL-4, IFN-γ and Foxp3-positive cells were located around ischemic lesions of the ipsilateral hemisphere. CONCLUSIONS IL-23p19 knockdown prevents delayed cerebral ischemic injury by dampening the ischemia-induced inflammation, and is a promising approach for clinically managing ischemic stroke.
Collapse
Affiliation(s)
- Y Zheng
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - D Zhong
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - H Chen
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - S Ma
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - Y Sun
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - M Wang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - Q Liu
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China
| | - G Li
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilong Jiang Province, PR China.
| |
Collapse
|
342
|
Huang L, Wong S, Snyder EY, Hamblin MH, Lee JP. Human neural stem cells rapidly ameliorate symptomatic inflammation in early-stage ischemic-reperfusion cerebral injury. Stem Cell Res Ther 2014; 5:129. [PMID: 25418536 PMCID: PMC4445985 DOI: 10.1186/scrt519] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 11/04/2014] [Indexed: 01/03/2023] Open
Abstract
Introduction Clinically, a good deal of injury from stroke results from ischemic-reperfusion. There is a loss of cerebral parenchyma and its associated cells, disruption of neuronal connections, compromise of the blood-brain barrier, and inflammation. We tested whether exogenously engrafted human neural stem cells could migrate rapidly and extensively to damaged regions, following transplantation into a neurogenic site where migration cues are already underway during stroke onset, then counteract a number of these pathological processes. Methods One day post-injury, we injected human neural stem cells (hNSCs) into the ipsilesional hippocampus of a mouse model of stroke with middle cerebral artery occlusion to induce focal ischemia followed by reperfusion (MCAO/R). The time frame for hNSC transplantation corresponded to upregulation of endogenous proinflammatory cytokines. We examined the effect of hNSC transplantation on pathological processes and behavioral dysfunction 48 hours post-injury. Results Twenty-four hours after transplantation, engrafted hNSCs had migrated extensively to the lesion, and infarct volume was reduced relative to MCAO/R controls. The behavioral deficits seen in MCAO/R controls were also significantly improved. Given this rapid response, we hypothesized that the mechanisms underlying therapeutic activity were anti-inflammatory rather than due to cell replacement. In support of this idea, in hNSC-transplanted mice we observed reduced microglial activation, decreased expression of proinflammatory factors (tumor necrosis factor-α, interleukin (IL)-6, IL-1β, monocyte chemotactic protein-1, macrophage inflammatory protein-1α) and adhesion molecules (intercellular adhesion molecule-1, vascular cell adhesion molecule-1), and amelioration of blood-brain barrier damage. Conclusions While long-term effects of engrafted hNSCs on the amelioration of ischemic stroke-induced behavioral dysfunction in a rodent model have been reported, our study is the first to show rapid, beneficial impacts on behavioral function (within 24 hours) upon early delivery of hNSCs into the hippocampus. Electronic supplementary material The online version of this article (doi:10.1186/scrt519) contains supplementary material, which is available to authorized users.
Collapse
|
343
|
A Smoothened receptor agonist is neuroprotective and promotes regeneration after ischemic brain injury. Cell Death Dis 2014; 5:e1481. [PMID: 25341035 PMCID: PMC4649529 DOI: 10.1038/cddis.2014.446] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/08/2014] [Accepted: 09/09/2014] [Indexed: 12/26/2022]
Abstract
Ischemic stroke occurs as a result of blood supply interruption to the brain causing tissue degeneration, patient disabilities or death. Currently, treatment of ischemic stroke is limited to thrombolytic therapy with a narrow time window of administration. The sonic hedgehog (Shh) signaling pathway has a fundamental role in the central nervous system development, but its impact on neural cell survival and tissue regeneration/repair after ischemic stroke has not been well investigated. Here we report the neuroprotective properties of a small-molecule agonist of the Shh co-receptor Smoothened, purmorphamine (PUR), in the middle cerebral artery occlusion model of ischemic stroke. We found that intravenous administration of PUR at 6 h after injury was neuroprotective and restored neurological deficit after stroke. PUR promoted a transient upregulation of tissue-type plasminogen activator in injured neurons, which was associated with a reduction of apoptotic cell death in the ischemic cortex. We also observed a decrease in blood–brain barrier permeability after PUR treatment. At 14 d postinjury, attenuation of inflammation and reactive astrogliosis was found in PUR-treated animals. PUR increased the number of newly generated neurons in the peri-infarct and infarct area and promoted neovascularization in the ischemic zone. Notably, PUR treatment did not significantly alter the ischemia-induced level of Gli1, a Shh target gene of tumorigenic potential. Thus our study reports a novel pharmacological approach for postischemic treatment using a small-molecule Shh agonist, providing new insights into hedgehog signaling-mediated mechanisms of neuroprotection and regeneration after stroke.
Collapse
|
344
|
Prakash A, Chopra K, Medhi B. Granulocyte-colony stimulating factor improves Parkinson's disease associated with co-morbid depression: an experimental exploratory study. Indian J Pharmacol 2014; 45:612-5. [PMID: 24347771 PMCID: PMC3847253 DOI: 10.4103/0253-7613.121374] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 04/26/2013] [Accepted: 09/18/2013] [Indexed: 11/23/2022] Open
Abstract
Introduction: The present study was designed to evaluate the effect of granulocyte-colony stimulating factor (G-CSF) in the treatment of Parkinson's disease (PD), the second most common neurodegenerative disease characterized by muscle and movement disorder, often associated with depression. PD is very difficult to treat. Hence, the present study was aimed to evaluate the effect of G-CSF in PD associated with depression. Materials and Methods: Adult Wistar male rats weighing about 180-250 g were selected and divided into five groups in parallel designed method namely; control group (n = 5); sham operated group (n = 5); Vehicle group (n = 5); G-CSF group (70 μg/kg, s.c.) (n = 5) and L-DOPA group (n = 5). The rats were treated with 6-hydroxydopamine (6-OHDA) on day 0 and then treatment was continued for 14 day of L-DOPA/carbidopa, whereas G-CSF (70 μg/kg, s.c.) was given from day 1 to 6. Thereafter, adhesive removal and forced swim tests were conducted to evaluate the behavioral outcome of G-CSF treatment. The finding was correlated and analyzed with Nissl staining findings for the final conclusion. Results: The behavioral parameters were assessed and found to be ameliorate the symptoms of Parkinson's and reduced the depression like behavior in PD. The histological findings were supported the behavioral findings and showed pathological improvement. Conclusion: As a preliminary work, the present study first time suggested that G-CSF have a potential role in PD and associated depression.
Collapse
Affiliation(s)
- Ajay Prakash
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kanwaljit Chopra
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
345
|
Llovera G, Roth S, Plesnila N, Veltkamp R, Liesz A. Modeling stroke in mice: permanent coagulation of the distal middle cerebral artery. J Vis Exp 2014:e51729. [PMID: 25145316 PMCID: PMC4692348 DOI: 10.3791/51729] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stroke is the third most common cause of death and a main cause of acquired adult disability in developed countries. Only very limited therapeutical options are available for a small proportion of stroke patients in the acute phase. Current research is intensively searching for novel therapeutic strategies and is increasingly focusing on the sub-acute and chronic phase after stroke because more patients might be eligible for therapeutic interventions in a prolonged time window. These delayed mechanisms include important pathophysiological pathways such as post-stroke inflammation, angiogenesis, neuronal plasticity and regeneration. In order to analyze these mechanisms and to subsequently evaluate novel drug targets, experimental stroke models with clinical relevance, low mortality and high reproducibility are sought after. Moreover, mice are the smallest mammals in which a focal stroke lesion can be induced and for which a broad spectrum of transgenic models are available. Therefore, we describe here the mouse model of transcranial, permanent coagulation of the middle cerebral artery via electrocoagulation distal of the lenticulostriatal arteries, the so-called “coagulation model”. The resulting infarct in this model is located mainly in the cortex; the relative infarct volume in relation to brain size corresponds to the majority of human strokes. Moreover, the model fulfills the above-mentioned criteria of reproducibility and low mortality. In this video we demonstrate the surgical methods of stroke induction in the “coagulation model” and report histological and functional analysis tools.
Collapse
Affiliation(s)
- Gemma Llovera
- Institute for Stroke and Dementia Research, University Hospital Munich; Munich Cluster for Systems Neurology (SyNergy)
| | - Stefan Roth
- Institute for Stroke and Dementia Research, University Hospital Munich; Munich Cluster for Systems Neurology (SyNergy)
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, University Hospital Munich; Munich Cluster for Systems Neurology (SyNergy)
| | - Roland Veltkamp
- Department of Neurology, University Heidelberg; Imperial College, Charing Cross Hospital
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Hospital Munich; Munich Cluster for Systems Neurology (SyNergy);
| |
Collapse
|
346
|
Mao-Ying QL, Kavelaars A, Krukowski K, Huo XJ, Zhou W, Price TJ, Cleeland C, Heijnen CJ. The anti-diabetic drug metformin protects against chemotherapy-induced peripheral neuropathy in a mouse model. PLoS One 2014; 9:e100701. [PMID: 24955774 PMCID: PMC4067328 DOI: 10.1371/journal.pone.0100701] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 05/27/2014] [Indexed: 12/05/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) characterized by loss of sensory sensitivity and pain in hands and feet is the major dose-limiting toxicity of many chemotherapeutics. At present, there are no FDA-approved treatments for CIPN. The anti-diabetic drug metformin is the most widely used prescription drug in the world and improves glycemic control in diabetes patients. There is some evidence that metformin enhances the efficacy of cancer treatment. The aim of this study was to test the hypothesis that metformin protects against chemotherapy-induced neuropathic pain and sensory deficits. Mice were treated with cisplatin together with metformin or saline. Cisplatin induced increased sensitivity to mechanical stimulation (mechanical allodynia) as measured using the von Frey test. Co-administration of metformin almost completely prevented the cisplatin-induced mechanical allodynia. Co-administration of metformin also prevented paclitaxel-induced mechanical allodynia. The capacity of the mice to detect an adhesive patch on their hind paw was used as a novel indicator of chemotherapy-induced sensory deficits. Co-administration of metformin prevented the cisplatin-induced increase in latency to detect the adhesive patch indicating that metformin prevents sensory deficits as well. Moreover, metformin prevented the reduction in density of intra-epidermal nerve fibers (IENFs) in the paw that develops as a result of cisplatin treatment. We conclude that metformin protects against pain and loss of tactile function in a mouse model of CIPN. The finding that metformin reduces loss of peripheral nerve endings indicates that mechanism underlying the beneficial effects of metformin includes a neuroprotective activity. Because metformin is widely used for treatment of type II diabetes, has a broad safety profile, and is currently being tested as an adjuvant drug in cancer treatment, clinical translation of these findings could be rapidly achieved.
Collapse
Affiliation(s)
- Qi-Liang Mao-Ying
- Neuroimmunology Laboratory, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Annemieke Kavelaars
- Neuroimmunology Laboratory, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Karen Krukowski
- Neuroimmunology Laboratory, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Xiao-Jiao Huo
- Neuroimmunology Laboratory, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Wenjun Zhou
- Neuroimmunology Laboratory, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, United States of America
| | - Charles Cleeland
- Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Cobi J. Heijnen
- Neuroimmunology Laboratory, Department of Symptom Research, University of Texas M.D. Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
347
|
Liu NK, Zhang YP, Zou J, Verhovshek T, Chen C, Lu QB, Walker CL, Shields CB, Xu XM. A semicircular controlled cortical impact produces long-term motor and cognitive dysfunction that correlates well with damage to both the sensorimotor cortex and hippocampus. Brain Res 2014; 1576:18-26. [PMID: 24905625 DOI: 10.1016/j.brainres.2014.05.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/22/2014] [Accepted: 05/28/2014] [Indexed: 11/29/2022]
Abstract
Animal models of traumatic brain injury (TBI) are essential for testing novel hypotheses and therapeutic interventions. Unfortunately, due to the broad heterogeneity of TBI in humans, no single model has been able to reproduce the entire spectrum of these injuries. The controlled cortical impact (CCI) model is one of the most commonly used models of contusion TBI. However, behavioral evaluations have revealed transient impairment in motor function after CCI in rats and mice. Here we report a new semicircular CCI (S-CCI) model by increasing the impact tip area to cover both the motor cortex and hippocampal regions in adult mice. Mice were subjected to S-CCI or CCI using an electromagnetic impactor (Impactor One, MyNeuroLab; semicircular tip: 3mm radius; CCI tip diameter: 3mm). We showed that S-CCI, at two injury severities, significantly decreased the neuroscore and produced deficits in performance on a rotarod device for the entire duration of the study. In contrast, the CCI induced motor deficits only at early stages after the injury, suggesting that the S-CCI model produces long-lasting motor deficits. Morris water maze test showed that both CCI and S-CCI produced persisting memory deficits. Furthermore, adhesive removal test showed significant somatosensory and motor deficits only in the S-CCI groups. Histological analysis showed a large extent of cortical contusion lesions, including both the sensory and motor cortex, and hippocampal damage in the S-CCI. These findings collectively suggest that the current model may offer sensitive, reliable, and clinically relevant outcomes for assessments of therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Department of Anatomy and Cell Biology, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yi-Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, Louisville, KY 40202, USA
| | - Jian Zou
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Department of Anatomy and Cell Biology, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tom Verhovshek
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Department of Anatomy and Cell Biology, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chen Chen
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Department of Anatomy and Cell Biology, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Qing-Bo Lu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Department of Anatomy and Cell Biology, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chandler L Walker
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Department of Anatomy and Cell Biology, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Department of Anatomy and Cell Biology, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
348
|
Li H, Zhang N, Lin HY, Yu Y, Cai QY, Ma L, Ding S. Histological, cellular and behavioral assessments of stroke outcomes after photothrombosis-induced ischemia in adult mice. BMC Neurosci 2014; 15:58. [PMID: 24886391 PMCID: PMC4039545 DOI: 10.1186/1471-2202-15-58] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/22/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Following the onset of focal ischemic stroke, the brain experiences a series of alterations including infarct evolvement, cellular proliferation in the penumbra, and behavioral deficits. However, systematic study on the temporal and spatial dependence of these alterations has not been provided. RESULTS Using multiple approaches, we assessed stroke outcomes by measuring brain injury, dynamic cellular and glial proliferation, and functional deficits at different times up to two weeks after photothrombosis (PT)-induced ischemic stroke in adult mice. Results from magnetic resonance imaging (MRI) and Nissl staining showed a maximal infarction, and brain edema and swelling 1-3 days after PT. The rate of Bromodeoxyuridine (Brdu)-labeled proliferating cell generation is spatiotemporal dependent in the penumbra, with the highest rate in post ischemic days 3-4, and higher rate of proliferation in the region immediate to the ischemic core than in the distant region. Similar time-dependent generation of proliferating GFAP+ astrocytes and Iba1+ microglia/macrophage were observed in the penumbra. Using behavioral tests, we showed that PT resulted in the largest functional deficits during post ischemic days 2-4. CONCLUSION Our study demonstrated that first a few days is a critical period that causes brain expansion, cellular proliferation and behavioral deficits in photothrombosis-induced ischemic model, and proliferating astrocytes only have a small contribution to the pools of proliferating cells and reactive astrocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, 134 Research Park Drive, Columbia, MO 65211, USA.
| |
Collapse
|
349
|
Venna VR, Li J, Hammond MD, Mancini NS, McCullough LD. Chronic metformin treatment improves post-stroke angiogenesis and recovery after experimental stroke. Eur J Neurosci 2014; 39:2129-38. [PMID: 24649970 DOI: 10.1111/ejn.12556] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 02/06/2023]
Abstract
Metformin is currently the first-line treatment drug for type 2 diabetes. Metformin is a well-known activator of AMP-activated protein kinase (AMPK). In experimental studies, metformin has been shown to exert direct vascular effects by increasing vascular endothelial growth factor expression and improving microvascular density. As stroke is the leading cause of long-term disability and angiogenesis is implicated as an important mechanism in functional recovery, we hypothesized that chronic metformin treatment would improve post-stroke functional recovery by enhancing functional microvascular density. For this study, C57BL/6N male mice were subjected to a 60-min middle cerebral artery occlusion, and were given 50 mg/kg/day metformin beginning 24 h post-stroke for 3 weeks. Behavioral recovery was assessed using adhesive-tape removal and the apomorphine-induced turning test. The role of angiogenesis was assessed by counting vessel branch points from fluorescein-conjugated lectin-perfused brain sections. Importantly even if metformin treatment was initiated 24 h after injury it enhanced recovery and significantly improved stroke-induced behavioral deficits. This recovery occurred in parallel with enhanced angiogenesis and with restoration of endogenous cerebral dopaminergic tone and revascularization of ischemic tissue. We assessed if the effects on recovery and angiogenesis were mediated by AMPK. When tested in AMPK α-2 knockout mice, we found that metformin treatment did not have the same beneficial effects on recovery and angiogenesis, suggesting that metformin-induced angiogenic effects are mediated by AMPK. The results from this study suggest that metformin mediates post-stroke recovery by enhancing angiogenesis, and these effects are mediated by AMPK signaling.
Collapse
Affiliation(s)
- Venugopal R Venna
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | | | |
Collapse
|
350
|
Zhang YP, Cai J, Shields LBE, Liu N, Xu XM, Shields CB. Traumatic brain injury using mouse models. Transl Stroke Res 2014; 5:454-71. [PMID: 24493632 DOI: 10.1007/s12975-014-0327-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 12/09/2013] [Accepted: 01/05/2014] [Indexed: 12/14/2022]
Abstract
The use of mouse models in traumatic brain injury (TBI) has several advantages compared to other animal models including low cost of breeding, easy maintenance, and innovative technology to create genetically modified strains. Studies using knockout and transgenic mice demonstrating functional gain or loss of molecules provide insight into basic mechanisms of TBI. Mouse models provide powerful tools to screen for putative therapeutic targets in TBI. This article reviews currently available mouse models that replicate several clinical features of TBI such as closed head injuries (CHI), penetrating head injuries, and a combination of both. CHI may be caused by direct trauma creating cerebral concussion or contusion. Sudden acceleration-deceleration injuries of the head without direct trauma may also cause intracranial injury by the transmission of shock waves to the brain. Recapitulation of temporary cavities that are induced by high-velocity penetrating objects in the mouse brain are difficult to produce, but slow brain penetration injuries in mice are reviewed. Synergistic damaging effects on the brain following systemic complications are also described. Advantages and disadvantages of CHI mouse models induced by weight drop, fluid percussion, and controlled cortical impact injuries are compared. Differences in the anatomy, biomechanics, and behavioral evaluations between mice and humans are discussed. Although the use of mouse models for TBI research is promising, further development of these techniques is warranted.
Collapse
Affiliation(s)
- Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, 210 East Gray Street, Suite 1102, Louisville, KY, 40202, USA,
| | | | | | | | | | | |
Collapse
|